WO2016184963A1 - Traitement de patients souffrant d'une infection par le vih - Google Patents

Traitement de patients souffrant d'une infection par le vih Download PDF

Info

Publication number
WO2016184963A1
WO2016184963A1 PCT/EP2016/061273 EP2016061273W WO2016184963A1 WO 2016184963 A1 WO2016184963 A1 WO 2016184963A1 EP 2016061273 W EP2016061273 W EP 2016061273W WO 2016184963 A1 WO2016184963 A1 WO 2016184963A1
Authority
WO
WIPO (PCT)
Prior art keywords
hiv
immunogenic compound
peptide
group
cooh
Prior art date
Application number
PCT/EP2016/061273
Other languages
English (en)
Inventor
Joël Crouzet
Raphaël Ho Tsong Fang
Original Assignee
Innavirvax
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 Innavirvax filed Critical Innavirvax
Publication of WO2016184963A1 publication Critical patent/WO2016184963A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • 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
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6037Bacterial toxins, e.g. diphteria toxoid [DT], tetanus toxoid [TT]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6081Albumin; Keyhole limpet haemocyanin [KLH]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/62Medicinal preparations containing antigens or antibodies characterised by the link between antigen and carrier
    • A61K2039/627Medicinal preparations containing antigens or antibodies characterised by the link between antigen and carrier characterised by the linker
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • This invention relates to the treatment and/or prevention of an HIV infection or a HIV-related condition in an individual.
  • HIV Human Immunodeficiency Virus
  • Current therapies have succeeded in controlling the disease but long-term use of Anti-Retroviral Therapy (ART) is limited due to the nature of the viral replication cycle of those viruses, but also by issues of side effects.
  • ART Anti-Retroviral Therapy
  • HIV virions start multiplying again as soon as antiretroviral therapy (ART) is interrupted, which typically means daily, life-long treatment for patients.
  • ART antiretroviral therapy
  • retroviruses such as HIV are characterized by proviral latency, which is the ability of a virus to lie dormant within a cell. Latency is the phase of the viral replication cycle in which, after initial infection, proliferation of virus particles ceases without full eradication.
  • proviral latency is associated to the appearance of so-called "reservoirs" within the host, which are generally difficult to reach, and which are also one of the main reasons of the difficulty to provide with a sterilizing cure and/or a functional cure for HIV.
  • the invention relates to an immunogenic compound comprising an antigenic peptide derived from the HIV gp41 protein, for use for treating an HIV-infected individual under anti-retro viral treatment, in the course of anti-retro viral treatment interruption.
  • the invention relates to an immunogenic compound as defined above, for treating or preventing or delaying the rebound of HIV viral loads, including HIV RNA levels, or a HIV-related condition in said individual.
  • the invention relates to an immunogenic compound as defined above, for reducing the amplitude of the rebound of HIV viral loads, including HIV RNA levels, or a HIV-related condition in said individual.
  • the invention relates to an immunogenic compound as defined above, for reducing the likelihood of rebound of HIV viral loads, including HIV RNA levels, or a HIV-related condition in said individual.
  • the invention further relates to vaccine compositions comprising said immunogenic compound, for the uses described herein
  • the invention relates to a vaccine composition
  • a vaccine composition comprising said immunogenic compound, for use for treating an HIV-infected individual under anti- retroviral treatment, in the course of anti-retroviral treatment interruption.
  • Figure 1 Immunization Schedule.
  • A. Schedule for three injections (DO, week 4 and week 8) at 0.1 ⁇ g and 20 ⁇ g of antigenic peptide equivalent.
  • B. Schedule for three injections (DO, week 4 and week 8) at 1 ⁇ g and 10 ⁇ g of antigenic peptide equivalent, and an additional booster injection at week 24.
  • follow-up visits are scheduled at week 48, week 60, week 72 and week 84.
  • Figure 2 Dose-dependent Immunogenicity at week 12.
  • Dose- dependent evolution over time P 0.003.
  • Figure 3 Variation of the HIV DNA from baseline over time at week 60 and 84, in placebo vs non responder vs responder patients.
  • the y-axis represents the HIV DNA change from baseline in log copies per 10 6 Peripheral Blood Mononuclear Cells (PBMC). From left to right: placebo group, non-responder patients, responder patients.
  • PBMC Peripheral Blood Mononuclear Cells
  • Figure 4 Evolution of HIV DNA per million of PBMCs over time, in Responders vs Non Responders + placebo.
  • the y-axis represents the HIV DNA change from baseline in log copies per 10 6 Peripheral Blood Mononuclear Cells (PBMC).
  • the x- axis represents time, at 12, 36, 60 and 84 weeks after the first administration of the immunogenic compound.
  • Non Responders + placebos are represented by "o”.
  • Responders are represented by "+”.
  • p values of ANOVA test for differences between the groups are shown, p corresponds to the significance of the changes from baseline
  • Figure 5 Anti-3S Dose-Dependent Change of Total HIV DNA over time at week 12, 36, 60 and 84.
  • the y-axis represents the HIV DNA change from baseline in log copies per 10 6 Peripheral Blood Mononuclear Cells (PBMC).
  • the x-axis represents the anti-3S total antibodies at the corresponding time, expressed in log A.U. A. At week 12.
  • B. At week 36.
  • C. At week 60.
  • D. At week 84.
  • Figure 6 Variation of the CD4/CD8 ratio over time, in placebo vs non responder vs responder patients.
  • the y-axis represents the CD4/CD8 ratio.
  • the x-axis represents for each patient the measurements corresponding, from left to right, to the baseline (week 0) and at week 24.
  • Figure 7 Variation of the CD4 and CD8% at week 24 after injection of the immunogenic compound.
  • the present invention has for purpose to meet the aforementioned needs.
  • the present invention provides anti-HIV immunogenic compounds, and compositions thereof, comprising antigenic peptides derived from the gp41 protein of HIV, for treating an HIV-infected individual under anti-retroviral treatment, in the course of anti-retroviral treatment interruption
  • the invention relates to such immunogenic compounds, and/or compositions thereof, for use as a medicament; which includes compositions in the form of pharmaceutical compositions and/or vaccine compositions.
  • the invention also relates to such immunogenic compounds, and/or compositions thereof, for use as a medicament, or for the preparation of a medicament, for treating an HIV-infected individual under anti-retroviral treatment, in the course of anti- retroviral treatment interruption.
  • HIV-1 gp41 is composed of three domains, an extracellular domain (ectodomain), a transmembrane domain and an intracellular domain (endodomain).
  • the gp41 ectodomain contains three major functional regions, i.e., the fusion peptide located at the N-terminus of gp41, followed by two 4-3 heptad repeats adjacent to the N- and C- terminal portions of the gp41 ectodomain, designated NHR (N-terminal heptad repeat) and CHR (C-terminal heptad repeat), respectively.
  • N- and C-terminal repeats are also named as "HR1" and "HR2". Both NHR and CHR regions function as essential structures required for conformational changes during the process of membrane fusion between HIV-1 and CD4+ T cells.
  • WO2013/179262 taught immunogenic compounds derived from a HIV gp41 peptide, for treating a condition caused by the infection of an individual with a HIV virus. This document showed that such immunogenic compounds were able to raise antibodies having the ability to reduce the NK-induced CD4 T cell lysis in individuals infected with an HIV virus.
  • SEROCO cohorts consist of confirmed HIV patients, of which the contamination date is known or of which seropositivity has been first established less than two years (24 months), or even less than one year (12 months) before the inclusion, and who were AIDS-free at enrollment and had no prior antiretroviral therapy.
  • immunogenic compounds comprising an antigenic peptide derived from the gp41 protein of a HIV virus can reduce Peripheral Blood Mononuclear Cells (PBMC)-associated HIV DNA levels in HIV-infected individuals.
  • PBMC Peripheral Blood Mononuclear Cells
  • immunogenic compounds comprising an antigenic peptide derived from the gp41 protein of a HIV virus can maintain or restore T cell homeostasis and thus high level of CD4 + T cell count.
  • PBMCs Peripheral Blood Mononuclear Cells
  • the total cell-associated HIV DNA has been recently proposed as a standardized clinical marker for measuring HIV reservoirs in HIV-infected individuals. Most noticeably, it has been recently determined that this value represents the global capacity of the reservoirs to produce virus over time.
  • HIV DNA is a distinct marker from HIV RNA. HIV DNA is expressed as a number of copies per 10 6 PBMCs, and is indicative of the integration of the HIV genome in cellular "reservoirs", which includes CD4 + T cells. HIV-DNA levels are positively correlated to HIV-RNA in patients receiving ART.
  • the inventors have further shown in the examples that the administration of an immunogenic compound comprising an antigenic peptide derived from gp41 in HIV- infected individuals, lead to an increase of the CD4/CD8 ratio in the responder group, 24 weeks after the first injection.
  • the CD4/CD8 is established as an indicator of immune system health, and thus its restoration and/or increase is indicative of immune restoration (Serrano-Villar et al, PLOS Pathogens, vol. 10, 2014).
  • the term “comprising” encompasses “consisting of.
  • the terms “patient” and “individual” encompass humans and non-human animals, which includes non-human mammals, and/or any organism that is prone to HIV infection.
  • « preventing » also encompasses « reducing the likelihood of occurrence » or « reducing the likelihood of reoccurrence»
  • an "anti-retroviral agent” or “anti-retroviral treatment” relates to the administration of a compound, or combination of compounds, acting against an HIV infection.
  • the invention relates to an immunogenic compound comprising an antigenic peptide derived from the gp41 protein of an HIV virus, for use for treating an HIV-infected individual under anti-retroviral treatment, in the course of anti-retroviral treatment interruption.
  • an "individual under anti-retroviral treatment” relates to HIV- infected individuals who have been administered, or alternatively who are being administered at least one drug in the context of an anti-retroviral treatment for treating said HIV infection, thereby defining a reference anti-retroviral treatment.
  • an HIV-infected individual "in the course of anti-retroviral treatment interruption” encompasses HIV-infected individuals under anti-retroviral treatment, and wherein the said anti-retroviral treatment was effective and led to a control of the HIV infection, as assessed by a low PBMC-associated HIV DNA value, a low viral load value and a high level of CD4+ T cells.
  • These individuals which define a specific population of HIV-infected individuals among the general population of HIV-infected individuals who receive an anti-retroviral treatment, may thus be selected for undergoing an interruption of the anti-retroviral treatment which is replaced by a treatment with an immunogenic compound comprising an antigenic peptide derived from the gp41 protein.
  • the HIV-infected individuals which are eligible to an anti- retroviral treatment interruption are characterized by one or more of the following physiological parameters:
  • PBMC Peripheral Blood Mononuclear Cells
  • the immunogenic compound comprising an antigenic peptide derived from the HIV gp41 protein is administered to the HIV-infected individuals at a time period when the anti-retroviral treatment is still also administered.
  • the administration of the said immunogenic compound raises, in said HIV-infected individuals, an antibody response directed against the antigenic peptide derived from the gp41 protein, which antibody response has been shown by the inventors to cause a reduction in the PBMC-associated DNA, and thus a reduction of the HIV reservoir in the said HIV-infected individuals.
  • the detection of an antibody response directed against the said antigenic peptide derived from the HIV gp41 protein may be conventionally performed, for example by using an ELISA assay.
  • the results of the ELISA assay are analyzed against a control curve generated by using a serial of anti- antigenic peptide antibodies at known dilutions or at known concentrations.
  • the anti-retroviral treatment may be stopped or interrupted in these specific HIV-infected individuals when an antibody response against the antigenic peptide derived from the HIV gp41 protein has been raised and or when the threshold of marker(s), e.g. (i) PBMC-associated HIV DNA level, (ii) HIV RNA level and (iii) CD4+ T cells level, is (are) reached.
  • the threshold of marker(s) e.g. (i) PBMC-associated HIV DNA level, (ii) HIV RNA level and (iii) CD4+ T cells level
  • the said immunogenic compound is administered only once during a time period wherein the HIV-infected individual is still administered with the anti-retroviral treatment.
  • the said immunogenic compound is administered a plurality of times during a time period wherein the HIV-infected individual is still administered with the anti-retroviral treatment.
  • the said immunogenic compound is administered a number of times ranging from 2 to 6 during a time period wherein the HIV-infected individual is still administered with the anti-retroviral treatment.
  • the said immunogenic compound may be administered twice, with the second administration being performed about four weeks subsequent to the first administration, during a time period wherein the HIV-infected individual is still administered with the anti-retro viral treatment.
  • the said immunogenic compound may be administered three times, with the second administration being performed about four weeks subsequent to the first administration and the third administration being performed about four weeks subsequent to the second administration, during a time period wherein the HIV-infected individual is still administered with the anti-retroviral treatment.
  • the said immunogenic compound may be administered according to the following treatment schedule:
  • T2 Tl + about four weeks :3 rd administration of the immunogenic compound
  • - T3 T2 + about twelve weeks : 4 th administration of the immunogenic compound
  • - T4 T3 + about twelve weeks :5 th administration of the immunogenic compound
  • T5 T4 + about twelve weeks: 6 th administration of the immunogenic compound.
  • an interruption of the anti- retroviral treatment may be performed at various times during the administration schedule, including :
  • the anti-retroviral treatment is stopped or interrupted, once an antibody response directed against the antigenic peptide derived from the HIV gp41 protein is detected in the serum of the said HIV-infected individual.
  • the immunogenic compound comprising the antigenic peptide derived from the gp41 protein may be administered at appropriate time intervals following the anti-retroviral treatment interruption.
  • one or more subsequent administrations of the immunogenic compound comprising the antigenic peptide derived from the gp41 protein may be performed at time intervals ranging from 3 months to 24 months following the anti-retroviral treatment interruption, which includes at time intervals ranging from 6 months to 12 months following the anti-retroviral treatment interruption.
  • the HIV-infected individual is subject to a regular monitoring of one or more of the following physiological parameters including (i) the level of PBMC-associated HIV DNA, (ii) the level of the HIV RNA and (iii) the level of CD4+ T cells.
  • a further administration of an immunogenic compound comprising an antigenic peptide derived from the HIV gp41 protein may be decided in the HIV-infected individuals having stopped an antiretroviral treatment when one or more of the following physiological changes is detected or determined:
  • the said further administration of the immunogenic compound that is decided is aimed at maintaining or increasing the immune response against the antigenic peptide derived from the HIV gp41 protein and thus maintaining or increasing the viral control by the HIV-infected individual.
  • the administration of an immunogenic compound comprising an antigenic peptide derived from the HIV gp41 protein drastically reduce the PBMC-associated HIV DNA level
  • the inventors believe that the administration of the said compound may lead, at least for some of the HIV-infected individuals treated therewith, to a total eradication of the HIV virus and thus to a complete cure to the said individuals.
  • the invention relates to immunogenic compounds comprising an antigenic peptide derived from the gp41 protein of HIV for treating, preventing or delaying the rebound of HIV viral load in the HIV-infected individual in the course of treatment interruption.
  • H7V viral load or "HIV viral titer” generally refers to:
  • - the activity or concentration of a HIV-related protein in a plasma sample which may for example include determining the reverse transcriptase (RT) activity in said plasma sample.
  • RT reverse transcriptase
  • maintaining a low viral load encompasses maintaining an
  • HIV RNA viral load under the detectable level or under a threshold of 40 copies per mL, or alternatively delaying an increase of the viral load by at least two weeks (or more) after interruption of the anti-retro viral treatment.
  • the HIV viral load test is used primarily to monitor HIV infection over time.
  • RNA HIV nucleic acid
  • the invention relates to an immunogenic compound comprising an antigenic peptide derived from the gp41 protein of an HIV virus, for use for treating an HIV-infected individual under anti-retroviral treatment, and then terminating said anti-retroviral treatment; preferably wherein:
  • the HIV viral load is low or decreased
  • HIV viral load, CD4+ cell count and PBMC-associated HIV DNA level are compared to a reference value determined at the time of the administration of said immunogenic compound.
  • the immunogenic compounds of the invention can be administered to HIV-infected individuals in the course of anti-retroviral treatment interruption for reducing the PBMC-associated HIV DNA level in an individual.
  • the invention relates to an immunogenic compound comprising an antigenic peptide derived from the gp41 protein of an HIV virus for its use as defined above, for treating or preventing or delaying a rebound of HIV viral loads in said individual.
  • the invention relates to an immunogenic compound comprising an antigenic peptide derived from the gp41 protein of an HIV virus for its use as defined above, for reducing the likelihood of rebound of HIV viral loads in said individual.
  • the invention relates to a vaccine composition, comprising at least one immunogenic compound comprising an antigenic peptide derived from the gp41 protein of an HIV virus.
  • the invention relates to a vaccine composition
  • a vaccine composition comprising at least one immunogenic compound of the invention, for use for treating an HIV-infected individual under anti-retroviral treatment, in the course of anti-retroviral treatment interruption.
  • An immunogenic compound comprising an antigenic peptide derived from the gp41 protein of a HIV virus can also be used for the preparation of any composition, in particular any pharmaceutical composition and/or vaccine composition for a use as defined above.
  • HIV infection encompasses the infection of a host animal, particularly a human host, by the HIV virus, including type 1 human immunodeficiency virus (HIV-1).
  • HIV may include HIV-I, HIV-2 and all forms, subtypes, clades and variations thereof, which includes HIV-I strains belonging to the HIV-I B subtype, HIV-I C subtype, and HIV-I recombinants.
  • the HIV virus is HIV-I.
  • HIV-1 can be used herein to refer to any strains, forms, subtypes, clades and variations in the HIV- 1 family.
  • HIV-related condition encompasses any symptom or set of symptoms commonly found in HIV-infected patients, including symptoms belonging to conditions associated with AIDS (Acquired Immune Deficiency Syndrome).
  • a carrier of HIV-1 may be identified by any methods known in the art. For example, a person can be identified as an HIV- 1 carrier on the basis that the person is anti- HIV-1 antibody positive, or is HIV-1 -positive, or has symptoms of AIDS. That is, "treating HIV-1 infection” should be understood as treating a patient who is at any one of the several stages of HIV infection progression, in particular HIV-1, infection progression.
  • HIV-infected individuals which are specifically considered by the invention are HIV-infected individuals under anti-retroviral treatment, in the course of treatment interruption.
  • HIV-infected individuals which are under anti-retroviral treatment for treating said HIV infection, thereby defining a reference anti-retroviral treatment; wherein said anti- retroviral treatment will be subsequently interrupted for a known or unknown amount of time, and will preferably be definitively interrupted.
  • HIV-infected individuals in the course of treatment interruption differ from the broader category of HIV-infected individuals under retroviral treatment, because of specific markers of HIV infection, typically favourable levels of markers including (i) a low PBMC-associated DNA level, (ii) a low viral load and (iii) a high CD4+ T cells level, as described elsewhere in the present specification.
  • An anti-retroviral treatment may also comprise the administration of at least one drug selected from: ART (Antiretroviral Therapy), and HAART (Highly Active Antiretro viral Therapy).
  • ART Antiretroviral Therapy
  • HAART Highly Active Antiretro viral Therapy
  • ART and HAART are known in the Art and generally relate to combinations of two, three or more antiretroviral medicines.
  • antiretroviral medicines encompass:
  • nucleoside/nucleotide reverse transcriptase inhibitors also called nucleoside analogs, such as abacavir, emtricitabine, and tenofovir;
  • NRTIs n- nucleoside reverse transcriptase inhibitors
  • protease inhibitors such as atazanavir, darunavir, and ritonavir
  • entry inhibitors such as enfuvirtide and maraviroc
  • (v) integrase inhibitors such as dolutegravir and raltegravir.
  • anti-retroviral treatments include, in a non-limitative manner, the administration of at least one drug selected from: Zidovudine, Lamivudine, Emtricitabine, Didanosine, Stavudine, Abacavir, Zalcitabine, Tenofivir, Racivir, Amdoxovir, Apricitabine, Elvucitabine, Efavirenz, Nevirapine, Etravirine, Delavirdine, Rilpvirine, Tenofovir, Fosalvudine, Amprenavir, Tipranavir, Indinavir, Saquinavir, Fosamprenavir, Ritonavir, Darunavir, Atazanavir, Nelfinavir, Lopinavir, Raltegravir, Elvitegravir, Dolutegravir, Enfuvirtide, Maraviroc, Vicriviroc, and combinations thereof.
  • it may be an HIV-infected individual under anti-retroviral treatment, in the course of anti-retroviral treatment interruption; wherein said anti- retrovival treatment includes the administration of at least one drug selected from: ART (Antiretro viral Therapy) and HAART (Highly Active Antiretro viral Therapy).
  • ART Antiretro viral Therapy
  • HAART Highly Active Antiretro viral Therapy
  • the first stage is referred herein as the "acute infection stage, or the "acute primary infection syndrome” (which can be asymptomatic or associated with an influenza- like illness with fevers, malaise, diarrhoea and neurologic symptoms such as headache).
  • This stage generally corresponds to the period which extends from day 1 to about 1 to 3 months after HIV infection.
  • large amounts of virus are produced, thereby increasing the viral load (or HIV RNA level) in the HIV-infected individual.
  • the CD4 cell count may also decrease rapidly to low levels.
  • the viral load or HIV RNA
  • the median of HIV-DNA levels in individuals with a primary infection is generally higher than at a chronic state.
  • a "primo-infected" individual refers in particular to an HIV- infected individual in the early stages of the infection, who has generally been contaminated within a period of less than one year, which includes less than 6 months or even less than 3 months.
  • the HIV-infected individual in the course of treatment interruption is a primo- infected individual.
  • the second stage is referred herein as the "clinical latency stage" or
  • asymptomatic infection This stage generally corresponds to the stage where the HIV- infected individual is asymptomatic, and where the disease enters into its chronic state. However, the HIV virus continues to reproduce at detectable levels. The viral load (or HIV RNA) generally remains stabilized and the CD4+ T cell count gradually declines. This stage is highly variable in length, and depends on multiple factors, such as the nature of the HIV strain, the efficacy of a treatment, host factors and so on. It may last for years, in particular if the individual is under antiretroviral treatment.
  • the third stage is referred herein as the "AIDS phase". It corresponds to the symptomatic stage, wherein the HIV-infected individual exhibits an HIV-related condition, including symptoms belonging to the condition that is generally known as AIDS. It also corresponds to the stage, where the viral load increases up to high levels, while the CD4 + T cell count drops below the level of 200 cells/mm 3 . This stage also corresponds to a life-threatening stage, in the absence of treatment.
  • HIV-infected individuals belonging to the above-mentioned stages are considered by the invention.
  • the HIV-infected individuals in the course of anti-retroviral treatment interruption can be further characterized based on distinct biological markers at the time of the administration of said immunogenic compound.
  • PBMC Peripheral Blood Mononuclear Cells
  • the "HIV viral load” relates to the number of copies of HIV RNA per mL of blood plasma and is expressed in HIV RNA copies per mL of blood plasma, according to known methods, which includes nucleic acid-based tests such as reverse- transcriptase polymerase chain reaction (RT-PCR), branched DNA (bDNA), or nucleic acid sequence-based amplification (NASBA) analysis.
  • RT-PCR reverse- transcriptase polymerase chain reaction
  • bDNA branched DNA
  • NASBA nucleic acid sequence-based amplification
  • HIV viral load test is ordered when a person is first diagnosed.
  • test results function as a baseline measurement that shows how actively the virus is reproducing.
  • the HIV viral load test is then performed over time and compared to said baseline measurement or to a reference value, in order to assess a relative variation of the HIV viral load.
  • conventional methods for determining HIV viral load include: (i) providing a whole blood sample obtained from a patient;
  • RNA iii determining the number of copies of HIV RNA per milliliter of plasma, for example, by reverse-transcriptase polymerase chain reaction (RT-PCR), branched DNA (bDNA), or nucleic acid sequence-based amplification (NASBA) analysis.
  • RT-PCR reverse-transcriptase polymerase chain reaction
  • bDNA branched DNA
  • NASBA nucleic acid sequence-based amplification
  • step (iv) optionally comparing the result obtained at step (iii) with a reference value and/or a baseline measurement.
  • HIV RNA level is low (for example, below 500 copies/mL of plasma), this indicates that the anti- retroviral treatment regimen is effective, i.e. that the virus may not be actively replicating and the disease may progress more slowly.
  • a low viral load is usually below 500 HIV RNA copies/mL of plasma; which includes between 20 and 500 copies/mL of plasma, or 40 to 500 copies/mL of plasma, depending on the type and sensitivity of the test that is used. This result indicates that HIV is not actively reproducing and that the risk of disease progression is low.
  • a low viral load may consist of a viral load below 500 HIV RNA copies/mL of plasma; which includes below 450, 400, 350, 300, 250, 200, 150, 100, 50, 40, 30, 20, 10, 9, 8, 7, 6, 5, 4, 3, 2, and 1 copies/mL.
  • a high viral load may consist of a viral load above 1000 HIV RNA copies/mL of plasma; which includes above 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000 and 10000 copies/mL of plasma.
  • An undetectable viral load for routine methods is generally below 40 copies/mL of plasma, which includes 20 copies/mL of plasma, in particular when measured with a method and/or kits selected from: COBAS® AmpliPrep/COBAS® TaqMan® HIV-1 Test and COBAS® AMPLICOR HIV-1 MONITOR Test sold by Roche Molecular Diagnostic or NucliSENS EasyQ®HIV-l sold by Biomerieux Diagnostics.
  • a method and/or kits selected from: COBAS® AmpliPrep/COBAS® TaqMan® HIV-1 Test and COBAS® AMPLICOR HIV-1 MONITOR Test sold by Roche Molecular Diagnostic or NucliSENS EasyQ®HIV-l sold by Biomerieux Diagnostics.
  • an undetectable viral load in a patient with diagnosed HIV infection does not mean that the patient is cured; it means only that the level of HIV RNA is currently below the limit of detection of the technique. What is more, an undetectable viral load does not necessarily rule out the presence of HIV in latent reservoirs.
  • Changes in viral load are generally more important during HIV monitoring than obtaining a single test result.
  • An increasing viral load indicates either that the infection is getting worse or that the virus has developed resistance to the drugs that are being used for therapy and are no longer effective.
  • a decreasing viral load indicates improvement, treatment effectiveness, and a decrease in HIV replication.
  • a restored CD4+ T cell count may correspond to a physiological
  • CD4+ T cell count which is generally equal or superior to 350 CD4+ cells/mm 3 , which includes equal or superior to 500 CD4+ cells/mm 3 of plasma, which generally varies between 500 and 1500 CD4+ T cells/mm 3 of plasma, though it may be lower for some individuals.
  • a restored CD4+ T cell count may correspond to an increase in the CD4+ T cell count, compared to the CD4+ T cell count in said patient prior to said treatment.
  • a low CD4+ T cell count includes a CD4+ T cell count inferior to 350 / mm 3 in blood plasma, which includes inferior to 350; 300; 250; 200; 150, 100 and 50 / mm 3 in blood plasma.
  • a high CD4+ T cell count includes a CD4+ T cell count above 350 / mm 3 in blood plasma, which includes above 350, 400; 450; 500; 550, 600, 650 and 700 / mm 3 in blood plasma.
  • a "low HIV reservoir” may in particular correspond to a low PBMC-associated HIV DNA level, which includes below 1000 copies per million of Peripheral Blood Mononuclear Cells (PBMCs).
  • PBMCs Peripheral Blood Mononuclear Cells
  • below 1000 copies per million of PBMCs encompasses below 500, 100, 50, 10, 5, and 1 HIV DNA copies per million of Peripheral Blood Mononuclear Cells (PBMCs).
  • a low PBMC-associated HIV DNA level may thus consist of an HIV DNA level below 1000 HIV DNA copies per million of PBMCs; which includes below 500, 450, 400, 350, 300, 250, 200, 150, 100, 50, 40, 30, 20, 10, 9, 8, 7, 6, 5, 4, 3, 2, and 1 copie(s) per million of PBMCs.
  • a high PBMC-associated HIV DNA level includes above 1000 HIV DNA copies per million of Peripheral Blood Mononuclear Cells (PBMCs); which includes above 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, 10000, 50000 and 100 000 HIV DNA copies per million of PBMCs.
  • PBMCs Peripheral Blood Mononuclear Cells
  • PBMCs are known in the Art and refer to blood cells having a round nucleus, which includes lymphocytes (including T cells), monocytes and macrophages.
  • Protocols for isolating PBMCs are known in the Art.
  • PBMCs are separated from whole blood on FicoU-Hypaque so as to obtain a PBMC extract; the amount of total DNA is then quantified by real-time PCR from said PBMC extract.
  • the PBMC-associated HIV DNA level is preferably determined according to the protocols disclosed in Rouzioux et al. ("Quantification of total HIV1- DNA in peripheral blood mononuclear cells"; Methods Mol. Biol; 1087:261-70; 2014), and as further detailed in the Material & Methods section.
  • HIV-DNA levels are then reported in copies per million of PBMCs for total
  • the HIV-infected individuals in the course of an anti-retroviral treatment interruption has a low or decreased viral load, optionally below 40 HIV RNA copies / mL of blood plasma, at the time of the administration of said immunogenic compound.
  • the HIV-infected individuals in the course of an anti-retroviral treatment interruption has a low or decreased Peripheral Blood Mononuclear Cells (PBMC)-associated HIV DNA level, optionally below 1000 copies per 10 6 PBMCs, at the time of the administration of said immunogenic compound.
  • PBMC Peripheral Blood Mononuclear Cells
  • the HIV-infected individuals in the course of an anti-retroviral treatment interruption has a high CD4+ cell count, optionally above 350 per mm 3 of blood plasma at the time of the administration of said immunogenic compound.
  • HIV-infected individuals in the course of anti-retro viral treatment interruption are characterized by:
  • HIV-infected individuals in the course of anti-retro viral treatment interruption are characterized by:
  • PBMC Peripheral Blood Mononuclear Cells
  • Immunogenic compounds of the invention consist of compounds comprising an antigenic peptide derived from the gp41 protein of a HIV virus.
  • the antigenic peptide is a peptide derived from the gp41 protein and located between the N-terminal heptad repeat 1 (HR1) and the HR2 regions.
  • the peptide sequence SEQ ID N°l is provided, which corresponds to an antigenic peptide fragment of gp41 located between the N-terminal heptad repeat 1 (HR1) and the HR2 regions.
  • the antigenic peptide is a peptide derived from the gp41 protein, and comprising at least one fragment of gp41 derived from the amino acid sequence SEQ ID N°l or 2; or of a functionally equivalent sequence thereof.
  • the immunogenic compound comprises an antigenic peptide derived from the gp41 protein of a HIV virus; said antigenic peptide comprising 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 (preferably consecutive) amino acids from the amino acid sequence SEQ ID N°l, or of a functionally equivalent sequence thereof.
  • the antigenic peptide is a peptide derived from the gp41 protein and located between the HRl and HR2 regions, and comprising at least 1, 2, 3, 4, 5 or 6 amino acids of the well-conserved 3S motif, also referred herein as the "SWSNKS" motif (SEQ ID N°2) when considering a reference gp41 protein isolated from HIV strains, including naturally-occurring and no n- naturally occuring HIV strains, such as R5 and X4 HIV-1 strains.
  • SWSNKS well-conserved 3S motif
  • antigenic peptides derived from the gp41 protein of strains obtained from HIV-infected patients are considered as functionally equivalent sequences in the sense of the invention, such as the ones disclosed in Curriu et al. ( « Viremic HIV infected individuals with high CD4 T cells and functional envelope proteins show anti-gp41 antibodies with unique specificity and function; PLoS one ;7(2) ; 2012).
  • the antigenic peptide derived from the gp41 protein of a HIV virus comprises the amino acid sequence:
  • - X 2 is selected from the group consisting of W (Tryptophane) and A (Alanine),
  • - X3 is selected from the group consisting of K (Lysine) and R (Arginine),
  • the antigenic peptide derived from the gp41 protein of a HIV virus comprises at least 6 consecutive amino acids derived from the said gp41 protein, which includes at least 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50 consecutive amino acids derived from the said gp41 protein, or of a functionally equivalent sequence thereof.
  • the antigenic peptide derived from the gp41 protein of a HIV virus comprises from 5 to 100 amino acids of the said gp41 protein; which includes 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67?
  • the antigenic peptide derived from the gp41 protein of a HIV virus as defined above is preferably at least 6 amino acids long.
  • Immunogenic compounds suitable for the invention are notably disclosed in WO2004/070385, WO2005/076001, WO 2012140620 and WO2013/179262.
  • the immunogenic compound is a compound as defined above, wherein the said antigenic peptide derived from gp41 is of the formula (I) :
  • PepNt consists of a polypeptide having an amino acid length varying from 0 to
  • - CORE consists of a polypeptide comprising the amino acid sequence:
  • - X 2 is selected from the group consisting of W (Tryptophane) and A (Alanine),
  • - X3 is selected from the group consisting of K (Lysine) and R (Arginine),
  • - X 4 is selected from the group consisting of S (Serine) and T (Threonine), and
  • PepCt consists of a polypeptide having an amino acid length varying from 0 to 20 amino acid residues and located at the C-terminal end of the polypeptide of formula (I).
  • the antigenic peptide derived from the gp41 protein of a HIV virus, as defined above, or alternatively the CORE of said antigenic peptide of formula (I), comprises the amino acid sequence selected from:
  • NH2-SWSNKA- COOH SEQ ID N° 6
  • « PepNt » consists of a peptide having from 1 to 10 amino acid residues in length, which includes from 1 to 5 amino acid residues in length.
  • Nt is a peptide having 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acid residues in length.
  • « PepNt » comprises or consists of the amino acid sequence NH2-PWNA-COOH (SEQ ID N°7).
  • « PepCt » consists of a peptide having from 1 to 10 amino acid residues in length, which includes from 1 to 5 amino acid residues in length.
  • Ct is a peptide having 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acid residues in length. In some embodiments, Ct has 5 or 6 amino acid residues in length.
  • « PepCt » comprises or consists of the amino acid sequence NH2-LDDIW-COOH (SEQ ID N° 8).
  • the said antigenic peptide is of the following formula (II) : NH 2 - [Nt]y-P-W-N-Xi-S-X 2 -S-N-X3 -X4-X5 -X6-X7-I-W-[Ct] z -COOH (II), wherein:
  • - y is an integer meaning 0 or 1
  • - Nt consists of a peptide having from 1 to 16 amino acids in length
  • - Ct consists of a peptide having from 1 to 15 amino acids in length
  • - Xi is an amino acid selected from the group consisting of A (Alanine), T (threonine), S (Serine) and N (Asparagine),
  • - X 2 is an amino acid selected from the group consisting of W (Tryptophane) and A (Alanine),
  • - X3 is selected from the group consisting of K (Lysine) and R (Arginine),
  • - X 4 is selected from the group consisting of S (Serine) and T (Threonine),
  • - X5 is selected from the group consisting of L (Leucine), Y (Tyrosine) and Q
  • Glutamine - X 6 is selected from the group consisting of D (Aspartic acid), N (Asparagine), E (Glutamic acid), S (Serine), G (Glycine) and K (Lysine),
  • - X 7 is selected from the group consisting of D (Aspartic acid), Q (Glutamine), L (Leucine), A (Alanine), K (Lysine) and E (Glutamic acid).
  • the antigenic peptide derived from gp41 may comprise at least one fragment of any one of any one of 3S regions of gp41 disclosed in Curriu et al. ("Viremic HIV infected individuals with high CD4 T cells and functional envelope proteins show anti-gp41 antibodies with unique specificity and function"; PLoS one ;7(2) ; 2012), especially in Table 1 and Figure 3A of Curriu.
  • the antigenic peptide may be selected from the group consisting of:
  • - m is an integer meaning 0 or 1
  • - n is an integer meaning 0 or 1
  • - Al is an amino acid residue
  • - A2 is an amino acid residue.
  • the antigenic peptide is selected from the group consisting of the following formulae (Ilia) and (Illb) :
  • - m is an integer meaning 0 or 1
  • - n is an integer meaning 0 or 1
  • - Al is an amino acid residue
  • - A2 is an amino acid residue.
  • the immunogenic compound further includes amino acids which are not derived from gp41.
  • the C- and/or N-terminal ends of a peptide of formula (I) could deviate from the natural sequences expressly specified herein by modification of the terminal NH 2 - group and/or COOH-group and/or by modification of a NH 2 group and/or a COOH group of a lateral chain of an amino acid residue contained therein.
  • These groups may for instance be acylated, acetylated, amidated or modified to provide a binding site for a carrier molecule.
  • amino acids may be added to the N-terminal end of the antigenic peptide such as cysteines.
  • the immunogenic compound comprises an antigenic peptide selected from the group consisting of
  • C-PWNASASNKSLDDIW (SEQ ID NO 10)
  • SEQ ID NO 10 The 3S peptide of SEQ ID N°9 was previously identified as a candidate anti- HIV antigen by Vieillard et al. (Vieillard et al., 2008, PNAS, Vol. 105 (6) : 2100-2104).
  • any one of the above-mentioned antigenic peptides is linked to a carrier molecule.
  • carrier molecules used for generating an immunogenic product comprising a polypeptide of formula (I) linked to a carrier molecule are well in the general knowledge of the one skilled in the art.
  • the function of the carrier molecule is to provide cytokine help (or T-cell help) in order to enhance the immune response against HIV- 1.
  • the carrier molecule to which the peptide is optionally bound can be selected from a wide variety of known carriers.
  • carrier molecules for vaccine purposes encompass proteins such as human or bovine serum albumin and keyhole limpet haemocyanin (KLH) and fatty acids.
  • KLH keyhole limpet haemocyanin
  • Other embodiments of carrier molecules to which an antigenic peptide of formula (I) may be covalently linked include bacterial toxins or toxoids, such as diphtheria, cholera, E. coli heat labile or tetanus toxoids, the N.
  • meningitidis outer membrane protein European patent application n° EP0372501
  • synthetic peptides European patent applications n° EP0378881 and n° EP0427347
  • heat shock proteins PCT application n° W093/17712
  • Pertussis proteins PCT application n° W098/58668
  • protein D from H. influenzae PCT application n° WO00/56360.
  • toxin A or B from C. difficile International patent application WO00/61761.
  • the carrier molecule is an amine- containing carrier protein.
  • the antigenic peptide is covalently bound to the carrier molecule by its N-terminal end amino acid residue.
  • the carrier molecule is a CRM197 protein, of sequence SEQ ID N° 11, as described for instance in WO2013/179262.
  • the CRM 197 protein consists of a non- toxic mutant of the well- known diphtheria toxin, which mutant was initially described by Uchida et al. (1973, J. Biol. Chem., Vol. 248 : 3838-3844).
  • the CRM197 mutant protein was initially described as the translation product of the mutant tox97 gene where a G— »A transition led to the substitution of the glycine (G) residue at position 52 of the wild-type diphtheria toxin with a glutamic acid residue (E).
  • the amount of antigenic peptide linked to one carrier molecule is measured preferably by Amino Acid Analysis.
  • This method is the methodology conventionally used to determine the amino acid composition of proteins. Proteins are macromolecules consisting of covalently bonded amino acid residues organized as a linear polymer. The peptide bonds are broken upon incubation under acid condition leading to the release of amino acids. An amino acid analysis is then performed on the product of the hydrolysis.
  • Amino Acid Analysis is preferably used to determine the rate of coupling of the antigenic peptide on a carrier molecule such as CRM197. This was possible since some amino acids may be both present on the carrier molecule and the grafted peptides and others may be only present on the carrier molecule. Based on the results of the amino acids present, a calculation allowed to determine the coupling ratio of the peptide of formula (I) onto CRM 197.
  • RP-HPLC reverse phase high pressure liquid chromatography
  • this instrument has a pre- or post-column derivatization capability and the detector is an ultraviolet-visible or fluorescence detector depending on the derivatization method used.
  • An integrator is used for the transforming the analog signal from the detector and for quantitation of each amino acid.
  • the quantification method of reference herein of the peptide-protein conjugates is performed by integration of the monomer peak at 215 nm obtained by isocratic SE-HPLC in 50 mM phosphate buffer pH6.8 using a Phenomenex column (BioSep SEC S2000) at a defined flow.
  • the determination of the protein content can also be made by other methods such as BCA assay using the BCA Protein Assay Kit (Thermo Scientific), which is not a reference method herein.
  • This kit is a detergent-compatible bicinchoninic acid formulation for the colorimetric detection and quantification of total protein.
  • BSA is used as the reference material in the protein determination.
  • Absorbance at 562nm is linear with increasing protein concentrations. The inventors have shown that quantifying the peptide- protein conjugates by this method lead to a reduced quantification value in respect to the reference method described above, i.e. a value of around 0.8 times the quantification value found with the reference method described above.
  • the said antigenic peptide is covalently bound to the carrier molecule through a linker moiety.
  • the said restricted family of linker agents encompasses, or even consists of, the linker agents named GMBS, sulfo-GMBS, SMPB and sulfo-SMPB.
  • the said linker agent is selected form the group consisting of GMBS ( ⁇ -[ ⁇ - maleimidobutyryl-oxy]succinimide ester), Sulfo-GMBS (N-[Y-maleimidobutyryl- oxy]sulfosuccinimide ester), SMPB (succinimidyl 4-[/?-maleimidophenyi]butyrate) and Sulfo-SMPB (sulfo succinimidyl 4-[/?-maleimidophenyl]butyrate).
  • GMBS ⁇ -[ ⁇ - maleimidobutyryl-oxy]succinimide ester
  • Sulfo-GMBS N-[Y-maleimidobutyryl- oxy]sulfosuccinimide ester
  • SMPB succinimidyl 4-[/?-maleimidophenyi]butyrate
  • Sulfo-SMPB sulfo succinimidyl 4-[/?-maleimidoph
  • GMBS N- hydroxysuccinimide
  • Sulfo-GMBS Sulfo-GMBS
  • SMPB Sulfo-SMPB
  • GMBS, Sulfo-GMBS, SMPB and Sulfo-SMPB consist of heterobifunctional linker agents that contain both a N- hydroxysuccinimide (NHS) ester group and a maleimide group.
  • NHS N- hydroxysuccinimide
  • Conjugation using GMBS, Sulfo-GMBS, SMPB or Sulfo-SMPB is usually performed by a two-step procedure.
  • the amine-containing protein e.g. CRM197
  • the linker agent e.g. a several-fold molar excess of the linker agent at pH 7-9 to form amide bonds, followed by removal of excess non-reacted linker agent, usually by desalting or dialysis.
  • the sulfhydryl-containing molecule e.g. peptide of formula (I)
  • the maleimide groups already attached to the first protein e.g. free maleimide groups of the linker chain that is already covalently linked to CRM197
  • SMPB or Sulfo-SMPB as linker agents for covalently linking peptides of formula (I) to the amine-containing carrier protein, in particular the CRM 197 carrier protein, leads to a conjugate of formula (VII) below:
  • - Rl consists of one reactive group of the amine-containing carrier protein, and wherein the NH group attached thereto derives from (i) the alpha amino group located at the N-terminal end of the amine-containing carrier protein or (ii) a lateral chain amino group from a Lysine (K) amino acid residue of the amine-containing carrier protein.
  • - R2 consists of a peptide of formula (I), and wherein the sulphur (S) atom attached thereto derives from a sulfhydryl (SH) group of a cysteine residue located at the N-terminal end or at the C-terminal end of a peptide of formula (I).
  • S sulphur
  • SH sulfhydryl
  • the sulfhydryl moiety could be part of an unnatural amino acid, or any other molecule present at the end of the peptide of formula (I).
  • GMBS or Sulfo-GMBS as linker agents for covalently linking peptides of formula (I) to the amine-containing carrier protein, in particular the CRM 197 carrier, protein leads to a conjugate of formula (VIII) below:
  • - Rl consists of one reactive group of the amine-containing carrier protein, and wherein the NH group attached thereto derives from (i) the alpha amino group located at the N-terminal end of the amine-containing carrier proteinor (ii) a lateral chain amino group from a Lysine (K) amino acid residue of the amine-containing carrier protein.
  • - R2 consists of a peptide of formula (I), and wherein the sulphur (S) atom attached thereto derives from a sulfhydryl (SH) group of a cysteine residue located at the N-terminal end or at the C-terminal end of a peptide of formula (I).
  • S sulphur
  • SH sulfhydryl
  • the sulfhydryl moiety could be part of an unnatural amino acid, or any other molecule present at the end of the peptide of formula (I).
  • an amine-containing carrier protein such as the CRM197 protein, comprises a plurality of reactive groups Rl, so that a plurality of peptides of formula (I) may be linked to CRM 197 in a conjugate of formula (VII) or (VIII).
  • an immunogenic compound as defined above are those wherein a plurality of reactive groups of the amine-containing carrier protein, in particular CRM 197, are covalently linked to an antigenic peptide, which peptide generally possesses a cysteine residue at its N-terminal end, according to the covalent linkage represented by formula (VII) or (VIII) above.
  • a mean number of antigenic peptides ranging from 2 to 20 are covalently linked to one molecule of CRM 197.
  • a mean number of from 5 to 10 antigenic peptides, which includes a mean number of from 7-8 peptides of formula (I) are covalently linked to one molecule of CRM 197.
  • This invention also relates to compositions comprising an immunogenic compound as defined above, in combination with one or more immuno adjuvant substances.
  • composition as defined herein which comprises an immunogenic compound as defined above, and which further comprises one or more immuno -adjuvant substances may also be termed an "immunogenic composition” or alternatively a “vaccine composition” in the present specification.
  • an immunogenic composition according to the invention there is no substantial distinction to be made between an immunogenic composition according to the invention and a vaccine composition according to the invention, beyond the terms employed to designate such compositions, excepted that the features of the vaccine composition shall comply with the technical requirements of the various drug agencies for the grant of marketing authorizations for human or veterinary use.
  • an immunogenic composition according to the invention may not comply to the requirements of drug agencies while being usable for administration to animals, e.g. for producing antibodies in a given individual, wherein the generated antibodies are expected to exert a preventive or a therapeutic effect in the HIV-infected individual.
  • compositions according to the invention encompass both (i) immunogenic compositions and (ii) vaccine compositions.
  • an immuno adjuvant may be selected form the group consisting of (i) mineral salts, (ii) emulsions, (iii) microbial natural or synthetic derivatives, (iv) combination adjuvants, (v) cytokine-derived or accessory molecules- derived adjuvants, and (vi) particulate formulations.
  • emulsion-based immunoadjuvants are also selected from the group consisting of MontanideTM (SEPPIC) adjuvants, including Montanide ISA 51 which is a water- in-oil stabilized emulsion, and (4) ISA-720 which is a stabilized composition comprising water and squalene.
  • SEPPIC MontanideTM
  • ISA 51 which is a water- in-oil stabilized emulsion
  • ISA-720 which is a stabilized composition comprising water and squalene.
  • Immunogenic compositions of the invention preferably include a pharmaceutically acceptable carrier.
  • Suitable pharmaceutically acceptable carriers and/or diluents include any and all conventional solvents, dispersion media, fillers, solid carriers, aqueous solutions, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like.
  • Suitable pharmaceutically acceptable carriers include, for example, one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof.
  • Pharmaceutically acceptable carriers may further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody.
  • auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody.
  • the preparation and use of pharmaceutically acceptable carriers is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, use thereof in the immunogenic compositions of the present invention is contemplated.
  • Such immunogenic compounds, and compositions thereof can be administered parenterally, e.g., by injection, either by subcutaneous, intradermal or intramuscular route, as well as orally or intranasally.
  • Other modes of administration employ oral formulations, pulmonary formulations, suppositories, and transdermal applications, for example, without limitation.
  • Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like, without limitation.
  • the immunogenic compounds, and compositions thereof are administered parenterally, and most preferably intramuscularly.
  • an amount of antigenic peptide is expressed as "antigenic peptide equivalent" , which consists of the amount of antigenic peptide that is contained in the considered immunogenic compound material.
  • the antigenic peptide equivalent when the antigenic peptide is coupled to a carrier molecule, the antigenic peptide equivalent only corresponds to the amount of antigenic peptide present and does not include the amount of other ingredients that are effectively administered.
  • an immunogenic compound, or composition thereof, used according to the invention comprises an antigenic peptide of the invention in an amount which is adapted to the administration of from 10 ng to 10 mg of the said antigenic peptide to an individual in need thereof, when expressed as an antigenic peptide equivalent.
  • an equivalent amount of an antigenic peptide of from 10 ng to 10 mg encompasses an amount of peptide of about 20 ng, 30 ng, 40 ng, 50 ng, 60 ng, 70 ng, 80 ng, 90 ng, 100 ng, 150 ng, 200 ng, 250 ng, 300 ng, 350 ng, 400 ng, 450 ng, 500 ng, 550 ng, 600 ng, 700 ng, 800 ng, 900 ng, 1 ⁇ g, 2 ⁇ g, 3 ⁇ g, 4 ⁇ g, 5 ⁇ g, 6 ⁇ g, 7 ⁇ g, 8 ⁇ g, 9 ⁇ g, 10 ⁇ g, 20 ⁇ g, 30 ⁇ g, 40 ⁇ g, 50 ⁇ g, 60 ⁇ g, 70 ⁇ g, 80 ⁇ g, 90 ⁇ g, 100 ⁇ g, 110 ⁇ g, 120 ⁇ g, 130 ⁇ g, 140 ⁇ g, 150 ⁇ g, 160 ⁇ g, 170 ⁇ g, 180 ⁇
  • the antigenic peptide is administered in an equivalent amount ranging from 1 ⁇ g to 80 ⁇ g.
  • the equivalent amount of antigenic peptide may be of about 1 ⁇ g, 5 ⁇ g, 10 ⁇ g, 20 ⁇ g, 30 ⁇ g, 40 ⁇ g, 50 ⁇ g, 60 ⁇ g, 70 ⁇ g, or 80 ⁇ g.
  • an immunogenic compound, or composition thereof, used according to the invention comprises an antigenic peptide of the invention in an equivalent amount which is adapted to multiple parenteral administrations, which includes 1, 2, 3, 4, 5 and 6 administrations, and preferably 6 doses per year.
  • the first cycle is composed of three administrations every 4 weeks.
  • the immunogenic compound, or composition thereof is administered, as a booster injection, about every 12 weeks after the first cycle, which includes being administered, as a booster injection, in a period ranging from 8 to 48 weeks after the latest administration, which includes 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47 and 48 weeks after.
  • the immunogenic compound is administered parenterally, for example intramuscularly, in 3 to 6 administrations, and preferably 6 doses within a year, wherein the fourth, fifth and sixth administration is administered, as a booster injection, in a period ranging from 8 to 48 weeks after the latest administration.
  • the immunogenic compound may also be administered in combinattion with other compounds, including Latency Reversing Agents (LRA) or other compounds that have either a shock, or a kill effect, and compounds behaving as immune boost
  • LRA Latency Reversing Agents
  • the immunogenic compound may be further administered in combination with
  • Latency Reversing Agents such as agents can be selected in a group consisting of histone deacetylase inhibitors (HDACi), including romidepsin, panobinostat, vorinostat, givnostat, belinostat, sirtuin inhibitors, NF-KB-inducing agents, protein kinase C agonists, T cell activators and TLR agonists, immune checkpoint inhibitors selected in a group comprising PD-1 inhibitors and PDL-1 inhibitors (including Pembrolizumab, Nivolumab, Pidilizumab, BMS 936559, MPDL3280A), LAG-3 inhibitors, TIGIT inhibitors, and CTLA-4 inhibitors, pro-apoptotic and cell differentiating molecules including JQl, Nutlin3, Disulfiram, aphidicolin, Wnt small molecules inhibitors and Notch inhibitors.
  • HDACi histone deacetylase inhibitors
  • romidepsin including
  • the immunogenic compound may also be administered in combination withat least one drug selected from one or more immunotherapy inducing an HIV-antigen specific immune response with either a Thl and or a Th2 type immune response, and other immunotherapies acting more as immune boost and including IL-15, IL-7 or revlimid could be also used in synergy.
  • Example 1 Preparation of an immunogenic compound derived from gp41, and compositions thereof.
  • the following immunogenic compounds or conjugates were synthesized. There were derived from KLH and CRM197 using either MBS or SMPB as crosslinker molecules.
  • the used peptide was the 3S peptide consisting of SEQ ID N°2 with either an additional cysteine residue at its amino-terminus end or at its carboxy-terminus end.
  • the peptide which is termed "Nter(Cys)-3S" above comprises the 3S reference peptide of SEQ ID N°2 herein.
  • sulfo-SMPB Sulfo- (Succinimidyl-4-(p-maleimidophenyl) Butyrate)
  • sulfo-MBS Sulfo-(m- Maleimidobenzoyl-N-hydroxysuccinimide) ester
  • These molecules consist of a maleimide moiety linked by a polyethylene chain to an ester of N-hydroxysuccinimide (Cross-linking of protein by w-maleimido alkanoyl N-hydroxysuccinimido esters. Partis M.D and al. Journal of Protein Chemistry, vol.2, No 3, 1983).
  • the succinimide moiety can react with amino groups of the protein. Once this reaction has occurred, the maleimide moiety reacts with sulfhydryl groups of the 3S peptides. They are different in length, 7,3 A for sulfo-
  • the coupling reaction was a two-step reaction.
  • the first step was the activation of the CRM 197 with the cross-linker. 15 milligrams of linker, diluted in dimethyl sulfoxide were added to 20 milligrams of CRM 197 in a volume of 5-20 mL of conjugation buffer (PBS 10 mM pH 7-pH 7.4) and mixed gently for 30-90 min at room temperature (Protective immunogenicity of two synthetic peptides selected from the amino acid sequence of Bordetella pertussis toxin subunit SI. Askelof P. and al. PNAS, vol.87, pp 1347-1351, February 1990).
  • conjugation buffer PBS 10 mM pH 7-pH 7.4
  • the immuno-conjugates were then purified by size exclusion chromatography.
  • the immuno-conjugates were analyzed using an amino acid analysis (AAA) to determine the peptide/CRM197 ratio.
  • AAA amino acid analysis
  • the CRM197-3S peptide was lyophilized with a lyoprotector (Lyophilisation and development of solid protein pharmaceuticals. Wang W. International Journal of Pharmaceutics 203 (2000) 1- 60; Fundamentals of freeze-drying. Nail S.L and al. Pharm Biotechnol. 2002; 14:281-360).
  • AAA amino acid analysis
  • VAC-3S is a sterile suspension for intramuscular injection containing the 3S drug substance adsorbed on aluminium hydroxide in buffered isotonic saline. The manufacturing of VAC-3S was performed in compliance with the GMP.
  • the 3S drug substance is formulated at concentrations ranging from 1 to 40 ⁇ g/mL of 3S16Nter peptide equivalent with aluminium hydroxide (1 mg/mL of Al 3+ ions) provided by Brenntag (Alhydrogel 85 2%-Ph Eur), 150 mM sodium chloride (European pharmacopoeia) and 1 mM sodium phosphate (European pharmacopoeia). Products for injection are used for the formulation of the vaccine. The final pH is at 6.8. VAC-3S contains no preservative.
  • the vaccine After shaking, the vaccine is a homogeneous white suspension ready to use. If needed to achieve the needed concentration of 3S16Nter the vaccine is diluted in aluminium hydroxide (1 mg/mL of Al 3+ ions) provided by Brenntag (Alhydrogel 85 2%- Ph Eur), 150 mM sodium chloride (European pharmacopoeia) and 1 mM sodium phosphate (European pharmacopoeia), equilibrated at pH 6.8. The vaccine could be injected intramuscularly in the deltoid. A sterile syringe with sterile needle is used for injection. Patients should receive at least 6 doses of 0.5 mL each, with a minimum interval of 2 to 4 weeks between vaccinations.
  • Example 2 Preparation of an immunogenic compound derived from gp41, and compositions thereof, for clinical use.
  • the following immunogenic compound or conjugate was synthesized. It was derived from CRM197 using SMPB as crosslinker molecule (as shown in example 1).
  • the used peptide was a mutated 3S (m3S) peptide consisting of SEQ ID N°13 (NH 2 - PWNASASNKSLDDIW-COOH) with an additional cysteine residue at its amino- terminus end to allow the chemical coupling the cross linker leading to CRM197-SMPB- Nter(Cys)-m3S
  • the peptide which is termed "Nter(Cys)-m3S" above consists of the 3S peptide of SEQ ID N°14 herein.
  • the heterobifunctional cross-linker sulfo-SMPB Sulfo-(Succinimidyl-4-(p- maleimidophenyl) Butyrate) was used.
  • These molecules consist of a maleimide moiety linked by a polyethylene chain to an ester of N-hydroxysuccinimide (Cross-linking of protein by w-maleimido alkanoyl N-hydroxysuccinimido esters. Partis M.D and al. Journal of Protein Chemistry, vol.2, No 3, 1983).
  • the succinimide moiety can react with amino groups of the protein. Once this reaction has occurred, the maleimide moiety reacts with sulfhydryl groups of the 3S peptides. They are different in length, 7,3 A for sulfo-
  • the coupling reaction was a two-step reaction.
  • the first step was the activation of the CRM 197 with the cross-linker. 15 milligrams of linker, diluted in dimethyl sulfoxide were added to 20 milligrams of CRM 197 in a volume of 5-20 ml of conjugation buffer (PBS 10 mM pH7-pH7.4) and mixed gently for 30-90 min at room temperature (Protective immunogenicity of two synthetic peptides selected from the amino acid sequence of Bordetella pertussis toxin subunit SI. Askelof P. and al. PNAS, vol.87, pp 1347-1351, February 1990).
  • VAC-3S is a sterile suspension for intramuscular injection containing the 3S drug substance adsorbed on aluminium hydroxide in buffered isotonic saline.
  • the manufacturing of VAC-3S was performed in compliance with the GMP.
  • the 3S drug substance is formulated at the concentration of 0.02 mg/mL of 3S16Nter peptide, comprising the antigenic peptide of sequence SEQ ID N°9, equivalent in 0.5 mL with aluminium hydroxide (1 mg/mL of Al 3+ ions) provided by Brenntag (Alhydrogel 85 2 -Ph Eur), 150 mM sodium chloride (European pharmacopoeia) and 1 mM sodium phosphate (European pharmacopoeia). Products for injection are used for the formulation of the vaccine. The final pH is at 6.8. VAC-3S contains no preservative.
  • the vaccine After shaking, the vaccine is a homogeneous white suspension ready to use.
  • the vaccine could be injected intramuscularly in the deltoid.
  • a sterile syringe with sterile needle is used for injection.
  • Patients should receive at least 6 doses of 0.5 mL each, with a minimum interval of 2 to 4 weeks between vaccinations.
  • the four patient populations are further detailed in Table 1 here below.
  • the administration protocol is also detailed in figures 1A and IB.
  • N 8; 7 M / 1 F for modified As Treated (mA r ) population ( patient replaced after 1 vaccination).
  • Example 3 Assessment of the dose-dependent immunogenicity at week 12 based on the amount of immunogenic compound that is administered.
  • the ELISA assay was designed to perform the measurement of total Ig antibodies that would recognize the peptides of SEQ ID N°9, also called anti-3S peptide antibodies.
  • the anti-3S IgG antibody titers were determined by an Enzyme-Linked Immunosorbent Assay (ELISA).
  • the antigen coated to the Nunc Maxisorp micro plates is a 3S peptide conjugated to bovine serum albumin (BSA) with a different linker than the one used for the synthesis of the immuno-conjugates: SMCC (succinimidyl-4-(N- maleimidomethyl)cyclohexane-l-carboxylate) (produced from Imject® Maleimide Activated BSA Protein Kits purchased from Thermo Fisher Scientific, Waltham, USA) .
  • BSA bovine serum albumin
  • the anti-m3S IgG antibodies are revealed by a colorimetric reaction using a goat anti- mouse IgG (Fc), conjugated to the HorseRadish Peroxydase (HRP) (Jackson Immunoresearch, West Grove, USA), and the HRP substrate: the tetramethylbenzidine (TMB) (Sigma, Missouri, USA).
  • Fc goat anti- mouse IgG
  • HRP substrate the tetramethylbenzidine
  • Results are provided in figure 2, which show that the percentage of responsive patients increases with a higher amounts of immunogenic compounds, at week 12.
  • the HIV DNA level is measured using the protocols described in Rouzioux et al. ("Quantification of total HIV 1 -DNA in peripheral blood mononuclear cells"; Methods Mol. Biol; 1087:261-70; 2014).
  • the HIV DNA level can be expressed either in log copies per 10 6 PBMC cells or in log copies per mL of whole blood. 1.1 Isolation of total DNA from patient EDTA blood samples.
  • PBMC pellets are stored at -80°C until use.
  • the total DNA is isolated from PBMC (Peripheral Blood Mononuclear Cells).
  • PBMC pellets are prepared by Ficoll-Hypaque gradient, while cell pellets are obtained from whole blood after centrifugation (2,500 rpm, 10 min) and plasma decantation. Purified CD4+ T cells, cell pellet, or even whole blood can be used.
  • Total DNA is extracted from PBMCs using a QIAamp DNA mini kit (QIAGEN, Courtaboeuf, France) according to the manufacturer's instructions, to obtain 100 ⁇ _, of eluate.
  • the kit NucleoSpin® Blood (Macherey-Nagel) is preferred for DNA extraction from cell pellets and whole blood samples.
  • the protocol is set up as described previously.
  • Figure 3 provides evidence of a decrease in the HIV-DNA levels in the responder group at weeks 60 and 84, which thus provides evidence of a decrease in the HIV latent reservoirs.
  • This link is confirmed by Figures 5A, 5B, 5C and 5D showing at the 12, 36, 60 and 84 weeks an inverse correlation between the level of anti-3S antibodies in patients participating to the trial and level of HIV DNA change from baseline (in Log of copies per million of PBMC)..
  • FIG. 5A, 5B, 5C and 5D showing at the 12, 36, 60 and 84 weeks an inverse correlation between the level of anti-3S antibodies in patients participating to the trial and level of HIV DNA change from baseline (in Log of copies per million of PBMC).
  • Figures 6 and 7 also reports a significant increase of the CD4/CD8 ratio in Responders and not the non-Responders and Placebo groups, at week 24 and thus immune restoration since an increase of the CD4/CD8 ratio corresponds to improved immune functions (Serrano-Villar et ah, PLOS Pathogens, vol. 10, 2014).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Virology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Biophysics (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Veterinary Medicine (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne un composé immunogène comprenant un peptide antigénique dérivé de la protéine gp41 du VIH, et destiné à être utilisé dans le traitement d'un patient souffrant d'une infection par le VIH, soumis à un traitement anti-rétroviral et au cours de l'interruption du traitement anti-rétroviral.
PCT/EP2016/061273 2015-05-19 2016-05-19 Traitement de patients souffrant d'une infection par le vih WO2016184963A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP15305751 2015-05-19
EPEP15305751 2015-05-19

Publications (1)

Publication Number Publication Date
WO2016184963A1 true WO2016184963A1 (fr) 2016-11-24

Family

ID=53189758

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2016/061273 WO2016184963A1 (fr) 2015-05-19 2016-05-19 Traitement de patients souffrant d'une infection par le vih

Country Status (1)

Country Link
WO (1) WO2016184963A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
WO2022180263A1 (fr) 2021-02-26 2022-09-01 Curavac Europe Protéine porteuse pour antigene peptidique
WO2022180262A1 (fr) 2021-02-26 2022-09-01 Curavac Europe Vaccin pour un traitement therapeutique ou prophylactique de la myastenie grave
WO2023280833A1 (fr) * 2021-07-05 2023-01-12 Diaccurate Nouveaux antigènes et vaccins

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0372501A2 (fr) 1988-12-07 1990-06-13 BEHRINGWERKE Aktiengesellschaft Antigènes synthétiques, procédé pour leur préparation et leur utilisation
EP0378881A1 (fr) 1989-01-17 1990-07-25 ENIRICERCHE S.p.A. Peptides synthétiques et leur utilisation comme support universel pour la préparation de conjugués immunogènes convenant au développement de vaccins synthétiques
EP0427347A1 (fr) 1989-11-10 1991-05-15 ENIRICERCHE S.p.A. Peptides synthétiques utiles comme porteurs universels pour la préparation des conjugués immunogéniques el leur emploi dans le développment des vaccins synthétiques
WO1993017712A2 (fr) 1992-03-06 1993-09-16 Biocine Spa Composes conjugues obtenus a partir de proteines du choc thermique et d'oligosaccharides ou de polysaccharides
WO1998058668A2 (fr) 1997-06-20 1998-12-30 Microbiological Research Authority VACCIN CONTENANT UN ANTIGENE DE $i(BORDETELLA PERTUSSIS)
WO2000056360A2 (fr) 1999-03-19 2000-09-28 Smithkline Beecham Biologicals S.A. Vaccin
WO2002058726A1 (fr) * 2001-01-26 2002-08-01 The Immune Response Corporation Procede de traitement d'un individu infecte par le vih en associant l'immunisation a une interruption structuree du traitement antiretroviral
WO2004070385A1 (fr) 2003-02-06 2004-08-19 Institut National De La Sante Et De La Recherche Medicale (Inserm) Procede pour evaluer in vitro l'etat de progression d'une infection par le virus vih chez un individu
WO2005076001A2 (fr) 2004-02-06 2005-08-18 Inserm (Institut National De La Sante Et De La Recherche Medicale) Polypeptide derive de gp41, composition de vaccin comprenant ce polypeptide et utilisations de celle-ci pour traiter une personne infectee par un virus vih
WO2012140620A1 (fr) 2011-04-15 2012-10-18 Institut National De La Sante Et De La Recheche Medicale Compositions destinées au traitement prophylactique et/ou thérapeutique d'une infection par un virus vih-1
WO2013179262A1 (fr) 2012-05-31 2013-12-05 Innavirvax Composés immunogènes comprenant le peptide gp41 du vih couplé à la protéine porteuse crm197

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0372501A2 (fr) 1988-12-07 1990-06-13 BEHRINGWERKE Aktiengesellschaft Antigènes synthétiques, procédé pour leur préparation et leur utilisation
EP0378881A1 (fr) 1989-01-17 1990-07-25 ENIRICERCHE S.p.A. Peptides synthétiques et leur utilisation comme support universel pour la préparation de conjugués immunogènes convenant au développement de vaccins synthétiques
EP0427347A1 (fr) 1989-11-10 1991-05-15 ENIRICERCHE S.p.A. Peptides synthétiques utiles comme porteurs universels pour la préparation des conjugués immunogéniques el leur emploi dans le développment des vaccins synthétiques
WO1993017712A2 (fr) 1992-03-06 1993-09-16 Biocine Spa Composes conjugues obtenus a partir de proteines du choc thermique et d'oligosaccharides ou de polysaccharides
WO1998058668A2 (fr) 1997-06-20 1998-12-30 Microbiological Research Authority VACCIN CONTENANT UN ANTIGENE DE $i(BORDETELLA PERTUSSIS)
WO2000056360A2 (fr) 1999-03-19 2000-09-28 Smithkline Beecham Biologicals S.A. Vaccin
WO2002058726A1 (fr) * 2001-01-26 2002-08-01 The Immune Response Corporation Procede de traitement d'un individu infecte par le vih en associant l'immunisation a une interruption structuree du traitement antiretroviral
WO2004070385A1 (fr) 2003-02-06 2004-08-19 Institut National De La Sante Et De La Recherche Medicale (Inserm) Procede pour evaluer in vitro l'etat de progression d'une infection par le virus vih chez un individu
WO2005076001A2 (fr) 2004-02-06 2005-08-18 Inserm (Institut National De La Sante Et De La Recherche Medicale) Polypeptide derive de gp41, composition de vaccin comprenant ce polypeptide et utilisations de celle-ci pour traiter une personne infectee par un virus vih
WO2012140620A1 (fr) 2011-04-15 2012-10-18 Institut National De La Sante Et De La Recheche Medicale Compositions destinées au traitement prophylactique et/ou thérapeutique d'une infection par un virus vih-1
WO2013179262A1 (fr) 2012-05-31 2013-12-05 Innavirvax Composés immunogènes comprenant le peptide gp41 du vih couplé à la protéine porteuse crm197

Non-Patent Citations (22)

* Cited by examiner, † Cited by third party
Title
ALEXANDRA TRKOLA ET AL: "Delay of HIV-1 rebound after cessation of antiretroviral therapy through passive transfer of human neutralizing antibodies", NATURE MEDICINE, vol. 11, no. 6, 1 June 2005 (2005-06-01), pages 615 - 622, XP055003068, ISSN: 1078-8956, DOI: 10.1038/nm1244 *
ASKELOF P., PNAS, vol. 87, February 1990 (1990-02-01), pages 1347 - 1351
C. PETITDEMANGE ET AL: "A Single Amino-Acid Change in a Highly Conserved Motif of gp41 Elicits HIV-1 Neutralization and Protects Against CD4 Depletion", CLINICAL INFECTIOUS DISEASES, vol. 57, no. 5, 21 May 2013 (2013-05-21), US, pages 745 - 755, XP055223877, ISSN: 1058-4838, DOI: 10.1093/cid/cit335 *
CARDOSO ROSA M F ET AL: "Broadly neutralizing anti-HIV antibody 4E10 recognizes a helical conformation of a highly conserved fusion-associated motif in gp41", IMMUNITY, CELL PRESS, US, vol. 22, no. 2, 1 February 2005 (2005-02-01), pages 163 - 173, XP009104606, ISSN: 1074-7613, DOI: 10.1016/J.IMMUNI.2004.12.011 *
CURRIU ET AL.: "Viremic HIV infected individuals with high CD4 T cells and functional envelope proteins show anti-gp41 antibodies with unique specificity and function", PLOS ONE, vol. 7, no. 2, 2012, XP055226669, DOI: doi:10.1371/journal.pone.0030330
HILL ET AL., PNAS, vol. ILL, no. 37, 2014, pages 13475 - 13480
KATLAMA ET AL., LANCET, vol. 381, no. 9883, pages 2109 - 2117
KAY, TRENDS BIOTECHNOL, 2003, pages 420 - 3
MARTA CURRIU ET AL: "Viremic HIV Infected Individuals with High CD4 T Cells and Functional Envelope Proteins Show Anti-gp41 Antibodies with Unique Specificity and Function", PLOS ONE, vol. 7, no. 2, 1 February 2012 (2012-02-01), pages e30330, XP055226669, DOI: 10.1371/journal.pone.0030330 *
MATTSON G.: "A practical approach to crosslinking", MOLECULAR BIOLOGY REPORTS, vol. 17, 1993, pages 167 - 183, XP009033258, DOI: doi:10.1007/BF00986726
MATTSON G: "A practical approach to crosslinking", MOLECULAR BIOLOGY REPORTS, vol. 17, 1993, pages 167 - 183, XP009033258, DOI: doi:10.1007/BF00986726
NAHAS DD ET AL., BIOCONJ CHEM, vol. 19, no. 1, 12 December 2007 (2007-12-12), pages 322 - 6
NAIL S.L: "Fundamentals of freeze-drying", PHARM BIOTECHNOL., vol. 14, 2002, pages 281 - 360, XP001181137
PARTIS M.D: "Cross-linking of protein by w-maleimido alkanoyl N-hydroxysuccinimido esters", JOURNAL OF PROTEIN CHEMISTRY, vol. 2, no. 3, 1983, XP000563178, DOI: doi:10.1007/BF01025358
ROUZIOUX ET AL.: "Quantification of total HIV1-DNA in peripheral blood mononuclear cells", METHODS MOL. BIOL, vol. 1087, 2014, pages 261 - 70
SERRANO-VILLAR ET AL., PLOS PATHOGENS, vol. 10, 2014
SERRANO-VILLAR ET AL., PLOS PATHOGENS, vol. 10, no. 5, 2014
UCHIDA ET AL., J. BIOL. CHEM., vol. 248, 1973, pages 3838 - 3844
VIEILLARD ET AL., JAIDS, vol. 61, no. 3, 2012, pages 403 - 5
VIEILLARD ET AL., PNAS, vol. 105, no. 6, 2008, pages 2100 - 2104
VIEILLARD VINCENT ET AL: "A vaccine strategy against AIDS: an HIV gp41 peptide immunization prevents NKp44L expression and CD4+ T cell depletion in SHIV-infected macaques", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, NATIONAL ACADEMY OF SCIENCES, US, vol. 105, no. 6, 12 February 2008 (2008-02-12), pages 2100 - 2104, XP002510400, ISSN: 0027-8424, DOI: 10.1073/PNAS.0711629105 *
WANG W.: "Lyophilisation and development of solid protein pharmaceuticals", INTERNATIONAL JOURNAL OF PHARMACEUTICS, vol. 203, 2000, pages 1 - 60

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
US10385130B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US11535670B2 (en) 2016-05-11 2022-12-27 Huyabio International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
WO2022180263A1 (fr) 2021-02-26 2022-09-01 Curavac Europe Protéine porteuse pour antigene peptidique
WO2022180262A1 (fr) 2021-02-26 2022-09-01 Curavac Europe Vaccin pour un traitement therapeutique ou prophylactique de la myastenie grave
BE1029149B1 (fr) * 2021-02-26 2022-09-26 Nicolas Havelange Protéine porteuse pour antigene peptidique
BE1029148B1 (fr) * 2021-02-26 2022-09-26 Curavac Europe Vaccin pour un traitement therapeutique ou prophylactique de la myastenie grave
WO2023280833A1 (fr) * 2021-07-05 2023-01-12 Diaccurate Nouveaux antigènes et vaccins

Similar Documents

Publication Publication Date Title
US10335482B2 (en) Method of inducing an anti-HIV-1 immune response comprising administering a C5/TM-GP41 peptide dimer
WO2016184963A1 (fr) Traitement de patients souffrant d'une infection par le vih
AU2013269120B2 (en) Immunogenic compounds comprising HIV gp41 peptide coupled to CRM197 carrier protein
WO2016184962A1 (fr) Traitement de patients souffrant d'une infection par le vih
US8603965B2 (en) Pharmaceutical composition for the prophylaxis and treatment of HIV infection and its use
KR20100075483A (ko) 장산재 핵 요소 폴리펩티드 조성물 및 그것의 사용방법
JP2010506926A (ja) Ii−Key増強ワクチンの効力
WO2015007337A1 (fr) Procédé de vaccination contre le vih
WO2016184973A1 (fr) Traitement de patients souffrant d'une infection par le vih
ES2912099T3 (es) Composiciones para prevenir y/o tratar una infección por un virus VIH-1
WO2015036061A1 (fr) Nouvelles compositions
JP2014512353A (ja) 抗ウイルス組成物
US9682137B2 (en) Mutant human and simian immunodeficiency virus ENV proteins with reduced immunosuppressive properties
US20160130305A1 (en) Dengue virus (dv) polypeptide sequences, t cell epitopes and methods and uses thereof
Kityo et al. Therapeutic immunization in HIV infected Ugandans receiving stable antiretroviral treatment: a Phase I safety study
US20160375127A1 (en) Method of producing an inactivated lentivirus, especially HIV, vaccine, kit and method of use
US9636396B2 (en) Mutant human and simian immunodeficiency virus ENV proteins with reduced immunosuppressive properties
WO2015036062A1 (fr) Compositions d'un nouveau type
CA2851852A1 (fr) Nouvelles compositions

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: 16725809

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: 16725809

Country of ref document: EP

Kind code of ref document: A1