WO2013174988A1 - Methods for predicting and monitoring treatment response in hcv- and hcv/hiv-infected subjects - Google Patents
Methods for predicting and monitoring treatment response in hcv- and hcv/hiv-infected subjects Download PDFInfo
- Publication number
- WO2013174988A1 WO2013174988A1 PCT/EP2013/060770 EP2013060770W WO2013174988A1 WO 2013174988 A1 WO2013174988 A1 WO 2013174988A1 EP 2013060770 W EP2013060770 W EP 2013060770W WO 2013174988 A1 WO2013174988 A1 WO 2013174988A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- hcv
- treatment
- responder
- subject
- expression level
- Prior art date
- Legal status (The legal status 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 status listed.)
- Ceased
Links
Classifications
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/569—Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
- G01N33/56983—Viruses
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/569—Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
- G01N33/56983—Viruses
- G01N33/56988—HIV or HTLV
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
- G01N33/6863—Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
- G01N33/6863—Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
- G01N33/6869—Interleukin
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/005—Assays involving biological materials from specific organisms or of a specific nature from viruses
- G01N2333/08—RNA viruses
- G01N2333/15—Retroviridae, e.g. bovine leukaemia virus, feline leukaemia virus, feline leukaemia virus, human T-cell leukaemia-lymphoma virus
- G01N2333/155—Lentiviridae, e.g. visna-maedi virus, equine infectious virus, FIV, SIV
- G01N2333/16—HIV-1, HIV-2
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/005—Assays involving biological materials from specific organisms or of a specific nature from viruses
- G01N2333/08—RNA viruses
- G01N2333/18—Togaviridae; Flaviviridae
- G01N2333/183—Flaviviridae, e.g. pestivirus, mucosal disease virus, bovine viral diarrhoea virus, classical swine fever virus (hog cholera virus) or border disease virus
- G01N2333/186—Hepatitis C; Hepatitis NANB
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/435—Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
- G01N2333/52—Assays involving cytokines
- G01N2333/521—Chemokines
- G01N2333/522—Alpha-chemokines, e.g. NAP-2, ENA-78, GRO-alpha/MGSA/NAP-3, GRO-beta/MIP-2alpha, GRO-gamma/MIP-2beta, IP-10, GCP-2, MIG, PBSF, PF-4 or KC
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/435—Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
- G01N2333/52—Assays involving cytokines
- G01N2333/521—Chemokines
- G01N2333/523—Beta-chemokines, e.g. RANTES, I-309/TCA-3, MIP-1alpha, MIP-1beta/ACT-2/LD78/SCIF, MCP-1/MCAF, MCP-2, MCP-3, LDCF-1or LDCF-2
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/435—Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
- G01N2333/52—Assays involving cytokines
- G01N2333/54—Interleukins [IL]
- G01N2333/5406—IL-4
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/435—Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
- G01N2333/52—Assays involving cytokines
- G01N2333/54—Interleukins [IL]
- G01N2333/5412—IL-6
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/435—Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
- G01N2333/52—Assays involving cytokines
- G01N2333/54—Interleukins [IL]
- G01N2333/5418—IL-7
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/435—Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
- G01N2333/52—Assays involving cytokines
- G01N2333/54—Interleukins [IL]
- G01N2333/5421—IL-8
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2800/00—Detection or diagnosis of diseases
- G01N2800/52—Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis
Definitions
- IP- 10 has its general meaning in the art and refers to interferon gamma- inducible protein 10 (IP- 10). Predicting methods during the course of the treatment
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Immunology (AREA)
- Molecular Biology (AREA)
- Chemical & Material Sciences (AREA)
- Urology & Nephrology (AREA)
- Biomedical Technology (AREA)
- Hematology (AREA)
- Cell Biology (AREA)
- Medicinal Chemistry (AREA)
- Pathology (AREA)
- Biotechnology (AREA)
- Food Science & Technology (AREA)
- Virology (AREA)
- Physics & Mathematics (AREA)
- Analytical Chemistry (AREA)
- Biochemistry (AREA)
- General Health & Medical Sciences (AREA)
- General Physics & Mathematics (AREA)
- Microbiology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Tropical Medicine & Parasitology (AREA)
- AIDS & HIV (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Description
METHODS FOR PREDICTING AND MONITORING TREATMENT RESPONSE IN HCV- AND HCV/HIV-INFECTED SUBJECTS
FIELD OF THE INVENTION:
The present invention relates to methods and compositions for predicting and monitoring treatment response in HCV- and HCV/HIV-infected subjects.
BACKGROUND OF THE INVENTION:
Infection by hepatitis C virus (HCV) and HIV shared common routes of transmission and thus co-infection with the two viruses is frequent [1]. HCV infection is seen in 15-30% of HIV-infected patients in Western Europe, with around 25 000 HIV-infected individuals also chronically infected by HCV in France [1, 2]. Since the introduction of highly active antiretroviral therapy (HAART), HCV-related liver disease has become a leading cause of morbidity and mortality in HIV-infected individuals [3-5].
Faster progression to cirrhosis was observed in patients co-infected with HCV and
HIV by comparison with individuals infected with HCV alone [6]. HIV/HCV co-infection is also associated with higher HCV viral levels in serum [7]. Finally, HAART -related adverse events are more frequent in HCV/HIV co-infected individuals, which increase difficulty to optimally treat HIV infection in those patients [8-11].
HCV eradication is therefore a suitable objective in HIV/HCV co-infected individuals.
Current standard therapy for HCV/HIV co-infected patients is PeglNFa in combination with ribavirin (RBV). Combination of RBV with PeglNFa has resulted in an approximately 50% rate of recovery in patients infected with HCV genotype 1 [12, 13]. HCV/HIV co-infected individuals have a lower response to therapy [14-17]. Interferons directly activate immune cells and have multiple functions limiting virus replication.
The effect of PeglNFa on cytokine production and cell activation is probably highly complex throughout multiple regulatory networks and with large inter-individual differences. Therapy efficacy against HCV seems to be dependent on the patient's immunological status at baseline.
By contrast, data on the impact of PeglNFa-RBV treatment, which is an active immunotherapy, on T cell activation and cytokine network remains poorly characterized. Likewise immunological changes associated to sustained virological response needs to be explored. Thus, the association between multiple cytokines, T cells activation and treatment
outcomes are largely unknown. Characterizing and monitoring the immunological determinants of sustained virological response among HIV/HCV co-infected patients, particularly after PeglNFa-RBV treatment initiation may be helpful in optimizing therapeutic outcome.
SUMMARY OF THE INVENTION:
The present invention relates to methods and compositions for predicting and monitoring treatment response in HCV- and HCV/HIV-infected subjects. DETAILED DESCRIPTION OF THE INVENTION:
The HCV treatment responsiveness was investigated by inventors in HCV- and HCV/HIV-infected subjects using microbeads-based multiplex immunoassay to simultaneously measure secretion level of 25 cytokines alongside with the level of CD8+ T cell activation. Immunological response to HCV treatment was evaluated at baseline and after initiation of PeglNFa-RBV therapy. The inventors found that IL-8, IP- 10 and MCP-1 pretreatment levels were higher in the non-responder subjects. The inventors also demonstrated that cytokine levels change in serum samples obtained before and 4 weeks after initiation of treatment. The inventors demonstrated that MCP-1, IL-4, IL-6, IL-7 and IP- 10 levels statistically increase in responder patients after four weeks of therapy whereas their values decreased or remained stable in the non-responder patients. The inventors also found that the IL-8 and MIP-Ιβ concentration levels remain stable or increase in responder subjects whereas their values tend to decrease in non-responder subjects four weeks after initiation of treatment by comparison with pretreatment levels. Therefore, measuring cytokines levels before and 4 weeks after initiation of HCV treatment constitutes new approaches for monitoring HCV or HCV/HIV treatment efficacy and for maintaining or reorienting such a treatment.
Definitions The term "subject" denotes a mammal. In a preferred embodiment of the invention, a subject refers to any subject (preferably human) afflicted with HCV infection. In a particular embodiment, the subject may also be infected by HIV.
The term "HCV infection" has its general meaning in the art and refers to a subject infected by hepatitis C virus.
The term "HCV/HIV co-infection" has its general meaning in the art and refers to a subject co-infected by hepatitis C virus and human immunodeficiency virus.
The term "interferon-alpha" or "IFN-alpha" refers to a family of related polypeptides that inhibit viral replication and cellular proliferation and modulate immune response. The term "IFN-alpha" includes naturally occurring IFN-alpha; synthetic IFN-alpha; derivatized IFN-alpha (e. g., PEGylated IFN-alpha, glycosylated IFN-alpha, and the like); and analogs of naturally occurring or synthetic IFN-alpha; essentially any IFN-alpha that has antiviral properties, as described for naturally occurring IFN-alpha. Suitable interferons alpha include, but are not limited to, naturally-occurring IFN-alpha (including, but not limited to, naturally occurring IFN-alpha2a, IFN-alpha2b) or recombinant interferon alpha. The term "IFN-alpha" also encompasses derivatives of IFN-alpha that are derivatized (e. g., are chemically modified) to alter certain properties such as serum half-life. As such, the term "IFN-alpha" includes glycosylated IFN-alpha; IFN-alpha derivatized with polyethylene glycol ("PEGylated IFN-alpha" or "PEG-IFN"); and the like. PEGylated IFN-alpha, and methods for making same, is discussed in, e. g., U. S. Patent Nos. 5,382, 657; 5,981, 709; and 5,951, 974. PEGylated IFN-alpha encompasses conjugates of PEG and any of the above-described IFN- alpha molecules, including, but not limited to, PEG conjugated to interferon alpha-2a (Roferon,Hoffman La-Roche, Nutley, N. J.), interferon alpha 2b (Intron, Schering-Plough, Madison, N. J. ), interferon alpha-2c (Berofor Alpha, Boehringer Ingelheim, Ingelheim, Germany); and consensus interferon as defined by determination of a consensus sequence of naturally occurring interferons alpha (Infergen, InterMune, Inc. , Brisbane,Calif).
The term "ribavirin" denotes the l-B-D-ribofuranosyl-l,2,4-triazole-3-carboxamide compound. Ribavirin is a nucleoside analog available from ICN Pharmaceuticals, Inc., Costa Mesa,Calif., and is described in the Merck Index, compound No. 8199, Eleventh Edition. Its manufacture and formulation is described in U. S. Pat. No. 4,211,771.
The term "HCV treatment" relates to any type of HCV therapy undergone by the HCV-infected subjects. Typically, said treatment may be a PeglNFa and ribavirine treatment, a protease inhibitor treatment, a replicase inhibitor treatment, a polymerase inhibitor
treatment, a helicase inhibitor treatment, a therapeutic vaccine-based treatment, a RNA interference-based treatment, an entry inhibitor treatment, an immunotherapy treatment, an immunomodulatory treatment or a combination of any of these treatments. The term "HCV/HIV treatment" relate to any type of HCV/HIV therapy undergone by the HCV/HIV co-infected subjects, including highly active antiretroviral therapy (HAART), interferon, peginterferon-a (PeglNFa), ribavirin. Typically, said treatment may be a PeglNFa and ribavirine treatment.
The term "responder" refers to a subject with a sustained virological response (SVR) when serum HCV RNA was undetectable 24 weeks after completing treatment. The term "responder" refers to HCV infected or HCV/HIV subject that will respond to HCV or HCV/HIV treatment. The term "non-responder" refers to a subject without a sustained virological response (Non-SVR) when HCV RNA decrease was < 2 log copies/mL at week 12 or when HCV RNA was detectable at the end of treatment. The term "non-responder" refers to HCV infected or HCV/HIV subject that will not respond to HCV or HCV/HIV treatment.
The term "IL-8" has its general meaning in the art and refers to interleukin-8 (IL-8). The term "MCP-1" has its general meaning in the art and refers to monocyte chemoattractant protein-1 (MCP-1).
The term "MIP-Ιβ" has its general meaning in the art and refers to macrophage inflammatory protein (ΜΙΡ-1β).
The term "IL-4" has its general meaning in the art and refers to interleukin-4 (IL-4).
The term "IL-6" has its general meaning in the art and refers to interleukin-6 (IL-6). The term "IL-7" has its general meaning in the art and refers to interleukin-7 (IL-7).
The term "IP- 10" has its general meaning in the art and refers to interferon gamma- inducible protein 10 (IP- 10).
Predicting methods during the course of the treatment
In one embodiment, the present invention relates to a method for determining whether a HCV-infected subject is a responder or is a non-responder to a HCV treatment which comprises the steps of:
i) measuring the expression level of at least one cytokine selected from the group consisting of IL-7, IL-8, ΜΙΡ-Ι β, MCP-1, IL-4, IL-6, and IP-10 in a blood sample obtained from said subject before treatment,
ii) measuring the expression level of at least one cytokine selected from the group consisting of IL-7, IL-8, ΜΙΡ-Ιβ, MCP-1, IL-4, IL-6, and IP-10 in a blood sample obtained from said subject after 1, 2, 3 or 4 weeks of treatment,
iii) and comparing the expression levels measured at step ii) with the expression levels measured at step i) wherein a difference between said expression levels is indicative that said subject is a responder or a non-responder.
In a particular embodiment, the cytokine expression level of step ii) is measured after 4 weeks of treatment.
Typically, when the expression level of IL-7 determined at step ii) is higher than the expression level of IL-7 determined at step i), then it is concluding that the subject is a responder to treatment, and accordingly, when the expression level of IL-7 determined at step ii) is equal or lower than the expression level of IL-7 determined at step i), then it is concluding that the subject is a non-responder to treatment. Typically, when the expression level of IL-8 determined at step ii) is higher than the expression level of IL-8 determined at step i), then it is concluding that the subject is a responder to treatment, and accordingly, when the expression level of IL-8 determined at step ii) is equal or lower than the expression level of IL-8 determined at step i), then it is concluding that the subject is a non-responder to treatment.
Typically, when the expression level of MIP-Ι β determined at step ii) is higher than the expression level of MIP-Ιβ determined at step i), then it is concluding that the subject is a responder to treatment, and accordingly, when the expression level of MIP-Ιβ determined at
step ii) is equal or lower than the expression level of MIP-Ιβ determined at step i), then it is concluding that the subject is a non-responder to treatment.
Typically, when the expression level of MCP-1 determined at step ii) is higher than the expression level of MCP-1 determined at step i), then it is concluding that the subject is a responder to treatment, and accordingly, when the expression level of MCP-1 determined at step ii) is equal or lower than the expression level of MCP-1 determined at step i), then it is concluding that the subject is a non-responder to treatment.
Typically, when the expression level of IL-4 determined at step ii) is higher than the expression level of IL-4 determined at step i), then it is concluding that the subject is a responder to treatment, and accordingly, when the expression level of IL-4 determined at step ii) is equal or lower than the expression level of IL-4 determined at step i), then it is concluding that the subject is a non-responder to treatment.
Typically, when the expression level of IL-6 determined at step ii) is higher than the expression level of IL-6 determined at step i), then it is concluding that the subject is a responder to treatment, and accordingly, when the expression level of IL-6 determined at step ii) is equal or lower than the expression level of IL-6 determined at step i), then it is concluding that the subject is a non-responder to treatment.
Typically, when the expression level of IP- 10 determined at step ii) is higher than the expression level of IP- 10 determined at step i), then it is concluding that the subject is a responder to treatment, and accordingly, when the expression level of IP- 10 determined at step ii) is equal or lower than the expression level of IP- 10 determined at step i), then it is concluding that the subject is a non-responder to treatment.
In some embodiments, the expression levels of 2, 3, 4, 5, 6, or 7 cytokines are measured.
In a particular embodiment, the expression level of IL-8 and ΜΙΡ-Ιβ, IL-8 and MCP-1 or MIP-Ιβ and MCP-1 are measured.
In a particular embodiment, the expression level of IL-8, MIP-Ι β and MCP-1 are measured.
In a particular embodiment, the expression levels of IL-8, ΜΙΡ-Ιβ, MCP-1, IL-4, IL-6, IL-7 and IP- 10 are measured.
Predicting methods before the initiation of the treatment
In a further embodiment, the present invention relates to a method for determining whether a HCV-infected subject will be a responder or a non-responder to a HCV treatment which comprises the steps of:
(i) measuring the expression level of at least one cytokine selected from the group consisting of IL-8, ΜΙΡ-Ιβ, MCP-1, IL-4, IL-6, IL-7 and IP-10 in a blood sample obtained from said subject before the treatment,
(ii) comparing the expression level measured at step i) with a reference value,
(iii) detecting differential in the cytokine expression level between the blood sample and the reference value is indicative that said subject will be a responder or a non-responder.
In a particular embodiment, the present invention comprises measurement of IL-8 expression level.
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 cytokine expression levels obtained according to the method of the invention with a defined threshold value. In one embodiment of the present invention, the threshold value is derived from the cytokine expression level (or ratio, or score) determined in a blood sample derived from one or more subjects who are responders to HCV or HCV/HIV
treatment. In one embodiment of the present invention, the threshold value may also be derived from cytokine expression level (or ratio, or score) determined in a blood sample derived from one or more subjects who are non-responders to HCV or HCV/HIV treatment. Furthermore, retrospective measurement of the cytokine expression levels (or ratio, or scores) in properly banked historical subject samples may be used in establishing these threshold values.
In one embodiment of the invention, the reference value may consist in expression level measured in a blood sample associated with a responder subject or in a blood sample associated with a non-responder subject.
Typically, when the expression level determined for IL-8, MCP-1 or IP- 10 is lower than the corresponding reference value it is concluded that the HCV-infected subject will be a responder to HCV treatment, and accordingly, when the expression level determined for IL-8, MCP-1 or IP- 10 is higher than the corresponding reference value it is concluded that the HCV-infected subject will be a non responder to HCV treatment.
In some embodiments, the expression levels of 2, 3, 4, 5, 6, or 7 cytokines are measured.
In a particular embodiment, the expression levels of IL-8 and IP- 10, IL-8 and MCP-1 or IP- 10 and MCP-1 are measured.
In a particular embodiment, the expression levels of IL-8, IP- 10 and MCP-1 are measured.
In a particular embodiment, the expression levels of IL-8, ΜΙΡ-Ιβ, MCP-1, IL-4, IL-6, IL-7 and IP- 10 are measured. In another particular embodiment, a score which is a composite of the expression levels of the different cytokines may be also determined and compared to a reference value wherein a difference between said score and said reference value is indicative whether said subject is a responder or a non-responder to HCV or HCV/HIV treatment.
In a particular embodiment, the score may be generated by a computer program.
Methods for determining the expression level of a cytokine Methods for measuring the expression level of a cytokine in a blood sample may be assessed by any of a wide variety of well-known methods from one of skill in the art for detecting expression of a protein including, but not limited to, enzyme immunoassay (EIA), radioimmunoassay (RIA), Western blot analysis, ELISA, Luminex, ELISPOT and enzyme linked immunoabsorbant assay.
Said analysis can be assessed by contacting the blood sample with a binding partner capable of selectively interacting with the cytokine present in the blood sample. The binding partner may be an antibody that may be polyclonal or monoclonal, preferably monoclonal (e.g., a radio-labeled, chromophore- labeled, fluorophore-labeled, or enzyme-labeled antibody), an antibody derivative (e.g., an antibody conjugate with a substrate or with the protein or ligand of a protein of a protein/ligand pair (e.g., biotin-streptavidin)), or an antibody fragment (e.g., a single-chain antibody, an isolated antibody hypervariable domain, etc.) which binds specifically to the protein translated from the gene encoding for IL-7, IL-8, ΜΙΡ-Ιβ, MCP-1 , IL-4, IL-6, and IP- 10. In another embodiment, the binding partner may be an aptamer.
Polyclonal antibodies of the invention or a fragment thereof can be raised according to known methods by administering the appropriate antigen or epitope to a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, and mice, among others. Various adjuvants known in the art can be used to enhance antibody production. Although antibodies useful in practicing the invention can be polyclonal, monoclonal antibodies are preferred.
Monoclonal antibodies of the invention or a fragment thereof can be prepared and isolated using any technique that provides for the production of antibody molecules by continuous cell lines in culture. Techniques for production and isolation include but are not limited to the hybridoma technique originally described by Ko filer and Milstein (1975); the human B-cell hybridoma technique (Cote et al, 1983); and the EBV-hybridoma technique (Cole et al. 1985).
Alternatively, techniques described for the production of single chain antibodies (see e.g. U.S. Pat. No. 4,946,778) can be adapted to produce anti-cytokine, single chain antibodies. Antibodies useful in practicing the present invention also include anti-cytokine fragments including but not limited to F(ab')2 fragments, which can be generated by pepsin digestion of an intact antibody molecule, and Fab fragments, which can be generated by reducing the disulfide bridges of the F(ab')2 fragments. Alternatively, Fab and/or scFv expression libraries can be constructed to allow rapid identification of fragments having the desired specificity to cytokine. For example, phage display of antibodies may be used. In such a method, single- chain Fv (scFv) or Fab fragments are expressed on the surface of a suitable bacteriophage, e. g., M13. Briefly, spleen cells of a suitable host, e. g., mouse, that has been immunized with a protein are removed. The coding regions of the VL and VH chains are obtained from those cells that are producing the desired antibody against the protein. These coding regions are then fused to a terminus of a phage sequence. Once the phage is inserted into a suitable carrier, e. g., bacteria, the phage displays the antibody fragment. Phage display of antibodies may also be provided by combinatorial methods known to those skilled in the art. Antibody fragments displayed by a phage may then be used as part of an immunoassay.
In another embodiment, the binding partner may be an aptamer. Aptamers are a class of molecule that represents an alternative to antibodies in term of molecular recognition. Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity. Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library, as described in Tuerk C. 1997. The random sequence library is obtainable by combinatorial chemical synthesis of DNA. In this library, each member is a linear oligomer, eventually chemically modified, of a unique sequence. Possible modifications, uses and advantages of this class of molecules have been reviewed in Jayasena S.D., 1999. Peptide aptamers consist of conformationally constrained antibody variable regions displayed by a platform protein, such as E. coli Thioredoxin A, that are selected from combinatorial libraries by two hybrid methods (Colas et al, 1996).
The binding partners of the invention such as antibodies or aptamers, may be labelled with a detectable molecule or substance, such as a fluorescent molecule, a radioactive molecule or any others labels known in the art. Labels are known in the art that generally provide (either directly or indirectly) a signal.
As used herein, the term "labelled", with regard to the antibody, is intended to encompass direct labelling of the antibody or aptamer by coupling (i.e., physically linking) a detectable substance, such as a radioactive agent or a fluorophore (e.g. fluorescein isothiocyanate (FITC) or phycoerythrin (PE) or Indocyanine (Cy5)) to the antibody or aptamer, as well as indirect labelling of the probe or antibody by reactivity with a detectable substance. An antibody or aptamer of the invention may be labelled with a radioactive molecule by any method known in the art. For example radioactive molecules include but are not limited radioactive atom for scintigraphic studies such as 1123, 1124, Inl l l, Re 186, Rel88.
The afore mentioned assays generally involve the binding of the binding partner (ie. antibody or aptamer) to a solid support. Solid supports which can be used in the practice of the invention include substrates such as nitrocellulose (e. g., in membrane or microtiter well form); polyvinylchloride (e. g., sheets or microtiter wells); polystyrene latex (e.g., beads or microtiter plates); polyvinylidene fluoride; diazotized paper; nylon membranes; activated beads, magnetically responsive beads.
In a particular embodiment, an ELISA method can be used, wherein the wells of a microtiter plate are coated with a set of antibodies which recognize said cytokine(s). A blood sample containing or suspected of containing said cytokine(s) is then added to the coated wells. After a period of incubation sufficient to allow the formation of antibody-antigen complexes, the plate(s) can be washed to remove unbound moieties and a detectably labelled secondary binding molecule added. The secondary binding molecule is allowed to react with any captured sample marker protein, the plate washed and the presence of the secondary binding molecule detected using methods well known in the art.
In one embodiment, an Enzyme-linked immunospot (ELISpot) method may be used. Typically, the blood sample is transferred to a plate which has been coated with the desired anti-cytokine capture antibodies. Revelation is carried out with biotinylated secondary Abs and standard colorimetric or fluorimetric detection methods such as streptavidin-alkaline phosphatase and NBT-BCIP and the spots counted.
In one embodiment, when multi- cytokine expression measurement is required, use of beads bearing binding partners of interest may be preferred. In a particular embodiment, the
bead may be a cytometric bead for use in flow cytometry. Such beads may for example correspond to BD™ Cytometric Beads commercialized by BD Biosciences (San Jose, California). Typically cytometric beads may be suitable for preparing a multiplexed bead assay. A multiplexed bead assay, such as, for example, the BD(TM) Cytometric Bead Array, is a series of spectrally discrete beads that can be used to capture and quantify soluble antigens. Typically, beads are labelled with one or more spectrally distinct fluorescent dyes, and detection is carried out using a multiplicity of photodetectors, one for each distinct dye to be detected. A number of methods of making and using sets of distinguishable beads have been described in the literature. These include beads distinguishable by size, wherein each size bead is coated with a different target-specific antibody (see e.g. Fulwyler and McHugh, 1990, Methods in Cell Biology 33:613-629), beads with two or more fluorescent dyes at varying concentrations, wherein the beads are identified by the levels of fluorescence dyes (see e.g. European Patent No. 0 126,450), and beads distinguishably labelled with two different dyes, wherein the beads are identified by separately measuring the fluorescence intensity of each of the dyes (see e.g. U.S. patent Nos. 4,499,052 and 4,717,655). Both one-dimensional and two- dimensional arrays for the simultaneous analysis of multiple antigens by flow cytometry are available commercially. Examples of one-dimensional arrays of singly dyed beads distinguishable by the level of fluorescence intensity include the BD<-™-) Cytometric Bead Array (CBA) (BD Biosciences, San Jose, Calif.) and Cyto-Plex(TM) Flow Cytometry microspheres (Duke Scientific, Palo Alto, Calif). An example of a two-dimensional array of beads distinguishable by a combination of fluorescence intensity (five levels) and size (two sizes) is the QuantumPlex1'™^ microspheres (Bangs Laboratories, Fisher, Ind.). An example of a two-dimensional array of doubly-dyed beads distinguishable by the levels of fluorescence of each of the two dyes is described in Fulton et al. (1997, Clinical Chemistry 43(9): 1749-1756). The beads may be labelled with any fluorescent compound known in the art such as e.g. FITC (FL1), PE (FL2), fiuorophores for use in the blue laser (e.g. PerCP, PE-Cy7, PE-Cy5, FL3 and APC or Cy5, FL4), fiuorophores for use in the red, violet or UV laser (e.g. Pacific blue, pacific orange). In another particular embodiment, bead is a magnetic bead for use in magnetic separation. Magnetic beads are known to those of skill in the art. Typically, the magnetic bead is preferably made of a magnetic material selected from the group consisting of metals (e.g. ferrum, cobalt and nickel), an alloy thereof and an oxide thereof. In another particular embodiment, bead is bead that is dyed and magnetized.
Kits of the invention
The invention also relates to a kit for performing the methods as above described, wherein said kit comprises means for measuring the expression level of at least one cytokine selected from the group consisting of IL-7, IL-8, ΜΙΡ-Ιβ, MCP-1 , IL-4, IL-6, and IP- 10 that are indicative of subject responder to HCV or HCV/HIV treatment. Typically the kit may include an antibody, or a set of antibodies as above described. In a particular embodiment, the antibody or set of antibodies are labelled as above described. The kit may also contain other suitably packaged reagents and materials needed for the particular detection protocol, including solid-phase matrices, if applicable, and standards.
Methods of treatment
The method of the invention allows to define a subgroup of subjects who are responder or non responder to the HCV treatment.
A further object of the invention relates to a method for the treatment of HCV- infection in a subject in need thereof comprising the steps of:
a) determining whether a HCV-infected subject is a responder or a non-responder to HCV treatment by performing the method according to the invention,
b) administering the HCV treatment, if said subject has been considered as a responder.
A further object of the invention relates to a PeglNFa and ribavirine for use in the treatment of HCV infection in a subject in need thereof, wherein the subject was being classified as responder by the method as above described.
The invention will be further illustrated by the following examples. However, these examples should not be interpreted in any way as limiting the scope of the present invention. EXAMPLE:
Multicytokine response to Peg-Interferon-a-based therapy strongly influences HCV responsiveness in HIV co-infected patients.
Material & Methods
Patient samples
Thirty patients co-infected by HCV and HIV and followed at Montpellier University Hospital were included in this study after providing written informed consent. Ethics approval was obtained from our Institutional Review Board. Chronic hepatitis was proved by presence of serum HCV antibodies and detectable viral RNA. HCV genotype, HCV and HIV-1 viral loads, CD4+ T cell count, and liver enzyme levels were all determined using standard procedures.
Based on the treatment effect, HCV/HIV co-infected patients were ranged into two distinct groups: i) those with a sustained viro logical response (SVR) when serum HCV RNA was undetectable 24 weeks after completing therapy, and ii) those without a sustained viro logical response (Non-SVR) when HCV RNA decrease was < 2 log copies/mL at week 12 or when HCV RNA was detectable at the end of treatment (Table 1). Patient relapsing in 24 weeks period after therapeutic cessation were exclude from the study.
All (n=30) R (n=19) NR (n=ll)
Age 45 44 47
CD4 T (cells/μΐ) 350 (50-1076) 378(128-1076) 328 (50-715)
Nadir CD4 T
(cells/μΐ) 122.5 (32-598) 124 (33-598) 119.5 (32-324)
ALAT (UI/1) 48.5 (13-172) 40.5 (13-172) 56.5 (24-133)
ASAT (UI/1) 40 (20-224) 37 (20-224) 54 (31-106)
CV VHC SO
(Ul/ml) 915000 311000 4830000
CV VHC S4 (UI/1) 27000 270 1215500
CV VIH SO (UI/1) 0 0 0
Gen VHC 1/2/3/4 19/1/6/1 9/1/6/0 10/0/0/1
Ratio CD8CD38
S4/S0 1.34 (0.15-4.5) 1.27 (0.15-4.5) 1.46 (0.8-4.3)
Table 1: Patient characteristics
Routine laboratory testing
The inventors detected antibodies to HCV in serum samples via third generation enzyme-linked immunoassays (Cobas; Roche Laboratories). Serum levels of HCV RNA were
detected using the COBAS AMPLICOR assays (Roche Diagnostic Systems), which amplify HCV RNA by reverse transcriptase-polymerase chain reaction. The lower limit of the assay was 50 IU/mL. HCV genotype was determined using INNO-LiPA HCV II test (Innogenetics). Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were quantified using standard methods.
Quantitation of serum cytokine concentrations
Cytokines and chemokines were quantified in serum samples obtained before initiation of treatment, and 4 weeks after the start of PeglNFa-based therapy. A multiplexed microbead assay was used in this purposed according to manufacturer's instructions (cytokine twenty- five-plex kit, Life Technologies Ltd, Paisley, UK) and a FIDIS™ apparatus (BMD, Marne La Vallee, France). Twenty-five cytokines were quantified: IL-Ι β, IL-1RA, IL-2, IL-2R, IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12p40/70, IL-13, IL-15, IL-17, EOTAXIN, GM-CSF, IFN-a, IFN-γ, IP-10, MCP-1 , MIG, MIP-la, ΜΙΡ-Ι β, RANTES, and TNF-a. Data were analyzed using the MLX-Booster program (BMD) and standard curves. Mean concentrations (pg/ml) of cytokines and chemokines were all superior to the detection limits, defined as the mean background value plus 2 SD.
Analysis of CD8 /CD38bri ht T cells
Activation of CD8+ T cells was assessed by flow cytometry analysis on EDTA-treated fresh whole blood using FC 500 apparatus (Beckman Coulter, Miami, Florida). The expression of CD38bnght on CD8+ T cells was explored by using two-color staining with anti- CD8 and anti-CD38 conjugated to fluorescein isothiocyanate (FITC) and phycoerythrin (PE), respectively (Beckman Coulter) as previously described [18]. The positive threshold for CD38bright analysis was established using the CellQuant CD38/CD8 kit for quantitation of CD38 cell surface expression (BioCytex, Marseille, France) and was defined as 8,500 CD38 binding sites/cells. The CD8+/CD38bnght values were expressed as the percentage of CD38bnght cells from the CD8+ T cell populations. Statistical Analysis
The Mann- Whitney U test and Kruskal-Wallis test were used to analyze continuous variables where appropriate. The Friedman test was used to evaluate changes in serum cytokine levels over time . A P value < .05 was considered statistically significant.
Multivariate analyses were performed using a stepwise logistic regression model. Statistical analyses were done using SPSS software version 18.0J.
Results
Clinical characteristics of patients at baseline
Of the 30 HCV/HIV co-infected patients receiving peglFNa and RBV therapy, 19 (63%) have SVR with prolonged suppression of HCV plasma RNA after therapeutic cessation and 11 (37%) have non-SVR. Before treatment initiation, the median AST and ALT levels were significantly lower in the SVR group than those in the non-SVR group (37 vs 54 IU/L). Likewise the inventors observed a lower HCV RNA viral load in the SVR group compared to the non-SVR group (311000 vs 4830000 IU/L).
Pretreatment levels of soluble and cell inflammatory markers
Twenty five cytokines secretion levels were quantitated in serum samples prior to anti- HCV therapy by multiplex assays (Table 2). Two of soluble markers were statistically associated with therapeutic response at baseline. IP- 10 levels were higher in the non-SVR group than in the SVR group: 64.3 (33.0) pg/ml vs 132.0 (58.9) pg/ml, respectively, P = 0.021 . Individuals from the non-SVR group had also higher MCP-1 pretreatment concentrations: 347.9.3 pg/ml (91.2) vs 442.6 (145.7) pg/ml, respectively, P = 0.0009. Serum concentrations of IL-8 observed in patients of the non-SVR group also tend to be higher than those observed in the SVR group: 22.5 (73.8) pg/ml vs 128.1 (707.8) pg/ml, respectively P = 0.106.
SVR pg/ml NSVR pg/ml p-value
Cytokines mean [CV] mean [CV] (Wilcoxon)
ILlbeta 12.9 [34.24] 10.84 [4.904] 0.093
IL1RA 1556 [198] 1506 [123] 0.779
IL2 6.296 [2.427] 5.866 [2.628] 0.425
IL2R 837 [812.27] 884.1 [155.6] 0.101
IL4 8 [177.95] 8 [173.5] 0.338
IL5 2.352 [0.589] 2.352 [0] 0.616
IL6 4.666 [2.25] 5 [23.017] 0.353
IL7 18.91 [2.38] 20.88 [4.32] 0.142
IL8 22.51 [73.85] 128.1 [707.79] 0.106
IL10 3.608 [1.18] 3.092 [2.061] 0.777
IL12p40.70 321.5 [55.8] 318.3 [56.5] 0.948
IL13 11 [4.125] 11 [2.755] 0.702
IL15 33.21 [9.48] 33.21 [3.16] 0.863
IL17 24.89 [9.34] 24 [8] 0.73
TNFa 8.866 [2.606] 8.866 [1.167] 0.762
IFNg 5.268 [1.371] 6.146 [0.878] 0.621
GM.CSF 18.46 [9.23] 15.38 [13.84] 0.482
MlPla 119.4 [1.8] 120.6 [6.8] 0.262
MlPlb 150.4 [24.3] 150.4 [23.6] 0.88
IP10 64.27 [32.98] 132 [58.89] 0.021
MIG 458.6 [5.9] 458.6 [4] 0.497
EOTAXIN 75.54 [30.71] 72.96 [29.4] 0.813
RANTES 10540 [6413] 9350 [10922] 0.683
MCP1 347.9 [91.2] 442.6 [145.7] 0.009
IFNa 143 [21.3] 143 [1.4] 0.477
Table 2: Pretreatment Cytokine concentration in serum from SVR and NSVR patients
Cytokine changes after therapeutic initiation of HC V treatment
As a consequence of PeglNFa administration, IFN-a rose in the two groups. This enhancement of INF-a in serum was not different in SVR vs. non-SVR patients. PeglNFa therapy induced also a significant increased of the levels of 7 immune soluble markers in serum of SVR and non-SVR patients after four weeks of treatment: IL-Ιβ, IL-1RA, IL-2, IL- 5, IL-12, IL-13, and Eotaxin. By contrast, five cytokines and chemokines levels rose only in the SVR group: IL-4, IL-6, IL-7, IP-10, and MCP-1. Finally, when results were analyzed
without consideration of the therapeutic HCV response, the inventors also observed an increase in IL-10 and RA TES concentration (Table 3).
Comparison of the cytokine level changes induced by PeglNFa therapy in the SVR and the non-SVR group.
The ratios of cytokines concentrations at four weeks and at baseline were compared in SVR and non-SVR patients (Table 3). A robust increased of MCP-1 and ΜΙΡ-1β concentration was induced by the PeglNFa-based therapy in the SVR group whereas the value decreased or remained stable in the non-SVR group. Likewise in most of patients responding to anti-HCV treatment, the IL-8 concentration remains stable after one month period of PeglNFa-based therapy, whereas it is declined in non-SVR patients.
All patients SVR NSVR
p- p-
Cytokine Median [95%IC] value Median [95%IC] value Median [95%IC] p-value
ILlbeta 2.05 [1.21 -4.86] 2e-04 4.42 [1.15-6.17] 0.0053 1.32 [1.11 -4.09] 0.0068
IL1RA 1.38 [1.3 - 1.54] le-04 1.43 [1.32 - 1.63] 3e-04 1.35 [0.99 - 1.57] 0.0537
IL2 1.3 [1.07 - 1.54] 0.0066 1.3 [1 - 1.65] 0.0546 1.29 [1 -6.1] 0.042
IL2R 1.26 [0.79 - 6.03] 0.2621 1.38 [0.83 - 8.44] 0.0955 0.93 [0.42 - 7.78] 0.8311
IL4 16.73 [1.19 - 19.42] 0.0104 16.81 [1.2- 19.6] 0.0058 1.21 [0.49-21.1] 0.7596
IL5 1.34 [1.23 - 1.47] <0.001 1.31 [1.16- 1.52] 0.001 1.37 [1.18- 1.77] 0.0067
IL6 1.65 [1.23 - 7.08] 0.0013 2.11 [1.33 - 10.58] 0.001 1.23 [0.69 - 6.36] 0.5195
IL7 1.12 [1.02- 1.5] 0.0237 1.14 [1.03 - 1.63] 0.0124 1.03 [0.89-4.1] 0.6247
IL8 0.95 [0.56-2.39] 0.9032 1.26 [0.68-3.13] 0.49 0.58 [0.09-2.9] 0.1748
IL10 1.43 [1.02 -2.35] 0.0417 1.67 [0.92-3.15] 0.0948 1.21 [0.89 - 3.45] 0.2061
IL12p40.70 1.44 [1.31 - 1.61] <0.001 1.48 [1.29- 1.8] <0.001 1.42 [1.22- 1.66] 0.001
IL13 1.99 [1.61 - 8.89] <0.001 2.24 [1.63-9.6] 8e-04 1.66 [1.38 - 12.04] 0.0038
IL15 1.13 [0.95 - 1.34] 0.1812 1.19 [0.97- 1.49] 0.08 0.97 [0.81 -4.72] 0.8984
IL17 1.22 [0.99-4.76] 0.0799 1.9 [0.99-5.18] 0.0663 1.04 [0.76-4.74] 0.8385
TNFa 0.86 [0.76- 1.12] 0.2286 0.94 [0.78- 1.21] 0.8596 0.77 [0.68 - 14.39] 0.0674
IFNg 1.04 [0.78- 1.21] 0.5784 1.03 [0.78 - 1.22] 0.732 1.06 [0.73 - 1.41] 0.6889
GM.CSF 1.24 [0.96- 1.57] 0.0712 1.34 [0.89-2] 0.1025 1.16 [0.8- 1.44] 0.624
MlPla 1.06 [0.74- 1.16] 0.7096 1.08 [0.73 - 1.24] 0.6026 0.99 [0.64- 1.16] 0.9188
MlPlb 1.08 [0.71 -2.54] 0.7922 1.37 [0.84- 5.52] 0.1232 0.56 [0.1 - 1.09] 0.083
IP10 1.46 [1.18- 1.94] 6e-04 1.6 [1.19-2.56] 0.0033 1.24 [0.91 - 1.96] 0.1016
MIG 1.07 [0.77 - 1.09] 0.3762 1.04 [0.76- 1.1] 0.5062 1.08 [0.73 - 1.24] 0.6247
EOTAXIN 1.95 [1.53 -2.45] <0.001 2.24 [1.57 -2.98] <0.001 1.55 [1.16-2.23] 0.0068
RANTES 1.33 [1.01 - 1.89] 0.0427 1.31 [0.97 - 3.19] 0.0874 1.4 [0.73 - 2.04] 0.2783
MCP1 1.23 [1.04 - 1.47] 0.0145 1.42 [1.2 - 1.97] le-04 0.87 [0.69 - 1.24] 0.5771
IFNa 4.89 [3.85 - 6.12] <0.001 5.06 [3.83 - 6.71] <0.001 4.6 [2.95 - 8.51] 0.001
Table 3: Cytokines fold changes after therapeutic initiation of HCV treatment. Ratios of S4/S0 concentrations are indicated overall and according to the HCV response (SVR and NSVR) for each cytokines
Changes in T cell activation over the 3 months period of therapeutic initiation.
Pretreatment CD8+ T cell activation was more elevated in non-SVR patients compared to patients belonging to the SVR group. Under PeglNFa-based therapy, an increased of CD38bnght expression was observed at the surface of CD8 T+ cells in the two groups of patients. This is the direct effect of the immunologically active therapy on T cell activation. However, no significant differences in the increase of CD8+ T cell activation were observed between the two groups of patients suggesting that this marker was not associated with a better PeglNFa immune response. 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.
[1] Alter MJ. Epidemiology of viral hepatitis and HIV co-infection. J Hepatol 2006;44(1 Suppl):S6-9.
[2] Larsen C, Pialoux G, Salmon D, Antona D, Le Strat Y, Piroth L, Pol S, Rosenthal E, Neau D, Delarocque Astagneau E. Prevalence of hepatitis C and hepatitis B infection in the HIV- infected population for France, 2004. Euro Surveill 2008;13(22).
[3] Bica I, McGovern B, Dhar R, Stone D, McGowan K, Scheib R, Snydman DR. Increasing mortality due to end-stage liver disease in patients with human immunodeficiency virus infection. Clin Infect Dis 2001;32(3):492-7.
[4] Pineda JA, Garcia-Garcia JA, Aguilar-Guisado M, Rios-Villegas MJ, Ruiz-Morales J, Rivero A, del Valle J, Luque R, Rodriguez-Bano J, Gonzalez- Serrano M, Camacho A, Macias
J, Grilo I, Gomez-Mateos JM. Clinical progression of hepatitis C virus-related chronic liver disease in human immunodeficiency virus-infected patients undergoing highly active antiretroviral therapy. Hepatology 2007;46(3):622-30.
[5] Martin-Carbonero L, Soriano V, Valencia E, Garcia- Samaniego J, Lopez M, Gonzalez- Lahoz J. Increasing impact of chronic viral hepatitis on hospital admissions and mortality among HIV-infected patients. AIDS Res Hum Retroviruses 2001 ;17( 16): 1467-71.
[6] Benhamou Y, Bochet M, Di Martino V, Charlotte F, Azria F, Coutellier A, Vidaud M, Bricaire F, Opolon O, Katlama C, Poynard T. Liver fibrosis progression in human immunodeficiency virus and hepatitis C virus coinfected patients. The Multivirc Group. Hepatology 1999;30(4): 1054-8.
[7] Eyster ME, Fried MW, Di Bisceglie AM, Goedert JJ. Increasing hepatitis C virus RNA levels in hemophiliacs: relationship to human immunodeficiency virus infection and liver disease. Blood 1994;84(4): 1020-3.
[8] Aceti A, Pasquazzi C, Zechini B, De Bac C. Hepatotoxicity development during antiretroviral therapy containing protease inhibitors in patients with HIV: the role of hepatitis B and C virus infection. J Acquir Immune Defic Syndr 2002;29(l):41-8.
[9] Sulkowski MS. Hepatotoxicity associated with antiretroviral therapy containing HIV-1 protease inhibitors. Semin Liver Dis 2003;23(2): 183-94.
[10] Sulkowski MS, Mehta SH, Chaisson RE, Thomas DL, Moore RD. Hepatotoxicity associated with protease inhibitor-based antiretroviral regimens with or without concurrent ritonavir. AIDS 2004;18(17):2277-84.
[11] Sulkowski MS, Thomas DL, Mehta SH, Chaisson RE, Moore RD. Hepatotoxicity associated to nevirapine or efavirenz-containing antiretroviral therapy: role of hepatitis C and B infections. Hepatology 2002;35(1): 182-9.
[12] Manns MP, McHutchison JG, Gordon SC, Rustgi VK, Shiffman M, Reindollar R, Goodman ZD, Koury K, Ling M, Albrecht JK. Peginterferon alfa-2b plus ribavirin compared with interferon alfa-2b plus ribavirin for initial treatment of chronic hepatitis C: a randomized trial. Lancet 2001;358:958-65.
[13] Fried MW, Shiffman ML, Reddy KR, Smith C, Marinos G, Goncales FL, Haussinger D, Diago M, Carosi G, Dhumeaux D, Craxi A, Lin A, Hoffman J, Yu J. Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection. N Engl J Med 2002;347(13):975-82.
[14] Carrat F, Bani-Sadr F, Pol S, Rosenthal E, Lunel-Fabiani F, Benzekri A. Pegylated interferon alfa-2b vs standard interferon alfa-2b, plus ribavirin, for chronic hepatitis C in HIV-infected patients: a randomized controlled trial. JAMA 2004; 292(23):2839-48.
[15] Chung RT, Andersen J, Volberding P, Robbins GK, Liu T, Sherman KE, Peters MG, Koziel MJ, Bhan AK, Alston B, Colquhoun D, Nevin T, Harb G, van der Horst C, AIDS Clinical Trials Group A5071 Study Team. Peginterferon alfa-2a plus ribavirin versus interferon alfa-2a plus ribavirin for chronic hepatitis C in HIV-coinfected persons. N Engl J Med 2004;351(5):451-9.
[16] Perez-Olmeda M, Nunez M, Romero M, Gonzalez J, Castro A, Arribas JR, Pedreira J, Barreiro P, Garcia-Samaniego J, Martin-Carbonero L, Jimenez-Nacher I, Soriano V. Pegylated IFN-alpha2b plus ribavirin as therapy for chronic hepatitis C in HIV-infected patients. AIDS 2003;17(7): 1023-8.
[17] Torriani FJ, Rodriguez-Torres M, Rockstroh JK, Lissen E, Gonzalez-Garcia J, Lazzarin A, Carosi G, Sasadeusz J, Katlama C, Montaner J, Sette H Jr, Passe S, De Pamphilis J, Duff F, Schrenk UM, Dieterich DT, APRICOT Study Group. Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection in HIV-infected patients. N Engl J Med 2004;351(5):438-50.
[18] Tuaillon E, Al Tabaa Y, Baillat V, Segondy M, Picot MC, Reynes J, Vendrell JP. Close association of CD8+/CD38 bright with HIV-1 replication and complex relationship with CD4+ T-cell count. Cytometry B Clin Cytom 2009;76(4):249-60.
Claims
1. A method for determining whether a HCV-infected subject is a responder or is a non- responder to a HCV treatment which comprises the steps of:
i) measuring the expression level of at least one cytokine selected from the group consisting of IL-7, IL-8, ΜΙΡ-Ιβ, MCP-1 , IL-4, IL-6, and IP-10 in a blood sample obtained from said subject before treatment,
ii) measuring the expression level of at least one cytokine selected from the group consisting of IL-7, IL-8, ΜΙΡ-Ιβ, MCP-1 , IL-4, IL-6, and IP-10 in a blood sample obtained from said subject after 1, 2, 3 or 4 weeks of treatment,
iii) and comparing the expression levels measured at step ii) with the expression levels measured at step i) wherein a difference between said expression levels is indicative that said subject is a responder or a non-responder.
2. The method according to claim 1 wherein the expression level of at least one cytokine of step ii) is measured after 4 weeks of treatment.
3. The method according to any one of claims 1 or 2 wherein said method comprises measuring the expression level of IL-8, MIP-Ιβ and MCP-1.
4. A method for determining whether a HCV-infected subject will be a responder or a non-responder to a HCV treatment which comprises the steps of:
(i) measuring the expression level of at least one cytokine selected from the group consisting of IL-8, ΜΙΡ-Ιβ, MCP-1 , IL-4, IL-6, IL-7 and IP-10 in a blood sample obtained from said subject before the treatment,
(ii) comparing the expression level measured at step i) with a reference value,
(iii) detecting differential in the cytokine expression level between the blood sample and the reference value is indicative that said subject will be a responder or a non-responder.
5. The method according to claim 4 wherein said method comprises measuring the expression level of IL-8, IP-10 and MCP-1.
6. The method according to any one of claims 1, 2 or 4 wherein said method comprises measuring the expression level of IL-8.
7. The method according to any one of claims 1 to 6 wherein said subject is also infected by HIV.
8. A kit for performing the methods according to any one of claims 1 to 7 wherein said kit comprises means for measuring the expression level of at least one cytokine selected from the group consisting of IL-7, IL-8, ΜΙΡ-Ιβ, MCP-1, IL-4, IL-6, and IP-10.
9. A method for treating HCV-infection in a subject in need thereof comprising the steps of:
a) determining whether a HCV-infected subject is a responder or a non-responder to HCV treatment by performing the method according to any one of claims 1 to 7,
b) administering the HCV treatment, if said subject has been considered as a responder.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP12305573 | 2012-05-24 | ||
| EP12305573.3 | 2012-05-24 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2013174988A1 true WO2013174988A1 (en) | 2013-11-28 |
Family
ID=48579025
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/EP2013/060770 Ceased WO2013174988A1 (en) | 2012-05-24 | 2013-05-24 | Methods for predicting and monitoring treatment response in hcv- and hcv/hiv-infected subjects |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2013174988A1 (en) |
Citations (11)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US4211771A (en) | 1971-06-01 | 1980-07-08 | Robins Ronald K | Treatment of human viral diseases with 1-B-D-ribofuranosyl-1,2,4-triazole-3-carboxamide |
| EP0126450A2 (en) | 1983-05-19 | 1984-11-28 | Ioannis Dr. Tripatzis | Particle and method for the detection of antigens and/or antibodies using this particle |
| US4499052A (en) | 1982-08-30 | 1985-02-12 | Becton, Dickinson And Company | Apparatus for distinguishing multiple subpopulations of cells |
| US4717655A (en) | 1982-08-30 | 1988-01-05 | Becton, Dickinson And Company | Method and apparatus for distinguishing multiple subpopulations of cells |
| US4946778A (en) | 1987-09-21 | 1990-08-07 | Genex Corporation | Single polypeptide chain binding molecules |
| US5382657A (en) | 1992-08-26 | 1995-01-17 | Hoffmann-La Roche Inc. | Peg-interferon conjugates |
| US5951974A (en) | 1993-11-10 | 1999-09-14 | Enzon, Inc. | Interferon polymer conjugates |
| US5981709A (en) | 1997-12-19 | 1999-11-09 | Enzon, Inc. | α-interferon-polymer-conjugates having enhanced biological activity and methods of preparing the same |
| WO2000019209A1 (en) * | 1998-09-28 | 2000-04-06 | Diacrin, Inc. | Determining resistance to treatment for hepatitis c virus |
| WO2011058084A1 (en) * | 2009-11-14 | 2011-05-19 | F. Hoffmann-La Roche Ag | Biomarkers for predicting rapid response to hcv treatment |
| WO2011067195A1 (en) * | 2009-12-02 | 2011-06-09 | F. Hoffmann-La Roche Ag | Biomarkers for predicting sustained response to hcv treatment |
-
2013
- 2013-05-24 WO PCT/EP2013/060770 patent/WO2013174988A1/en not_active Ceased
Patent Citations (11)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US4211771A (en) | 1971-06-01 | 1980-07-08 | Robins Ronald K | Treatment of human viral diseases with 1-B-D-ribofuranosyl-1,2,4-triazole-3-carboxamide |
| US4499052A (en) | 1982-08-30 | 1985-02-12 | Becton, Dickinson And Company | Apparatus for distinguishing multiple subpopulations of cells |
| US4717655A (en) | 1982-08-30 | 1988-01-05 | Becton, Dickinson And Company | Method and apparatus for distinguishing multiple subpopulations of cells |
| EP0126450A2 (en) | 1983-05-19 | 1984-11-28 | Ioannis Dr. Tripatzis | Particle and method for the detection of antigens and/or antibodies using this particle |
| US4946778A (en) | 1987-09-21 | 1990-08-07 | Genex Corporation | Single polypeptide chain binding molecules |
| US5382657A (en) | 1992-08-26 | 1995-01-17 | Hoffmann-La Roche Inc. | Peg-interferon conjugates |
| US5951974A (en) | 1993-11-10 | 1999-09-14 | Enzon, Inc. | Interferon polymer conjugates |
| US5981709A (en) | 1997-12-19 | 1999-11-09 | Enzon, Inc. | α-interferon-polymer-conjugates having enhanced biological activity and methods of preparing the same |
| WO2000019209A1 (en) * | 1998-09-28 | 2000-04-06 | Diacrin, Inc. | Determining resistance to treatment for hepatitis c virus |
| WO2011058084A1 (en) * | 2009-11-14 | 2011-05-19 | F. Hoffmann-La Roche Ag | Biomarkers for predicting rapid response to hcv treatment |
| WO2011067195A1 (en) * | 2009-12-02 | 2011-06-09 | F. Hoffmann-La Roche Ag | Biomarkers for predicting sustained response to hcv treatment |
Non-Patent Citations (24)
| Title |
|---|
| "Merck Index" |
| ACETI A; PASQUAZZI C; ZECHINI B; DE BAC C: "Hepatotoxicity development during antiretroviral therapy containing protease inhibitors in patients with HIV: the role of hepatitis B and C virus infection", J ACQUIR IMMUNE DEFIC SYNDR, vol. 29, no. 1, 2002, pages 41 - 8 |
| ALTER MJ: "Epidemiology of viral hepatitis and HIV co-infection", J HEPATOL, vol. 44, no. 1, 2006, pages 6 - 9 |
| ANGELES JIMENEZ-SOUSA MARIA ET AL: "Increased Th1, Th17 and pro-fibrotic responses in hepatitis C-infected patients are down-regulated after 12 weeks of treatment with pegylated interferon plus ribavirin", EUROPEAN CYTOKINE NETWORK, JOHN LIBBEY EUROTEXT LTD, FR, vol. 21, no. 2, 1 June 2010 (2010-06-01), pages 84 - 91, XP002615303, ISSN: 1148-5493 * |
| BENHAMOU Y; BOCHET M; DI MARTINO V; CHARLOTTE F; AZRIA F; COUTELLIER A; VIDAUD M; BRICAIRE F; OPOLON 0; KATLAMA C: "Liver fibrosis progression in human immunodeficiency virus and hepatitis C virus coinfected patients", THE MULTIVIRC GROUP. HEPATOLOGY, vol. 30, no. 4, 1999, pages 1054 - 8 |
| BICA I; MCGOVERN B; DHAR R; STONE D; MCGOWAN K; SCHEIB R; SNYDMAN DR: "Increasing mortality due to end-stage liver disease in patients with human immunodeficiency virus infection", CLIN INFECT DIS, vol. 32, no. 3, 2001, pages 492 - 7 |
| CARRAT F; BANI-SADR F; POL S; ROSENTHAL E; LUNEL-FABIANI F; BENZEKRI A: "Pegylated interferon alfa-2b vs standard interferon alfa-2b, plus ribavirin, for chronic hepatitis C in HIV-infected patients: a randomized controlled trial", JAMA, vol. 292, no. 23, 2004, pages 2839 - 48 |
| CHUNG RT; ANDERSEN J; VOLBERDING P; ROBBINS GK; LIU T; SHERMAN KE; PETERS MG; KOZIEL MJ; BHAN AK; ALSTON B: "AIDS Clinical Trials Group A5071 Study Team. Peginterferon alfa-2a plus ribavirin versus interferon alfa-2a plus ribavirin for chronic hepatitis C in HIV-coinfected persons", N ENGL J MED, vol. 351, no. 5, 2004, pages 451 - 9 |
| EYSTER ME; FRIED MW; DI BISCEGLIE AM; GOEDERT JJ: "Increasing hepatitis C virus RNA levels in hemophiliacs: relationship to human immunodeficiency virus infection and liver disease", BLOOD, vol. 84, no. 4, 1994, pages 1020 - 3 |
| FELD J J ET AL: "Ribavirin Improves Early Responses to Peginterferon Through Improved Interferon Signaling", GASTROENTEROLOGY, ELSEVIER, PHILADELPHIA, PA, vol. 139, no. 1, 1 July 2010 (2010-07-01), pages 154 - 162.e4, XP027202923, ISSN: 0016-5085, [retrieved on 20100317] * |
| FRIED MW; SHIFFMAN ML; REDDY KR; SMITH C; MARINOS G; GONQALES FL; HAUSSINGER D; DIAGO M; CAROSI G; DHUMEAUX D: "Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection", N ENGL J MED, vol. 347, no. 13, 2002, pages 975 - 82 |
| FULTON ET AL., CLINICAL CHEMISTRY, vol. 43, no. 9, 1997, pages 1749 - 1756 |
| FULWYLER; MCHUGH, METHODS IN CELL BIOLOGY, vol. 33, 1990, pages 613 - 629 |
| LARSEN C; PIALOUX G; SALMON D; ANTONA D; LE STRAT Y; PIROTH L; POL S; ROSENTHAL E; NEAU D; DELAROCQUE ASTAGNEAU E: "Prevalence of hepatitis C and hepatitis B infection in the HIV-infected population for France, 2004", EURO SURVEILL 2008, vol. 13, no. 22, 2004 |
| MANNS MP; MCHUTCHISON JG; GORDON SC; RUSTGI VK; SHIFFMAN M; REINDOLLAR R; GOODMAN ZD; KOURY K; LING M; ALBRECHT JK: "Peginterferon alfa-2b plus ribavirin compared with interferon alfa-2b plus ribavirin for initial treatment of chronic hepatitis C: a randomized trial", LANCET, vol. 358, 2001, pages 958 - 65, XP004806199, DOI: doi:10.1016/S0140-6736(01)06102-5 |
| MARTIN-CARBONERO L; SORIANO V; VALENCIA E; GARCIA-SAMANIEGO J; LOPEZ M; GONZALEZ-LAHOZ J: "Increasing impact of chronic viral hepatitis on hospital admissions and mortality among HIV-infected patients", AIDS RES HUM RETROVIRUSES, vol. 17, no. 16, 2001, pages 1467 - 71 |
| PEREZ-OLMEDA M; NUNEZ M; ROMERO M; GONZALEZ J; CASTRO A; ARRIBAS JR; PEDREIRA J; BARREIRO P; GARCIA-SAMANIEGO J; MARTIN-CARBONERO: "Pegylated IFN-alpha2b plus ribavirin as therapy for chronic hepatitis C in HIV-infected patients", AIDS, vol. 17, no. 7, 2003, pages 1023 - 8 |
| PINEDA JA; GARCIA-GARCIA JA; AGUILAR-GUISADO M; RIOS-VILLEGAS MJ; RUIZ-MORALES J; RIVERO A; DEL VALLE J; LUQUE R; RODRIGUEZ-BANO J: "Clinical progression of hepatitis C virus-related chronic liver disease in human immunodeficiency virus-infected patients undergoing highly active antiretroviral therapy", HEPATOLOGY, vol. 46, no. 3, 2007, pages 622 - 30 |
| STEPHANIE BEQ ET AL: "Altered Thymic Function during Interferon Therapy in HCV-Infected Patients", PLOS ONE, vol. 7, no. 4, 16 April 2012 (2012-04-16), pages e34326 - e34326, XP055035949, ISSN: 1932-6203, DOI: 10.1371/journal.pone.0034326 * |
| SULKOWSKI MS: "Hepatotoxicity associated with antiretroviral therapy containing HIV-1 protease inhibitors", SEMIN LIVER DIS, vol. 23, no. 2, 2003, pages 183 - 94 |
| SULKOWSKI MS; MEHTA SH; CHAISSON RE; THOMAS DL; MOORE RD: "Hepatotoxicity associated with protease inhibitor-based antiretroviral regimens with or without concurrent ritonavir", AIDS, vol. 18, no. 17, 2004, pages 2277 - 84 |
| SULKOWSKI MS; THOMAS DL; MEHTA SH; CHAISSON RE; MOORE RD: "Hepatotoxicity associated to nevirapine or efavirenz-containing antiretroviral therapy: role of hepatitis C and B infections", HEPATOLOGY, vol. 35, no. 1, 2002, pages 182 - 9 |
| TORRIANI FJ; RODRIGUEZ-TORRES M; ROCKSTROH JK; LISSEN E; GONZALEZ-GARCIA J; LAZZARIN A; CAROSI G; SASADEUSZ J; KATLAMA C; MONTANER: "APRICOT Study Group. Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection in HIV-infected patients", N ENGL J MED, vol. 351, no. 5, 2004, pages 438 - 50 |
| TUAILLON E; AL TABAA Y; BAILLAT V; SEGONDY M; PICOT MC; REYNES J; VENDRELL JP: "Close association of CD8+/CD38 bright with HIV-1 replication and complex relationship with CD4+ T-cell count", CYTOMETRY B CLIN CYTOM, vol. 76, no. 4, 2009, pages 249 - 60 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Tsoukas et al. | Markers predicting progression of human immunodeficiency virus-related disease | |
| Mehta et al. | Serum albumin as a prognostic indicator for HIV disease progression | |
| US20230204568A1 (en) | Measuring trail by lateral flow immunoassay | |
| Kalantar-Zadeh et al. | Diagnostic discordance for hepatitis C virus infection in hemodialysis patients | |
| Becquart et al. | Acute dengue virus 2 infection in Gabonese patients is associated with an early innate immune response, including strong interferon alpha production | |
| de Paula et al. | Reduction of inflammation and T cell activation after 6 months of cART initiation during acute, but not in early chronic HIV-1 infection | |
| Hutchinson et al. | Multiplex analysis of cytokines in the blood of cynomolgus macaques naturally infected with Ebola virus (Reston serotype) | |
| Aghemo et al. | Quantification of core antigen monitors efficacy of direct-acting antiviral agents in patients with chronic hepatitis C virus infection | |
| Biancotto et al. | A highly sensitive and dynamic immunofluorescent cytometric bead assay for the detection of HIV-1 p24 | |
| Montanheiro et al. | The elevated interferon gamma production is an important immunological marker in HAM/TSP pathogenesis | |
| Bello et al. | Assessment of a hepatitis C virus antibody assay in saliva for epidemiological studies | |
| Neuman et al. | Predictors of sustained response to alpha interferon therapy in chronic hepatitis C | |
| Hasegawa et al. | Th1 cytokine endotype discriminates and predicts severe complications in COVID-19 | |
| BRIGGS et al. | A 90-kDa protein serum marker for the prediction of progression to AIDS in a cohort of HIV-1+ homosexual men | |
| Groopman et al. | Lack of evidence of prolonged human immunodeficiency virus infection before antibody seroconversion | |
| EP2678683B1 (en) | A method for the prognosis of progression of the hiv disease | |
| WO2013174988A1 (en) | Methods for predicting and monitoring treatment response in hcv- and hcv/hiv-infected subjects | |
| Nguyen et al. | Cytokine response associated with Hepatitis C virus clearance in HIV coinfected patients initiating peg interferon-α based therapy | |
| WO1997039358A1 (en) | In vitro prognostic test for progressors and non-progressors after hiv infection | |
| Grant et al. | Clinical and immunologic predictors of death after an acute opportunistic infection: results from ACTG A5164 | |
| Molina et al. | Quantification of HIV-1 virus load under zidovudine therapy in patients with symptomatic HIV infection: relation to disease progression | |
| Ra et al. | Viral, Immunologic, and laboratory parameters in patients with and without post-acute sequelae of SARS-CoV-2 infection (PASC) | |
| RU2226694C2 (en) | Method for differential diagnostics of viral hepatitis c | |
| Thomé et al. | Persistent immunological repercussions of late antiretroviral initiation in children living with HIV | |
| McCloskey et al. | Response to superantigen stimulation in peripheral blood mononuclear cells from children perinatally infected with human immunodeficiency virus and receiving highly active antiretroviral therapy |
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: 13727830 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: 13727830 Country of ref document: EP Kind code of ref document: A1 |