WO2005023867A1 - Method for the treatment or prophylaxis of tuberculosis - Google Patents

Method for the treatment or prophylaxis of tuberculosis Download PDF

Info

Publication number
WO2005023867A1
WO2005023867A1 PCT/GB2004/003830 GB2004003830W WO2005023867A1 WO 2005023867 A1 WO2005023867 A1 WO 2005023867A1 GB 2004003830 W GB2004003830 W GB 2004003830W WO 2005023867 A1 WO2005023867 A1 WO 2005023867A1
Authority
WO
WIPO (PCT)
Prior art keywords
ivig
infection
immunoglobulin
tuberculosis
scig
Prior art date
Application number
PCT/GB2004/003830
Other languages
French (fr)
Inventor
Stephen Jolles
Ricardo Tascon
Douglas Lowrie
Vangelis Stavropoulos
Original Assignee
Medical Research Council
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
Priority claimed from GB0320987A external-priority patent/GB0320987D0/en
Priority claimed from GB0322288A external-priority patent/GB0322288D0/en
Priority claimed from GB0402358A external-priority patent/GB0402358D0/en
Application filed by Medical Research Council filed Critical Medical Research Council
Priority to EP04768377A priority Critical patent/EP1664119A1/en
Publication of WO2005023867A1 publication Critical patent/WO2005023867A1/en
Priority to US11/371,262 priority patent/US7687060B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/06Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies from serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • C07K16/1289Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria from Mycobacteriaceae (F)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Definitions

  • the present invention relates to the treatment and/or prophylaxis of tuberculosis.
  • the invention relates to the treatment or prophylaxis of Mycobacterium tuberculosis infection by the administration of plasma immunoglobulin.
  • Plasma immunoglobulin is a blood product prepared from the serum of between 1000- 15000 donors per batch, and referred to as intravenous immunoglobulin (IVIg), subcutaneous immunoglobulin (SCIg) or intramuscular immunoglobulin (L Ig) according to the specific formulation relating to the intended route of administration. Functionally, IVIg, SCIg and LMIg are equivalent. These immunoglobulins are the treatment of choice for patients with antibody deficiencies. In this indication, IVIg is used at a 'replacement dose' of 200-400 mg/kg body weight, given approximately three weekly. In contrast, 'high dose' IVIg (hdlVIg), given most frequently at 2 g/kg/month, is used as an 'immunomodulatory' agent in an increasing number of immune and inflammatory disorders (1).
  • IVIg intravenous immunoglobulin
  • SCIg subcutaneous immunoglobulin
  • L Ig intramuscular immunoglobulin
  • Plasma immunoglobulin may be considered to have four separate mechanistic components: 1) actions mediated by the variable regions F(ab') 2 ; 2) actions of Fc on a range of Fc receptors (FcR); 3) actions mediated by complement binding within the Fc fragment; and 4) immunomodulatory substances other than antibody in the plasma preparations.
  • IVIg intravenous immunoglobulin
  • SCIg subcutaneous immunoglobulin
  • LMIg intramuscular immunoglobulin
  • the mycobacterial infection is M. tuberculosis infection.
  • the medicament according to the invention is advantageously administered to subject(s) suffering from or at risk from tuberculosis.
  • Intravenous immunoglobulin IVIg
  • SCIg subcutaneous immunoglobulin
  • LMIg intramuscular immunoglobulin
  • IVIg Intravenous immunoglobulin
  • the plasma immunoglobulin referred to herein as IVIg is administered intravenously and is intravenous immunoglobulin.
  • the IVIg is human IVIg and preferably comprises about 90% IgG, preferably about 95% IgG.
  • Suitable IVIg preparations are known in the art (see Table 1), and include OCTAGAM® and GAMMANORM® manufactured by Octapharma, Vienna, Austria. IVIg preparations known in the art are indicated for and licensed for administration in conditions of immunodeficiency, such as CVL , XLA, Wiskott Aldrich syndrome, as well as for their immunomodulatory properties, for example in Guillain Barre syndrome.
  • IVIg Immunoglobulin may be administered intramuscularly, intravenously, subcutaneously or nebulised, though it is more usually given intravenously. For example, a vein may be catheterised for the administration procedure.
  • IVIg may also be administered subcutaneously. This method of administration is used less frequently and can cause discomfort at the site. Some IVIg prescriptions may be ordered at slower rates continuously over several days. The subcutaneous method is better suited for these slower rates to maintain the Ig level. However, this method is generally not suitable for high dose administration.
  • IVIg usually is given intravenously over several hours, typically for several consecutive days.
  • a typical course of therapy may require 2-6 hour infusions for 4-5 consecutive days.
  • IVIg is advantageously administered at a "high dose", that is at a dose superior to the replacement dose.
  • Replacement dose is typically 200-400mg/kg, given three-weekly.
  • high dose IVIg is given at at least 1 g/kg/month, and advantageously about 2g/kg/month.
  • High-dose administration is associated with immunomodulatory use of IVIg, for example as indicated for Guillain- Barre syndrome, where a dose of 0.4g/kg/day for 3-7 days is advocated.
  • the invention moreover provides a method for treating a mycobacterial infection in a subject in need thereof, comprising administering to said subject intravenous immunoglobulin (IVIg).
  • IVIg intravenous immunoglobulin
  • the invention provides a method for preventing a mycobacterial infection in a subject, comprising administering to said subject intravenous immunoglobulin (IVIg).
  • IVIg intravenous immunoglobulin
  • the mycobacterial infection is M. tuberculosis infection.
  • the invention provides IVIg and a vaccine for combined use in the treatment or prevention of infection.
  • the invention provides IVIg and a vaccine for simultaneous, simultaneous separate or sequential use in the treatment or prevention of infection.
  • the effect of IVIg on the immune response to M. tuberculosis infection may be seen in terms of a cross-priming phenomenon, in which macrophages infected by M. tuberculosis cross-prime dendritic cells (DCs) or professional antigen presenting cells (APCs), for example through apoptosis of the macrophages and blebbing of vesicles comprising M. tuberculosis bacilli, proteins derived from M. tuberculosis or surface components of M. tuberculosis-infected cells.
  • DCs dendritic cells
  • APCs professional antigen presenting cells
  • Uptake of vesicles or macrophages by DCs may be enhanced through interaction between Fc receptors (FcR) on DCs and antibodies present in IVIg which are specific for the M. tuberculosis-xeldXe ⁇ components present therein.
  • FcR Fc receptors
  • the DCs or APCs thus cross-primed are able to initiate a CD4 + and/or CD8 + T-cell response against M. tuberculosis.
  • the invention provides the use of IVIg to potentiate the use of any vaccine.
  • the invention is particularly indicated for use against intracellular infectious organisms, which may evade DCs and APCs though multiplying in different cell types.
  • the vaccine is a vaccine against an intracellular infectious organism.
  • the vaccine may be a conventional vaccine based on killed, attenuated or similar organisms; in the case of TB, the vaccine may be the BCG (Bacillus Calmette-Guerin) vaccine, a DNA vaccine, a recombinant protein or subunit vaccine, or the like.
  • BCG Bacillus Calmette-Guerin
  • the IVIg may be replaced with a more specific immunoglobulin preparation.
  • immunoglobulin preparation for example, immune serum from BCG-vaccinated or DNA-vaccinated individuals, or a polyclonal or monoclonal anti- . tuberculosis IgG may be used to facilitate uptake of blebbed macrophage vesicles by FcR on DCs or APCs. More specific Ig preparations are especially suited to pulmonary delivery as described above.
  • the dose used may be lowered with respect to the standard IVIg dose.
  • doses of 0.1- l ⁇ g/kg, 1-lO ⁇ g/kg, 10-100 ⁇ g/kg or more are envisaged, administered over a period of one to several days in a series of repeated administrations.
  • FIG. 1 The effect of IVIg on the growth of M. tuberculosis in macrophages.
  • Murine C57BL/6 bone marrow derived macrophages were infected with M. tuberculosis for 6 hours, washed and maintained in either control medium or medium containing IVIg. Colony counts were obtained on days 0, 3 and 6. There was no difference in colony counts between the groups.
  • FIG. 1 The effect of IVIg on the growth of M. tuberculosis in mice. Colony counts of M. tuberculosis in the spleens and lungs taken from animals at different time points show a significant reduction in IVIg treated animals compared to controls. This effect seems to increase over time and is slightly greater in mice treated with IVIg on days 3 and 5 post infection. Each point represents five mice and error bars are standard error of the mean.
  • Figure 3 Lung Histology.
  • Late Treatment Colony counts of M. tuberculosis in the spleens and lungs taken from animals at different time points show that late treatment of animals by injection of octagam 105 days post- infection effectively reduces the M. tuberculosis load to that seen when the animals are treated at an early stage. This suggests an effective therapeutic outcome with iVig treatment.
  • Colony counts were obtained from the lungs and spleens of control or IVIg treated nude mice. Each point represents 5 mice and the experiment was repeated twice. There is no difference in the colony counts between control and IVIg treated nude mice, suggesting that the mechanism of action of IVIg may act through T cells.
  • mice were infected with Mtb and treated with saline control, O.lg/kg, 0.5g/kg, lg/kg, 1.5g/kg or 2g/kg of IVIg and colony counts obtained from the lungs and spleens. Each point represents five mice and error bars are standard error of the mean. The results show that the greatest reduction in colony counts was achieved with a dose of 2g/kg.
  • IVIg is a plasma product comprising pooled plasma from at least two individuals and comprising at least about 90% IgG.
  • IVIg is prepared from the pooled plasma of 1000 or more individuals.
  • IVIg is by its nature highly polyclonal, and comprises many antibody specificities, as well as non-antibody immunoactive molecules such as cytokines.
  • the term "IVIg” includes intravenous immunoglobulin (IVIg), subcutaneous immunoglobulin (SCIg) and intramuscular immunoglobulin (LMIg).
  • Treatment is the modulation of an existing condition such as the attenuation of an existing infection.
  • an infection is "treated” as described herein if the titre of the infectious organism is reduced by a factor of 10 in the subject, and preferably a factor of 100 or more.
  • Titre can be assayed by a standard colony formation assays for M. tuberculosis.
  • an invention is "treated” is a measurable microbiological or therapeutic benefit observable in the patient.
  • Prevention or prophylaxis is the inhibition or attenuation of an infection acquired after the administration of the treatment.
  • a subject becomes infected with M. tuberculosis subsequent to treatment with IVIg, and optionally BCG, DNA or other vaccine, attenuation or prevention of the infection is observed.
  • an infection is "attenuated” as described herein if the titre of the infectious organism is reduced by a factor of 10 in the subject, and preferably a factor of 100 or more.
  • Titre can be assayed by a standard colony formation assays for M. tuberculosis.
  • attenuation is defined in terms of a measurable microbiological or therapeutic benefit in the patient.
  • Vaccine as referred to herein is a preparation which elicits immunological protection against a pathogen.
  • the pathogen is an intracellular organism.
  • the vaccine may be a conventional vaccine, that is a vaccine consisting of non-pathogenic organisms which are homologous with the potential pathogen, or attenuated and/or killed pathogens, which elicit an immune response due to the presence of antigens similar or identical to those of the pathogen, but without the pathogenic effects.
  • it may be a DNA vaccine, which comprises a DNA molecule encoding one or more antigenic epitopes characteristic of the pathogen, or a peptide or subunit vaccine which comprises peptide or protein components characteristic the pathogen.
  • such components may be produced recombinantly.
  • Intracellular organism when applied to pathogens herein, denotes that the organism is present within the cells of the infected individual. Generally, it is not present outside said cells and effectively "hides” from the immune system by concealing itself within infected cells.
  • mice C57BL/6 and BALB/c mice aged 8-12 weeks obtained from the SPF unit NLMR were used throughout these studies .
  • a 250 ml Dubos containing 10ml of albumin Dubos supplement (Difco, Laboratories, Surrey, United Kingdom) was inoculated with 1 ml of TB culture obtained from original stock and incubated in a 37oC rotating incubator for five days. The OD reading at 600nm found to be 0.380. This was diluted to an OD of 0.02 in saline.
  • mice were held in a restraining box.
  • the tail of the mouse was warmed with a heat lamp and swabbed with gauze soaked in 70% ethanol.
  • a 1ml syringe with a 25-G needle was filled with the innoculum and any air bubbles removed.
  • the lateral tail vein was visualised and the needle was inserted parallel to the vein 2 to 4 mm into the lumen.
  • Each mouse received 0.2ml of TB of concentration 2.00x10 5 .
  • the infection was monitored by removing the lungs and spleens of infected mice at various intervals; the baseline level of infection of each tissue was estimated by harvesting organs from the mice 18 h after infection and determining viable counts.
  • the tissues were weighed and homogenised by shaking with 2-mm-diameter glass beads in chilled saline with a Mini-Bead Beater (Biospec Products, Bartlesville, Okla.), and 10- fold dilutions of the suspension were plated onto Dubos 7H11 agar with Dubos oleic albumic complex supplement (Difco Laboratories, Surrey, United Kingdom). Numbers of CFU were determined after the plates had been incubated at 37°C for approximately 20 days.
  • the mouse was restrained manually. A 1ml syringe with a 22-G needle was filled with Octagam 5% liquid and any air bubbles were removed. The needle then was inserted into the lower right quadrant of the abdomen, avoiding the abdominal midline and 0.5ml injected into each mouse 6 hours post infection and repeated on day 2.
  • Macrophages were grown in 12 well plates, 1 plate per day for day 0, 4, 7, 9 & 11. Each plate contained 8 wells (lml/well) 4 control wells and 4 IVIg treated wells (Gammanorm). Medium was cLMDM with 10% L929 cell supernatant containing either 25mg/ml IVIg or saline. Human albumin (Sigma-Aldrich, St Louis, MO) was used as a control.
  • Group 1 Given 0.5 ml IVIg (Octagam) injected ip then 9 h later given 50 ⁇ l DNA injected into the muscle tissue of each hind leg (100 ⁇ l total). The DNA was 1 mg/ml plasmid in buffered saline, expressing mycobacterial hsp65 from CMV immediate early gene promoter(2-4). 24 h after the first dose of IVIg they received another 0.5 ml IVIg ip. Then 21 d and 42 d after the first DNA injections the DNA injections were repeated. Three weeks after the last DNA injections the mice were challenged by iv injection of M. tuberculosis H37Rv. Four weeks after infection, mice were killed for counts of live bacteria in lungs and spleens.
  • Group 3 High dose TVIg as in Group 1 but without DNA.
  • Group 4 Low dose LVIg as in Group 2 but without DNA.
  • Group 5 Untreated controls.
  • Group 6 DNA treatment as in Group 1 but without IVIg treatment.
  • Group 7 BCG vaccinated, 50 ⁇ l sc at the time of first IVIg treatment.
  • Staining of cells obtained from lungs was performed using directly conjugated anti TCR PE, anti CD4 FITC, anti CD8 FITC, anti CD44 PE and anti LFN ⁇ PE from Pharmingen.
  • Octagam and Gammanorm both pooled normal IgG obtained from healthy donors were used. Octagam was used for the in vivo experiments unaltered and Gammanorm for the in vitro studies.
  • the Gammanorm was dialysed twice in either RPMI 1640 (Gibco) or LMDM (Gibco) before use to remove stabilising agents and filter sterilised, or used unaltered.
  • mice were given a lethal dose of anaesthetic (Sagatal) i.p.; the lungs perfused with 1ml 10% neutral buffered formalin solution (Sigma) via the trachea in situ and a further 1ml on removal of the lungs before being placed in 10ml 10% neutral buffered formalin for 3 days to await histology. After dehydration in a graded series of ethanol and clearing in xylene, the lungs were embedded in fibrowax (BDH). Sections of 6 ⁇ m thickness were stained with haematoxylin and eosin.
  • serial lung sections were deparaffinized and incubated with 0.1 g/ml of rabbit anti- mouse iNOS (BD Transduction Laboratories). Bound antibodies were detected with biotinylated, affinity-purified anti-rabbit immunoglobulin as the secondary antibody, and after washing, the sections were incubated with avidin-coupled biotinylated horseradish peroxidase with aminobenzidine as substrate (VECTASTALN ABS Kit, VECTOR Laboratories) according to the manufacturer's instructions.
  • mice following infection with Mycobacterium tuberculosis reduced the colony counts in lungs and spleen by 1000 fold and was associated with a more lymphocyte predominant appearance of lung granulomas than in control mice.
  • IVIg did not affect the growth of M. tuberculosis in murine bone marrow derived macrophages in vitro.
  • Fc interactions through FcR ⁇ lLB which delivers a negative signal through an ITIM motif have been shown to downregulate macrophage function (5). It has also been shown that IVIg inhibits dendritic cell maturation (6) and this may be important as immature DCs have an antigen sampling role while mature DCs loose this ability and become functionally more potent antigen presenting cells.
  • Cross priming is an important mechanism for antigen presentation in mycobacterial infection (7), if the action of IVIg is through enhancement of cross priming it would be beneficial to temporarily maintain DCs in an immature state in which they were able to take up antigen and for these two events to coincide. It has been shown that cross priming using antibody immune complex coated apoptotic tumour cells enhances anti tumour immunity and tumour rejection (8). A further potential immunomodulatory mechanism might be to alter the numbers of regulatory T cells (CD4 CD25, IL-10) present which have been shown in a Leishmania model to allow persistence of infection.
  • the IVIg used (Octagam) is produced from plasma derived from donors in Austria, Germany and the US and will contain antibodies from BCG vaccinated individuals. It has been shown that the efficacy of BCG given in trials in parts of south India and southern Africa has little benefit (9, 10) while in most cases in the developed world there has been demonstrable efficacy. This has been explained by differences in environmental factors such as prior exposure to other environmental mycobacteria in parts of south India and southern Africa. It is possible that LVIg imposes a western antibody binding site repertoire, which facilitates the response to M.
  • Fine P Group KPT. Randomised controlled trial of single BCG, repeated BCG, or combined BCG and killed Mycobacterium leprae vaccine for prevention of leprosy and tuberculosis in Malawi. Karonga Prevention Trial Group. Lancet 1996; 348: 17-24.
  • Tripathy SP Trial of BCG vaccines in South India for tuberculosis prevention: First Report. Bulletin of the World Health Organization 1979; 57: 819-827.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The invention provides the use of intravenous immunoglobulin (IVIg) in the preparation of a medicament for the treatment and/or prophylaxis of mycobacterial infection.

Description

METHOD FOR THE TREATMENT OR PROPHYLAXIS OF TUBERCULOSIS
The present invention relates to the treatment and/or prophylaxis of tuberculosis. In particular, the invention relates to the treatment or prophylaxis of Mycobacterium tuberculosis infection by the administration of plasma immunoglobulin.
infection with Mycobacterium Tuberculosis is a major global health problem with one third of the world's population infected. There is only one licensed vaccine (BCG) with variable efficacy. In view of the global seriousness of the problem, the World Health Organisation declared it to be a Global Emergency in 1993. Two million people die every year, constituting 26% of avoidable adult deaths world-wide.
Plasma immunoglobulin is a blood product prepared from the serum of between 1000- 15000 donors per batch, and referred to as intravenous immunoglobulin (IVIg), subcutaneous immunoglobulin (SCIg) or intramuscular immunoglobulin (L Ig) according to the specific formulation relating to the intended route of administration. Functionally, IVIg, SCIg and LMIg are equivalent. These immunoglobulins are the treatment of choice for patients with antibody deficiencies. In this indication, IVIg is used at a 'replacement dose' of 200-400 mg/kg body weight, given approximately three weekly. In contrast, 'high dose' IVIg (hdlVIg), given most frequently at 2 g/kg/month, is used as an 'immunomodulatory' agent in an increasing number of immune and inflammatory disorders (1).
Plasma immunoglobulin may be considered to have four separate mechanistic components: 1) actions mediated by the variable regions F(ab')2; 2) actions of Fc on a range of Fc receptors (FcR); 3) actions mediated by complement binding within the Fc fragment; and 4) immunomodulatory substances other than antibody in the plasma preparations.
It is likely that these components act concurrently, however different mechanisms may be important in different settings. In some cases more than one mechanism is operative or current understanding does not allow accurate categorisation. Summary of the Invention
We have found that subjects treated with hdlVIg following infection with M. tuberculosis have significantly lower colony counts in the lungs and spleen; and moreover that IVIg in vitro does not inhibit the growth of TB in macrophages. These results indicate that IVIg improves the immunological response to M. tuberculosis.
According to a first aspect of the invention, therefore, there is provided the use of intravenous immunoglobulin (IVIg), subcutaneous immunoglobulin (SCIg) or intramuscular immunoglobulin (LMIg) in the preparation of a medicament for the treatment and/or prophylaxis of mycobacterial infection.
Preferably, the mycobacterial infection is M. tuberculosis infection. The medicament according to the invention is advantageously administered to subject(s) suffering from or at risk from tuberculosis.
Intravenous immunoglobulin (IVIg), subcutaneous immunoglobulin (SCIg) or intramuscular immunoglobulin (LMIg) are functionally equivalent and referred to herein, for convenience, as IVIg. In a preferred embodiment, the plasma immunoglobulin referred to herein as IVIg is administered intravenously and is intravenous immunoglobulin.
Advantageously, the IVIg is human IVIg and preferably comprises about 90% IgG, preferably about 95% IgG. Suitable IVIg preparations are known in the art (see Table 1), and include OCTAGAM® and GAMMANORM® manufactured by Octapharma, Vienna, Austria. IVIg preparations known in the art are indicated for and licensed for administration in conditions of immunodeficiency, such as CVL , XLA, Wiskott Aldrich syndrome, as well as for their immunomodulatory properties, for example in Guillain Barre syndrome. IVIg Immunoglobulin may be administered intramuscularly, intravenously, subcutaneously or nebulised, though it is more usually given intravenously. For example, a vein may be catheterised for the administration procedure.
IVIg may also be administered subcutaneously. This method of administration is used less frequently and can cause discomfort at the site. Some IVIg prescriptions may be ordered at slower rates continuously over several days. The subcutaneous method is better suited for these slower rates to maintain the Ig level. However, this method is generally not suitable for high dose administration.
Because of the large doses required for immunomodulation, high dose IVIg usually is given intravenously over several hours, typically for several consecutive days. A typical course of therapy may require 2-6 hour infusions for 4-5 consecutive days.
For treatment and/or prophylaxis of TB, IVIg is advantageously administered at a "high dose", that is at a dose superior to the replacement dose. Replacement dose is typically 200-400mg/kg, given three-weekly. Typically, high dose IVIg is given at at least 1 g/kg/month, and advantageously about 2g/kg/month. High-dose administration is associated with immunomodulatory use of IVIg, for example as indicated for Guillain- Barre syndrome, where a dose of 0.4g/kg/day for 3-7 days is advocated.
The invention moreover provides a method for treating a mycobacterial infection in a subject in need thereof, comprising administering to said subject intravenous immunoglobulin (IVIg).
Further, the invention provides a method for preventing a mycobacterial infection in a subject, comprising administering to said subject intravenous immunoglobulin (IVIg).
Advantageously, the mycobacterial infection is M. tuberculosis infection.
In a further embodiment, the invention provides IVIg and a vaccine for combined use in the treatment or prevention of infection. Thus, the invention provides IVIg and a vaccine for simultaneous, simultaneous separate or sequential use in the treatment or prevention of infection.
Although not wishing to be bound by theory, the effect of IVIg on the immune response to M. tuberculosis infection may be seen in terms of a cross-priming phenomenon, in which macrophages infected by M. tuberculosis cross-prime dendritic cells (DCs) or professional antigen presenting cells (APCs), for example through apoptosis of the macrophages and blebbing of vesicles comprising M. tuberculosis bacilli, proteins derived from M. tuberculosis or surface components of M. tuberculosis-infected cells. Uptake of vesicles or macrophages by DCs may be enhanced through interaction between Fc receptors (FcR) on DCs and antibodies present in IVIg which are specific for the M. tuberculosis-xeldXeά components present therein. The DCs or APCs thus cross-primed are able to initiate a CD4+ and/or CD8+ T-cell response against M. tuberculosis.
A similar approach is of benefit in other infectious diseases (e.g. leprosy etc) and tumours which require a T cell mediated immune response which can be augmented by improved cross priming.
Accordingly, the invention provides the use of IVIg to potentiate the use of any vaccine. The invention is particularly indicated for use against intracellular infectious organisms, which may evade DCs and APCs though multiplying in different cell types. Advantageously, therefore, the vaccine is a vaccine against an intracellular infectious organism.
The vaccine may be a conventional vaccine based on killed, attenuated or similar organisms; in the case of TB, the vaccine may be the BCG (Bacillus Calmette-Guerin) vaccine, a DNA vaccine, a recombinant protein or subunit vaccine, or the like.
It is possible to deliver intact IgG into the lungs using a nebuliser (Aerogen Inc. Mountain View, CA, USA) which does not destroy the function of the IgG protein. Given that M. tuberculosis is spread via the respiratory route and the majority of infections are pulmonary it may be advantageous to deliver IVIg into the lungs to enhance the immune response locally. Moreover, pulmonary delivery of vaccines, such as the BCG TB vaccine, may be combined with pulmonary delivery of IVIg to provide a potentiated vaccine for uptake via the pulmonary route. Typically, lower doses of IVIg are sufficient when delivered by the pulmonary route.
Where the IVIg is acting to facilitate cross-priming of DCs and/or APCs, the IVIg may be replaced with a more specific immunoglobulin preparation. For example, immune serum from BCG-vaccinated or DNA-vaccinated individuals, or a polyclonal or monoclonal anti- . tuberculosis IgG may be used to facilitate uptake of blebbed macrophage vesicles by FcR on DCs or APCs. More specific Ig preparations are especially suited to pulmonary delivery as described above.
Where immunoglobulin preparations are used which are more specific than IVIg, the dose used may be lowered with respect to the standard IVIg dose. For example, doses of 0.1- lμg/kg, 1-lOμg/kg, 10-100μg/kg or more are envisaged, administered over a period of one to several days in a series of repeated administrations.
Description of the Figures
Figure 1. The effect of IVIg on the growth of M. tuberculosis in macrophages. Murine C57BL/6 bone marrow derived macrophages were infected with M. tuberculosis for 6 hours, washed and maintained in either control medium or medium containing IVIg. Colony counts were obtained on days 0, 3 and 6. There was no difference in colony counts between the groups.
Figure 2. The effect of IVIg on the growth of M. tuberculosis in mice. Colony counts of M. tuberculosis in the spleens and lungs taken from animals at different time points show a significant reduction in IVIg treated animals compared to controls. This effect seems to increase over time and is slightly greater in mice treated with IVIg on days 3 and 5 post infection. Each point represents five mice and error bars are standard error of the mean. Figure 3. Lung Histology.
Lung sections of control and IVIg treated mice at 40x and lOOx magnification stained with haematoxylin and eosin at day 42 following infection. The lungs obtained from IVIg treated mice contain granulomata which compared to controls exhibit a considerably more dense lymphocytic infiltrate, while the ratio of macrophages to lymphocytes is greater in controls. This suggests a more vigorous immunological response in the treated animals.
Figure 4. Late Treatment Colony counts of M. tuberculosis in the spleens and lungs taken from animals at different time points show that late treatment of animals by injection of octagam 105 days post- infection effectively reduces the M. tuberculosis load to that seen when the animals are treated at an early stage. This suggests an effective therapeutic outcome with iVig treatment.
Figure 5. Control with Albumin
Colony counts of M. tuberculosis in the spleens and lungs taken from animals at different time points show that human albumin has no effect. This confirms that the observed phenomenon is not related to a mouse anti-human response.
Figure 6. The effect of IVIg in Nude mice infected with Mtb.
Colony counts were obtained from the lungs and spleens of control or IVIg treated nude mice. Each point represents 5 mice and the experiment was repeated twice. There is no difference in the colony counts between control and IVIg treated nude mice, suggesting that the mechanism of action of IVIg may act through T cells.
Figure 7. Dose response of IVIg in Mtb infected mice.
Mice were infected with Mtb and treated with saline control, O.lg/kg, 0.5g/kg, lg/kg, 1.5g/kg or 2g/kg of IVIg and colony counts obtained from the lungs and spleens. Each point represents five mice and error bars are standard error of the mean. The results show that the greatest reduction in colony counts was achieved with a dose of 2g/kg. Detailed Description of the Invention
IVIg, as defined herein, is a plasma product comprising pooled plasma from at least two individuals and comprising at least about 90% IgG. Advantageously, it is prepared from the pooled plasma of 1000 or more individuals. IVIg is by its nature highly polyclonal, and comprises many antibody specificities, as well as non-antibody immunoactive molecules such as cytokines. The term "IVIg" includes intravenous immunoglobulin (IVIg), subcutaneous immunoglobulin (SCIg) and intramuscular immunoglobulin (LMIg).
Treatment, as defined herein, is the modulation of an existing condition such as the attenuation of an existing infection. Thus, the treatment of a mycobacterial infection by the administration of IVIg presupposes that the subject to be treated has a mycobacterial infection prior to the said administration. In an experimental context, an infection is "treated" as described herein if the titre of the infectious organism is reduced by a factor of 10 in the subject, and preferably a factor of 100 or more. Titre can be assayed by a standard colony formation assays for M. tuberculosis. In the context of therapy, an invention is "treated" is a measurable microbiological or therapeutic benefit observable in the patient.
Prevention or prophylaxis is the inhibition or attenuation of an infection acquired after the administration of the treatment. Thus, if a subject becomes infected with M. tuberculosis subsequent to treatment with IVIg, and optionally BCG, DNA or other vaccine, attenuation or prevention of the infection is observed. In an experimental context, an infection is "attenuated" as described herein if the titre of the infectious organism is reduced by a factor of 10 in the subject, and preferably a factor of 100 or more. Titre can be assayed by a standard colony formation assays for M. tuberculosis. In a therapeutic context, attenuation is defined in terms of a measurable microbiological or therapeutic benefit in the patient.
Vaccine as referred to herein is a preparation which elicits immunological protection against a pathogen. Preferably, the pathogen is an intracellular organism. The vaccine may be a conventional vaccine, that is a vaccine consisting of non-pathogenic organisms which are homologous with the potential pathogen, or attenuated and/or killed pathogens, which elicit an immune response due to the presence of antigens similar or identical to those of the pathogen, but without the pathogenic effects. Alternatively, it may be a DNA vaccine, which comprises a DNA molecule encoding one or more antigenic epitopes characteristic of the pathogen, or a peptide or subunit vaccine which comprises peptide or protein components characteristic the pathogen. Typically, such components may be produced recombinantly.
Intracellular organism, when applied to pathogens herein, denotes that the organism is present within the cells of the infected individual. Generally, it is not present outside said cells and effectively "hides" from the immune system by concealing itself within infected cells.
Mice.
C57BL/6 and BALB/c mice aged 8-12 weeks obtained from the SPF unit NLMR were used throughout these studies .
M. Tuberculosis culture for intravenous infections.
A 250 ml Dubos containing 10ml of albumin Dubos supplement (Difco, Laboratories, Surrey, United Kingdom) was inoculated with 1 ml of TB culture obtained from original stock and incubated in a 37oC rotating incubator for five days. The OD reading at 600nm found to be 0.380. This was diluted to an OD of 0.02 in saline.
Intravenous infections of mice
Mice were held in a restraining box. The tail of the mouse was warmed with a heat lamp and swabbed with gauze soaked in 70% ethanol. A 1ml syringe with a 25-G needle was filled with the innoculum and any air bubbles removed. The lateral tail vein was visualised and the needle was inserted parallel to the vein 2 to 4 mm into the lumen. Each mouse received 0.2ml of TB of concentration 2.00x105.
The infection was monitored by removing the lungs and spleens of infected mice at various intervals; the baseline level of infection of each tissue was estimated by harvesting organs from the mice 18 h after infection and determining viable counts. The tissues were weighed and homogenised by shaking with 2-mm-diameter glass beads in chilled saline with a Mini-Bead Beater (Biospec Products, Bartlesville, Okla.), and 10- fold dilutions of the suspension were plated onto Dubos 7H11 agar with Dubos oleic albumic complex supplement (Difco Laboratories, Surrey, United Kingdom). Numbers of CFU were determined after the plates had been incubated at 37°C for approximately 20 days.
Intraperitoneal administration of compounds in mice
The mouse was restrained manually. A 1ml syringe with a 22-G needle was filled with Octagam 5% liquid and any air bubbles were removed. The needle then was inserted into the lower right quadrant of the abdomen, avoiding the abdominal midline and 0.5ml injected into each mouse 6 hours post infection and repeated on day 2.
Generation of bone marrow derived macrophages.
Media 500ml Iscoves modified Dulbeccos medium (LMDM), 5% (25ml) heat-inactivated FCS, 2mM L-glutamine, 50uM 2-mercaptoethanol, 10% L929 cell supernatant (source of M-CSF).
On Day 0 bone marrow from C57B1/6 mice was removed. Cells were washed counted and resuspended to 2xl06 cells/ml in cLMDM with 10% L929 cell supernatant. The cells were plated into 12 well plates at lml/well and incubated at 37°C. On day 3 the non- adherent cells and medium were removed and fresh medium containing 10% L929 cell supernatant added. This was repeated on day 5 and the cells infected on day 6.
Growth of H37Rv in bone marrow derived macrophages. Macrophages were grown in 12 well plates, 1 plate per day for day 0, 4, 7, 9 & 11. Each plate contained 8 wells (lml/well) 4 control wells and 4 IVIg treated wells (Gammanorm). Medium was cLMDM with 10% L929 cell supernatant containing either 25mg/ml IVIg or saline. Human albumin (Sigma-Aldrich, St Louis, MO) was used as a control.
On day 0 the number of macrophages per well was counted and all wells infected with H37Rv at multiplicity of infection (MOI) 2:1 and incubated at 37°C for 6hrs in medium not containing IVIg. Following this the medium was removed and replaced with fresh cLMDM containing either 25mg/ml IVIg or human albumin control. Four plates were returned to the incubator and day 0 colony forming units (cfu) performed to establish a base line for growth.
Preparation of CFU plates.
The supernatant was removed from each well and retained. Each well was washed three times with 1ml PBS and 1ml PBS with lOOul 2% saponin added to each well, and incubated at 37°C for a minimum of lhr.
1 in 10 serial dilutions of each well and each supernatant were prepared in 900ul saline to 10-4, (neat, 10, 100, 1000, 10000 fold dilutions). 20ul of each dilution was applied to 7H11 plates with albumin, these were divided into 5 sections(duplicate plate per well/supernatant), allowed to dry, wrapped, inverted and incubated at 37°C for 14-21 days before counting (no colonies x dilution factor = cfu/ml).
The effects of IVIg on protective response to DNA vaccination against TB challenge.
Seven groups of 8 Balb/c females, 6-8 wk.
Group 1: Given 0.5 ml IVIg (Octagam) injected ip then 9 h later given 50 μl DNA injected into the muscle tissue of each hind leg (100 μl total). The DNA was 1 mg/ml plasmid in buffered saline, expressing mycobacterial hsp65 from CMV immediate early gene promoter(2-4). 24 h after the first dose of IVIg they received another 0.5 ml IVIg ip. Then 21 d and 42 d after the first DNA injections the DNA injections were repeated. Three weeks after the last DNA injections the mice were challenged by iv injection of M. tuberculosis H37Rv. Four weeks after infection, mice were killed for counts of live bacteria in lungs and spleens.
Group 2: As above but the dose of IVIg was reduced to 0.2 ml instead of 1 ml total.
Group 3: High dose TVIg as in Group 1 but without DNA.
Group 4: Low dose LVIg as in Group 2 but without DNA. Group 5: Untreated controls.
Group 6: DNA treatment as in Group 1 but without IVIg treatment.
Group 7: BCG vaccinated, 50 μl sc at the time of first IVIg treatment.
Statistics.
Data are the geometric means and the standard errors of the means of five mice. Graphs were plotted using SigmaPlot 8.0 software.
Antibodies and Reagents.
Staining of cells obtained from lungs was performed using directly conjugated anti TCR PE, anti CD4 FITC, anti CD8 FITC, anti CD44 PE and anti LFNγPE from Pharmingen.
Therapeutic Intravenous Immunoglobulin.
Octagam and Gammanorm (Octapharma, Vienna, Austria) both pooled normal IgG obtained from healthy donors were used. Octagam was used for the in vivo experiments unaltered and Gammanorm for the in vitro studies. The Gammanorm was dialysed twice in either RPMI 1640 (Gibco) or LMDM (Gibco) before use to remove stabilising agents and filter sterilised, or used unaltered.
Lung Histology.
Mice were given a lethal dose of anaesthetic (Sagatal) i.p.; the lungs perfused with 1ml 10% neutral buffered formalin solution (Sigma) via the trachea in situ and a further 1ml on removal of the lungs before being placed in 10ml 10% neutral buffered formalin for 3 days to await histology. After dehydration in a graded series of ethanol and clearing in xylene, the lungs were embedded in fibrowax (BDH). Sections of 6 μm thickness were stained with haematoxylin and eosin. For immunohistochemical localisation of iNOS, serial lung sections were deparaffinized and incubated with 0.1 g/ml of rabbit anti- mouse iNOS (BD Transduction Laboratories). Bound antibodies were detected with biotinylated, affinity-purified anti-rabbit immunoglobulin as the secondary antibody, and after washing, the sections were incubated with avidin-coupled biotinylated horseradish peroxidase with aminobenzidine as substrate (VECTASTALN ABS Kit, VECTOR Laboratories) according to the manufacturer's instructions.
Results Administration of human pooled IVIg to mice following infection with Mycobacterium tuberculosis reduced the colony counts in lungs and spleen by 1000 fold and was associated with a more lymphocyte predominant appearance of lung granulomas than in control mice. When IVIg was given on days three and five following infection rather than 6 hours and 24 hours the reduction in colony counts was slightly greater. IVIg did not affect the growth of M. tuberculosis in murine bone marrow derived macrophages in vitro.
The reduction of M. tuberculosis colony counts when IVIg was given on days three and five post infection suggests that it is unlikely that M. tuberculosis is being opsonised and killed in the circulation as the organism becomes intracellular within hours following infection. In support of this is the fact that the difference in the colony counts from control increases with time and at a time when there would have been a mouse anti human response. Taken with the lack of effect of IVIg on the growth of M. tuberculosis in macrophages in vitro this suggests that the improved control of infection is immunological in nature. The lung histology shows a more lymphocytic infiltrate within granulomas in treated mice.
The mechanisms of action of IVIg given at immunomodulatory doses are complex and incompletely understood. The possibilities for the benefit observed in mice infected with M. tuberculosis may be divided into two major subsets based on the structure of the antibody molecule and are mechanisms dependent on Fc or the variable binding site regions of F(ab)2. It would be possible to test the role of Fc interactions in a number of ways. Fc could be given alone having digested F(ab)2 using papain, as a control or monoclonal human myeloma IgGl could be used as mis would have only one binding specificity and therefore not reflect the repertoire present in polyclonal IVIg. The avidity of an Fc reagent could be increased to enhance this modality using Fc tetramers. Another possibility would be the use of pre-treatment with anti murine Fc to block Fc interactions. These approaches might delineate which part of the antibody molecule was most important. Fc interactions through FcRγlLB, which delivers a negative signal through an ITIM motif have been shown to downregulate macrophage function (5). It has also been shown that IVIg inhibits dendritic cell maturation (6) and this may be important as immature DCs have an antigen sampling role while mature DCs loose this ability and become functionally more potent antigen presenting cells. Cross priming is an important mechanism for antigen presentation in mycobacterial infection (7), if the action of IVIg is through enhancement of cross priming it would be beneficial to temporarily maintain DCs in an immature state in which they were able to take up antigen and for these two events to coincide. It has been shown that cross priming using antibody immune complex coated apoptotic tumour cells enhances anti tumour immunity and tumour rejection (8). A further potential immunomodulatory mechanism might be to alter the numbers of regulatory T cells (CD4 CD25, IL-10) present which have been shown in a Leishmania model to allow persistence of infection.
It would be possible to determine if particular F(ab)2 variable region binding site interactions are important. The serum of mice treated with DNA vaccination, BCG, or M. tuberculosis could be used to augment vaccination with DNA or BCG. These interactions may play a role in cross priming which has been shown to be important for protection against M. tuberculosis.
The data suggest a novel means by which to enhance the immune response to M. tuberculosis using an agent, which is already licensed for human use. The IVIg used (Octagam) is produced from plasma derived from donors in Austria, Germany and the US and will contain antibodies from BCG vaccinated individuals. It has been shown that the efficacy of BCG given in trials in parts of south India and southern Africa has little benefit (9, 10) while in most cases in the developed world there has been demonstrable efficacy. This has been explained by differences in environmental factors such as prior exposure to other environmental mycobacteria in parts of south India and southern Africa. It is possible that LVIg imposes a western antibody binding site repertoire, which facilitates the response to M. tuberculosis and potentially even vaccination efficacy in mice maintained in an SPF environment. All publications mentioned in the present specification are herein incorporated by reference. Various modifications and variations of the described methods and system of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention which are apparent to those skilled in molecular biology or related fields are intended to be within the scope of the following claims.
References.
1. Sewell WA, Jolles S. Immunomodulatory action of intravenous immunoglobulin. Immunology 2002; 107: 387-93.
2. Tascon RE, Colston MJ, Ragno S, Stavropoulos E, Gregory D, Lowrie DB. Vaccination against tuberculosis by DNA injection. Nat Med 1996; 2: 888-92.
3. Lowrie DB, Tascon RE, Bonato VL, Lima VM, Faccioli LH, Stavropoulos E, et al. Therapy of tuberculosis in mice by DNA vaccination. Nature 1999; 400: 269-71.
4. Lowrie DB. DNA vaccines against tuberculosis. Curr Opin Mol Ther 1999; 1 : 30- 3. 5. Samuelsson A, Towers TL, Ravetch JV. Anti-inflammatory activity of IVIG mediated through the inhibitory Fc receptor. Science 2001; 291: 484-6. 6. Bayry J, Lacroix-Desmazes S, Carbonneil C, Misra N, Donkova V, Pashov A, et al. Inhibition of maturation and function of dendritic cells by intravenous immunoglobulin. Blood 2003; 101: 758-65. 7. Schaible UE, Winau F, Sieling PA, Fischer K, Collins HL, Hagens K, et al. Apoptosis facilitates antigen presentation to T lymphocytes through MHC-I and CDl in tuberculosis. Nat Med 2003; 9: 1039-46.
8. Akiyama K, Ebihara S, Yada A, Matsumura K, Aiba S, Nukiwa T, et al. Targeting apoptotic tumor cells to Fc gamma R provides efficient and versatile vaccination against tumors by dendritic cells. J Immunol 2003; 170: 1641-8.
9. Fine P, Group KPT. Randomised controlled trial of single BCG, repeated BCG, or combined BCG and killed Mycobacterium leprae vaccine for prevention of leprosy and tuberculosis in Malawi. Karonga Prevention Trial Group. Lancet 1996; 348: 17-24.
10. Tripathy SP. Trial of BCG vaccines in South India for tuberculosis prevention: First Report. Bulletin of the World Health Organization 1979; 57: 819-827.
Table 1 IGIV INFORMATION CHART March, 2002
Figure imgf000017_0001
All Information is derived from manufacturer's sales brochures and package inserts SD = Solvent Detergent, Past. = Pastuπzation Table 1 IGIV INFORMATION CHART March, 2002
Figure imgf000018_0001
All information is derived from manufacturer's sales brochures and package inserts. SD = Solvent Detergent, Past. = Pastunzation Table 1 IGIV INFORMATION CHART March, 2O02
Figure imgf000019_0001
All information is derived from manufacturer's sales brochures and package inserts. SD = Solvent Detergent, Past. = Pastunzation Table 1 IGIV INFORMATION CHART March, 2002
Figure imgf000020_0001
All information is derived from manufacturer's sales brochures and package inserts. SD = Solvent Detergent, Past. = Pastunzation Table 1 IGIV INFORMATION CHART March, 2002
Figure imgf000021_0001
All information is derived from manufacturer's sales brochures and package inserts. SD = Solvent Detergent, Past. = Pasturization

Claims

Claims
1. Use of intravenous immunoglobulin (IVIg), subcutaneous immunoglobulin (SCIG) or intramuscular immunoglobulin (IMIG) in the preparation of a medicament for the treatment and/or prophylaxis of infections with intracellular organisms.
2. Use according to claim 1, wherein the IVIg, IMIg or SCIg is at least 90% IgG.
3. Use according to claim 1 or claim 2 wherein the IVIg, IMIg or SCIg is human IVIg, IMIg or SCIg.
4. Use according to any preceding claim, wherein the IVIg, IMIg or SCIg is administered intravenously.
5. Use according to any preceding claim, wherein the IVIg, IMIg or SCIg is administered at a dose of 2g/kg/month.
6. Use according to claim 5, wherein the IVIg, IMIg or SCIg is administered in a course of intravenous injections over a period of 2 or more days as a single dose or in repeated doses.
7. Use according to any preceding claim, wherein the intracellular infection is a mycobacterial infection.
8. Use according to claim 7, wherein the infection is a M. tuberculosis infection.
9. A method for treating an infection by an intracellular organism, in a subject in need thereof, comprising administering to said subject intravenous immunoglobulin (IVIg), subcutaneous immunoglobulin (SCIG) or intramuscular immunoglobulin (IMIG).
10. A method for preventing an infection by an intracellular organism in a subject, comprising administering to said subject intravenous immunoglobulin (IVIg), subcutaneous immunoglobulin (SCIG) or intramuscular immunoglobulin (IMIG).
11. A method according to claim 9 or claim 10 wherein the infection is a mycobacterial infection.
12. A method according to claim 11, wherein the mycobacterium is M. tuberculosis.
13. An immunoglobulin preparation and a vaccine for simultaneous, simultaneous separate or sequential use in the treatment or prevention of infection with an intracellular organism, wherein said antibody preparation is specific for one or more components of the intracellular organism.
14. Use according to claim 13, wherein the immunoglobulin preparation is selected from the group consisting of an immune serum, a polyclonal IgG and a monoclonal IgG.
15. Use according to claim 13 or claim 14, wherein the intracellular organism is a mycobacterium.
16. Use according to claim 15, wherein the mycobacterium is M. tuberculosis.
PCT/GB2004/003830 2003-09-08 2004-09-08 Method for the treatment or prophylaxis of tuberculosis WO2005023867A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP04768377A EP1664119A1 (en) 2003-09-08 2004-09-08 Method for the treatment or prophylaxis of tuberculosis
US11/371,262 US7687060B2 (en) 2003-09-08 2006-03-08 Method for the treatment or prophylaxis of tuberculosis

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
GB0320987.1 2003-09-08
GB0320987A GB0320987D0 (en) 2003-09-08 2003-09-08 Method
GB0322288.2 2003-09-23
GB0322288A GB0322288D0 (en) 2003-09-23 2003-09-23 Method
GB0402358A GB0402358D0 (en) 2004-02-03 2004-02-03 Method
GB0402358.6 2004-02-03

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/371,262 Continuation US7687060B2 (en) 2003-09-08 2006-03-08 Method for the treatment or prophylaxis of tuberculosis

Publications (1)

Publication Number Publication Date
WO2005023867A1 true WO2005023867A1 (en) 2005-03-17

Family

ID=34279362

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2004/003830 WO2005023867A1 (en) 2003-09-08 2004-09-08 Method for the treatment or prophylaxis of tuberculosis

Country Status (3)

Country Link
US (1) US7687060B2 (en)
EP (1) EP1664119A1 (en)
WO (1) WO2005023867A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2435474A2 (en) 2009-05-27 2012-04-04 Baxter International Inc A method to produce a highly concentrated immunoglobulin preparation for subcutaneous use
WO2015015001A1 (en) 2013-08-01 2015-02-05 Suppremol Gmbh In vitro method for determining the stability of compositions comprising soluble fc gamma receptor(s)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016064955A1 (en) 2014-10-21 2016-04-28 The General Hospital Corporation Methods of diagnosis and treatment of tuberculosis and infection
US10421788B2 (en) 2015-09-10 2019-09-24 The General Hospital Corporation Synthesizing vaccines, immunogens, and antibodies

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004047865A1 (en) 2002-11-25 2004-06-10 Instituto Finlay. Centro De Investigacion Produccion De Vacunas Y Sueros. Compositions for the treatment of mycobacterial infections

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6531464B1 (en) * 1999-12-07 2003-03-11 Inotek Pharmaceutical Corporation Methods for the treatment of neurodegenerative disorders using substituted phenanthridinone derivatives
AT410637B (en) * 2001-06-01 2003-06-25 Igeneon Krebs Immuntherapie USE OF POLYCLONAL IMMUNOGLOBULINES

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004047865A1 (en) 2002-11-25 2004-06-10 Instituto Finlay. Centro De Investigacion Produccion De Vacunas Y Sueros. Compositions for the treatment of mycobacterial infections

Non-Patent Citations (20)

* Cited by examiner, † Cited by third party
Title
AKIYAMA K; EBIHARA S; YADA A; MATSUMURA K; AIBA S; NUKIWA T ET AL.: "Targeting apoptotic tumor cells to Fc gamma R provides efficient and versatile vaccination against tumors by dendritic cells", J IMMUNOL, vol. 1, no. 70, 2003, pages 1641 - 8
BAYRY J; LACROIX-DESMAZES S; CARBONNEIL C; MISRA N; DONKOVA V; PASHOV A ET AL.: "Inhibition of maturation and function of dendritic cells by intravenous immunoglobulin", BLOOD, vol. 101, 2003, pages 758 - 65
BUSSE ET AL., J. ALLERGY CLIN. IMMUNOL., vol. 109, 2002, pages 1001 - 1004
BUSSE PAULA JANE ET AL: "Efficacy of intravenous immunoglobulin in the prevention of pneumonia in patients with common variable immunodeficiency", JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY, vol. 109, no. 6, June 2002 (2002-06-01), pages 1001 - 1004, XP002302234, ISSN: 0091-6749 *
CASASSUS P ET AL: "[Prevention of opportunistic infections in HIV seropositive patients. Prevention of bacterial infections]", PRESSE MEDICALE (PARIS, FRANCE : 1983) 8 MAR 1997, vol. 26, no. 7, 8 March 1997 (1997-03-08), pages 340 - 343, XP009038459, ISSN: 0755-4982 *
CASASSUS, PRESSE MEDICAL, vol. 26, 1997, pages 340 - 343
FINE P; GROUP KPT: "Randomised controlled trial of single BCG, repeated BCG, or combined BCG and killed Mycobacterium leprae vaccine for prevention of leprosy and tuberculosis in Malawi. Karonga Prevention Trial Group", LANCET, vol. 348, 1996, pages 17 - 24
HEMMING VAL G: "Use of intravenous immunoglobulins for prophylaxis or treatment of infectious diseases", CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY, vol. 8, no. 5, September 2001 (2001-09-01), pages 859 - 863, XP002302233, ISSN: 1071-412X *
HEMMING, CLIN. DIAG. LAB. IMMUNOL., vol. 8, 2001, pages 859 - 863
LOWRIE DB.: "DNA vaccines against tuberculosis", CURR OPIN MOL THER, vol. 1, 1999, pages 30 - 3
LOWRIE DB; TASCON RE; BONATO VL; LIMA VM; FACCIOLI LH; STAVROPOULOS E ET AL.: "Therapy of tuberculosis in mice by DNA vaccination", NATURE, vol. 400, 1999, pages 269 - 71
ONDAR ET AL., ACTA HAEMATOL, vol. 96, 1996, pages 73 - 78
SAMUELSSON A; TOWERS TL; RAVETCH JV: "Anti-inflammatory activity of IVIG mediated through the inhibitory Fc receptor", SCIENCE, vol. 291, 2001, pages 484 - 6
SAWYER LEIGH A: "Antibodies for the prevention and treatment of viral diseases", ANTIVIRAL RESEARCH, vol. 47, no. 2, August 2000 (2000-08-01), pages 57 - 77, XP002302232, ISSN: 0166-3542 *
SAWYER, ANTIVIRAL RESEARCH, vol. 47, 2000, pages 57 - 77
SCHAIBLE UE; WINAU F; SIELING PA; FISCHER K; COLLINS HL; HAGENS K ET AL.: "Apoptosis facilitates antigen presentation to T lymphocytes through MHC-I and CD1 in tuberculosis", NAT MED, vol. 9, 2003, pages 1039 - 46
SEWELL WA; JOLLES S.: "Immunomodulatory action of intravenous immunoglobulin", IMMUNOLOGY, vol. 107, 2002, pages 387 - 93
TASCON RE; COLSTON MJ; RAGNO S; STAVROPOULOS E; GREGORY D; LOWRIE DB: "Vaccination against tuberculosis by DNA injection", NAT MED, vol. 2, 1996, pages 888 - 92
TRIPATHY SP.: "Trial of BCG vaccines in South India for tuberculosis prevention: First Report", BULLETIN OF THE WORLD HEALTH ORGANIZATION, vol. 57, 1979, pages 819 - 827
UNDAR LEVENT ET AL: "Tuberculosis-associated haemophagocytic syndrome: A report of two cases and a review of the literature", ACTA HAEMATOLOGICA (BASEL), vol. 96, no. 2, 1996, pages 73 - 78, XP009038454, ISSN: 0001-5792 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2435474A2 (en) 2009-05-27 2012-04-04 Baxter International Inc A method to produce a highly concentrated immunoglobulin preparation for subcutaneous use
US9175068B2 (en) 2009-05-27 2015-11-03 Baxalta Incorporated Method to produce a highly concentrated immunoglobulin preparation for subcutaneous use
US10125189B2 (en) 2009-05-27 2018-11-13 Baxalta Incorporated Method to produce a highly concentrated immunoglobulin preparation for subcutaneous use
US11242380B2 (en) 2009-05-27 2022-02-08 Takeda Pharmaceutical Company Limited Method to produce a highly concentrated immunoglobulin preparation for subcutaneous use
WO2015015001A1 (en) 2013-08-01 2015-02-05 Suppremol Gmbh In vitro method for determining the stability of compositions comprising soluble fc gamma receptor(s)
US20160195541A1 (en) * 2013-08-01 2016-07-07 Suppremol Gmbh In vitro method for determining the stability of compositions comprising soluble Fc gamma receptor(s)
US10866247B2 (en) * 2013-08-01 2020-12-15 Suppremol Gmbh In vitro method for determining the stability of compositions comprising soluble Fc gamma receptor(s)

Also Published As

Publication number Publication date
US7687060B2 (en) 2010-03-30
US20060275311A1 (en) 2006-12-07
EP1664119A1 (en) 2006-06-07

Similar Documents

Publication Publication Date Title
AU2017220184B2 (en) Methods for enhancing efficacy of a vaccine by administering an IL-4R antagonist
CA2769394C (en) Human monoclonal antibody against s. aureus derived alpha-toxin and its use in treating or preventing abscess formation
EP1137766B1 (en) Use of il-12 antibodies to treat psoriasis
Mody et al. Depletion of murine CD8+ T cells in vivo decreases pulmonary clearance of a moderately virulent strain of Cryptococcus neoformans
EP0744957B1 (en) Composition and method for preventing and treating inflammation with immunoglobulin a
KR102326482B1 (en) Treatment for dermatological pathologies
KR20140057635A (en) Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
Chace et al. Effect of anti-CD4 on CD4 subsets. I. Anti-CD4 preferentially deletes resting, naive CD4 cells and spares activated CD4 cells.
EP1230932B1 (en) Use of antibodies for anticancer vaccination
US20060275311A1 (en) Method for the treatment or prophylaxis of tuberculosis
McClelland et al. 3 Clinical Use of Immunoglobulins
WO2011133636A1 (en) COMBINATION THERAPY WITH CD4 LYMPHOCYTE DEPLETION AND mTOR INHIBITORS
US6410019B1 (en) Use of G class immunoglobulins for the topical treatment of atopic dermatitis
Sullivan Immunomodulation in allogeneic marrow transplantation: use of intravenous immune globulin to suppress acute graft‐versus‐host disease
Yang et al. Antibody-mediated protection against Staphylococcus aureus dermonecrosis: synergy of toxin neutralization and neutrophil recruitment
Camussone et al. Immune response and functional role of antibodies raised in heifers against a Staphylococcus aureus CP5 lysate and recombinant antigens vaccine formulated with Iscom Matrix adjuvant
Susuki et al. Effect of methylprednisolone in patients with Guillain-Barré syndrome
Pritchard Novel therapeutic approaches to Guillain-Barré syndrome
JP2006500386A (en) Anti-Yersinia vaccine comprising an antibody specific for the F. pestis F1 antigen and one or two of the antibodies specific for the B. pestis V antigen
US20100239595A1 (en) Equine antibodies against bacillus anthracis for passive immunization and treatment
US20220332805A1 (en) Treatment of Skin Blistering Diseases Using Antibodies
Munster et al. Effect of lymphocytotherapy on the course of experimental Pseudomonas sepsis.
Salinas-Carmona et al. Immunogenicity is unrelated to protective immunity when induced by soluble and particulate antigens from Nocardia brasiliensis in BALB/c mice
CN118234749A (en) Method for preventing graft rejection in xenografts
Reljic et al. Immunotherapy of tuberculosis with IgA and cytokines

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 11371262

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2004768377

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2004768377

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 11371262

Country of ref document: US