WO2022135753A1 - Methods for prognosis the humoral response of a subject prior to vaccination - Google Patents

Methods for prognosis the humoral response of a subject prior to vaccination Download PDF

Info

Publication number
WO2022135753A1
WO2022135753A1 PCT/EP2021/070524 EP2021070524W WO2022135753A1 WO 2022135753 A1 WO2022135753 A1 WO 2022135753A1 EP 2021070524 W EP2021070524 W EP 2021070524W WO 2022135753 A1 WO2022135753 A1 WO 2022135753A1
Authority
WO
WIPO (PCT)
Prior art keywords
gene
vaccine
subject
level
gene expression
Prior art date
Application number
PCT/EP2021/070524
Other languages
French (fr)
Inventor
Behazine Combadiere
Elena GONÇALVES
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Sorbonne Université
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by INSERM (Institut National de la Santé et de la Recherche Médicale), Sorbonne Université filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2022135753A1 publication Critical patent/WO2022135753A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/21Retroviridae, e.g. equine infectious anemia virus
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/5052Cells of the immune system involving B-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5256Virus expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55572Lipopolysaccharides; Lipid A; Monophosphoryl lipid A
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/575Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/555Interferons [IFN]
    • G01N2333/56IFN-alpha
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/91Transferases (2.)
    • G01N2333/912Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)

Definitions

  • the present invention relates to methods and kits for predicting the humoral response of a subject prior to vaccination. More specifically present invention relates to methods for assessing the humoral response of a subject to a vaccine through detection in a blood sample of specific RNAs.
  • the present invention relates to an in vitro method for assessing a subject’s humoral immune response to a vaccine, said method, comprising the step of (a) measuring in a sample obtained from said subject prior to the administration of said vaccine the level of one or more gene expression level selected from a group of genes consisting of: IGLV8-61, BLK, EBF1 and IFNal6 gene b) wherein the level of one or more gene expression of step a) is negatively correlated with the humoral immune response of said subject.
  • Another object of the invention relates to a kit, comprising:
  • - at least a mean for determining one or more gene expression level selected from a group of gene consisting of: IGLV8-61, BLK, EBF1 and IFNal6 gene and
  • Inventors work represents the first systems biology approach to investigate the volunteers' immune predisposition to respond to MVA-B vaccination, assessed by their blood transcriptome profile; specifically, that related to their B cell differentiation stages, and its conditioning by the human microbiota before vaccination. That is, they investigated the host gene expression in blood by a microarray approach and the skin and stool microbiota by using 16S ribosomal RNA sequencing both before vaccination. The objective was to examine their potential involvement in an effective MVA-B neutralizing antibody (Nabs) response during the CUTHIVAC 03 randomized phase lb clinical study. This trial immunized 10 HIV seronegative subjects aged from 18 to 45 years by the intramuscular route with MVA-HIV clade B vaccine. Inventors analyzed their baseline transcriptomic signature in blood and baseline bacterial abundance and diversity in skin and stool to assess their potential association with the intensity of the Nabs response.
  • Nabs MVA-B neutralizing antibody
  • the inventors found that gene expression of a group of genes of the invention (detected through mRNA nucleic acids in blood) are overexpressed at baseline in subjects with the lowest antibody response compared to the subjects with the highest antibody response, and therefore may be considered as good biomarkers to assess the humoral immune response of a subject to a vaccine (Figure 1C and 2A, 2B, 2C, 2D, 2E).
  • This minimal marker set identified by the inventors may be used as prognoses tool alone or in combination with bacterial abundance and diversity scores. These results thus set- up the basis for the development of a rapid functional specific prognostic test for predicting the humoral response of a subject prior to vaccination with an antigen.
  • the present invention relates to an in vitro method for assessing a subject’s humoral immune response to a vaccine, said method, comprising the step of (a) measuring in a sample obtained from said subject prior to the administration of said vaccine, the level of one or more gene expression level selected from a group of genes consisting of: IGLV8-61, BLK, EBF1 and IFNal6 gene b) wherein the level of one or more gene expression of step a) is negatively correlated with the humoral immune response of said subject.
  • the said method is performed in vitro or ex vivo.
  • a high level of one or more gene expression compared to said control reference value is predictive that said subject is a high risk to be a “low responder” to said vaccine
  • a low level of one or more gene expression compared to said control reference value is predictive that said subject is a high risk to be a “high responder” to said vaccine
  • subject refers to a mammalian, such as a rodent (e.g. a mouse or a rat), a feline, a canine or a primate. In a preferred embodiment, said subject is a human subject.
  • rodent e.g. a mouse or a rat
  • feline e.g. a feline
  • canine e.g. a canine
  • primate e.g. a mammalian
  • said subject is a human subject.
  • the subject according to the invention can be a healthy subject or a subject suffering from a given disease such as infectious disease prior to the vaccination of said subject with an antigen.
  • infectious disease refers to a condition in which an infectious organism or agent is present in a detectable amount in the blood or in a normally sterile tissue or normally sterile compartment of a subject.
  • Infectious organisms and agents include viruses, mycobacteria, bacteria, fungi, and parasites. The terms encompass both acute and chronic infections, as well as sepsis.
  • the infectious organism is a virus or a bacteria that is responsible of infection such as:
  • Adenoviridae ie Adenovirus
  • Herpesviridae Herpes simplex, type 1, Herpes simplex, type 2, Varicella-zoster virus, Epstein-Barr virus, Human cytomegalovirus, Human herpesvirus, type 8
  • Papillomaviridae Human papillomavirus
  • Polyomaviridae BK virus, JC virus
  • Poxviridae Smallpox
  • Hepadnaviridae Hepatitis B virus
  • Parvoviridae Parvovirus Bl 9
  • Astroviridae Human astrovirus
  • Caliciviridae Neorwalk virus
  • Picornaviridae coxsackievirus, hepatitis A virus, poliovirus rhinovirus
  • Coronaviridae severe acute respiratory syndrome-related coronavirus, strains: Severe acute respiratory syndrome virus, Severe acute respiratory syndrome coronavirus 2
  • Flavoviridae severe acute respiratory syndrome
  • the viral infection is, Coronaviridae (Severe acute respiratory syndrome-related coronavirus, strains: Severe acute respiratory syndrome virus, Severe acute respiratory syndrome coronavirus 2) and Retroviridae (Human immunodeficiency virus (HIV)).
  • Coronaviridae severe acute respiratory syndrome-related coronavirus, strains: Severe acute respiratory syndrome virus, Severe acute respiratory syndrome coronavirus 2
  • Retroviridae Human immunodeficiency virus (HIV)
  • the viral infection is Human immunodeficiency virus (HIV) and the vaccination with a MVA-B vaccine.
  • HIV Human immunodeficiency virus
  • MVA-B or “Modified Vaccinia Ankara B” refers to is a particular HIV vaccine created to give immune resistance to infection by the human immunodeficiency virus.
  • MVA-B was developed by the Spanish National Research Council's Biotechnology National Centre headed by Dr Mariano Esteban.
  • the vaccine is based on the Modified vaccinia Ankara (MV A) virus used during the 1970s to help eradicate the smallpox virus.
  • the B in the name "refers to HIV-B, the most common HIV subtype in Europe.
  • the Vaccinia viruses is re- engineered to express foreign genes which are vectors for production of recombinant proteins, the most common being a vaccine delivery system for antigens (Pavot V, et al (2017).
  • MVA Recombinant Virus Vaccines. Methods in Molecular Biology. 1581. Springer New York. pp. 97-119).
  • MVA-B recombinant HIV vaccine
  • Studies in mice and nonhuman primates have further demonstrated the safety of MVA under conditions of immune suppression.
  • MVA provides similar or higher levels of recombinant gene expression even in non- permissive cells.
  • humoral immune response means the immune response involving the transformation of B cells into plasma cells that produce and secrete antibodies to a specific antigen.
  • humoral immune response to a vaccine means detecting antibody response, in particular neutralizing antibody response, after immunization with an antigen, against a pathogen (infectious agent, or tumor cells).
  • Humoral immunity prevents infection and can be of different kinds. It is characterized, for example, by neutralizing antibodies responses induced after vaccination. These functional antibodies are the main actors of protective immunity, they are able to neutralize the ability of the virus to penetrate or replicate in cells.
  • Antibodies can belong to different immunoglobulin classes and subclasses including for example serum IgG and mucosal IgG and IgA. These Ig can also act in other ways by opsonization or complement activation, involving serum immune regulatory proteins for pathogen elimination. Other ways to defend oneself is antibody-dependent cell-mediated virus inhibition (ADCVI) and antibody-dependent cell-mediated-cytotoxicity (ADCC). This is one of the mechanisms by which antibodies act to limit and contain infection. It’s carried out by natural killer cells (NK) through different proteins: perforins (pore-forming proteins), granzymes (serine proteases), reactive oxygen intermediates, and cytokines. These cells lysis an infected cell labeled with antibody bound to antigen present on the membrane.
  • ADCVI antibody-dependent cell-mediated virus inhibition
  • ADCC antibody-dependent cell-mediated-cytotoxicity
  • protective humoral immunity means a humoral immune response that confers the essential component of protection against a pathogen.
  • sterilizing humoral immunity means a humoral immune response that prevents the establishment of any detectable infection by a pathogen.
  • long-lasting humoral immunity means that some aspect of humoral immunity is detectable three months after antigen administration, such as, for example, antibodies elicited by the antigen.
  • Suitable methods of antibody detection include, but are not limited to, such methods as ELISA, immunofluorescence (IF A), focus reduction neutralization tests (FRNT), immunoprecipitation, and Western blotting.
  • neutralizing humoral response means that the antibodies elicited during humoral immunity directly block the ability of a pathogen to infect cells.
  • the term "quick response” as used herein, means that protective humoral immunity is conferred within three weeks of antigen administration.
  • very quick response means that protective humoral immunity is conferred within one week of antigen administration.
  • a “responder to a vaccine” means that after vaccination of a subject, the serum antibody titer against the targeted antigen of said subject is sufficient to adequately clear the disease (infectious disease or tumor) and exhibit an efficient immune humoral activity. In such a case, then one can conclude that said patient has a greater proportion of immune B cells which induce an efficient immune humoral response in particular a neutralizing antibody response.
  • Tests for determining the serum antibody titer are well known to the person skilled in the art. For example, a GFP Neutralizing assay (as described in example section) to detected an anti-antigen neutralizing activity in a serum of subjects collected 8 days after vaccination based on GFP detection by flow cytometry can be used.
  • HI hemagglutinin inhibition
  • a variety of substances can be used as antigens in a compound or formulation, of immunogenic or vaccine type.
  • attenuated and inactivated viral and bacterial pathogens purified macromolecules, polysaccharides, toxoids, recombinant antigens, organisms containing a foreign gene from a pathogen, synthetic peptides, polynucleic acids (DNA, RNA), antibodies and tumor cells can be used to prepare (i) an immunogenic composition useful to induce an immune response in an individual or (ii) a vaccine useful for treating a pathological condition.
  • An antigen is additionally capable of being recognized by the immune system and/or being capable of inducing a humoral immune response and/or cellular immune response leading to the activation of B- and/or T-lymphocytes.
  • An antigen can have one or more epitopes or antigenic sites (B- and T- epitopes)
  • An isolated antigen can be prepared using a variety of methods well known in the art.
  • a gene encoding any immunogenic polypeptide can be isolated and cloned, for example, in bacterial, yeast, insect, reptile or mammalian cells using recombinant methods well known in the art and described, for example in Sambrook et al., Molecular cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York (1992) and in Ansubel et al., Current Protocols in Molecular Biology, John Wiley and Sons, Baltimore, MD (1998).
  • a number of genes encoding surface antigens from viral, bacterial and protozoan pathogens have been successfully cloned, expressed and used as antigens for vaccine development.
  • the major surface antigen of hepatitis B virus, HbsAg, the P subunit of choleratoxin, the enterotoxin of E. coh. the circumsporozoite protein of the malaria parasite, and a glycoprotein membrane antigen from Epstein-Barr virus, as well as tumor cell antigens have been expressed in various well known vector/host systems, purified and used in vaccines.
  • a pathologically aberrant cell may also be used in a vaccine composition according to the invention can be obtained from any source such as one or more individuals having a pathological condition or ex vivo or in vitro cultured cells obtained from one or more such individuals, including a specific individual to be treated with the resulting vaccine.
  • sample refers to any biological sample of a subject and can include, by way of example and not limitation, bodily fluids and/or tissue extracts such as homogenates or solubilized tissue obtained from a subject. Tissue extracts are obtained routinely from tissue biopsy.
  • the biological sample is a body fluid sample (such blood sample) or tissue biopsy of said subject.
  • the body fluid sample is a blood sample.
  • blood sample means a whole blood sample obtained from a subject (e.g. an individual for which it is interesting to determine whether a gene expression level can be identified).
  • the term “prior to the administration of (said) vaccine” means that the level of one or more gene expression level selected from a group of gene consisting of: IGLV8-61, BLK, EBF1 and IFNal6 gene, is measured at least 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, 0 days prior the administration of the vaccine.
  • the level of one or more gene expression level selected from a group of gene consisting of: IGLV8-61, BLK, EBF1 and IFNal6 gene is measured at least 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, 0 days prior the administration of the vaccine.
  • IFNal6 also known as “Interferon alpha-16” or “IFN alpha 16”
  • IFN alpha 16 has its general meaning in the art and refers to a cytokine that in humans is encoded by the IFNA16 gene (Gene ID: 3449). Produced by macrophages, IFN-alpha 16 have antiviral activities. Interferon alpha 16 stimulates the production of two enzymes: a protein kinase and an oligoadenylate synthetase. The sequence of said protein may be found with the NCBI Reference: NM_002173 and NP_002164.
  • IGLV8-61 also known as “Immunoglobulin lambda variable 8-61” has its general meaning in the art refers to a protein that in humans is encoded by the IGLV8-61gene (gene ID 28774). V region of the variable domain of immunoglobulin light chains that participates in the antigen recognition. Immunoglobulins, also known as antibodies, are membrane-bound or secreted glycoproteins produced by B lymphocytes. The antigen binding site is formed by the variable domain of one heavy chain, together with that of its associated light chain. Thus, each immunoglobulin has two antigen binding sites with remarkable affinity for a particular antigen. The variable domains are assembled by a process called V-(D)-J rearrangement and can then be subjected to somatic hypermutations which, after exposure to antigen and selection, allow affinity maturation for a particular antigen
  • the sequence of said gene may be found with the NCBI Reference: NG 000002 and NC_000022 (Reference GRCh38.pl3 Primary Assembly).
  • BLK Terosine-protein kinase BLK
  • B lymphocyte kinase B lymphocyte kinase
  • BLK proto-oncogene Src family tyrosine kinase
  • BLK gene encodes a nonreceptor tyrosine-kinase of the src family of proto- oncogenes that are typically involved in cell proliferation and differentiation.
  • the protein has a role in B-cell receptor signaling and B-cell development. The protein also stimulates insulin synthesis and secretion in response to glucose and enhances the expression of several pancreatic beta-cell transcription factors.
  • NM_001715/NM_001330465 isoform 1 and isoform 2
  • NP_001317394/NP_001706 isoform 1 and isoform 2
  • EBF1 also known as “Early B-Cell Factor 1” “or “Transcription factor COE1” has its general meaning in the art and refers to a protein that in humans is encoded by the EBF1 gene (Gene ID: 1879).
  • the transcription factor EBF1 controls the expression of key proteins required for B cell differentiation, signal transduction and function (Treiber, T. et al (2010). Immunity. 32 (5): 714-725.; Hagman J. et al (2012). Current Topics in Microbiology and Immunology. 356: 17-38).
  • the crucial role of this factor is shown in the regulation of expression of SLAM family co-receptors in B-cells (Schwartz, A. et al. (2016). Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms. 1859 (10): 1259-1268).
  • NM_001290360 and NP_001277289 isoform 1/ NM_024007 and NP_076870 (isoform 2)/ NMJ82708 and NP_874367 (isoform 3)/ NM_001324101 and NP_001311030 (isoform 4)/ NM_001324103 and NP_001311032 (isoform 5)/ NM_001324106 and NP_001311035 (isoform 6)/ NM_001324107 and NP_001311036 ((isoform 7)/ NM_001324108 and NP_001311037 (isoform 8)/ NM_001324109 and NP_001311038 (isoform 9)/ NM_001324111 and NP_001311040 (isoform 10)/ NM_001364155 and NP_001351084 (isoform 11)/ NM_001364
  • biomarkers gene expression level selected from a group of gene
  • subject humoral immune response to a vaccine
  • biomarkers gene expression level selected from a group of gene
  • the present invention provides an in vitro method for assessing a subject’s humoral immune response to a vaccine, comprising the step of (a) measuring in a sample obtained from said subject prior to the administration of said vaccine, the level of one or more gene expression level selected from a group of genes consisting of: IFNal6, IGLV8- 61, BLK, and EBF1 gene and b) wherein the level of one or more gene expression of step a) is negatively correlated with the humoral immune response of said subject.
  • a plurality of gene expression level biomarkers (i.e., one or more than one gene expression level biomarkers) is used in the method of prognosis.
  • the method of the invention may comprise steps of: measuring in the biological sample plurality of gene expression level biomarkers, between one, two, three; four, gene expression level biomarker selected from a group of gene consisting of: IFNal6, IGLV8-61, BLK, and EBF1 gene present in the biological sample.
  • the method of prognosis is performed using the four different gene expression level biomarkers including the IFNal6, IGLV8-61, BLK, and EBF1 gene.
  • Measuring the expression level of a gene can be performed by a variety of techniques well known in the art.
  • the expression level of a gene may be determined by determining the quantity of mRNA.
  • Methods for determining the quantity of mRNA are well known in the art.
  • the nucleic acid contained in the samples e.g., blood, cell or tissue prepared from the patient
  • the extracted mRNA is then detected by hybridization (e. g., Northern blot analysis, in situ hybridization) and/or amplification (e.g., RT-PCR).
  • LCR ligase chain reaction
  • TMA transcription- mediated amplification
  • SDA strand displacement amplification
  • NASBA nucleic acid sequence-based amplification
  • Nucleic acids having at least 10 nucleotides and exhibiting sequence complementarity or homology to the mRNA of interest herein find utility as hybridization probes or amplification primers. It is understood that such nucleic acids need not be identical, but are typically at least about 80% identical to the homologous region of comparable size, more preferably 85% identical and even more preferably 90-95% identical. In certain embodiments, it will be advantageous to use nucleic acids in combination with appropriate means, such as a detectable label, for detecting hybridization.
  • the nucleic acid probes include one or more labels, for example to permit detection of a target nucleic acid molecule using the disclosed probes.
  • a nucleic acid probe includes a label (e.g., a detectable label).
  • a “detectable label” is a molecule or material that can be used to produce a detectable signal that indicates the presence or concentration of the probe (particularly the bound or hybridized probe) in a sample.
  • a labeled nucleic acid molecule provides an indicator of the presence or concentration of a target nucleic acid sequence (e.g., genomic target nucleic acid sequence) (to which the labeled uniquely specific nucleic acid molecule is bound or hybridized) in a sample.
  • a label associated with one or more nucleic acid molecules can be detected either directly or indirectly.
  • a label can be detected by any known or yet to be discovered mechanism including absorption, emission and/ or scattering of a photon (including radio frequency, microwave frequency, infrared frequency, visible frequency and ultra-violet frequency photons).
  • Detectable labels include colored, fluorescent, phosphorescent and luminescent molecules and materials, catalysts (such as enzymes) that convert one substance into another substance to provide a detectable difference (such as by converting a colorless substance into a colored substance or vice versa, or by producing a precipitate or increasing sample turbidity), haptens that can be detected by antibody binding interactions, and paramagnetic and magnetic molecules or materials.
  • detectable labels include fluorescent molecules (or fluorochromes).
  • fluorescent molecules or fluorochromes
  • Numerous fluorochromes are known to those of skill in the art, and can be selected, for example from Life Technologies (formerly Invitrogen), e.g., see, The Handbook — A Guide to Fluorescent Probes and Labeling Technologies).
  • fluorophores that can be attached (for example, chemically conjugated) to a nucleic acid molecule (such as a uniquely specific binding region) are provided in U.S. Pat. No.
  • fluorophores include thiol -reactive europium chelates which emit at approximately 617 mn (Heyduk and Heyduk, Analyt. Biochem. 248:216-27, 1997; J. Biol. Chem. 274:3315- 22, 1999), as well as GFP, LissamineTM, di ethylaminocoumarin, fluorescein chlorotriazinyl, naphthofluorescein, 4,7-dichlororhodamine and xanthene (as described in U.S. Pat. No. 5,800,996 to Lee et al.) and derivatives thereof.
  • fluorophores known to those skilled in the art can also be used, for example those available from Life Technologies (Invitrogen; Molecular Probes (Eugene, Oreg.)) and including the ALEXA FLUOR® series of dyes (for example, as described in U.S. Pat. Nos. 5,696,157, 6, 130, 101 and 6,716,979), the BODIPY series of dyes (dipyrrometheneboron difluoride dyes, for example as described in U.S. Pat. Nos.
  • a fluorescent label can be a fluorescent nanoparticle, such as a semiconductor nanocrystal, e.g., a QUANTUM DOTTM (obtained, for example, from Life Technologies (QuantumDot Corp, Invitrogen Nanocrystal Technologies, Eugene, Oreg.); see also, U.S. Pat. Nos. 6,815,064; 6,682,596; and 6,649, 138).
  • Semiconductor nanocrystals are microscopic particles having size-dependent optical and/or electrical properties.
  • Semiconductor nanocrystals that can he coupled to a variety of biological molecules (including dNTPs and/or nucleic acids) or substrates by techniques described in, for example, Bruchez et al., Science 281 :20132016, 1998; Chan et al., Science 281 :2016- 2018, 1998; and U.S. Pat. No. 6,274,323. Formation of semiconductor nanocrystals of various compositions are disclosed in, e.g., U.S. Pat. Nos.
  • quantum dots that emit light at different wavelengths based on size (565 mn, 655 mn, 705 mn, or 800 mn emission wavelengths), which are suitable as fluorescent labels in the probes disclosed herein are available from Life Technologies (Carlshad, Calif.). Additional labels include, for example, radioisotopes (such as 3 H), metal chelates such as DOTA and DPTA chelates of radioactive or paramagnetic metal ions like Gd3+, and liposomes.
  • radioisotopes such as 3 H
  • metal chelates such as DOTA and DPTA chelates of radioactive or paramagnetic metal ions like Gd3+, and liposomes.
  • Detectable labels that can he used with nucleic acid molecules also include enzymes, for example horseradish peroxidase, alkaline phosphatase, acid phosphatase, glucose oxidase, beta-galactosidase, beta-glucuronidase, or beta-lactamase.
  • enzymes for example horseradish peroxidase, alkaline phosphatase, acid phosphatase, glucose oxidase, beta-galactosidase, beta-glucuronidase, or beta-lactamase.
  • an enzyme can he used in a metallographic detection scheme.
  • SISH silver in situ hybridization
  • Metallographic detection methods include using an enzyme, such as alkaline phosphatase, in combination with a water-soluble metal ion and a redox-inactive substrate of the enzyme. The substrate is converted to a redox-active agent by the enzyme, and the redoxactive agent reduces the metal ion, causing it to form a detectable precipitate.
  • Metallographic detection methods also include using an oxido-reductase enzyme (such as horseradish peroxidase) along with a water-soluble metal ion, an oxidizing agent and a reducing agent, again to form a detectable precipitate.
  • an oxido-reductase enzyme such as horseradish peroxidase
  • a water-soluble metal ion such as horseradish peroxidase
  • an oxidizing agent such as horseradish peroxidase
  • Probes made using the disclosed methods can be used for nucleic acid detection, such as ISH procedures (for example, fluorescence in situ hybridization (FISH), chromogenic in situ hybridization (CISH) and silver in situ hybridization (SISH)) or comparative genomic hybridization (CGH).
  • ISH procedures for example, fluorescence in situ hybridization (FISH), chromogenic in situ hybridization (CISH) and silver in situ hybridization (SISH)
  • CGH comparative genomic hybridization
  • ISH In situ hybridization
  • a sample containing target nucleic acid sequence e.g., genomic target nucleic acid sequence
  • a metaphase or interphase chromosome preparation such as a cell or tissue sample mounted on a slide
  • a labeled probe specifically hybridizable or specific for the target nucleic acid sequence (e.g., genomic target nucleic acid sequence).
  • the slides are optionally pretreated, e.g., to remove paraffin or other materials that can interfere with uniform hybridization.
  • the sample and the probe are both treated, for example by heating to denature the double stranded nucleic acids.
  • the probe (formulated in a suitable hybridization buffer) and the sample are combined, under conditions and for sufficient time to permit hybridization to occur (typically to reach equilibrium).
  • the chromosome preparation is washed to remove excess probe, and detection of specific labeling of the chromosome target is performed using standard techniques.
  • a biotinylated probe can be detected using fluorescein-labeled avidin or avidin-alkaline phosphatase.
  • fluorochrome detection the fluorochrome can be detected directly, or the samples can be incubated, for example, with fluorescein isothiocyanate (FITC)-conjugated avidin.
  • FITC fluorescein isothiocyanate
  • Amplification of the FITC signal can be affected, if necessary, by incubation with biotin-conjugated goat anti-avidin antibodies, washing and a second incubation with FITC-conjugated avidin.
  • samples can be incubated, for example, with streptavidin, washed, incubated with biotin-conjugated alkaline phosphatase, washed again and pre-equilibrated (e.g., in alkaline phosphatase (AP) buffer).
  • AP alkaline phosphatase
  • Numerous reagents and detection schemes can be employed in conjunction with FISH, CISH, and SISH procedures to improve sensitivity, resolution, or other desirable properties.
  • probes labeled with fluorophores including fluorescent dyes and QUANTUM DOTS®
  • fluorophores including fluorescent dyes and QUANTUM DOTS®
  • the probe can be labeled with a nonfluorescent molecule, such as a hapten (such as the following non-limiting examples: biotin, digoxigenin, DNP, and various oxazoles, pyrrazoles, thiazoles, nitroaryls, benzofurazans, triterpenes, ureas, thioureas, rotenones, coumarin, courmarin-based compounds, Podophyllotoxin, Podophyllotoxin-based compounds, and combinations thereof), ligand or other indirectly detectable moiety.
  • a hapten such as the following non-limiting examples: biotin, digoxigenin, DNP, and various oxazoles, pyrrazoles, thiazoles, nitroaryls, benzofurazans, triterpenes, ureas, thioureas, rotenones, coumarin, courmarin-based compounds, Podophyllotoxin, Podo
  • Probes labeled with such non-fluorescent molecules (and the target nucleic acid sequences to which they bind) can then be detected by contacting the sample (e.g., the cell or tissue sample to which the probe is bound) with a labeled detection reagent, such as an antibody (or receptor, or other specific binding partner) specific for the chosen hapten or ligand.
  • a labeled detection reagent such as an antibody (or receptor, or other specific binding partner) specific for the chosen hapten or ligand.
  • the detection reagent can be labeled with a fluorophore (e.g., QUANTUM DOT®) or with another indirectly detectable moiety, or can be contacted with one or more additional specific binding agents (e.g., secondary or specific antibodies), which can be labeled with a fluorophore.
  • the probe, or specific binding agent (such as an antibody, e.g., a primary antibody, receptor or other binding agent) is labeled with an enzyme that is capable of converting a fluorogenic or chromogenic composition into a detectable fluorescent, colored or otherwise detectable signal (e.g., as in deposition of detectable metal particles in SISH).
  • the enzyme can be attached directly or indirectly via a linker to the relevant probe or detection reagent. Examples of suitable reagents (e.g., binding reagents) and chemistries (e.g., linker and attachment chemistries) are described in U.S. Patent Application Publication Nos. 2006/0246524; 2006/0246523, and 2007/ 01 17153.
  • multiplex detection schemes can he produced to facilitate detection of multiple target nucleic acid sequences (e.g., genomic target nucleic acid sequences) in a single assay (e.g., on a single cell or tissue sample or on more than one cell or tissue sample).
  • a first probe that corresponds to a first target sequence can he labelled with a first hapten, such as biotin, while a second probe that corresponds to a second target sequence can be labelled with a second hapten, such as DNP.
  • the bound probes can he detected by contacting the sample with a first specific binding agent (in this case avidin labelled with a first fluorophore, for example, a first spectrally distinct QUANTUM DOT®, e.g., that emits at 585 mn) and a second specific binding agent (in this case an anti-DNP antibody, or antibody fragment, labelled with a second fluorophore (for example, a second spectrally distinct QUANTUM DOT®, e.g., that emits at 705 mn).
  • a first specific binding agent in this case avidin labelled with a first fluorophore, for example, a first spectrally distinct QUANTUM DOT®, e.g., that emits at 585 mn
  • a second specific binding agent in this case an anti-DNP antibody, or antibody fragment, labelled with a second fluorophore (for example, a second spectrally distinct QUANTUM DOT®,
  • Probes typically comprise single-stranded nucleic acids of between 10 to 1000 nucleotides in length, for instance of between 10 and 800, more preferably of between 15 and 700, typically of between 20 and 500.
  • Primers typically are shorter single-stranded nucleic acids, of between 10 to 25 nucleotides in length, designed to perfectly or almost perfectly match a nucleic acid of interest, to be amplified.
  • the probes and primers are “specific” to the nucleic acids they hybridize to, i.e. they preferably hybridize under high stringency hybridization conditions (corresponding to the highest melting temperature Tm, e.g., 50 % formamide, 5x or 6x SCC.
  • SCC is a 0.15 M NaCl, 0.015 M Na-citrate).
  • the nucleic acid primers or probes used in the above amplification and detection method may be assembled as a kit.
  • a kit includes consensus primers and molecular probes.
  • a preferred kit also includes the components necessary to determine if amplification has occurred.
  • the kit may also include, for example, PCR buffers and enzymes; positive control sequences, reaction control primers; and instructions for amplifying and detecting the specific sequences.
  • the methods of the invention comprise the steps of providing total RNAs extracted from blood and subjecting the RNAs to amplification and hybridization to specific probes, more particularly by means of a quantitative or semi- quantitative RT-PCR.
  • the expression level is determined by DNA chip analysis.
  • DNA chip or nucleic acid microarray consists of different nucleic acid probes that are chemically attached to a substrate, which can be a microchip, a glass slide or a microsphere-sized bead.
  • a microchip may be constituted of polymers, plastics, resins, polysaccharides, silica or silica-based materials, carbon, metals, inorganic glasses, or nitrocellulose.
  • Probes comprise nucleic acids such as cDNAs or oligonucleotides that may be about 10 to about 60 base pairs.
  • a sample from a test subject optionally first subjected to a reverse transcription, is labelled and contacted with the microarray in hybridization conditions, leading to the formation of complexes between target nucleic acids that are complementary to probe sequences attached to the microarray surface.
  • the labelled hybridized complexes are then detected and can be quantified or semi-quantified. Labelling may be achieved by various methods, e.g. by using radioactive or fluorescent labelling.
  • Many variants of the microarray hybridization technology are available to the man skilled in the art (see e.g. the review by Hoheisel, Nature Reviews, Genetics, 2006, 7:200- 210).
  • Expression level of a gene may be expressed as absolute expression level or normalized expression level.
  • expression levels are normalized by correcting the absolute expression level of a gene by comparing its expression to the expression of a gene that is not a relevant for determining the cancer stage of the patient, e.g., a housekeeping gene that is constitutively expressed.
  • Suitable genes for normalization include housekeeping genes such as the actin gene ACTB, ribosomal 18S gene, GUSB, PGK1, TBP, HPRT1 and TFRC.
  • TATA-binding protein TBP) and hypoxanthine phosphoribosyl transferase 1 (HPRTT) were used as reference genes in the present study. This normalization allows the comparison of the expression level in one sample, e.g., a patient sample, to another sample, or between samples from different sources.
  • Said reference control values may be determined in regard to the level of gene expression biomarker present in blood samples taken from one or more healthy subject(s) or in a control population.
  • the method according to the present invention comprises the step of comparing said level of humoral immune response to a vaccine -specific gene expression level biomarkers (“Biomarkerl”: IFNal6 gene and/or “Biomarker2”: IGLV8-61 gene and/or “Biomarker3”: BLK gene and/or “Biomarker4”: EBF1 gene) to a control reference value wherein a high level of “humoral immune response to a vaccine -specific” gene expression biomarkers (“Biomarkerl”: IFNal6 gene and/or “Biomarker2”: IGLV8-61 gene and/or “Biomarker3”: BLK gene and/or “Biomarker4”: EBF1 gene) compared to said control reference value is predictive of a high risk to be a “low responder” to said vaccine and a low “"humoral immune response to a vaccine -specific” gene expression biomarkers (Biomarkerl”: IFNal6 gene and/or “Biomarker2”: IGLV8-
  • the control reference value may depend on various parameters such as the method used to measure the level humoral immune response to a vaccine -specific gene expression level biomarkers (“Biomarkerl”: IFNal6 gene and/or “Biomarker2”: IGLV8-61 gene and/or “Biomarker3”: BLK gene and/or “Biomarker4”: EBF1 gene) or the gender of the subject.
  • Control reference values are easily determinable by the one skilled in the art, by using the same techniques as for determining the level of gene expression biomarker in a blood samples previously collected from the patient under testing.
  • a “reference value” can be a “threshold value” or a “cut-off value”. Typically, a “threshold value” or “cut-off value” can be determined experimentally, empirically, or theoretically.
  • a threshold value can also be arbitrarily selected based upon the existing experimental and/or clinical conditions, as would be recognized by a person of ordinary skilled in the art. The threshold value has to be determined in order to obtain the optimal sensitivity and specificity according to the function of the test and the benefit/risk balance (clinical consequences of false positive and false negative). Typically, the optimal sensitivity and specificity (and so the threshold value) can be determined using a Receiver Operating Characteristic (ROC) curve based on experimental data.
  • ROC Receiver Operating Characteristic
  • the person skilled in the art may compare the level of gene expression biomarkers (“Biomarkerl”: IFNal6 gene and/or “Biomarker2”: IGLV8-61 gene and/or “Biomarker3”: BLK gene and/or “Biomarker4”: EBF1 gene) with a defined threshold value.
  • the threshold value is derived from the gene expression level (or ratio, or score) determined in a blood sample derived from one or more subjects who are responders (to the method according to the invention).
  • the threshold value may also be derived from gene expression level (or ratio, or score) determined in a blood sample derived from one or more subjects or who are non-responders.
  • retrospective measurement of the gene expression level (or ratio, or scores) in properly banked historical subject samples may be used in establishing these threshold values.
  • the ROC curve analysis revealed that choosing a cut-off value at 3.5 unit expression of IFNal6 mRNA, and/or at 6.5 unit expression of IGLV8-61 mRNA and/or at 6 unit expression of BLK, and/or at 5.9 unit expression of EBF1 mRNA levels allowed to effectively discriminate responders from / non responders blood sample and which could be used as predetermined reference level for IFNal6, IGLV8-61, BLK, and/or EBF1.
  • “Risk” in the context of the present invention relates to the probability that an event will occur over a specific time period, as in humoral immune response of a subject to a vaccine, and can mean a subject's "absolute” risk or “relative” risk.
  • Absolute risk can be measured with reference to either actual observation post-measurement for the relevant time cohort, or with reference to index values developed from statistically valid historical cohorts that have been followed for the relevant time period.
  • Relative risk refers to the ratio of absolute risks of a subject compared either to the absolute risks of low-risk cohorts or an average population risk, which can vary by how clinical risk factors are assessed.
  • Odds ratios the proportion of positive events to negative events for a given test result, are also commonly used (odds are according to the formula p/(l-p) where p is the probability of event and (1- p) is the probability of no event) to no conversion.
  • Alternative continuous measures which may be assessed in the context of the present invention, include time to humoral immune response of a subject to a vaccine risk reduction ratios.
  • Risk evaluation in the context of the present invention encompasses making a prediction of the probability, odds, or likelihood that an event (humoral immune response of a subject to a vaccine) may occur, the rate of occurrence of the event or conversion from one state to another, i.e., from a “high responder” to said vaccine to “low responder” to said vaccine.
  • Risk evaluation can also comprise prediction of future clinical parameters, traditional laboratory risk factor values, or other indices of “humoral response”, such as cellular population determination in peripheral tissues, in serum or other fluid, either in absolute or relative terms in reference to a previously measured population.
  • the methods of the present invention may be used to make continuous or categorical measurements of the risk of an event (humoral immune response of a subject to a vaccine), thus prognosing and defining the risk spectrum of a category of subjects defined as being “high responder” to a vaccine.
  • the invention can be used to discriminate between normal and other subject cohorts at higher risk to be “high responder” to said vaccine.
  • the present invention may be used so as to help to discriminate those having “high responder” to a vaccine from “low responder” to said vaccine
  • the methods for assessing a subject’s humoral immune response to a vaccine may further comprise measuring, in a biological sample obtained from the subject, the level Eubacterium in stool sample and Prevotella in skin sample.
  • the level Eubacterium in stool sample and Prevotella in skin sample is performed by using 16S ribosomal sequencing.
  • kits for performing the methods of the invention comprise means for measuring the expression level of one or more gene expression level selected from a group of gene consisting of: IGLV8-61, BLK, EBF1 and IFNal6 gene of the invention in the sample obtained from the patient for use to assess a subject’s humoral immune response to a vaccine.
  • the present invention also relates to a kit of the invention comprising means for determining the expression level of one or more gene expression level selected from a group of gene consisting of IGLV8-61, BLK, EBF1 and IFNal6 gene.
  • the present invention relates to a kit for use to assess a subject’s humoral immune response to a vaccine, comprising:
  • - at least a means for determining the expression level of one or more gene expression level selected from a group of gene consisting of IGLV8-61, BLK, EBF1 and IFNal6 gene and
  • the kit for use comprising: - amplification primers and/or probe for determining the expression level one or more gene expression level selected from a group of gene consisting of IGLV8-61, BLK, EBF1 and IFNal6 gene,
  • kits may include probes, primers macroarrays or microarrays as above described.
  • the kit may comprise a set of probes as above defined, usually made of DNA, and that may be pre-labelled.
  • probes may be unlabelled and the ingredients for labelling may be included in the kit in separate containers.
  • the kit may further comprise hybridization reagents or other suitably packaged reagents and materials needed for the particular hybridization protocol, including solid-phase matrices, if applicable, and standards.
  • the kit of the invention may comprise amplification primers that may be pre- labelled or may contain an affinity purification or attachment moiety.
  • the kit may further comprise amplification reagents and also other suitably packaged reagents and materials needed for the particular amplification protocol.
  • the invention also relates to a method for vaccinating a subject in need thereof with an antigen wherein, prior to vaccination; the level of one or more gene expression level selected from a group of gene consisting of IFNal6, IGLV8-61, BLK and EBF1 gene obtained from said subject have been detected by one of method of the invention.
  • Another object of the present invention is a method for vaccinating in a subject comprising, prior to vaccination the steps of a) providing a blood sample from a subject, b) detecting the level of one or more gene expression level selected from a group of gene consisting of IFNal6, IGLV8-61, BLK and EBF1 gene obtained from said subject c) comparing the level determined in step b) with a reference value and if level determined at step b) is lower than the reference value, vaccinating the subject with antigen.
  • FIGURES
  • FIG. 1 Blood gene expression combined with host microbiota before vaccination shapes MVA-B responses
  • IGLV8, EBF1, and BLK minimal gene signature
  • FIG. 1 ROC gene Nab: IFNal6, IGLV8-61, BLK and EBF1
  • Amplifications were performed in triplicate 25-pL reactions, each containing 2.5 pL of non-diluted DNA template, 12.5 pL of KAPA HiFi HotStart Ready Mix (containing KAPA HiFi HotStart DNA Polymerase, buffer, MgC12, and dNTPs, KAPA Biosystems Inc., Wilmington, MA, USA), and 5 pL of each primer at 1 pM.
  • Thermal cycling conditions consisted of an initial denaturation step (3 minutes at 95°C), followed by 30 cycles of denaturation (30 seconds at 95°C), annealing (30 seconds at 55°C), and extension (30 seconds at 72°C). These were followed by a final extension step of 10 minutes at 72°C.
  • Sequencing was performed on an Illumina MiSeqTM platform (Illumina, Inc.) according to the manufacturer's specifications to generate a median of 30,644 paired-end sequences of -300 bp length in each direction (-61,289 reads per sample).
  • Mothur pipeline (Schloss et al., 2009) was used to bin 16S rDNA sequences into operational taxonomic units (OTUs) with a threshold of 97% sequence similarity. OTUs present in only a single sample were discarded. Rarefaction curves were represented by defining the maximum subsampling size as the number of sequences of the sample with the fewest sequences (2751 sequences for skin samples, and 1059 sequences for stool samples). Richness and diversity indexes were estimated by using the summary, single module implemented in mothur. For taxonomical analysis, 16S rDNA sequences were classified according to the GreenGenes database (McDonald et al., 2012) version 13.5.99.
  • Anti-MVA neutralizing activities were evaluated in serum collected at week 8 (w8) with an assay based on GFP detection by flow cytometry (Cosma et al., 2004; Earl et al., 2003). It used HeLa cells as targets and a recombinant strain of MVA expressing the enhanced Aequoriae GFP (Garcia et al., 2011). Serial dilutions of heat inactivated serum were performed in 96-well round-bottom tissue culture plates (TPP, Zurich, Switzerland) containing DMEM (Gibco, Invitrogen) supplemented with 2% fetal calf serum (PAA, Laboratories GmbH, Pashing, Austria).
  • DMEM Gibco, Invitrogen
  • MVAeGFP was then added to each well at a MOI of 0.25.
  • the plate was then incubated for 1 hour at 37°C until the addition of 1 x 10 5 HeLa cells. The incubation then continued for an additional 16 hours at 37°C, 0.5% CO2.
  • the cells were washed with PBS supplemented with 0.5% fetal calf serum and 2 mM EDTA and fixed with 2% formaldehyde.
  • GFP expression was analyzed with FACSCanto II and Diva software (BD Biosciences). The percentage of neutralization was defined as the ratio of the reduction in the number of GFP-expressing cells to the number of GFP-expressing cells in untreated control wells.
  • RNA samples of 2.5 mL were collected in PAXgene RNA tubes (PreAnalytix) twice from each volunteer two weeks before (w-2) and the day of the vaccination (wO). These tubes enable the preservation and stabilization of RNA (storage at -80°C).
  • Total RNA was extracted from whole blood according to the instructions in the handbook accompanying the PAXgene blood RNA Kit (PreAnalytiX, Hombrechtikon, Switzerland). RNA purity and integrity were assessed on the Agilent 2100 Bioanalyzer with the RNA 6000 Nano LabChip reagent set (Agilent, Palo Alto, CA, USA).
  • Samples for microarray hybridization were prepared as described in the Affymetrix GeneChip WT PLUS Reagent Kit User Manual (Affymetrix, Inc., Santa Clara, CA, USA). For hybridization (to Affymetrix Human Gene 2.1 ST Array Plates), washing, staining, and scanning took place in an Affymetrix GeneTitan system, controlled by the Affymetrix GeneChip Command Console software w4.2. Background signal correction was performed by applying the backgroundCorrect function from the limma package on the perfect match (PM) signals with R Software 3.3.1. The underlying model is the normal-exponential convolution model from RMA (chip intensity: addition of a signal exponentially distributed, chip noise: follows Gaussian distribution) (Irizarry et al., 2003).
  • the variance stabilizing transformation algorithm (justvsn function from the vsn package (Huber et al., 2002) was applied to the background corrected signal (monotonic transformation), and the signal then transformed back to its usual scale by exponentiation (base 2).
  • a quantile normalization (Bolstad et al., 2003) (normalize function from the affy package) was then applied to the variance- stabilized signal.
  • the probe signals for replicated arrays were averaged and a quantile normalization performed anew. In all, 24,768 probes were analyzed.
  • Microbiome samples were clustered according to their genus composition by a nonmetric multidimensional scaling (NMDS) approach based on ecological distance matrices calculated by Bray-Curtis dissimilarities, as implemented in R (Vegan, metaMDS, and ggplot2 packages).
  • NMDS ellipses were drawn based on a confidence interval (CI) of 0.95.
  • CI confidence interval
  • the heatmap was performed with values row-centered and scaled, Pearson correlation as the distance method and a dendrogram computed and reordered based on row means.
  • the heatmap, logistic regression analyses, and ROC curves were performed and generated with R.
  • Ingenuity® pathway analysis (IPA®) was used to perform functional enrichment analyses and identify new targets or candidate biomarkers within the context of biological systems. It provided the canonical pathways, molecular/cellular functions, and networks that were statistically overrepresented in the gene signatures.
  • MVA-HIV clade B MVA-HIV clade B
  • Prevotella in the skin at baseline was positively correlated with MVA-Nab response.
  • Prevotella is known to promote mucosal inflammation and to stimulate production of epithelial cell cytokines (Larsen, 2017).
  • Prevotella is also found in larger numbers in the skin of women aged 60-76 years than in that of women in their 20s and 30s and was enriched in all of the skin sites of the older group compared to the younger ones (Shibagaki et al., 2017).
  • Eubacterium abundance at baseline was positively correlated with the MVA-Nab response.
  • This family of bacteria is known to be associated with gut health (King et al., 2018; Le Bastard et al., 2018; Rinninella et al., 2019), and several of its species are higher in centenarians than in either young or elderly adults (Biagi et al., 2010).
  • the potential impact of the gut microbiota on vaccine immunogenicity has been already investigated with systemic vaccines (Huda et al., 2014) and with oral vaccines including those of rotavirus (RVV), polio, and cholera, mainly in infants/children living in low-income countries (Levine, 2010; Magwira and Taylor, 2018).
  • the BLK gene belongs to the family of protein tyrosine kinases src, and the B cells activation induces BLK gene product phosphorylation playing a key role in transmitting signals through surface immunoglobulins which supports the pro-B to pre-B transition and the signaling for growth arrest and apoptosis downstream of B-cell receptor (Burkhardt et al., 1991). BLK also plays a role in the development, differentiation, and activation of B cells and in the intracellular signaling pathway. BLK is detected in pro-B cells and persists in mature B cells but is absent in plasma cells.
  • EBF1 Triple protein tyrosine kinase (SFK)-deficient mice — BLK, LYN, and FYN — have impaired NFkB signaling and B cell development (Saijo et al., 2003).
  • EBF1 an early B cell factor 1
  • Heterozygosity of EBF1 results in the deregulation of at least eight transcription factors involved in lymphopoiesis and the deregulation of key proteins that play a crucial role in the survival, development, and differentiation of pro-B cells (Musa et al., 2018).
  • IGLV8 (variable domain) is a glycoprotein produced by B lymphocytes; its binding of a specific antigen triggers the clonal expansion and differentiation of B lymphocytes into immunoglobulin-secreting plasma cells.
  • the link between microbiota and host blood transcriptome has also been studied previously by Nakaya et al., who showed that TLR5 expression in blood 3 days after influenza vaccination was correlated with antibody response 28 days later (Nakaya et al., 2011). This correlation was significantly lower in TLR5 -deficient mice immunized with TIV compared to wild-type mice. As influenza vaccine does not stimulate TLR5 directly, however, Oh et al.
  • Table 1 Skin and stool genus diversity correlations with blood gene expression
  • Trimmomatic a flexible trimmer for Illumina sequence data. Bioinforma. Oxf. Engl. 30, 2114-2120.

Abstract

The present invention represents the first systems biology approach to investigate the volunteers' immune predisposition to respond to a vaccination (MVA-B), assessed by their blood transcriptome profile; specifically, that related to their B cell differentiation stages, and its conditioning by the human microbiota before vaccination. That is, inventors investigated the host gene expression in blood by a microarray approach and the skin and stool microbiota both before vaccination. The objective was to examine their potential involvement in an effective MVA-B neutralizing antibody (Nabs) response during a randomized phase Ib clinical study. This trial immunized 10 HIV seronegative subjects aged from 18 to 45 years by the intramuscular route with MVA-HIV clade B vaccine. Inventors found that gene expression of a group of genes (detected through mRNA nucleic acids in blood) are overexpressed at baseline in subjects with the lowest antibody response compared to the subjects with the highest antibody response, and therefore may be considered as good biomarkers to assess the humoral immune response of a subject to a vaccine. Accordingly, the present invention relates to methods and kits for predicting the humoral response of a subject prior to vaccination. More specifically present invention relates to methods for assessing the humoral response of a subject to a vaccine through detection in a blood sample of specific RNAs.

Description

METHODS FOR PROGNOSIS THE HUMORAL RESPONSE OF A SUBJECT PRIOR TO VACCINATION
FIELD OF THE INVENTION:
The present invention relates to methods and kits for predicting the humoral response of a subject prior to vaccination. More specifically present invention relates to methods for assessing the humoral response of a subject to a vaccine through detection in a blood sample of specific RNAs.
BACKGROUND OF THE INVENTION:
Systems biology has been successfully used to investigate the fundamental innate immune mechanisms orchestrating protective adaptive responses after the perturbation of vaccination against yellow fever (Gaucher et al., 2008; Querec et al., 2009), HIV (Ehrenberg et al., 2019), Ebola (Rechtien et al., 2017), and influenza (Nakaya et al., 2011). An important challenge, however, is to analyze individual baseline human health characteristics to help identify those at higher risk of infection despite vaccination. Until now, only a few studies have looked for candidate traits associated with vaccine responsiveness and partially predicting the humoral response to vaccination against influenza (Furman et al., 2014; Parvandeh et al., 2019; Team and Consortium, 2017; Tsang et al., 2014). No study has examined the interrelations between each individual's immunological state, their microbiota at baseline, and the impact of both on their vaccine-induced immune responses. As the most successful vaccines act through the production of antibodies (Plotkin, 2010), identifying specific individual characteristics of the genome at baseline should enhance our ability for dividing vaccinees into “high responders” or “low responders” in terms of humoral response (Tsang, 2015). Such predictive markers might serve as a potential diagnostic tool that assists vaccine development by taking into account the interindividual heterogeneity of immune responses.
Accordingly, there remains an unmet need in the art for specific and more rapid prognostic test for predicting the humoral response of a subject prior to vaccination, reflecting the immunological status of the patient. SUMMARY OF THE INVENTION:
The present invention relates to an in vitro method for assessing a subject’s humoral immune response to a vaccine, said method, comprising the step of (a) measuring in a sample obtained from said subject prior to the administration of said vaccine the level of one or more gene expression level selected from a group of genes consisting of: IGLV8-61, BLK, EBF1 and IFNal6 gene b) wherein the level of one or more gene expression of step a) is negatively correlated with the humoral immune response of said subject.
Another object of the invention relates to a kit, comprising:
- at least a mean for determining one or more gene expression level selected from a group of gene consisting of: IGLV8-61, BLK, EBF1 and IFNal6 gene and
- instructions for use, for use to assess a subject’s humoral immune response to a vaccine.
DETAILED DESCRIPTION OF THE INVENTION:
Prognostic methods according to the invention
Inventors work represents the first systems biology approach to investigate the volunteers' immune predisposition to respond to MVA-B vaccination, assessed by their blood transcriptome profile; specifically, that related to their B cell differentiation stages, and its conditioning by the human microbiota before vaccination. That is, they investigated the host gene expression in blood by a microarray approach and the skin and stool microbiota by using 16S ribosomal RNA sequencing both before vaccination. The objective was to examine their potential involvement in an effective MVA-B neutralizing antibody (Nabs) response during the CUTHIVAC 03 randomized phase lb clinical study. This trial immunized 10 HIV seronegative subjects aged from 18 to 45 years by the intramuscular route with MVA-HIV clade B vaccine. Inventors analyzed their baseline transcriptomic signature in blood and baseline bacterial abundance and diversity in skin and stool to assess their potential association with the intensity of the Nabs response.
In other words, the inventors found that gene expression of a group of genes of the invention (detected through mRNA nucleic acids in blood) are overexpressed at baseline in subjects with the lowest antibody response compared to the subjects with the highest antibody response, and therefore may be considered as good biomarkers to assess the humoral immune response of a subject to a vaccine (Figure 1C and 2A, 2B, 2C, 2D, 2E).
This minimal marker set identified by the inventors, may be used as prognoses tool alone or in combination with bacterial abundance and diversity scores. These results thus set- up the basis for the development of a rapid functional specific prognostic test for predicting the humoral response of a subject prior to vaccination with an antigen.
Thus, the present invention relates to an in vitro method for assessing a subject’s humoral immune response to a vaccine, said method, comprising the step of (a) measuring in a sample obtained from said subject prior to the administration of said vaccine, the level of one or more gene expression level selected from a group of genes consisting of: IGLV8-61, BLK, EBF1 and IFNal6 gene b) wherein the level of one or more gene expression of step a) is negatively correlated with the humoral immune response of said subject.
In some embodiments, the said method is performed in vitro or ex vivo.
In particular embodiments the prognostic method of the invention comprising the step of comparing said level of one or more gene expression to a control reference value wherein:
- a high level of one or more gene expression compared to said control reference value is predictive that said subject is a high risk to be a “low responder” to said vaccine
- a low level of one or more gene expression compared to said control reference value is predictive that said subject is a high risk to be a “high responder” to said vaccine
The term “subject” as used herein refers to a mammalian, such as a rodent (e.g. a mouse or a rat), a feline, a canine or a primate. In a preferred embodiment, said subject is a human subject.
The subject according to the invention can be a healthy subject or a subject suffering from a given disease such as infectious disease prior to the vaccination of said subject with an antigen.
As used herein, the term "infectious disease" refers to a condition in which an infectious organism or agent is present in a detectable amount in the blood or in a normally sterile tissue or normally sterile compartment of a subject. Infectious organisms and agents include viruses, mycobacteria, bacteria, fungi, and parasites. The terms encompass both acute and chronic infections, as well as sepsis. In a particular embodiment the infectious organism is a virus or a bacteria that is responsible of infection such as:
Example of viral infections: Adenoviridae (ie Adenovirus); Herpesviridae (Herpes simplex, type 1, Herpes simplex, type 2, Varicella-zoster virus, Epstein-Barr virus, Human cytomegalovirus, Human herpesvirus, type 8); Papillomaviridae (Human papillomavirus); Polyomaviridae (BK virus, JC virus); Poxviridae (Smallpox); Hepadnaviridae (Hepatitis B virus); Parvoviridae (Parvovirus Bl 9); Astroviridae (Human astrovirus) Caliciviridae (Norwalk virus); Picornaviridae (coxsackievirus, hepatitis A virus, poliovirus rhinovirus) ;Coronaviridae (Severe acute respiratory syndrome-related coronavirus, strains: Severe acute respiratory syndrome virus, Severe acute respiratory syndrome coronavirus 2); Flaviviridae (Hepatitis C virus, yellow fever virus, dengue virus, West Nile virus, TBE virus, Zika virus); Matonaviridae (Rubella virus); Hepeviridae (Hepatitis E virus); Retroviridae (Human immunodeficiency virus (HIV)); Orthomyxoviridae (Influenza virus); Arenaviridae (Lassa virus); Bunyaviridae (Crimean-Congo hemorrhagic fever virus, Hantaan virus); Filoviridae (Ebola virus, Marburg virus); Paramyxoviridae (Measles virus, Mumps virus, Parainfluenza virus); Pneumoviridae (Respiratory syncytial virus); Rhabdoviridae (Rabies virus) ; Unassigned (Hepatitis D); Reoviridae (Rotavirus, Orbivirus, Coltivirus, Banna virus)
In a particular embodiment the viral infection is, Coronaviridae (Severe acute respiratory syndrome-related coronavirus, strains: Severe acute respiratory syndrome virus, Severe acute respiratory syndrome coronavirus 2) and Retroviridae (Human immunodeficiency virus (HIV)).
In a particular embodiment, the viral infection is Human immunodeficiency virus (HIV) and the vaccination with a MVA-B vaccine.
The term “MVA-B” or “Modified Vaccinia Ankara B” refers to is a particular HIV vaccine created to give immune resistance to infection by the human immunodeficiency virus. MVA-B was developed by the Spanish National Research Council's Biotechnology National Centre headed by Dr Mariano Esteban. The vaccine is based on the Modified vaccinia Ankara (MV A) virus used during the 1970s to help eradicate the smallpox virus. The B in the name "refers to HIV-B, the most common HIV subtype in Europe. The Vaccinia viruses is re- engineered to express foreign genes which are vectors for production of recombinant proteins, the most common being a vaccine delivery system for antigens (Pavot V, et al (2017). Recombinant Virus Vaccines. Methods in Molecular Biology. 1581. Springer New York. pp. 97-119). The use of MVA as a recombinant HIV vaccine (MVA-B) is being tested in approximately 300 volunteers in several Phase I studies conducted by the International AIDS Vaccine Initiative. Studies in mice and nonhuman primates have further demonstrated the safety of MVA under conditions of immune suppression. Compared to replicating vaccinia viruses, MVA provides similar or higher levels of recombinant gene expression even in non- permissive cells.
As used herein the term “humoral immune response” means the immune response involving the transformation of B cells into plasma cells that produce and secrete antibodies to a specific antigen. In the context of the present invention “assessing a subject’s humoral immune response to a vaccine” means detecting antibody response, in particular neutralizing antibody response, after immunization with an antigen, against a pathogen (infectious agent, or tumor cells). Humoral immunity prevents infection and can be of different kinds. It is characterized, for example, by neutralizing antibodies responses induced after vaccination. These functional antibodies are the main actors of protective immunity, they are able to neutralize the ability of the virus to penetrate or replicate in cells. Antibodies can belong to different immunoglobulin classes and subclasses including for example serum IgG and mucosal IgG and IgA. These Ig can also act in other ways by opsonization or complement activation, involving serum immune regulatory proteins for pathogen elimination. Other ways to defend oneself is antibody-dependent cell-mediated virus inhibition (ADCVI) and antibody-dependent cell-mediated-cytotoxicity (ADCC). This is one of the mechanisms by which antibodies act to limit and contain infection. It’s carried out by natural killer cells (NK) through different proteins: perforins (pore-forming proteins), granzymes (serine proteases), reactive oxygen intermediates, and cytokines. These cells lysis an infected cell labeled with antibody bound to antigen present on the membrane.
The term "protective humoral immunity" as used herein, means a humoral immune response that confers the essential component of protection against a pathogen.
The term "sterilizing humoral immunity" as used herein, means a humoral immune response that prevents the establishment of any detectable infection by a pathogen.
The term "long-lasting humoral immunity" as used herein, means that some aspect of humoral immunity is detectable three months after antigen administration, such as, for example, antibodies elicited by the antigen. Suitable methods of antibody detection include, but are not limited to, such methods as ELISA, immunofluorescence (IF A), focus reduction neutralization tests (FRNT), immunoprecipitation, and Western blotting.
The term "neutralizing humoral response" as used herein, means that the antibodies elicited during humoral immunity directly block the ability of a pathogen to infect cells.
The term "quick response" as used herein, means that protective humoral immunity is conferred within three weeks of antigen administration. The term "very quick response" as used herein, means that protective humoral immunity is conferred within one week of antigen administration.
A “responder to a vaccine” means that after vaccination of a subject, the serum antibody titer against the targeted antigen of said subject is sufficient to adequately clear the disease (infectious disease or tumor) and exhibit an efficient immune humoral activity. In such a case, then one can conclude that said patient has a greater proportion of immune B cells which induce an efficient immune humoral response in particular a neutralizing antibody response. Tests for determining the serum antibody titer are well known to the person skilled in the art. For example, a GFP Neutralizing assay (as described in example section) to detected an anti-antigen neutralizing activity in a serum of subjects collected 8 days after vaccination based on GFP detection by flow cytometry can be used.
Another example, according to the US FDA Guidance for Industry document, having a hemagglutinin inhibition (HI) antibody titer of 1 :40 against influenza (seroprotection) or a fourfold increase in HI titer following influenza vaccination (sero-conversion) are considered protective.
A "vaccine composition" or a “vaccine”, once it has been administered to a subject or an animal, elicits a protective immune response against said one or more antigen(s) which is (are) comprised herein. Accordingly, the vaccine composition according to the present invention, once it has been administered to the subject or the animal, induces a protective immune response against, for example, a microorganism, or to efficaciously protect the subject or the animal against infection.
A variety of substances can be used as antigens in a compound or formulation, of immunogenic or vaccine type. For example, attenuated and inactivated viral and bacterial pathogens, purified macromolecules, polysaccharides, toxoids, recombinant antigens, organisms containing a foreign gene from a pathogen, synthetic peptides, polynucleic acids (DNA, RNA), antibodies and tumor cells can be used to prepare (i) an immunogenic composition useful to induce an immune response in an individual or (ii) a vaccine useful for treating a pathological condition.
An antigen is additionally capable of being recognized by the immune system and/or being capable of inducing a humoral immune response and/or cellular immune response leading to the activation of B- and/or T-lymphocytes. An antigen can have one or more epitopes or antigenic sites (B- and T- epitopes)
Therefore, a wide variety of antigens to produce a vaccine composition useful for inducing an immune response in an individual can be used.
Those skilled in the art will be able to select an antigen appropriate for treating a particular pathological condition and will know how to determine whether an isolated antigen is favored in a particular vaccine formulation.
An isolated antigen can be prepared using a variety of methods well known in the art. A gene encoding any immunogenic polypeptide can be isolated and cloned, for example, in bacterial, yeast, insect, reptile or mammalian cells using recombinant methods well known in the art and described, for example in Sambrook et al., Molecular cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York (1992) and in Ansubel et al., Current Protocols in Molecular Biology, John Wiley and Sons, Baltimore, MD (1998). A number of genes encoding surface antigens from viral, bacterial and protozoan pathogens have been successfully cloned, expressed and used as antigens for vaccine development. For example, the major surface antigen of hepatitis B virus, HbsAg, the P subunit of choleratoxin, the enterotoxin of E. coh. the circumsporozoite protein of the malaria parasite, and a glycoprotein membrane antigen from Epstein-Barr virus, as well as tumor cell antigens, have been expressed in various well known vector/host systems, purified and used in vaccines.
A pathologically aberrant cell may also be used in a vaccine composition according to the invention can be obtained from any source such as one or more individuals having a pathological condition or ex vivo or in vitro cultured cells obtained from one or more such individuals, including a specific individual to be treated with the resulting vaccine.
As used herein, the term “sample “ or "biological sample" as used herein refers to any biological sample of a subject and can include, by way of example and not limitation, bodily fluids and/or tissue extracts such as homogenates or solubilized tissue obtained from a subject. Tissue extracts are obtained routinely from tissue biopsy. In a particular embodiment regarding the prognostic method according to the invention, the biological sample is a body fluid sample (such blood sample) or tissue biopsy of said subject.
In preferred embodiments, the body fluid sample is a blood sample. The term “blood sample” means a whole blood sample obtained from a subject (e.g. an individual for which it is interesting to determine whether a gene expression level can be identified).
The term “prior to the administration of (said) vaccine” means that the level of one or more gene expression level selected from a group of gene consisting of: IGLV8-61, BLK, EBF1 and IFNal6 gene, is measured at least 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, 0 days prior the administration of the vaccine. Preferably the level of one or more gene expression level selected from a group of gene consisting of: IGLV8-61, BLK, EBF1 and IFNal6 gene, is measured at least 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, 0 days prior the administration of the vaccine.
As used herein, the term " IFNal6" also known as “Interferon alpha-16” or “IFN alpha 16”, has its general meaning in the art and refers to a cytokine that in humans is encoded by the IFNA16 gene (Gene ID: 3449). Produced by macrophages, IFN-alpha 16 have antiviral activities. Interferon alpha 16 stimulates the production of two enzymes: a protein kinase and an oligoadenylate synthetase. The sequence of said protein may be found with the NCBI Reference: NM_002173 and NP_002164. As used herein, the term "IGLV8-61", also known as “Immunoglobulin lambda variable 8-61” has its general meaning in the art refers to a protein that in humans is encoded by the IGLV8-61gene (gene ID 28774). V region of the variable domain of immunoglobulin light chains that participates in the antigen recognition. Immunoglobulins, also known as antibodies, are membrane-bound or secreted glycoproteins produced by B lymphocytes. The antigen binding site is formed by the variable domain of one heavy chain, together with that of its associated light chain. Thus, each immunoglobulin has two antigen binding sites with remarkable affinity for a particular antigen. The variable domains are assembled by a process called V-(D)-J rearrangement and can then be subjected to somatic hypermutations which, after exposure to antigen and selection, allow affinity maturation for a particular antigen
The sequence of said gene may be found with the NCBI Reference: NG 000002 and NC_000022 (Reference GRCh38.pl3 Primary Assembly).
As used herein, the term BLK (Tyrosine-protein kinase BLK) also known as “B lymphocyte kinase” “BLK proto-oncogene” or “Src family tyrosine kinase” has its general meaning in the art and refers to an enzyme that in humans is encoded by the B LK gene (Gene ID: 640). BLK gene encodes a nonreceptor tyrosine-kinase of the src family of proto- oncogenes that are typically involved in cell proliferation and differentiation. The protein has a role in B-cell receptor signaling and B-cell development. The protein also stimulates insulin synthesis and secretion in response to glucose and enhances the expression of several pancreatic beta-cell transcription factors.
The sequence of said protein may be found with the NCBI Reference: NM_001715/NM_001330465 (isoform 1 and isoform 2) and NP_001317394/NP_001706 (isoform 1 and isoform 2).
As used herein, the term EBF1 also known as “Early B-Cell Factor 1” “or “Transcription factor COE1” has its general meaning in the art and refers to a protein that in humans is encoded by the EBF1 gene (Gene ID: 1879). The transcription factor EBF1 controls the expression of key proteins required for B cell differentiation, signal transduction and function (Treiber, T. et al (2010). Immunity. 32 (5): 714-725.; Hagman J. et al (2012). Current Topics in Microbiology and Immunology. 356: 17-38). The crucial role of this factor is shown in the regulation of expression of SLAM family co-receptors in B-cells (Schwartz, A. et al. (2016). Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms. 1859 (10): 1259-1268).
The sequence of said protein may be found with the NCBI Reference: NM_001290360 and NP_001277289 (isoform 1)/ NM_024007 and NP_076870 (isoform 2)/ NMJ82708 and NP_874367 (isoform 3)/ NM_001324101 and NP_001311030 (isoform 4)/ NM_001324103 and NP_001311032 (isoform 5)/ NM_001324106 and NP_001311035 (isoform 6)/ NM_001324107 and NP_001311036 ((isoform 7)/ NM_001324108 and NP_001311037 (isoform 8)/ NM_001324109 and NP_001311038 (isoform 9)/ NM_001324111 and NP_001311040 (isoform 10)/ NM_001364155 and NP_001351084 (isoform 11)/ NM_001364156 and NP_001351085 (isoform 13)/ NM_001364157 and NP_001351086 (isoform 14)/ NM_001364158 and NP_001351087 (isoform 15)/ NM_001364159 and NP_001351088 (isoform 16).
The inventors have found biomarkers (gene expression level selected from a group of gene) associated with subject’s humoral immune response to a vaccine and have identified 4 biomarkers which could be used separately or in combination.
Thus, in one aspect, the present invention provides an in vitro method for assessing a subject’s humoral immune response to a vaccine, comprising the step of (a) measuring in a sample obtained from said subject prior to the administration of said vaccine, the level of one or more gene expression level selected from a group of genes consisting of: IFNal6, IGLV8- 61, BLK, and EBF1 gene and b) wherein the level of one or more gene expression of step a) is negatively correlated with the humoral immune response of said subject.
In preferred embodiments, a plurality of gene expression level biomarkers (i.e., one or more than one gene expression level biomarkers) is used in the method of prognosis. In other words, the method of the invention may comprise steps of: measuring in the biological sample plurality of gene expression level biomarkers, between one, two, three; four, gene expression level biomarker selected from a group of gene consisting of: IFNal6, IGLV8-61, BLK, and EBF1 gene present in the biological sample.
In particular embodiments, the method of prognosis is performed using the four different gene expression level biomarkers including the IFNal6, IGLV8-61, BLK, and EBF1 gene.
Measuring the expression level of a gene can be performed by a variety of techniques well known in the art.
Typically, the expression level of a gene may be determined by determining the quantity of mRNA. Methods for determining the quantity of mRNA are well known in the art. For example, the nucleic acid contained in the samples (e.g., blood, cell or tissue prepared from the patient) is first extracted according to standard methods, for example using lytic enzymes or chemical solutions or extracted by nucleic-acid-binding resins following the manufacturer's instructions. The extracted mRNA is then detected by hybridization (e. g., Northern blot analysis, in situ hybridization) and/or amplification (e.g., RT-PCR).
Other methods of Amplification include ligase chain reaction (LCR), transcription- mediated amplification (TMA), strand displacement amplification (SDA) and nucleic acid sequence-based amplification (NASBA).
Nucleic acids having at least 10 nucleotides and exhibiting sequence complementarity or homology to the mRNA of interest herein find utility as hybridization probes or amplification primers. It is understood that such nucleic acids need not be identical, but are typically at least about 80% identical to the homologous region of comparable size, more preferably 85% identical and even more preferably 90-95% identical. In certain embodiments, it will be advantageous to use nucleic acids in combination with appropriate means, such as a detectable label, for detecting hybridization.
Typically, the nucleic acid probes include one or more labels, for example to permit detection of a target nucleic acid molecule using the disclosed probes. In various applications, such as in situ hybridization procedures, a nucleic acid probe includes a label (e.g., a detectable label). A “detectable label” is a molecule or material that can be used to produce a detectable signal that indicates the presence or concentration of the probe (particularly the bound or hybridized probe) in a sample. Thus, a labeled nucleic acid molecule provides an indicator of the presence or concentration of a target nucleic acid sequence (e.g., genomic target nucleic acid sequence) (to which the labeled uniquely specific nucleic acid molecule is bound or hybridized) in a sample. A label associated with one or more nucleic acid molecules (such as a probe generated by the disclosed methods) can be detected either directly or indirectly. A label can be detected by any known or yet to be discovered mechanism including absorption, emission and/ or scattering of a photon (including radio frequency, microwave frequency, infrared frequency, visible frequency and ultra-violet frequency photons). Detectable labels include colored, fluorescent, phosphorescent and luminescent molecules and materials, catalysts (such as enzymes) that convert one substance into another substance to provide a detectable difference (such as by converting a colorless substance into a colored substance or vice versa, or by producing a precipitate or increasing sample turbidity), haptens that can be detected by antibody binding interactions, and paramagnetic and magnetic molecules or materials.
Particular examples of detectable labels include fluorescent molecules (or fluorochromes). Numerous fluorochromes are known to those of skill in the art, and can be selected, for example from Life Technologies (formerly Invitrogen), e.g., see, The Handbook — A Guide to Fluorescent Probes and Labeling Technologies). Examples of particular fluorophores that can be attached (for example, chemically conjugated) to a nucleic acid molecule (such as a uniquely specific binding region) are provided in U.S. Pat. No. 5,866, 366 to Nazarenko et al., such as 4-acetamido-4'-isothiocyanatostilbene-2,2' disulfonic acid, acridine and derivatives such as acridine and acridine isothiocyanate, 5-(2'-aminoethyl) aminonaphthalene- 1 -sulfonic acid (EDANS), 4-amino -N- [3 vinylsulfonyl)phenyl]naphthalimide-3,5 disulfonate (Lucifer Yellow VS), N-(4-anilino-l- naphthyl)maleimide, antllranilamide, Brilliant Yellow, coumarin and derivatives such as coumarin, 7-amino-4-methylcoumarin (AMC, Coumarin 120), 7-amino-4- trifluoromethylcouluarin (Coumarin 151); cyanosine; 4',6-diarninidino-2-phenylindole (DAPI); 5',5"dibromopyrogallol-sulfonephthalein (Bromopyrogallol Red); 7 -diethylamino -3 (4'-isothiocyanatophenyl)-4-methylcoumarin; diethylenetriamine pentaacetate; 4,4'- diisothiocyanatodihydro-stilbene-2,2'-disulfonic acid; 4,4'-diisothiocyanatostilbene-2,2'- disulforlic acid; 5-[dimethylamino] naphthalene- 1 -sulfonyl chloride (DNS, dansyl chloride); 4-(4'-dimethylaminophenylazo)benzoic acid (DABCYL); 4-dimethylaminophenylazophenyl- 4'-isothiocyanate (DABITC); eosin and derivatives such as eosin and eosin isothiocyanate; erythrosin and derivatives such as erythrosin B and erythrosin isothiocyanate; ethidium; fluorescein and derivatives such as 5-carboxyfluorescein (FAM), 5-(4,6diclllorotriazin-2- yDaminofluorescein (DTAF), 2'7'dimethoxy-4'5'-dichloro-6-carboxyfluorescein (JOE), fluorescein, fluorescein isothiocyanate (FITC), and QFITC Q(RITC); 2',7'-difluorofluorescein (OREGON GREEN®); fluorescamine; IR144; IR1446; Malachite Green isothiocyanate; 4- methylumbelliferone; ortho cresolphthalein; nitrotyrosine; pararosaniline; Phenol Red; B- phycoerythrin; o-phthaldialdehyde; pyrene and derivatives such as pyrene, pyrene butyrate and succinimidyl 1 -pyrene butyrate; Reactive Red 4 (Cibacron Brilliant Red 3B-A); rhodamine and derivatives such as 6-carboxy-X-rhodamine (ROX), 6-carboxyrhodamine (R6G), lissamine rhodamine B sulfonyl chloride, rhodamine (Rhod), rhodamine B, rhodamine 123, rhodamine X isothiocyanate, rhodamine green, sulforhodamine B, sulforhodamine 101 and sulfonyl chloride derivative of sulforhodamine 101 (Texas Red); N,N,N',N' -tetramethyl - 6-carboxyrhodamine (TAMRA); tetramethyl rhodamine; tetramethyl rhodamine isothiocyanate (TRITC); riboflavin; rosolic acid and terbium chelate derivatives. Other suitable fluorophores include thiol -reactive europium chelates which emit at approximately 617 mn (Heyduk and Heyduk, Analyt. Biochem. 248:216-27, 1997; J. Biol. Chem. 274:3315- 22, 1999), as well as GFP, LissamineTM, di ethylaminocoumarin, fluorescein chlorotriazinyl, naphthofluorescein, 4,7-dichlororhodamine and xanthene (as described in U.S. Pat. No. 5,800,996 to Lee et al.) and derivatives thereof. Other fluorophores known to those skilled in the art can also be used, for example those available from Life Technologies (Invitrogen; Molecular Probes (Eugene, Oreg.)) and including the ALEXA FLUOR® series of dyes (for example, as described in U.S. Pat. Nos. 5,696,157, 6, 130, 101 and 6,716,979), the BODIPY series of dyes (dipyrrometheneboron difluoride dyes, for example as described in U.S. Pat. Nos. 4,774,339, 5,187,288, 5,248,782, 5,274,113, 5,338,854, 5,451,663 and 5,433,896), Cascade Blue (an amine reactive derivative of the sulfonated pyrene described in U.S. Pat. No. 5,132,432) and Marina Blue (U.S. Pat. No. 5,830,912).
In addition to the fluorochromes described above, a fluorescent label can be a fluorescent nanoparticle, such as a semiconductor nanocrystal, e.g., a QUANTUM DOTTM (obtained, for example, from Life Technologies (QuantumDot Corp, Invitrogen Nanocrystal Technologies, Eugene, Oreg.); see also, U.S. Pat. Nos. 6,815,064; 6,682,596; and 6,649, 138). Semiconductor nanocrystals are microscopic particles having size-dependent optical and/or electrical properties. When semiconductor nanocrystals are illuminated with a primary energy source, a secondary emission of energy occurs of a frequency that corresponds to the handgap of the semiconductor material used in the semiconductor nanocrystal. This emission can he detected as colored light of a specific wavelength or fluorescence. Semiconductor nanocrystals with different spectral characteristics are described in e.g., U.S. Pat. No. 6,602,671. Semiconductor nanocrystals that can he coupled to a variety of biological molecules (including dNTPs and/or nucleic acids) or substrates by techniques described in, for example, Bruchez et al., Science 281 :20132016, 1998; Chan et al., Science 281 :2016- 2018, 1998; and U.S. Pat. No. 6,274,323. Formation of semiconductor nanocrystals of various compositions are disclosed in, e.g., U.S. Pat. Nos. 6,927, 069; 6,914,256; 6,855,202; 6,709,929; 6,689,338; 6,500,622; 6,306,736; 6,225,198; 6,207,392; 6,114,038; 6,048,616; 5,990,479; 5,690,807; 5,571,018; 5,505,928; 5,262,357 and in U.S. Patent Publication No. 2003/0165951 as well as PCT Publication No. 99/26299 (published May 27, 1999). Separate populations of semiconductor nanocrystals can he produced that are identifiable based on their different spectral characteristics. For example, semiconductor nanocrystals can he produced that emit light of different colors based on their composition, size or size and composition. For example, quantum dots that emit light at different wavelengths based on size (565 mn, 655 mn, 705 mn, or 800 mn emission wavelengths), which are suitable as fluorescent labels in the probes disclosed herein are available from Life Technologies (Carlshad, Calif.). Additional labels include, for example, radioisotopes (such as 3 H), metal chelates such as DOTA and DPTA chelates of radioactive or paramagnetic metal ions like Gd3+, and liposomes.
Detectable labels that can he used with nucleic acid molecules also include enzymes, for example horseradish peroxidase, alkaline phosphatase, acid phosphatase, glucose oxidase, beta-galactosidase, beta-glucuronidase, or beta-lactamase.
Alternatively, an enzyme can he used in a metallographic detection scheme. For example, silver in situ hybridization (SISH) procedures involve metallographic detection schemes for identification and localization of a hybridized genomic target nucleic acid sequence. Metallographic detection methods include using an enzyme, such as alkaline phosphatase, in combination with a water-soluble metal ion and a redox-inactive substrate of the enzyme. The substrate is converted to a redox-active agent by the enzyme, and the redoxactive agent reduces the metal ion, causing it to form a detectable precipitate. (See, for example, U.S. Patent Application Publication No. 2005/0100976, PCT Publication No. 2005/ 003777 and U.S. Patent Application Publication No. 2004/ 0265922). Metallographic detection methods also include using an oxido-reductase enzyme (such as horseradish peroxidase) along with a water-soluble metal ion, an oxidizing agent and a reducing agent, again to form a detectable precipitate. (See, for example, U.S. Pat. No. 6,670,113).
Probes made using the disclosed methods can be used for nucleic acid detection, such as ISH procedures (for example, fluorescence in situ hybridization (FISH), chromogenic in situ hybridization (CISH) and silver in situ hybridization (SISH)) or comparative genomic hybridization (CGH).
In situ hybridization (ISH) involves contacting a sample containing target nucleic acid sequence (e.g., genomic target nucleic acid sequence) in the context of a metaphase or interphase chromosome preparation (such as a cell or tissue sample mounted on a slide) with a labeled probe specifically hybridizable or specific for the target nucleic acid sequence (e.g., genomic target nucleic acid sequence). The slides are optionally pretreated, e.g., to remove paraffin or other materials that can interfere with uniform hybridization. The sample and the probe are both treated, for example by heating to denature the double stranded nucleic acids. The probe (formulated in a suitable hybridization buffer) and the sample are combined, under conditions and for sufficient time to permit hybridization to occur (typically to reach equilibrium). The chromosome preparation is washed to remove excess probe, and detection of specific labeling of the chromosome target is performed using standard techniques. For example, a biotinylated probe can be detected using fluorescein-labeled avidin or avidin-alkaline phosphatase. For fluorochrome detection, the fluorochrome can be detected directly, or the samples can be incubated, for example, with fluorescein isothiocyanate (FITC)-conjugated avidin. Amplification of the FITC signal can be affected, if necessary, by incubation with biotin-conjugated goat anti-avidin antibodies, washing and a second incubation with FITC-conjugated avidin. For detection by enzyme activity, samples can be incubated, for example, with streptavidin, washed, incubated with biotin-conjugated alkaline phosphatase, washed again and pre-equilibrated (e.g., in alkaline phosphatase (AP) buffer). For a general description of in situ hybridization procedures, see, e.g., U.S. Pat. No. 4,888,278.
Numerous procedures for FISH, CISH, and SISH are known in the art. For example, procedures for performing FISH are described in U.S. Pat. Nos. 5,447,841; 5,472,842; and 5,427,932; and for example, in Pirlkel et al., Proc. Natl. Acad. Sci. 83:2934-2938, 1986; Pinkel et al., Proc. Natl. Acad. Sci. 85:9138-9142, 1988; and Lichter et al., Proc. Natl. Acad. Sci. 85:9664-9668, 1988. CISH is described in, e.g., Tanner et al., Am. .1. Pathol. 157: 1467- 1472, 2000 and U.S. Pat. No. 6,942,970. Additional detection methods are provided in U.S. Pat. No. 6,280,929.
Numerous reagents and detection schemes can be employed in conjunction with FISH, CISH, and SISH procedures to improve sensitivity, resolution, or other desirable properties. As discussed above probes labeled with fluorophores (including fluorescent dyes and QUANTUM DOTS®) can be directly optically detected when performing FISH. Alternatively, the probe can be labeled with a nonfluorescent molecule, such as a hapten (such as the following non-limiting examples: biotin, digoxigenin, DNP, and various oxazoles, pyrrazoles, thiazoles, nitroaryls, benzofurazans, triterpenes, ureas, thioureas, rotenones, coumarin, courmarin-based compounds, Podophyllotoxin, Podophyllotoxin-based compounds, and combinations thereof), ligand or other indirectly detectable moiety. Probes labeled with such non-fluorescent molecules (and the target nucleic acid sequences to which they bind) can then be detected by contacting the sample (e.g., the cell or tissue sample to which the probe is bound) with a labeled detection reagent, such as an antibody (or receptor, or other specific binding partner) specific for the chosen hapten or ligand. The detection reagent can be labeled with a fluorophore (e.g., QUANTUM DOT®) or with another indirectly detectable moiety, or can be contacted with one or more additional specific binding agents (e.g., secondary or specific antibodies), which can be labeled with a fluorophore.
In other examples, the probe, or specific binding agent (such as an antibody, e.g., a primary antibody, receptor or other binding agent) is labeled with an enzyme that is capable of converting a fluorogenic or chromogenic composition into a detectable fluorescent, colored or otherwise detectable signal (e.g., as in deposition of detectable metal particles in SISH). As indicated above, the enzyme can be attached directly or indirectly via a linker to the relevant probe or detection reagent. Examples of suitable reagents (e.g., binding reagents) and chemistries (e.g., linker and attachment chemistries) are described in U.S. Patent Application Publication Nos. 2006/0246524; 2006/0246523, and 2007/ 01 17153.
It will be appreciated by those of skill in the art that by appropriately selecting labelled probe-specific binding agent pairs, multiplex detection schemes can he produced to facilitate detection of multiple target nucleic acid sequences (e.g., genomic target nucleic acid sequences) in a single assay (e.g., on a single cell or tissue sample or on more than one cell or tissue sample). For example, a first probe that corresponds to a first target sequence can he labelled with a first hapten, such as biotin, while a second probe that corresponds to a second target sequence can be labelled with a second hapten, such as DNP. Following exposure of the sample to the probes, the bound probes can he detected by contacting the sample with a first specific binding agent (in this case avidin labelled with a first fluorophore, for example, a first spectrally distinct QUANTUM DOT®, e.g., that emits at 585 mn) and a second specific binding agent (in this case an anti-DNP antibody, or antibody fragment, labelled with a second fluorophore (for example, a second spectrally distinct QUANTUM DOT®, e.g., that emits at 705 mn). Additional probes/binding agent pairs can be added to the multiplex detection scheme using other spectrally distinct fluorophores. Numerous variations of direct, and indirect (one step, two step or more) can he envisioned, all of which are suitable in the context of the disclosed probes and assays.
Probes typically comprise single-stranded nucleic acids of between 10 to 1000 nucleotides in length, for instance of between 10 and 800, more preferably of between 15 and 700, typically of between 20 and 500. Primers typically are shorter single-stranded nucleic acids, of between 10 to 25 nucleotides in length, designed to perfectly or almost perfectly match a nucleic acid of interest, to be amplified. The probes and primers are “specific” to the nucleic acids they hybridize to, i.e. they preferably hybridize under high stringency hybridization conditions (corresponding to the highest melting temperature Tm, e.g., 50 % formamide, 5x or 6x SCC. SCC is a 0.15 M NaCl, 0.015 M Na-citrate).
The nucleic acid primers or probes used in the above amplification and detection method may be assembled as a kit. Such a kit includes consensus primers and molecular probes. A preferred kit also includes the components necessary to determine if amplification has occurred. The kit may also include, for example, PCR buffers and enzymes; positive control sequences, reaction control primers; and instructions for amplifying and detecting the specific sequences.
In a particular embodiment, the methods of the invention comprise the steps of providing total RNAs extracted from blood and subjecting the RNAs to amplification and hybridization to specific probes, more particularly by means of a quantitative or semi- quantitative RT-PCR.
In another preferred embodiment, the expression level is determined by DNA chip analysis. Such DNA chip or nucleic acid microarray consists of different nucleic acid probes that are chemically attached to a substrate, which can be a microchip, a glass slide or a microsphere-sized bead. A microchip may be constituted of polymers, plastics, resins, polysaccharides, silica or silica-based materials, carbon, metals, inorganic glasses, or nitrocellulose. Probes comprise nucleic acids such as cDNAs or oligonucleotides that may be about 10 to about 60 base pairs. To determine the expression level, a sample from a test subject, optionally first subjected to a reverse transcription, is labelled and contacted with the microarray in hybridization conditions, leading to the formation of complexes between target nucleic acids that are complementary to probe sequences attached to the microarray surface. The labelled hybridized complexes are then detected and can be quantified or semi-quantified. Labelling may be achieved by various methods, e.g. by using radioactive or fluorescent labelling. Many variants of the microarray hybridization technology are available to the man skilled in the art (see e.g. the review by Hoheisel, Nature Reviews, Genetics, 2006, 7:200- 210).
Expression level of a gene may be expressed as absolute expression level or normalized expression level. Typically, expression levels are normalized by correcting the absolute expression level of a gene by comparing its expression to the expression of a gene that is not a relevant for determining the cancer stage of the patient, e.g., a housekeeping gene that is constitutively expressed. Suitable genes for normalization include housekeeping genes such as the actin gene ACTB, ribosomal 18S gene, GUSB, PGK1, TBP, HPRT1 and TFRC. TATA-binding protein TBP) and hypoxanthine phosphoribosyl transferase 1 (HPRTT) were used as reference genes in the present study. This normalization allows the comparison of the expression level in one sample, e.g., a patient sample, to another sample, or between samples from different sources.
The man skilled in the art also understands that the same technique of assessment of the expression level of a gene should of course be used for obtaining the reference value and thereafter for assessment of the expression level of a gene of a patient subjected to the method of the invention.
Said reference control values may be determined in regard to the level of gene expression biomarker present in blood samples taken from one or more healthy subject(s) or in a control population.
In one embodiment, the method according to the present invention comprises the step of comparing said level of humoral immune response to a vaccine -specific gene expression level biomarkers (“Biomarkerl”: IFNal6 gene and/or “Biomarker2”: IGLV8-61 gene and/or “Biomarker3”: BLK gene and/or “Biomarker4”: EBF1 gene) to a control reference value wherein a high level of “humoral immune response to a vaccine -specific” gene expression biomarkers (“Biomarkerl”: IFNal6 gene and/or “Biomarker2”: IGLV8-61 gene and/or “Biomarker3”: BLK gene and/or “Biomarker4”: EBF1 gene) compared to said control reference value is predictive of a high risk to be a “low responder” to said vaccine and a low “"humoral immune response to a vaccine -specific” gene expression biomarkers (Biomarkerl”: IFNal6 gene and/or “Biomarker2”: IGLV8-61 gene and/or “Biomarker3”: BLK gene and/or “Biomarker4”: EBF1 gene) compared to said control reference value is predictive of a high risk to be a “high responder” to said vaccine.
The control reference value may depend on various parameters such as the method used to measure the level humoral immune response to a vaccine -specific gene expression level biomarkers (“Biomarkerl”: IFNal6 gene and/or “Biomarker2”: IGLV8-61 gene and/or “Biomarker3”: BLK gene and/or “Biomarker4”: EBF1 gene) or the gender of the subject.
Control reference values are easily determinable by the one skilled in the art, by using the same techniques as for determining the level of gene expression biomarker in a blood samples previously collected from the patient under testing.
A “reference value” can be a “threshold value” or a “cut-off value”. Typically, a "threshold value" or "cut-off value" can be determined experimentally, empirically, or theoretically. A threshold value can also be arbitrarily selected based upon the existing experimental and/or clinical conditions, as would be recognized by a person of ordinary skilled in the art. The threshold value has to be determined in order to obtain the optimal sensitivity and specificity according to the function of the test and the benefit/risk balance (clinical consequences of false positive and false negative). Typically, the optimal sensitivity and specificity (and so the threshold value) can be determined using a Receiver Operating Characteristic (ROC) curve based on experimental data. Preferably, the person skilled in the art may compare the level of gene expression biomarkers (“Biomarkerl”: IFNal6 gene and/or “Biomarker2”: IGLV8-61 gene and/or “Biomarker3”: BLK gene and/or “Biomarker4”: EBF1 gene) with a defined threshold value. In one embodiment of the present invention, the threshold value is derived from the gene expression level (or ratio, or score) determined in a blood sample derived from one or more subjects who are responders (to the method according to the invention). In one embodiment of the present invention, the threshold value may also be derived from gene expression level (or ratio, or score) determined in a blood sample derived from one or more subjects or who are non-responders. Furthermore, retrospective measurement of the gene expression level (or ratio, or scores) in properly banked historical subject samples may be used in establishing these threshold values.
For instance in the MVA-B vaccination study and with the expression level of IFNal6, IGLV8-61, BLK, and/or EBF1 gene assessed by microarray hybridation, the ROC curve analysis revealed that choosing a cut-off value at 3.5 unit expression of IFNal6 mRNA, and/or at 6.5 unit expression of IGLV8-61 mRNA and/or at 6 unit expression of BLK, and/or at 5.9 unit expression of EBF1 mRNA levels allowed to effectively discriminate responders from / non responders blood sample and which could be used as predetermined reference level for IFNal6, IGLV8-61, BLK, and/or EBF1.
"Risk" in the context of the present invention, relates to the probability that an event will occur over a specific time period, as in humoral immune response of a subject to a vaccine, and can mean a subject's "absolute" risk or "relative" risk. Absolute risk can be measured with reference to either actual observation post-measurement for the relevant time cohort, or with reference to index values developed from statistically valid historical cohorts that have been followed for the relevant time period. Relative risk refers to the ratio of absolute risks of a subject compared either to the absolute risks of low-risk cohorts or an average population risk, which can vary by how clinical risk factors are assessed. Odds ratios, the proportion of positive events to negative events for a given test result, are also commonly used (odds are according to the formula p/(l-p) where p is the probability of event and (1- p) is the probability of no event) to no conversion. Alternative continuous measures, which may be assessed in the context of the present invention, include time to humoral immune response of a subject to a vaccine risk reduction ratios.
"Risk evaluation," or "evaluation of risk" in the context of the present invention encompasses making a prediction of the probability, odds, or likelihood that an event (humoral immune response of a subject to a vaccine) may occur, the rate of occurrence of the event or conversion from one state to another, i.e., from a “high responder” to said vaccine to “low responder” to said vaccine. Risk evaluation can also comprise prediction of future clinical parameters, traditional laboratory risk factor values, or other indices of “humoral response”, such as cellular population determination in peripheral tissues, in serum or other fluid, either in absolute or relative terms in reference to a previously measured population. The methods of the present invention may be used to make continuous or categorical measurements of the risk of an event (humoral immune response of a subject to a vaccine), thus prognosing and defining the risk spectrum of a category of subjects defined as being “high responder” to a vaccine. In the categorical scenario, the invention can be used to discriminate between normal and other subject cohorts at higher risk to be “high responder” to said vaccine. In other embodiments, the present invention may be used so as to help to discriminate those having “high responder” to a vaccine from “low responder” to said vaccine
Typically, the methods for assessing a subject’s humoral immune response to a vaccine may further comprise measuring, in a biological sample obtained from the subject, the level Eubacterium in stool sample and Prevotella in skin sample.
In a particular embodiment the level Eubacterium in stool sample and Prevotella in skin sample is performed by using 16S ribosomal sequencing.
Kit for performing the method of the invention
A further object of the invention relates to kits for performing the methods of the invention, wherein said kits comprise means for measuring the expression level of one or more gene expression level selected from a group of gene consisting of: IGLV8-61, BLK, EBF1 and IFNal6 gene of the invention in the sample obtained from the patient for use to assess a subject’s humoral immune response to a vaccine.
Accordingly, the present invention also relates to a kit of the invention comprising means for determining the expression level of one or more gene expression level selected from a group of gene consisting of IGLV8-61, BLK, EBF1 and IFNal6 gene.
In one embodiment, the present invention relates to a kit for use to assess a subject’s humoral immune response to a vaccine, comprising:
- at least a means for determining the expression level of one or more gene expression level selected from a group of gene consisting of IGLV8-61, BLK, EBF1 and IFNal6 gene and
- instructions for use.
In a particular embodiment, the kit for use comprising: - amplification primers and/or probe for determining the expression level one or more gene expression level selected from a group of gene consisting of IGLV8-61, BLK, EBF1 and IFNal6 gene,
- instructions for use.
The kits may include probes, primers macroarrays or microarrays as above described. For example, the kit may comprise a set of probes as above defined, usually made of DNA, and that may be pre-labelled. Alternatively, probes may be unlabelled and the ingredients for labelling may be included in the kit in separate containers. The kit may further comprise hybridization reagents or other suitably packaged reagents and materials needed for the particular hybridization protocol, including solid-phase matrices, if applicable, and standards. Alternatively the kit of the invention may comprise amplification primers that may be pre- labelled or may contain an affinity purification or attachment moiety. The kit may further comprise amplification reagents and also other suitably packaged reagents and materials needed for the particular amplification protocol.
Therapeutic method
The invention also relates to a method for vaccinating a subject in need thereof with an antigen wherein, prior to vaccination; the level of one or more gene expression level selected from a group of gene consisting of IFNal6, IGLV8-61, BLK and EBF1 gene obtained from said subject have been detected by one of method of the invention.
Another object of the present invention is a method for vaccinating in a subject comprising, prior to vaccination the steps of a) providing a blood sample from a subject, b) detecting the level of one or more gene expression level selected from a group of gene consisting of IFNal6, IGLV8-61, BLK and EBF1 gene obtained from said subject c) comparing the level determined in step b) with a reference value and if level determined at step b) is lower than the reference value, vaccinating the subject with antigen.
The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention. FIGURES:
Figure 1: Blood gene expression combined with host microbiota before vaccination shapes MVA-B responses
A. Investigation of the blood gene expression (w-2, wO) correlated with MVA-Nab response (w8) and host genus diversity (wO), for skin and stool. The Spearman correlation test was applied with P<0.05 defined as statistically significant. B. Table shows the significant correlation coefficients and P-values for each of the three genes with the abundance of the genus correlated with MVA-Nab response. C. ROC curves show the specificity and the sensitivity of the logistic regression models, i.e., the proportion of correctly predicted responders and nonresponders, respectively. The logistic regression is based on the expression of the minimal gene signature (IGLV8, EBF1, and BLK) and the abundance of Eubacterium and Prevotella, respectively, in stool and skin.
Figure 2: ROC gene Nab: IFNal6, IGLV8-61, BLK and EBF1
Among 154 genes associated with NAb (Figure 1A), we found that IFNal6 was associated with B cell differentiation and proliferation. ROC curves show the specificity and the sensitivity of the logistic regression models, i.e., the proportion of correctly predicted responders and nonresponders, respectively. A. The logistic regression based on single level of IGLV8, B. EBF1, C. BLK and D. IFNal6. E. The logistic regression is based on the expression of 4 genes IGLV8, EBF1, IFNal6 and BLK.
EXAMPLE:
Methods:
Skin and feces sampling
For each individual, skin swab samples from the deltoid muscle region (-5-20 cm below the vaccine administration site) were collected before the vaccination (wO). Skin samples were collected with Catch-All™ Sample Collection Swab kits moistened with SCF-1 solution. The skin surface was sampled for 30 seconds by firmly swabbing the cotton tip back and forth -50 times. The cotton tip was stored in sterile tubes with MoBio solution at -80°C until DNA extraction. Fecal samples for each participant were collected in sterile fecal collection tubes the day before the vaccination, matching the skin sample time points. All samples were stored at 4-5°C until their reception at the IMP ACTA clinical trial site, where they were cryopreserved at -80°C. All samples were shipped on dry ice to the IrsiCaixa AIDS Research Institute for DNA extraction, amplification, and sequencing. DNA extraction and amplicon sequencing from skin and fecal samples
DNA extraction was performed with the DNA Extraction kit from Epicentre Technologies© (Madison, WI, USA). Six aliquots of buffer solution from the DNA extraction kit were used as negative controls. To amplify the variable V3-V4 region from the 16S rRNA gene, we used the primer pair described in the MiSeq™ rRNA Amplicon Sequencing protocol, which already has the Illumina adapter overhang nucleotide sequences added to the 16S rRNA V3-V4-specific primers, i.e., 16S F 5’-(TCG GCA GCG TCA GAT GTG TAT AAG AGA CAG CCT ACG GGN GGC WGC AG)-3’(SEQ ID N°6, Table 2) and 16S R 5’-(GTC TCG TGG GCT CGG AGA TGT GTA TAA GAG ACA GGA CTA CHV GGG TAT CTA ATC C) -3’(SEQ ID N°7, Table 2). Amplifications were performed in triplicate 25-pL reactions, each containing 2.5 pL of non-diluted DNA template, 12.5 pL of KAPA HiFi HotStart Ready Mix (containing KAPA HiFi HotStart DNA Polymerase, buffer, MgC12, and dNTPs, KAPA Biosystems Inc., Wilmington, MA, USA), and 5 pL of each primer at 1 pM. Thermal cycling conditions consisted of an initial denaturation step (3 minutes at 95°C), followed by 30 cycles of denaturation (30 seconds at 95°C), annealing (30 seconds at 55°C), and extension (30 seconds at 72°C). These were followed by a final extension step of 10 minutes at 72°C. Once the desired amplicon was confirmed in 1% agarose gel electrophoresis, all three replicates were pooled and stored at -30°C until the sequencing library was prepared. After amplified DNA templates were cleaned up for non-DNA molecules and Illumina sequencing adapters and dual indices attached with the Nextera XT Index Kit (Illumina, Inc.), the corresponding PCR amplification program was run, as described in the MiSeq 16S rRNA Amplicon Sequencing protocol. After a second round of cleanup, amplicons were quantified with the Quant-iT™ PicoGreen® dsDNA Assay Kit (Invitrogen, Carlsbad, MA, USA) and diluted in equimolar concentrations (4 nM) for further pooling. Sequencing was performed on an Illumina MiSeq™ platform (Illumina, Inc.) according to the manufacturer's specifications to generate a median of 30,644 paired-end sequences of -300 bp length in each direction (-61,289 reads per sample).
Sequence quality control and microbiota analyses
The quality of MiSeq raw sequences was assessed with the FastQC software (A. and Bittencourt a, 2010) (http://www.bioinformatics.babraham.ac.uk/proiects/fastqc/). Sequences were trimmed with Trimmommatic (Bolger et al., 2014), with a cutoff value of Q30 for both ends, a minimum mean threshold of Q20 for 30-bp-sliding window across sequences, and a minimum read length of 250 bp. After quality control, 28 samples including controls (n=8) and volunteers (n=10, 5 women and 5 men) for skin and stools, were further analyzed. Mothur pipeline (Schloss et al., 2009) was used to bin 16S rDNA sequences into operational taxonomic units (OTUs) with a threshold of 97% sequence similarity. OTUs present in only a single sample were discarded. Rarefaction curves were represented by defining the maximum subsampling size as the number of sequences of the sample with the fewest sequences (2751 sequences for skin samples, and 1059 sequences for stool samples). Richness and diversity indexes were estimated by using the summary, single module implemented in mothur. For taxonomical analysis, 16S rDNA sequences were classified according to the GreenGenes database (McDonald et al., 2012) version 13.5.99.
MVA-GFP Neutralizing antibody assay
Anti-MVA neutralizing activities were evaluated in serum collected at week 8 (w8) with an assay based on GFP detection by flow cytometry (Cosma et al., 2004; Earl et al., 2003). It used HeLa cells as targets and a recombinant strain of MVA expressing the enhanced Aequoriae GFP (Garcia et al., 2011). Serial dilutions of heat inactivated serum were performed in 96-well round-bottom tissue culture plates (TPP, Zurich, Switzerland) containing DMEM (Gibco, Invitrogen) supplemented with 2% fetal calf serum (PAA, Laboratories GmbH, Pashing, Austria). MVAeGFP was then added to each well at a MOI of 0.25. The plate was then incubated for 1 hour at 37°C until the addition of 1 x 105 HeLa cells. The incubation then continued for an additional 16 hours at 37°C, 0.5% CO2. After trypsinization, the cells were washed with PBS supplemented with 0.5% fetal calf serum and 2 mM EDTA and fixed with 2% formaldehyde. GFP expression was analyzed with FACSCanto II and Diva software (BD Biosciences). The percentage of neutralization was defined as the ratio of the reduction in the number of GFP-expressing cells to the number of GFP-expressing cells in untreated control wells.
RNA extraction and data preprocessing for transcriptomic analysis
Whole blood samples of 2.5 mL were collected in PAXgene RNA tubes (PreAnalytix) twice from each volunteer two weeks before (w-2) and the day of the vaccination (wO). These tubes enable the preservation and stabilization of RNA (storage at -80°C). Total RNA was extracted from whole blood according to the instructions in the handbook accompanying the PAXgene blood RNA Kit (PreAnalytiX, Hombrechtikon, Switzerland). RNA purity and integrity were assessed on the Agilent 2100 Bioanalyzer with the RNA 6000 Nano LabChip reagent set (Agilent, Palo Alto, CA, USA). Samples for microarray hybridization were prepared as described in the Affymetrix GeneChip WT PLUS Reagent Kit User Manual (Affymetrix, Inc., Santa Clara, CA, USA). For hybridization (to Affymetrix Human Gene 2.1 ST Array Plates), washing, staining, and scanning took place in an Affymetrix GeneTitan system, controlled by the Affymetrix GeneChip Command Console software w4.2. Background signal correction was performed by applying the backgroundCorrect function from the limma package on the perfect match (PM) signals with R Software 3.3.1. The underlying model is the normal-exponential convolution model from RMA (chip intensity: addition of a signal exponentially distributed, chip noise: follows Gaussian distribution) (Irizarry et al., 2003). The variance stabilizing transformation algorithm (justvsn function from the vsn package (Huber et al., 2002) was applied to the background corrected signal (monotonic transformation), and the signal then transformed back to its usual scale by exponentiation (base 2). To make the chips comparable, a quantile normalization (Bolstad et al., 2003) (normalize function from the affy package) was then applied to the variance- stabilized signal. The probe signals for replicated arrays were averaged and a quantile normalization performed anew. In all, 24,768 probes were analyzed.
Statistical analyses
Microbiome samples were clustered according to their genus composition by a nonmetric multidimensional scaling (NMDS) approach based on ecological distance matrices calculated by Bray-Curtis dissimilarities, as implemented in R (Vegan, metaMDS, and ggplot2 packages). NMDS ellipses were drawn based on a confidence interval (CI) of 0.95. To determine significant factors that describe the community structure better, we used a multivariate ADONIS test with terms added sequentially. The associations between baseline genus abundance or genus diversity, blood gene expression, and MVA-Nab response were evaluated by using the Spearman rank correlation test with significance defined by C- value <0.05. The heatmap was performed with values row-centered and scaled, Pearson correlation as the distance method and a dendrogram computed and reordered based on row means. The heatmap, logistic regression analyses, and ROC curves were performed and generated with R. Ingenuity® pathway analysis (IPA®) was used to perform functional enrichment analyses and identify new targets or candidate biomarkers within the context of biological systems. It provided the canonical pathways, molecular/cellular functions, and networks that were statistically overrepresented in the gene signatures.
Ethics and community involvement
The study was conducted in accordance with the Declaration of Helsinki and the International Conference on Harmonization Good Clinical Practice guidelines and approved by the relevant regulatory and independent ethics committees. Each participant provided written informed consent before study entry. The study was registered and approved by the Peru regulatory authorities (IMP ACTA IRB 0037-2014-CE; Peru NIH 396-2014-OG-OGITT- OPE/INS).
Sequence and data availability
The normalized microarray data that support the finding of this study have been deposited in ArrayExpress with the accession code E-MT AB-9642.
Result
Study of host microbiota before vaccination and relation to post-vaccination humoral responses
The study included five men and five women (18-45 years old) vaccinated by the intramuscular route to assess the safety and immunogenicity of MVA-HIV clade B (MVA-B), results reported elsewhere (Sanchez & Goncalves, Frontiers in Immunology in press). Exploratory analysis of whole blood samples at two distinct time-points before vaccination (w-2 and wO) studied the gene expression profile and the skin and stool samples for microbiome analysis (wO) at baseline. As expected, the microbial composition differed between the skin and stool samples. In addition, the stool samples showed dissimilarities between men and women, but this comparison did not reach statistically significant differences (P < 0.097). The predominant microbial families relatively abundant in skin samples were Moraxellaceae, Staphylococcaceae and Pseudomonadaceae, whereas Ruminococcaceae, Lachnospiraceae, Prevotellaceae, and Bacteroidaceae were predominant in stool samples. The 16S RNA sequencing generated several metrics: richness (sobs: number of observed OTUs; chao: Chaol richness estimate; ace: Abundance-based coverage estimation) and diversity (Shannon: Shannon diversity index; sd invsimpson: inverse Simpson diversity index). The amplitude of the humoral response was defined by the MVA-specific IgG neutralizing antibodies measured in serum at w8 post-vaccination. We observed no correlation between the MVA-Nab response and the baseline indexes of diversity and richness in either skin or stool (data not shown). We did however find significant positive correlations between the abundance of both skin Prevotella (r = 0.76, P = 0.0159) (data not shown) and fecal Eubacterium (r = 0.68, P = 0.0351) (data not shown) at baseline with MVA-Nab response.
Whole blood gene expression and host microbiota before vaccination are associated with post-vaccination humoral responses
To improve our understanding of host molecular mechanisms potentially associated with skin and gut microbiota that may be involved in vaccine immunogenicity, we counted the number of genes at baselines that were correlated with the MVA-Nab response at w8. We confirmed that gene expression of the baseline samples did not differ between w-2 and wO using hierarchal clustering analysis (data not shown). Out of all samples, we found 154 significant genes correlated with the MVA-Nab response (P < 0.05) (Figure 1A). However, no correlation was observed between genus diversity and MVA-Nab response.
Next, we looked for a correlation between the microbiota diversity index and the genes (n=154) correlated at baseline with MVA-Nab responses. We found 22 genes for skin and 19 for stool that were correlated with at least one diversity index (Shannon or sd invsimpson) (Figure 1A and Table 1), including 10 common genes to the skin and stool samples. Among these genes, we observed one gene cluster positively correlated with MVA-Nab response and another negatively correlated with humoral response (data not shown). According to the IPA analysis, the negatively correlated genes appear to be involved in protein transmembrane transport, translation and transcription regulation, cell division, migration, proliferation, and differentiation, as well as in the oxidation reduction and metabolic processes. The positively correlated genes, on the other hand, appeared involved in cell homeostasis and migration, cell growth, proliferation, regulation of gene expression, the apoptotic process, exocytosis, and intracellular signal transduction. Interestingly, among the 10 common genes to the skin and stool samples we found the IGLV8-61, BLK, and EBF1 genes which are involved in antigen recognition, B cell development, proliferation, and differentiation, and in the positive regulation of transcription in B cell and B cell receptor signaling (data not shown). Surprisingly these three significant genes involved in B cell development stages were negatively correlated with the baseline abundance of Prevotella and Eubacterium, respectively for skin and stool (Figure IB). To assess the predictive power of this signature of three genes and each of the two microbial genera, we ran logistic regression models (Figure 1C). Use of the expression of the three genes and Prevotella abundance in the skin microbiota has an 85.42% chance, assessed by its area under the curve, of correctly predicting MVA-Nab responders, while with the three-gene signature and Eubacterium abundance in the stool microbiota there is an 89.58% chance of correctly predicting MVA-Nab responders (Figure 1C). These results suggest that advanced B lymphocyte differentiation before vaccination, potentially signaled by high expression of these three genes, and associated with low abundance of Prevotella or Eubacterium, is associated with poor MVA-Nab response. DISCUSSION
To our knowledge, this work is the first to investigate the crosstalk between pre- vaccination host gene expression in blood cells, skin and stool microbiota and their association with the intensity of ensuing post-vaccination Nab responses. The data may provide important guidance for future design and refinement of vaccine strategies aiming at the induction of neutralizing antibody-mediated immunity. The limitation of this study is the small number of individuals included. However, the strength of our work is the availability of two sets of gene expression data collected at baseline (w-2, wO) that is often absent in other studies. It is intriguing to discover three genes, all involved in B cell differentiation and proliferation correlated with humoral responses 2 months later. Further validation studies are necessary in the future.
First, we observed that the abundance of particular skin or stool bacteria were associated with the MVA-Nab response. Abundant Prevotella in the skin at baseline was positively correlated with MVA-Nab response. Prevotella is known to promote mucosal inflammation and to stimulate production of epithelial cell cytokines (Larsen, 2017). Prevotella is also found in larger numbers in the skin of women aged 60-76 years than in that of women in their 20s and 30s and was enriched in all of the skin sites of the older group compared to the younger ones (Shibagaki et al., 2017). In stool, we found that Eubacterium abundance at baseline was positively correlated with the MVA-Nab response. This family of bacteria is known to be associated with gut health (King et al., 2018; Le Bastard et al., 2018; Rinninella et al., 2019), and several of its species are higher in centenarians than in either young or elderly adults (Biagi et al., 2010). The potential impact of the gut microbiota on vaccine immunogenicity has been already investigated with systemic vaccines (Huda et al., 2014) and with oral vaccines including those of rotavirus (RVV), polio, and cholera, mainly in infants/children living in low-income countries (Levine, 2010; Magwira and Taylor, 2018). For example, bacteria related to Streptococcus bovis species were more abundant before vaccination in Ghanaian vaccine-responders than non-responders and were positively associated with RVV efficacy, whereas Bacteroides and Prevotella species were more common in the microbiome of nonresponders and correlated with a lack of RVV response (Harris et al., 2017). In Bangladeshi infants, the pre-vaccination presence of Bifidobacterium was positively associated with some adaptive immunological responses, such as CD4+ and CD8+ T-cell proliferative responses to BCG and tetanus toxoid vaccinations as well as specific IgG responses to tetanus toxoid and hepatitis B vaccines, whereas high levels of enteric pathogens such as Enterobacteriales and Pseudomonadales were associated with neutrophilia and poorer vaccine responses (Huda et al., 2014).
Secondly, we examined the pre-vaccination host blood genes that were correlated with MVA- Nab intensity. We then investigated microbiota abundance to decipher a minimal gene signature predictive of MVA-Nab responsiveness. Interestingly, within this signature we find BLK, IGLV8-61 and EBF1 involved in B cell development, proliferation and differentiation and in the positive regulation of transcription in B cells and B cell receptor signaling. The BLK gene belongs to the family of protein tyrosine kinases src, and the B cells activation induces BLK gene product phosphorylation playing a key role in transmitting signals through surface immunoglobulins which supports the pro-B to pre-B transition and the signaling for growth arrest and apoptosis downstream of B-cell receptor (Burkhardt et al., 1991). BLK also plays a role in the development, differentiation, and activation of B cells and in the intracellular signaling pathway. BLK is detected in pro-B cells and persists in mature B cells but is absent in plasma cells. Triple protein tyrosine kinase (SFK)-deficient mice — BLK, LYN, and FYN — have impaired NFkB signaling and B cell development (Saijo et al., 2003). EBF1, an early B cell factor 1, is one of the transcription factors essential for orchestrating the development of the B cell line. Heterozygosity of EBF1 results in the deregulation of at least eight transcription factors involved in lymphopoiesis and the deregulation of key proteins that play a crucial role in the survival, development, and differentiation of pro-B cells (Musa et al., 2018). IGLV8 (variable domain) is a glycoprotein produced by B lymphocytes; its binding of a specific antigen triggers the clonal expansion and differentiation of B lymphocytes into immunoglobulin-secreting plasma cells. The link between microbiota and host blood transcriptome has also been studied previously by Nakaya et al., who showed that TLR5 expression in blood 3 days after influenza vaccination was correlated with antibody response 28 days later (Nakaya et al., 2011). This correlation was significantly lower in TLR5 -deficient mice immunized with TIV compared to wild-type mice. As influenza vaccine does not stimulate TLR5 directly, however, Oh et al. demonstrated with germ-free or antibiotic- treatment that the commensal bacteria were the source of the TLR5 ligands responsible for enhancing immune response to TIV (Oh et al., 2014). It should be noted that in our study the three genes were negatively correlated with MVA-Nab response and microbial diversity of both skin and stool samples but also with the abundance of the Prevotella family in skin and the Eubacterium family in stool. The logistic regression based on the expression of these three genes and Prevotella and Eubacterium abundance for, respectively, skin and stool, highlights the predictive power of this signature for the MVA-Nab immune responses. These results propose that an advanced differentiation state of B lymphocytes before vaccination, potentially represented by a high expression of these three genes and associated with low genus abundance and diversity, might be associated with poor MVA-Nab response.
Table 1: Skin and stool genus diversity correlations with blood gene expression
Figure imgf000031_0001
Correlation coefficient and P-value for the relations between genus diversity (shannon: diversity shannon index, sd invsimpson: inverss simpson diversity index) and the genes correlated with MVA-Nab response in a. skin and b. stool. Three genes involved in B cell function and correlated in both skin and stool are highlighted. Table 2: mRNA used in the present invention:
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
REFERENCES:
Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure.
A., S., and Bittencourt a, S. (2010). FastQC: a quality control tool for high throughput sequence data.
Biagi, E., Nylund, L., Candela, M., Ostan, R., Bucci, L., Pini, E., Nikkila, J., Monti, D., Satokari, R., Franceschi, C., et al. (2010). Through Ageing, and Beyond: Gut Microbiota and Inflammatory Status in Seniors and Centenarians. PLoS ONE 5.
Bolger, A.M., Lohse, M., and Usadel, B. (2014). Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinforma. Oxf. Engl. 30, 2114-2120.
Bolstad, B.M., Irizarry, R.A., Astrand, M., and Speed, T.P. (2003). A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinforma. Oxf. Engl. 19, 185-193.
Burkhardt, A.L., Brunswick, M., Bolen, J.B., and Mond, J. J. (1991). Anti- immunoglobulin stimulation of B lymphocytes activates src-related protein-tyrosine kinases. Proc Natl Acad Sci USA 88, 7410-7414.
Cosma, A., Buhler, S., Nagaraj, R., Staib, C., Hammarin, A.-L., Wahren, B., Goebel, F.D., Erfle, V., and Sutter, G. (2004). Neutralization Assay Using a Modified Vaccinia Virus Ankara Vector Expressing the Green Fluorescent Protein Is a High-Throughput Method To Monitor the Humoral Immune Response against Vaccinia Virus. Clin. Diagn. Lab. Immunol. 11, 406-410.
Earl, P.L., Americo, J.L., and Moss, B. (2003). Development and Use of a Vaccinia Virus Neutralization Assay Based on Flow Cytometric Detection of Green Fluorescent Protein. J. Virol. 77, 10684-10688.
Ehrenberg, P.K., Shangguan, S., Issac, B., Alter, G., Geretz, A., Izumi, T., Bryant, C., Eller, M.A., Wegmann, F., Apps, R., et al. (2019). A vaccine-induced gene expression signature correlates with protection against SIV and HIV in multiple trials. Sci. Transl. Med. 11.
Furman, D., Jojic, V., Kidd, B., Shen-Orr, S., Price, J., Jarrell, J., Tse, T., Huang, H., Lund, P., Maecker, H.T., et al. (2014). Apoptosis and other immune biomarkers predict influenza vaccine responsiveness. Mol. Syst. Biol. 9, 659-659.
Garcia, F., Bemaldo de Quiros, J.C.L., Gomez, C.E., Perdiguero, B., Najera, J.L., Jimenez, V., Garcia-Arriaza, J., Guardo, A.C., Perez, I., Diaz-Brito, V., et al. (2011). Safety and immunogenicity of a modified pox vector-based HIV/AIDS vaccine candidate expressing Env, Gag, Pol and Nef proteins of HIV-1 subtype B (MVA-B) in healthy HIV- 1 -uninfected volunteers: A phase I clinical trial (RISVAC02). Vaccine 29, 8309-8316.
Gaucher, D., Therrien, R., Kettaf, N., Angermann, B.R., Boucher, G., Filali-Mouhim, A., Moser, J.M., Mehta, R.S., Drake, D.R., Castro, E., et al. (2008). Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses. J. Exp. Med. 205, 3119-3131.
Harris, V.C., Armah, G., Fuentes, S., Korpela, K.E., Parashar, U., Victor, J.C., Tate, J., de Weerth, C., Giaquinto, C., Wiersinga, W.J., et al. (2017). Significant Correlation Between the Infant Gut Microbiome and Rotavirus Vaccine Response in Rural Ghana. J. Infect. Dis. 215, 34-41.
Huber, W., von Heydebreck, A., Sultmann, H., Poustka, A., and Vingron, M. (2002). Variance stabilization applied to microarray data calibration and to the quantification of differential expression. Bioinforma. Oxf. Engl. 18 Suppl 1, S96-104.
Huda, M.N., Lewis, Z., Kalanetra, K.M., Rashid, M., Ahmad, S.M., Raqib, R., Qadri, F., Underwood, M.A., Mills, D.A., and Stephensen, C.B. (2014). Stool Microbiota and Vaccine Responses of Infants. Pediatrics 134, e362-e372.
Irizarry, R.A., Bolstad, B.M., Collin, F., Cope, L.M., Hobbs, B., and Speed, T.P. (2003). Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res. 31, el5.
King, C.H., Desai, H., Sylvetsky, A.C., LoTempio, J., Ayanyan, S., Carrie, J., Crandall, K., Fochtman, B., Gasparyan, L., Gulzar, N., et al. (2018). Baseline human gut microbiota
Larsen, J.M. (2017). The immune response to Prevotella bacteria in chronic inflammatory disease. Immunology 151, 363-374.
Le Bastard, Q., Ward, T., Sidiropoulos, D., Hillmann, B.M., Chun, C.L., Sadowsky, M.J., Knights, D., and Montassier, E. (2018). Fecal microbiota transplantation reverses antibiotic and chemotherapy-induced gut dysbiosis in mice. Sci. Rep. 8.
Lecuyer, E., Rakotobe, S., Lengline-Garnier, H., Lebreton, C., Picard, M., Juste, C., Fritzen, R., Eberl, G., McCoy, K.D., Macpherson, A.J., et al. (2014). Segmented Filamentous Bacterium Uses Secondary and Tertiary Lymphoid Tissues to Induce Gut IgA and Specific T Helper 17 Cell Responses. Immunity 40, 608-620.
Levine, M.M. (2010). Immunogenicity and efficacy of oral vaccines in developing countries: lessons from a live cholera vaccine. BMC Biol. 8, 129. Lindner, C., Wahl, B., Fdhse, L., Suerbaum, S., Macpherson, A. J., Prinz, I., and Pabst, O. (2012). Age, microbiota, and T cells shape diverse individual IgA repertoires in the intestine. J. Exp. Med. 209, 365-377.
Macpherson, A. J., Yilmaz, B., Limenitakis, J.P., and Ganal-Vonarburg, S.C. (2018). IgA Function in Relation to the Intestinal Microbiota. Annu. Rev. Immunol. 36, 359-381.
Magwira, C.A., and Taylor, M.B. (2018). Composition of gut microbiota and its influence on the immunogenicity of oral rotavirus vaccines. Vaccine 36, 3427-3433.
McDonald, D., Price, M.N., Goodrich, J., Nawrocki, E.P., DeSantis, T.Z., Probst, A., Andersen, G.L., Knight, R., and Hugenholtz, P. (2012). An improved Greengenes taxonomy with explicit ranks for ecological and evolutionary analyses of bacteria and archaea. ISME J. 6, 610-618.
Musa, Y.R., Boiler, S., Puchalska, M., Grosschedl, R., and Mittler, G. (2018). Comprehensive Proteomic Investigation of Ebfl Heterozygosity in Pro-B Lymphocytes Utilizing Data Independent Acquisition. J. Proteome Res. 17, 76-85.
Nakaya, H.I., Wrammert, J., Lee, E.K., Racioppi, L., Marie-Kunze, S., Haining, W.N., Means, A.R., Kasturi, S.P., Khan, N., Li, G.-M., et al. (2011). Systems biology of vaccination for seasonal influenza in humans. Nat. Immunol. 12, 786-795.
Oh, J.Z., Ravindran, R., Chassaing, B., Carvalho, F.A., Maddur, M.S., Bower, M., Hakimpour, P., Gill, K.P., Nakaya, H.I., Yarovinsky, F., et al. (2014). TLR5-Mediated Sensing of Gut Microbiota Is Necessary for Antibody Responses to Seasonal Influenza Vaccination. Immunity 41, 478-492.
Parvandeh, S., Poland, G.A., Kennedy, R.B., and McKinney, B.A. (2019). Multi-Level Model to Predict Antibody Response to Influenza Vaccine Using Gene Expression Interaction Network Feature Selection. Microorganisms 7.
Plotkin, S.A. (2010). Correlates of Protection Induced by Vaccination. Clin. Vaccine Immunol. 17, 1055-1065.
Querec, T.D., Akondy, R.S., Lee, E.K., Cao, W., Nakaya, H.I., Teuwen, D., Pirani, A., Gemert, K., Deng, J., Marzolf, B., et al. (2009). Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat. Immunol. 10, 116-125.
Rechtien, A., Richert, L., Lorenzo, H., Martrus, G., Hejblum, B., Dahlke, C., Kasonta, R., Zinser, M., Stubbe, H., Matschl, U., et al. (2017). Systems Vaccinology Identifies an Early Innate Immune Signature as a Correlate of Antibody Responses to the Ebola Vaccine rVSV- ZEBOV. Cell Rep. 20, 2251-2261. Rinninella, E., Raoul, P., Cintoni, M., Franceschi, F., Miggiano, G.A.D., Gasbarrini, A., and Mele, M.C. (2019). What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 7.
Saijo, K., Schmedt, C., Su, I. -hsin, Karasuyama, H., Lowell, C.A., Reth, M., Adachi, T., Patke, A., Santana, A., and Tarakhovsky, A. (2003). Essential role of Src-family protein tyrosine kinases in NF-KB activation during B cell development. Nat. Immunol. 4, 274-279.
Schloss, P.D., Westcott, S.L., Ryabin, T., Hall, J.R., Hartmann, M., Hollister, E.B., Lesniewski, R.A., Oakley, B.B., Parks, D.H., Robinson, C.J., et al. (2009). Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl. Environ. Microbiol. 75, 7537-7541.
Shibagaki, N., Suda, W., Clavaud, C., Bastien, P., Takayasu, L., lioka, E., Kurokawa, R., Yamashita, N., Hattori, Y., Shindo, C., et al. (2017). Aging-related changes in the diversity of women’s skin microbiomes associated with oral bacteria. Sci. Rep. 7, 1-10.
Team, H.-C.S.P., and Consortium, H.-I. (2017). Multicohort analysis reveals baseline transcriptional predictors of influenza vaccination responses. Sci. Immunol. 2.
Tsang, J.S. (2015). Utilizing population variation, vaccination, and systems biology to study human immunology. Trends Immunol. 36, 479-493.
Tsang, J.S., Schwartzberg, P.L., Kotliarov, Y., Biancotto, A., Xie, Z., Germain, R.N., Wang, E., Olnes, M.J., Narayanan, M., Golding, H., et al. (2014). Global analyses of human immune variation reveal baseline predictors of post-vaccination responses. Cell 157, 499-513.

Claims

CLAIMS:
1. An in vitro method for assessing a subject’s humoral immune response to a vaccine, said method, comprising the step of (a) measuring in a sample obtained from said subject prior to the administration of said vaccine, the level of one or more gene expression level selected from a group of gene consisting of: IFNal6, IGLV8-61, BLK, and EBF1 gene and b) wherein the level of one or more gene expression of step a) is negatively correlated with the humoral immune response of said subject to said vaccine.
2. The in vitro method according to claim 1 comprising the step of comparing said level of one or more gene expression to a control reference value wherein:
- a low level of one or more gene expression compared to said control reference value is predictive that said subject is a high risk to be a “high responder” to said vaccine.
3. The in vitro method according to any one of claim 1 to 2, wherein the sample is a blood sample.
4. The in vitro method according to any one of claim 1 or 3, wherein the level of one or more gene expression is determined at mRNA level.
5. The in vitro method according to any one of claim 1 or 4, wherein the level of the gene expression is determined with one, two, three, four gene selected from the group consisting of IGLV8-61, BLK, EBF1 and IFNal6 gene.
6. The in vitro method according to any one of claim 1 or 5, wherein the vaccine contain an antigen selected from the group consisting of attenuated and inactivated viral and bacterial pathogens, purified macromolecules, polysaccharides, toxoids, recombinant antigens, organisms containing a foreign gene from a pathogen, synthetic peptides, polynucleic acids (DNA, RNA), antibodies and tumor cells.
7. The in vitro method according to any one of claim 1 or 6, comprising an additional step consisting of measuring in a sample obtained from said subject the level Eubacterium in stool sample and Prevotella in skin sample.
8. The in vitro method according to claim 7, wherein the level Eubacterium in stool sample and Prevotella in skin sample is performed by using 16S ribosomal sequencing.
9. A kit, comprising:
- at least a mean for determining one or more gene expression level selected from a group of gene consisting of: IGLV8-61, BLK, EBF1 and IFNal6 gene and
- instructions for use, for use to assess a subject’s humoral immune response to a vaccine.
10. A kit for use according to claim 9, comprising:
- amplification primers and/or probe for determining one or more gene expression level selected from a group of gene consisting of: IFNal6, IGLV8-61, BLK, and EBF 1 gene,
- instructions for use.
11. Method for vaccinating a subject in need thereof with an antigen wherein, prior to vaccination, the level of one or more gene expression level selected from a group of gene consisting of: IFNal6IGLV8-61, BLK and EBF1 gene obtained from said subject, have been detected by one of the method of claim 1 to 8.
PCT/EP2021/070524 2020-12-21 2021-07-22 Methods for prognosis the humoral response of a subject prior to vaccination WO2022135753A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP20306634.5 2020-12-21
EP20306634 2020-12-21

Publications (1)

Publication Number Publication Date
WO2022135753A1 true WO2022135753A1 (en) 2022-06-30

Family

ID=74184385

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2021/070524 WO2022135753A1 (en) 2020-12-21 2021-07-22 Methods for prognosis the humoral response of a subject prior to vaccination

Country Status (1)

Country Link
WO (1) WO2022135753A1 (en)

Citations (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4774339A (en) 1987-08-10 1988-09-27 Molecular Probes, Inc. Chemically reactive dipyrrometheneboron difluoride dyes
US4888278A (en) 1985-10-22 1989-12-19 University Of Massachusetts Medical Center In-situ hybridization to detect nucleic acid sequences in morphologically intact cells
US5132432A (en) 1989-09-22 1992-07-21 Molecular Probes, Inc. Chemically reactive pyrenyloxy sulfonic acid dyes
US5187288A (en) 1991-05-22 1993-02-16 Molecular Probes, Inc. Ethenyl-substituted dipyrrometheneboron difluoride dyes and their synthesis
US5248782A (en) 1990-12-18 1993-09-28 Molecular Probes, Inc. Long wavelength heteroaryl-substituted dipyrrometheneboron difluoride dyes
US5262357A (en) 1991-11-22 1993-11-16 The Regents Of The University Of California Low temperature thin films formed from nanocrystal precursors
US5274113A (en) 1991-11-01 1993-12-28 Molecular Probes, Inc. Long wavelength chemically reactive dipyrrometheneboron difluoride dyes and conjugates
US5338854A (en) 1991-02-13 1994-08-16 Molecular Probes, Inc. Fluorescent fatty acids derived from dipyrrometheneboron difluoride dyes
US5427932A (en) 1991-04-09 1995-06-27 Reagents Of The University Of California Repeat sequence chromosome specific nucleic acid probes and methods of preparing and using
US5433896A (en) 1994-05-20 1995-07-18 Molecular Probes, Inc. Dibenzopyrrometheneboron difluoride dyes
US5447841A (en) 1986-01-16 1995-09-05 The Regents Of The Univ. Of California Methods for chromosome-specific staining
US5472842A (en) 1993-10-06 1995-12-05 The Regents Of The University Of California Detection of amplified or deleted chromosomal regions
US5505928A (en) 1991-11-22 1996-04-09 The Regents Of University Of California Preparation of III-V semiconductor nanocrystals
US5571018A (en) 1994-11-23 1996-11-05 Motorola, Inc. Arrangement for simulating indirect fire in combat training
US5690807A (en) 1995-08-03 1997-11-25 Massachusetts Institute Of Technology Method for producing semiconductor particles
US5696157A (en) 1996-11-15 1997-12-09 Molecular Probes, Inc. Sulfonated derivatives of 7-aminocoumarin
US5800996A (en) 1996-05-03 1998-09-01 The Perkin Elmer Corporation Energy transfer dyes with enchanced fluorescence
US5830912A (en) 1996-11-15 1998-11-03 Molecular Probes, Inc. Derivatives of 6,8-difluoro-7-hydroxycoumarin
US5866366A (en) 1997-07-01 1999-02-02 Smithkline Beecham Corporation gidB
WO1999026299A1 (en) 1997-11-13 1999-05-27 Massachusetts Institute Of Technology Highly luminescent color-selective materials
US5990479A (en) 1997-11-25 1999-11-23 Regents Of The University Of California Organo Luminescent semiconductor nanocrystal probes for biological applications and process for making and using such probes
US6048616A (en) 1993-04-21 2000-04-11 Philips Electronics N.A. Corp. Encapsulated quantum sized doped semiconductor particles and method of manufacturing same
US6114038A (en) 1998-11-10 2000-09-05 Biocrystal Ltd. Functionalized nanocrystals and their use in detection systems
US6130101A (en) 1997-09-23 2000-10-10 Molecular Probes, Inc. Sulfonated xanthene derivatives
US6207392B1 (en) 1997-11-25 2001-03-27 The Regents Of The University Of California Semiconductor nanocrystal probes for biological applications and process for making and using such probes
US6225198B1 (en) 2000-02-04 2001-05-01 The Regents Of The University Of California Process for forming shaped group II-VI semiconductor nanocrystals, and product formed using process
US6274323B1 (en) 1999-05-07 2001-08-14 Quantum Dot Corporation Method of detecting an analyte in a sample using semiconductor nanocrystals as a detectable label
US6280929B1 (en) 1986-01-16 2001-08-28 The Regents Of The University Of California Method of detecting genetic translocations identified with chromosomal abnormalities
US6306736B1 (en) 2000-02-04 2001-10-23 The Regents Of The University Of California Process for forming shaped group III-V semiconductor nanocrystals, and product formed using process
US6500622B2 (en) 2000-03-22 2002-12-31 Quantum Dot Corporation Methods of using semiconductor nanocrystals in bead-based nucleic acid assays
US6602671B1 (en) 1998-09-18 2003-08-05 Massachusetts Institute Of Technology Semiconductor nanocrystals for inventory control
US6649138B2 (en) 2000-10-13 2003-11-18 Quantum Dot Corporation Surface-modified semiconductive and metallic nanoparticles having enhanced dispersibility in aqueous media
US6670113B2 (en) 2001-03-30 2003-12-30 Nanoprobes Enzymatic deposition and alteration of metals
US6682596B2 (en) 2000-12-28 2004-01-27 Quantum Dot Corporation Flow synthesis of quantum dot nanocrystals
US6689338B2 (en) 2000-06-01 2004-02-10 The Board Of Regents For Oklahoma State University Bioconjugates of nanoparticles as radiopharmaceuticals
US6709929B2 (en) 2001-06-25 2004-03-23 North Carolina State University Methods of forming nano-scale electronic and optoelectronic devices using non-photolithographically defined nano-channel templates
US6716979B2 (en) 2000-08-04 2004-04-06 Molecular Probes, Inc. Derivatives of 1,2-dihydro-7-hydroxyquinolines containing fused rings
US6815064B2 (en) 2001-07-20 2004-11-09 Quantum Dot Corporation Luminescent nanoparticles and methods for their preparation
US20040265922A1 (en) 2003-06-24 2004-12-30 Ventana Medical Systems, Inc. Enzyme-catalyzed metal deposition for the enhanced in situ detection of immunohistochemical epitopes and nucleic acid sequences
US6855202B2 (en) 2001-11-30 2005-02-15 The Regents Of The University Of California Shaped nanocrystal particles and methods for making the same
US20050100976A1 (en) 2003-06-24 2005-05-12 Christopher Bieniarz Enzyme-catalyzed metal deposition for the enhanced detection of analytes of interest
US6942970B2 (en) 2000-09-14 2005-09-13 Zymed Laboratories, Inc. Identifying subjects suitable for topoisomerase II inhibitor treatment
US20060246524A1 (en) 2005-04-28 2006-11-02 Christina Bauer Nanoparticle conjugates
US20060246523A1 (en) 2005-04-28 2006-11-02 Christopher Bieniarz Antibody conjugates
US20070117153A1 (en) 2005-11-23 2007-05-24 Christopher Bieniarz Molecular conjugate

Patent Citations (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4888278A (en) 1985-10-22 1989-12-19 University Of Massachusetts Medical Center In-situ hybridization to detect nucleic acid sequences in morphologically intact cells
US6280929B1 (en) 1986-01-16 2001-08-28 The Regents Of The University Of California Method of detecting genetic translocations identified with chromosomal abnormalities
US5447841A (en) 1986-01-16 1995-09-05 The Regents Of The Univ. Of California Methods for chromosome-specific staining
US4774339A (en) 1987-08-10 1988-09-27 Molecular Probes, Inc. Chemically reactive dipyrrometheneboron difluoride dyes
US5132432A (en) 1989-09-22 1992-07-21 Molecular Probes, Inc. Chemically reactive pyrenyloxy sulfonic acid dyes
US5248782A (en) 1990-12-18 1993-09-28 Molecular Probes, Inc. Long wavelength heteroaryl-substituted dipyrrometheneboron difluoride dyes
US5338854A (en) 1991-02-13 1994-08-16 Molecular Probes, Inc. Fluorescent fatty acids derived from dipyrrometheneboron difluoride dyes
US5427932A (en) 1991-04-09 1995-06-27 Reagents Of The University Of California Repeat sequence chromosome specific nucleic acid probes and methods of preparing and using
US5187288A (en) 1991-05-22 1993-02-16 Molecular Probes, Inc. Ethenyl-substituted dipyrrometheneboron difluoride dyes and their synthesis
US5274113A (en) 1991-11-01 1993-12-28 Molecular Probes, Inc. Long wavelength chemically reactive dipyrrometheneboron difluoride dyes and conjugates
US5451663A (en) 1991-11-01 1995-09-19 Molecular Probes, Inc. Long wavelength chemically reactive dipyrrometheneboron difluoride dyes and conjugates
US5262357A (en) 1991-11-22 1993-11-16 The Regents Of The University Of California Low temperature thin films formed from nanocrystal precursors
US5505928A (en) 1991-11-22 1996-04-09 The Regents Of University Of California Preparation of III-V semiconductor nanocrystals
US6048616A (en) 1993-04-21 2000-04-11 Philips Electronics N.A. Corp. Encapsulated quantum sized doped semiconductor particles and method of manufacturing same
US5472842A (en) 1993-10-06 1995-12-05 The Regents Of The University Of California Detection of amplified or deleted chromosomal regions
US5433896A (en) 1994-05-20 1995-07-18 Molecular Probes, Inc. Dibenzopyrrometheneboron difluoride dyes
US5571018A (en) 1994-11-23 1996-11-05 Motorola, Inc. Arrangement for simulating indirect fire in combat training
US5690807A (en) 1995-08-03 1997-11-25 Massachusetts Institute Of Technology Method for producing semiconductor particles
US5800996A (en) 1996-05-03 1998-09-01 The Perkin Elmer Corporation Energy transfer dyes with enchanced fluorescence
US5696157A (en) 1996-11-15 1997-12-09 Molecular Probes, Inc. Sulfonated derivatives of 7-aminocoumarin
US5830912A (en) 1996-11-15 1998-11-03 Molecular Probes, Inc. Derivatives of 6,8-difluoro-7-hydroxycoumarin
US5866366A (en) 1997-07-01 1999-02-02 Smithkline Beecham Corporation gidB
US6130101A (en) 1997-09-23 2000-10-10 Molecular Probes, Inc. Sulfonated xanthene derivatives
WO1999026299A1 (en) 1997-11-13 1999-05-27 Massachusetts Institute Of Technology Highly luminescent color-selective materials
US6207392B1 (en) 1997-11-25 2001-03-27 The Regents Of The University Of California Semiconductor nanocrystal probes for biological applications and process for making and using such probes
US5990479A (en) 1997-11-25 1999-11-23 Regents Of The University Of California Organo Luminescent semiconductor nanocrystal probes for biological applications and process for making and using such probes
US6927069B2 (en) 1997-11-25 2005-08-09 The Regents Of The University Of California Organo luminescent semiconductor nanocrystal probes for biological applications and process for making and using such probes
US6602671B1 (en) 1998-09-18 2003-08-05 Massachusetts Institute Of Technology Semiconductor nanocrystals for inventory control
US6114038A (en) 1998-11-10 2000-09-05 Biocrystal Ltd. Functionalized nanocrystals and their use in detection systems
US6274323B1 (en) 1999-05-07 2001-08-14 Quantum Dot Corporation Method of detecting an analyte in a sample using semiconductor nanocrystals as a detectable label
US6306736B1 (en) 2000-02-04 2001-10-23 The Regents Of The University Of California Process for forming shaped group III-V semiconductor nanocrystals, and product formed using process
US6225198B1 (en) 2000-02-04 2001-05-01 The Regents Of The University Of California Process for forming shaped group II-VI semiconductor nanocrystals, and product formed using process
US6500622B2 (en) 2000-03-22 2002-12-31 Quantum Dot Corporation Methods of using semiconductor nanocrystals in bead-based nucleic acid assays
US20030165951A1 (en) 2000-03-22 2003-09-04 Quantum Dot Corporation Methods of using semiconductor nanocrystals in bead-based nucleic acid assays
US6689338B2 (en) 2000-06-01 2004-02-10 The Board Of Regents For Oklahoma State University Bioconjugates of nanoparticles as radiopharmaceuticals
US6716979B2 (en) 2000-08-04 2004-04-06 Molecular Probes, Inc. Derivatives of 1,2-dihydro-7-hydroxyquinolines containing fused rings
US6942970B2 (en) 2000-09-14 2005-09-13 Zymed Laboratories, Inc. Identifying subjects suitable for topoisomerase II inhibitor treatment
US6649138B2 (en) 2000-10-13 2003-11-18 Quantum Dot Corporation Surface-modified semiconductive and metallic nanoparticles having enhanced dispersibility in aqueous media
US6682596B2 (en) 2000-12-28 2004-01-27 Quantum Dot Corporation Flow synthesis of quantum dot nanocrystals
US6670113B2 (en) 2001-03-30 2003-12-30 Nanoprobes Enzymatic deposition and alteration of metals
US6709929B2 (en) 2001-06-25 2004-03-23 North Carolina State University Methods of forming nano-scale electronic and optoelectronic devices using non-photolithographically defined nano-channel templates
US6914256B2 (en) 2001-06-25 2005-07-05 North Carolina State University Optoelectronic devices having arrays of quantum-dot compound semiconductor superlattices therein
US6815064B2 (en) 2001-07-20 2004-11-09 Quantum Dot Corporation Luminescent nanoparticles and methods for their preparation
US6855202B2 (en) 2001-11-30 2005-02-15 The Regents Of The University Of California Shaped nanocrystal particles and methods for making the same
WO2005003777A2 (en) 2003-06-24 2005-01-13 Ventana Medical Systems, Inc. Enzyme-catalyzed metal deposition for the enhanced in situ detection of immunohistochemical epitopes and nucleic acid sequences
US20050100976A1 (en) 2003-06-24 2005-05-12 Christopher Bieniarz Enzyme-catalyzed metal deposition for the enhanced detection of analytes of interest
US20040265922A1 (en) 2003-06-24 2004-12-30 Ventana Medical Systems, Inc. Enzyme-catalyzed metal deposition for the enhanced in situ detection of immunohistochemical epitopes and nucleic acid sequences
US20060246524A1 (en) 2005-04-28 2006-11-02 Christina Bauer Nanoparticle conjugates
US20060246523A1 (en) 2005-04-28 2006-11-02 Christopher Bieniarz Antibody conjugates
US20070117153A1 (en) 2005-11-23 2007-05-24 Christopher Bieniarz Molecular conjugate

Non-Patent Citations (57)

* Cited by examiner, † Cited by third party
Title
"NCBI", Database accession no. NP _001351088
BIAGI, E.NYLUND, L.CANDELA, M.OSTAN, R.BUCCI, L.PINI, E.NIKKILA, J.MONTI, D.SATOKARI, R.FRANCESCHI, C. ET AL.: "Through Ageing, and Beyond: Gut Microbiota and Inflammatory Status in Seniors and Centenarians", PLOS ONE, vol. 5, 2010
BITTENCOURT A, S., FASTQC: A QUALITY CONTROL TOOL FOR HIGH THROUGHPUT SEQUENCE DATA, 2010
BOLGER, A.M.LOHSE, M.USADEL, B.: "Trimmomatic: a flexible trimmer for Illumina sequence data", BIOINFORMA. OXF ENGL., vol. 30, 2014, pages 2114 - 2120
BOLSTAD, B.M.IRIZARRY, R.A.ASTRAND, M.SPEED, T.P.: "A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinforma", OXF. ENGL., vol. 19, 2003, pages 185 - 193, XP008041261, DOI: 10.1093/bioinformatics/19.2.185
BRUCHEZ ET AL., SCIENCE, vol. 281, 1998, pages 20132016 - 2018
BURKHARDT, A.L.BRUNSWICK, M.BOLEN, J.B.MOND, J.J.: "Anti-immunoglobulin stimulation of B lymphocytes activates src-related protein-tyrosine kinases", PROC NATL ACAD SCI USA, vol. 88, 1991, pages 7410 - 7414
COSMA, A.BUHLER, S.NAGARAJ, R.STAIB, C.HAMMARIN, A.-L.WAHREN, B.GOEBEL, F.D.ERFLE, V.SUTTER, G.: "Neutralization Assay Using a Modified Vaccinia Virus Ankara Vector Expressing the Green Fluorescent Protein Is a High-Throughput Method To Monitor the Humoral Immune Response against Vaccinia Virus", CLIN. DIAGN. LAB. IMMUNOL., vol. 11, 2004, pages 406 - 410, XP009048927, DOI: 10.1128/CDLI.11.2.406-410.2004
EARL, P.L.AMERICO, J.L.MOSS, B.: "Development and Use of a Vaccinia Virus Neutralization Assay Based on Flow Cytometric Detection of Green Fluorescent Protein", J. VIROL., vol. 77, 2003, pages 10684 - 10688, XP002410211, DOI: 10.1128/JVI.77.19.10684-10688.2003
EHRENBERG, P.K.SHANGGUAN, S.ISSAC, B.ALTER, G.GERETZ, A.IZUMI, T.BRYANT, C.ELLER, M.A.WEGMANN, F.APPS, R. ET AL.: "A vaccine-induced gene expression signature correlates with protection against SIV and HIV in multiple trials", SCI. TRANSL. MED., vol. 11, 2019
EVELYN KP RICCIO ET AL: "Molecular and immunological tools for the evaluation of the cellular immune response in the neotropical monkey Saimiri sciureus, a non-human primate model for malaria research", MALARIA JOURNAL, BIOMED CENTRAL, LONDON, GB, vol. 14, no. 1, 18 April 2015 (2015-04-18), pages 166, XP021221374, ISSN: 1475-2875, DOI: 10.1186/S12936-015-0688-1 *
FURMAN, D.JOJIC, V.KIDD, B.SHEN-ORR, S.PRICE, J.JARRELL, J.TSE, T.HUANG, H.LUND, P.MAECKER, H.T. ET AL.: "Apoptosis and other immune biomarkers predict influenza vaccine responsiveness", MOL. SYST. BIOL., vol. 9, 2014, pages 659 - 659
GARCIA, F.BERNALDO DE QUIROS, J.C.L.GOMEZ, C.E.PERDIGUERO, B.NAJERA, J.L.JIMENEZ, V.GARCIA-ARRIAZA, J.GUARDO, A.C.PEREZ, I.DIAZ-BR: "Safety and immunogenicity of a modified pox vector-based HIV/AIDS vaccine candidate expressing Env, Gag, Pol and Nef proteins of HIV-1 subtype B (MVA-B) in healthy HIV-1-uninfected volunteers: A phase I clinical trial (RISVAC02", VACCINE, vol. 29, 2011, pages 8309 - 8316, XP028314378, DOI: 10.1016/j.vaccine.2011.08.098
GAUCHER, D.THERRIEN, R.KETTAF, N.ANGERMANN, B.R.BOUCHER, G.FILALI-MOUHIM, A.MOSER, J.M.MEHTA, R.S.DRAKE, D.R.CASTRO, E. ET AL.: "Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses", J. EXP. MED., vol. 205, 2008, pages 3119 - 3131
GONÇALVES ELENA ET AL: "B-Cell Gene Expression and Microbiota Prior Immunization Prole Vaccine Humoral Responsiveness", RESEARCH SQUARE, 23 July 2020 (2020-07-23), pages 1 - 18, XP055804751, Retrieved from the Internet <URL:https://assets.researchsquare.com/files/rs-46833/v1/90b40a1a-2565-42f8-8bd3-56b7a2a3f630.pdf> [retrieved on 20210517], DOI: 10.21203/rs.3.rs-46833/v1 *
HAGMAN J. ET AL., CURRENT TOPICS IN MICROBIOLOGY AND IMMUNOLOGY, vol. 356, 2012, pages 17 - 38
HARRIS, V.C.ARMAH, G.FUENTES, S.KORPELA, K.E.PARASHAR, U.VICTOR, J.C.TATE, J.DE WEERTH, C.GIAQUINTO, C.WIERSINGA, W.J. ET AL.: "Significant Correlation Between the Infant Gut Microbiome and Rotavirus Vaccine Response in Rural Ghana", J. INFECT. DIS., vol. 215, 2017, pages 34 - 41
HEYDUKHEYDUK, ANALYT. BIOCHEM., vol. 248, 1997, pages 216 - 27
HOHEISEL, NATURE REVIEWS, GENETICS, vol. 7, 2006, pages 200 - 210
HUANG YUNDA ET AL: "Predictors of durable immune responses six months after the last vaccination in preventive HIV vaccine trials", VACCINE, ELSEVIER, AMSTERDAM, NL, vol. 35, no. 8, 25 January 2017 (2017-01-25), pages 1184 - 1193, XP029913040, ISSN: 0264-410X, DOI: 10.1016/J.VACCINE.2016.09.053 *
HUBER, W.VON HEYDEBRECK, A.SULTMANN, H.POUSTKA, A.VINGRON, M.: "Variance stabilization applied to microarray data calibration and to the quantification of differential expression. Bioinforma", OXF. ENGL., vol. 18, no. 1, 2002, pages 96 - 104
HUDA, M.N.LEWIS, Z.KALANETRA, K.M.RASHID, M.AHMAD, S.M.RAQIB, R.QADRI, F.UNDERWOOD, M.A.MILLS, D.A.STEPHENSEN, C.B.: "Stool Microbiota and Vaccine Responses of Infants", PEDIATRICS, vol. 134, 2014, pages e362 - e372
IRIZARRY, R.A., BOLSTAD, B.M., COLLIN, F., COPE, L.M., HOBBS, B., SPEED, T.P: "Summaries of Affymetrix GeneChip probe level data", NUCLEIC ACIDS RES., vol. 31, 2003, pages e15, XP002460628, DOI: 10.1093/nar/gng015
J. BIOL. CHEM., vol. 274, 1999, pages 3315 - 22
KING, C.H.DESAI, H.SYLVETSKY, A.C.LOTEMPIO, J.AYANYAN, S.CARRIE, J.CRANDALL, K.FOCHTMAN, B.GASPARYAN, L.GULZAR, N. ET AL.: "Baseline human gut microbiota Larsen, J.M. (2017). The immune response to Prevotella bacteria in chronic inflammatory disease", IMMUNOLOGY, vol. 151, 2018, pages 363 - 374
LE BASTARD, Q.WARD, T.SIDIROPOULOS, D.HILLMANN, B.M.CHUN, C.L.SADOWSKY, M.J.KNIGHTS, D.MONTASSIER, E.: "Fecal microbiota transplantation reverses antibiotic and chemotherapy-induced gut dysbiosis in mice", SCI. REP., vol. 8, 2018
LECUYER, E.RAKOTOBE, S.LENGLINE-GARNIER, H.LEBRETON, C.PICARD, M.JUSTE, C.FRITZEN, R.EBERL, G.MCCOY, K.D.MACPHERSON, A.J. ET AL.: "Segmented Filamentous Bacterium Uses Secondary and Tertiary Lymphoid Tissues to Induce Gut IgA and Specific T Helper 17 Cell Responses", IMMUNITY, vol. 40, 2014, pages 608 - 620
LEVINE, M.M.: "Immunogenicity and efficacy of oral vaccines in developing countries: lessons from a live cholera vaccine", BMC BIOL., vol. 8, 2010, pages 129, XP021079357, DOI: 10.1186/1741-7007-8-129
LICHTER ET AL., PROC. NATL. ACAD. SCI., vol. 85, 1988, pages 9664 - 9668
LINDNER, C.WAHL, B.FOHSE, L.SUERBAUM, S.MACPHERSON, A.J.PRINZ, I.PABST, O.: "Age, microbiota, and T cells shape diverse individual IgA repertoires in the intestine", J. EXP. MED., vol. 209, 2012, pages 365 - 377
MACPHERSON, A.J.YILMAZ, B.LIMENITAKIS, J.P.GANAL-VONARBURG, S.C.: "IgA Function in Relation to the Intestinal Microbiota", ANNU. REV. IMMUNOL., vol. 36, 2018, pages 359 - 381
MAGWIRA, C.A.TAYLOR, M.B.: "Composition of gut microbiota and its influence on the immunogenicity of oral rotavirus vaccines", VACCINE, vol. 36, 2018, pages 3427 - 3433
MCDONALD, D.PRICE, M.N.GOODRICH, J.NAWROCKI, E.P.DESANTIS, T.Z.PROBST, A.ANDERSEN, G.L.KNIGHT, R.HUGENHOLTZ, P.: "An improved Greengenes taxonomy with explicit ranks for ecological and evolutionary analyses of bacteria and archaea", ISME J., vol. 6, 2012, pages 610 - 618
MUSA, Y.R.BOILER, S.PUCHALSKA, M.GROSSCHEDL, R.MITTLER, G.: "Comprehensive Proteomic Investigation of Ebfl Heterozygosity in Pro-B Lymphocytes Utilizing Data Independent Acquisition", J. PROTEOME RES., vol. 17, 2018, pages 76 - 85
NAKAYA, H.I.WRAMMERT, J.LEE, E.K.RACIOPPI, L.MARIE-KUNZE, S.HAINING, W.N.MEANS, A.R.KASTURI, S.P.KHAN, N.LI, G.-M. ET AL.: "Systems biology of vaccination for seasonal influenza in humans", NAT. IMMUNOL., vol. 12, 2011, pages 786 - 795, XP037135341, DOI: 10.1038/ni.2067
OH, J.Z.RAVINDRAN, R.CHASSAING, B.CARVALHO, F.A.MADDUR, M.S.BOWER, M.HAKIMPOUR, P.GILL, K.P.NAKAYA, H.I.YAROVINSKY, F. ET AL.: "TLR5-Mediated Sensing of Gut Microbiota Is Necessary for Antibody Responses to Seasonal Influenza Vaccination", IMMUNITY, vol. 41, 2014, pages 478 - 492
PARVANDEH, S.POLAND, G.A.KENNEDY, R.B.MCKINNEY, B.A.: "Multi-Level Model to Predict Antibody Response to Influenza Vaccine Using Gene Expression Interaction Network Feature Selection", MICROORGANISMS, vol. 7, 2019
PAVOT V ET AL.: "Methods in Molecular Biology", vol. 1581, 2017, SPRINGER, article "Recombinant Virus Vaccines", pages: 97 - 119
PIRLKEL ET AL., PROC. NATL. ACAD. SCI., vol. 83, 1986, pages 2934 - 2938
PLOTKIN, S.A.: "Correlates of Protection Induced by Vaccination", CLIN. VACCINE IMMUNOL., vol. 17, 2010, pages 1055 - 1065
QUEREC, T.D.AKONDY, R.S.LEE, E.K.CAO, W.NAKAYA, H.I.TEUWEN, D.PIRANI, A.GERNERT, K.DENG, J.MARZOLF, B. ET AL.: "Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans", NAT. IMMUNOL., vol. 10, 2009, pages 116 - 125, XP055183828, DOI: 10.1038/ni.1688
RECHTIEN, A.RICHERT, L.LORENZO, H.MARTRUS, G.HEJBLUM, B.DAHLKE, C.KASONTA, R.ZINSER, M.STUBBE, H.MATSCHL, U. ET AL.: "Systems Vaccinology Identifies an Early Innate Immune Signature as a Correlate of Antibody Responses to the Ebola Vaccine rVSV-ZEBOV", CELL REP., vol. 20, 2017, pages 2251 - 2261
RINNINELLA, E.RAOUL, P.CINTONI, M.FRANCESCHI, F.MIGGIANO, G.A.D.GASBARRINI, A.MELE, M.C.: "What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases", MICROORGANISMS, vol. 7, 2019
SAIJO, K.SCHMEDT, C.SU, I. -HSINKARASUYAMA, H.LOWELL, C.A.RETH, M.ADACHI, T.PATKE, A.SANTANA, A.TARAKHOVSKY, A.: "Essential role of Src-family protein tyrosine kinases in NF- B activation during B cell development", NAT. IMMUNOL., vol. 4, 2003, pages 274 - 279
SAMBROOK ET AL.: "Molecular cloning: A Laboratory Manual", 1992, COLD SPRING HARBOR LABORATORY
SCHLOSS, P.D.WESTCOTT, S.L.RYABIN, T.HALL, J.R.HARTMANN, M.HOLLISTER, E.B.LESNIEWSKI, R.A.OAKLEY, B.B.PARKS, D.H.ROBINSON, C.J. ET: "Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities", APPL. ENVIRON. MICROBIOL., vol. 75, 2009, pages 7537 - 7541, XP055154024, DOI: 10.1128/AEM.01541-09
SCHWARTZ, A. ET AL., BIOCHIMICA ET BIOPHYSICA ACTA (BBA) - GENE REGULATORY MECHANISMS, vol. 1859, no. 10, 2016, pages 1259 - 1268
SHIBAGAKI, N.SUDA, W.CLAVAUD, C.BASTIEN, P.TAKAYASU, L.IIOKA, E.KUROKAWA, R.YAMASHITA, N.HATTORI, Y.SHINDO, C. ET AL.: "Aging-related changes in the diversity of women's skin microbiomes associated with oral bacteria", SCI. REP., vol. 7, 2017, pages 1 - 10, XP055554307, DOI: 10.1038/s41598-017-10834-9
SHUZHAO LI ET AL: "Molecular signatures of antibody responses derived from a systems biology study of five human vaccines", NATURE IMMUNOLOGY, vol. 15, no. 2, 15 December 2013 (2013-12-15), pages 195 - 204, XP055199113, ISSN: 1529-2908, DOI: 10.1038/ni.2789 *
TANNER ET AL., AM..1. PATHOL., vol. 157, 2000, pages 1467 - 1472
TEAM, H.-C.S.P.CONSORTIUM, H.-I.: "Multicohort analysis reveals baseline transcriptional predictors of influenza vaccination responses", SCI. IMMUNOL., vol. 2, 2017, XP055527721, DOI: 10.1126/sciimmunol.aal4656
TREIBER, T. ET AL., IMMUNITY, vol. 32, no. 5, 2010, pages 714 - 725
TSANG, J.S.: "Utilizing population variation, vaccination, and systems biology to study human immunology", TRENDS IMMUNOL., vol. 36, 2015, pages 479 - 493
TSANG, J.S.SCHWARTZBERG, P.L.KOTLIAROV, Y.BIANCOTTO, A.XIE, Z.GERMAIN, R.N.WANG, E.OLNES, M.J.NARAYANAN, M.GOLDING, H. ET AL.: "Global analyses of human immune variation reveal baseline predictors of post-vaccination responses", CELL, vol. 157, 2014, pages 499 - 513
WACLECHE VANESSA SUE ET AL: "CD161 monocytes give rise to CD1031RALDH21TCF41 dendritic cells with unique transcriptional and immunological features", BLOOD ADVANCES, vol. 2, no. 21, 13 November 2018 (2018-11-13), pages 2862 - 2878, XP055805288 *
WIEBKE DEMASIUS ET AL: "Monitoring the immune response to vaccination with an inactivated vaccine associated to bovine neonatal pancytopenia by deep sequencing transcriptome analysis in cattle", VETERINARY RESEARCH, BIOMED CENTRAL LTD, LONDON, UK, vol. 44, no. 1, 7 October 2013 (2013-10-07), pages 93, XP021162933, ISSN: 1297-9716, DOI: 10.1186/1297-9716-44-93 *
YAN TAN ET AL: "Gene signatures related to B-cell proliferation predict influenza vaccine-induced antibody response : Clinical immunology", EUROPEAN JOURNAL OF IMMUNOLOGY, vol. 44, no. 1, 29 November 2013 (2013-11-29), Weinheim, pages 285 - 295, XP055379819, ISSN: 0014-2980, DOI: 10.1002/eji.201343657 *

Similar Documents

Publication Publication Date Title
EP3339451B1 (en) Primers for detecting influenza by using lamp, and use thereof
JP2017184642A (en) Dementia marker, evaluation method of dementia using same, evaluation reagent, and evaluation kit
US20090226895A1 (en) Method of detecting vibrio parahaemolyticus via real-time PCR-hybridization
EP4025345A1 (en) Systems, methods, and compositions for the rapid early-detection of host rna biomarkers of infection and early identification of covid-19 coronavirus infection in humans
WO2015190107A1 (en) Method for detecting vibrio parahaemolyticus, and carrier for detecting vibrio parahaemolyticus
US9617606B2 (en) Oligonucleotide for HIV detection, HIV detection kit, and HIV detection method
Gonçalves et al. Host transcriptome and microbiota signatures prior to immunization profile vaccine humoral responsiveness
KR102019804B1 (en) Primer set for detection of SFTSV and SFTSV detection method using the same
Wood et al. Variation in human herpesvirus 6B telomeric integration, excision, and transmission between tissues and individuals
GB2401175A (en) Detection of SARS virus by PCR
RU2471872C1 (en) Method for differentiating yersinia pestis strains of various subspecies and biovars by polymerase chain reaction and multilocus sequence typing
CN114729347A (en) High-sensitivity detection method for undifferentiated marker gene
JP2016019495A (en) Nucleic acid amplification technique
WO2018029452A1 (en) Flavivirus diagnostic assay
JP2006223180A (en) Method for detecting herpes virus gene by using multiplex pcr
WO2022135753A1 (en) Methods for prognosis the humoral response of a subject prior to vaccination
KR102030245B1 (en) Oligonucleotide set for detection of chikungunya virus and uses thereof
JP2017143810A (en) Biomarkers of mycobacteriosis or acid-fast bacterial infection
US20150140039A1 (en) Immunological markers for adjuvant therapy in melanoma
JP2007514440A (en) Sensitive and specific test to detect SARS coronavirus
KR102076341B1 (en) Composition for detecting severe fever with thrombocytopenia syndrome virus using LAMP and uses thereof
WO2022136252A1 (en) Methods for prognosis the humoral response of a subject prior to vaccination
TWI658048B (en) Specific primers for detection of photobacterium damselae subsp. piscicida and the method of use thereof
CA3211981A1 (en) Major histocompatibility complex single nucleotide polymorphisms
RU2737775C1 (en) Method for identifying yersinia pestis and yersinia pseudotuberculosis and simultaneous differentiation of yersinia pestis of main and central asian subspecies by multiplex pcr

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21749141

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21749141

Country of ref document: EP

Kind code of ref document: A1