WO2005027965A1 - Antibodies against annexins, use thereof for therapy and diagnosis. use of annexins for therapy and diagnosis. - Google Patents

Antibodies against annexins, use thereof for therapy and diagnosis. use of annexins for therapy and diagnosis. Download PDF

Info

Publication number
WO2005027965A1
WO2005027965A1 PCT/EP2004/010756 EP2004010756W WO2005027965A1 WO 2005027965 A1 WO2005027965 A1 WO 2005027965A1 EP 2004010756 W EP2004010756 W EP 2004010756W WO 2005027965 A1 WO2005027965 A1 WO 2005027965A1
Authority
WO
WIPO (PCT)
Prior art keywords
annexin
antibody
cells
fragment
apoptosis
Prior art date
Application number
PCT/EP2004/010756
Other languages
French (fr)
Inventor
Peter Krammer
Heiko Weyd
Original Assignee
Peter Krammer
Heiko Weyd
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 Peter Krammer, Heiko Weyd filed Critical Peter Krammer
Priority to EP04765595A priority Critical patent/EP1670510A1/en
Publication of WO2005027965A1 publication Critical patent/WO2005027965A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4721Lipocortins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • 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

  • ANTIBODIES AGAINST NNEXINS , USE THEREOF FOR THERAPY ND DIAGNOSIS , USE OF ANNEXINS FOR THERAPY AND DIAGNOSIS .
  • the present invention relates to anti-annexin antibodies and their uses as well as uses of theirs ligands.
  • Such annexins and anti-annexin antibodies are useful for detecting apoptosis and for the production of pharmaceutical compositions for the diagnosis and/or treatment of diseases linked to apoptosis, such as cancer, autoimmune diseases, cardiovascular and/or vascular diseases.
  • Apoptosis constitutes the most common form of cell death throughout the lifespan of an organism. During development and adulthood the removal of excess cells by apoptosis ensures tissue homeostasis. 1,2
  • apoptotic stimuli such as cytotoxic agents e.g. staurosporine, ⁇ -irradiation or the engagement of the CD95 (APO-1/Fas) receptor
  • cell death proceeds via activation of caspases, finally leading to the formation of membrane enclosed apoptotic bodies.
  • apoptotic cells have been reported to mediate anti-inflammatory signals to phagocytosing cells, thereby actively down-regulating the immune response. 9,10 Thus, in contrast to necrotic cell death, apoptosis causes little, if any, immune response.
  • Annexin I belongs to a well conserved family of lipid and calcium binding proteins with tissue- and development-specific expression. 11 Expression of annexin I can be detected in a variety of tissues including all cells of hematopoietic origin except erythrocytes, neuronal cells and epithelial as well as endothelial cells. 12 In contrast to other members of the annexin family like annexin IV and V, annexin I expression is absent in ceils of the hepato- pancreatic system in the adult organism. However, expression of annexin I in hepatocytes can be detected during early stages of embryogenesis.
  • annexin I expression can also be induced in adult hepatocytes after treatment with IL-6 and HGF and was reported to be strongly expressed in human hepatocellular carcinoma cells 13"15 . Therefore, expression of different members of the annexin family by a specific cell is well regulated and shows considerable plasticity. As determined so far, most if not all tissues express members of the annexin family to varying degrees. Notably, tissues in which expression of annexin I, II and IV cannot be detected show expression of annexin V instead, as for example astrocytes and skeletal muscle as well as cardiac muscle cells. 16,17
  • annexins differ considerably in their N-terminus. 11 While annexin V possesses an N-terminus of only 14 amino acids 18,19 , the N-terminus of annexin I consists of 40 amino acids and contains several target sequences for phosphorylation. 20 In a number of studies, annexin I has been implicated in the suppression of inflammation. While originally described as an intracellular mediator of glucocorticoid actions, 21,22 an extracellular form of annexin I has been shown to regulate neutrophil extravasation through interaction with the formyl peptide receptor.
  • mice deficient for annexin I show increased and aberrant inflammation, 24 adding further evidence to a role for annexin I in the control of inflammation.
  • Reported anti-inflammatory properties of annexin I have been linked to its unique N- terminus as well as to a sequence of its core domain, which shows homology to a class of anti inflammatory peptides, known as antiflammins.
  • 25 Neutrophils represent the largest entity of lymphocytes in the blood. Due to their short half-life, neutrophils also constitute the major load of apoptotic cells to be disposed of under physiological conditions. 17 Neutrophils also express high levels of endogenous annexin I, 18 which is translocated to the surface upon prolonged culture in vitro. Simultaneously, the N-terminus of annexin I is cleaved (Fig. 4).
  • DCs Dendritic cells
  • LPS lipopoly- saccharides
  • USP 5,658,877 discloses pharmaceutical compositions containing safe and effective amounts of annexin I and methods for treating lung disease by alleviating the effects of endotoxin.
  • USP 5,632,986 discloses a conjugate comprising an annexin covalently bound to urokinase.
  • the preferred annexin is annexin V.
  • USP 6,312,694 discloses methods for killing tumor vascular endothelial cells by administering a binding ligand comprising a cytotoxic agent operatively linked to a targeting agent.
  • a targeting agent comprising a cytotoxic agent operatively linked to a targeting agent.
  • One of the targeting agents mentioned is annexin V.
  • annexin V There is no hint of a connection between annexin V and apoptosis.
  • US 5,296,467 discloses pharmaceutical compositions consisting essentially of a vascular anticoagulant annexin, Ca 2+ and Zn ⁇ ⁇ 2+
  • Apoptosis is a mechanism which could be used to eliminate cells without the deleterious effects of necrosis. It would be highly desirable if this mechanism could be used for cancer therapy.
  • This object is solved by providing an anti-annexin antibody or a fragment thereof which is capable of binding specifically to an annexin present on a cell that is undergoing apoptosis.
  • an antibody generated to be specific for an annexin in particular anti-annexin I and further anti-annexin II, anti-annexin IV and anti-annexin V antibodies as well as antibodies against other members of the annexin family, can be used to detect apoptosis and to diagnose, monitor and/or treat a variety of conditions including cancer.
  • Such an anti-annexin antibody or fragment thereof preferably binds to annexin I or annexin V.
  • Further preferred antibodies of the present invention include anti- annexin I and further anti-annexin II, anti-annexin IV and anti-annexin V antibodies as well as antibodies against other members of the annexin family. Such antibodies can be used for example and without being limited thereto, for the detection of apoptosis in vitro, ex vivo or in vivo.
  • a preferred such antibody of the present invention is DAC5 or a fragment thereof, which is described in the experimental part.
  • DAC5 for "Detector of Apoptotic Cells Nr.5", recognized a protein on the surface of early apoptotic but not live Jurkat cells (Fig 1a). Binding kinetics of DAC5 resembled the kinetics of the widely used early apoptosis marker annexin V.
  • DAC5 binding occurred before membrane integrity of the cells was lost, as measured by propidium iodide (PI) uptake. This indicates that the antigen was localized on the cell surface. Binding was not restricted to the apoptotic stimulus nor to the Jurkat cell line, as binding occurred also after treatment with leucine zipper CD95 (LZ-CD95), which triggers the CD95 (APO-1/Fas) pathway, as well as upon ⁇ -irradiation (Fig 1b). Cells binding DAC5 upon apoptosis included primary human T cells, HeLa cells and the melanoma cell line A375 (data not shown). Using immunoprecipitation of Jurkat lysates resolved on a silverstained SDS-PAGE and subsequent mass spectroscopy, the antigen of DAC5 was identified as annexin I (Fig 2).
  • annexin I upon induction of apoptosis, annexin I is translocated to the outer cell membrane, where it binds to negatively charged phospholipids in a calcium dependent fashion. Consequently, we could detect increasing amounts of annexins I, II and IV in EDTA membrane-washes of early apoptotic cells (Fig. 3).
  • the antibody of the present invention allows the detection and also the monitoring of all stages of apoptosis, including early stages.
  • annexin I probably serves as an endogenous "anti-inflammation" signal on the surface of apoptotic cells.
  • annexin I prevents the activation of antigen presenting cells that have engulfed apoptotic cells.
  • annexin I helps to maintain tolerance towards self antigens derived from apoptotic cells and might serve as a powerful tool to control immune responses during transplantation and autoimmunity.
  • anti-annexin I antibodies such as DAC5 can be used to block the development of tolerance against tumors.
  • DAC5 might also facilitate the detection of apoptotic cells in vivo.
  • tumors were established in immune-compromised NOD/SCI D mice, using apoptosis sensitive (J16) and apoptosis resistant (Rapo) Jurkat clones, respectively.
  • Apoptosis in the sensitive tumor cells was induced through i.v. injection of the apoptosis inducing antibody anti APO-1. 26 24 h after injection tumors were isolated and tested for apoptosis by flowcytometry. Indeed, the sensitive tumor cell showed 30% apoptosis, while while the resistant tumor showed only background apoptosis levels (Fig 6).
  • DAC5-binding to the sensitive tumor in vivo the antibody was labeled with radioactive 131 l and tested for binding on apoptotic cells. As can be seen in Fig 7, 131 l DAC5 retained its capability to bind to apoptotic cells. To verify the potential of DAC5 to detect apoptotic cells in vivo, further studies are ongoing. In addition, a recent report from Oh ei al. showed the presence of annexin I on the outer surface of endothelial cells lining the vessels in the vicinity of tumors. Tumor therapy using an anti-annexin I antibody was successful in reducing tumor burden and survival of tumor bearing rats. 27 Therefore, anti-annexin I antibodies might as well be suitable for targeting therapeutic agents directly to tumors, even if tumor cells themselves do not express annexin I.
  • the antibody of the invention is preferably a monoclonal antibody.
  • the antibody of the invention can be a whole antibody, or it can be a fragment, such as a Fab or F(ab') 2 fragment.
  • the antibody or fragment according to a preferred embodiment of this invention is labeled.
  • the label should enable detection in vitro or in vivo or ex vivo, preferably in such a way that a diagnosis in vivo can be performed.
  • Labels which are suitable therefore are known to the man in the art as well as methods for detecting this label. Examples for such labels include radioactive labels, luminescent labels. An example is FITC.
  • the label can also be an effector molecule or a toxic substance, e.g. a cytotoxic effector molecule which is able to destroy cells, e.g. tumor cells.
  • a toxic substance e.g. a cytotoxic effector molecule which is able to destroy cells, e.g. tumor cells.
  • Examples are radioactive or toxic substances.
  • Such substances can be very specifically directed to the tumor locus by using the antibody according to the present invention, and such antibodies bind to tumor cells which due to apoptosis show annexin I or other annexins on their surface.
  • the effector substance which is coupled to the antibody can destroy not only the cell to which the antibody is coupled via annexin I, but also destroys cells in the environment of such apoptotic cells, which themselves are not yet apoptotic or do not surrender to apoptosis for whatever reason.
  • annexins in general, especially annexin I and further also annexin II, IV and V, as well as anti-annexin antibodies, such as anti-annexin I and further anti-annexin II, anti- annexin IV and anti-annexin V antibodies as well as antibodies against other members of the annexin family.
  • annexins are expressed on cells that are undergoing apoptosis. Such cells are often found in tissue affected by diseases such as cancer or tumor tissue, and tissue affected by autoimmune diseases such as rheumatoid arthritis, lupus erythematosus and multiple sclerosis, as well as diabetes and cardiovascular and vascular diseases.
  • anti-annexin antibodies can actually be used to modulate immune responses.
  • the use of the antibody according to the present invention in therapy and especially in tumor therapy as well as in support of commonly used tumor therapies can be mediated by the following mechanism: Cells showing annexin I on their surface block inflammatory signals within the immune system. Therefore, such cells are not destroyed efficiently by the immune apparatus.
  • antibodies bind to the annexin I molecules they block the anti-inflammatory signals that are presented to the immune effector cell. As a result, an inflammation can take place which itself supports the immune response in for example cancer therapy as well as support of an immune response against self-antigens.
  • the anti- annexin antibodies When such antibodies are brought in contact with cells or tissue containing cells that are undergoing apoptosis, the anti- annexin antibodies will bind to the annexins expressed on the apoptosing cells. This allows not only the use of such antibodies for closely monitoring and detecting apoptosis in vivo, in vitro and ex vivo, but also the specific targeting of such cells and/or tissues for diagnostic and/or therapeutic purposes. By way of such targeting, it is possible to modulate the immune system with regard to its responses to such cells.
  • anti-annexin antibodies directed to such cells will block such anti-inflammatory signals and thus elicit and/or increase the inflammatory response to the cells and also to the surrounding cells. This way, it is possible to use such anti-annexin antibodies to induce and/or increae an inflammatory response to tumor tissue.
  • anti-annexin antibodies which are coupled to an effector molecule which can be a toxic substance or a radioactive substance or a marker.
  • an effector molecule which can be a toxic substance or a radioactive substance or a marker.
  • the anti-annexin antibody will be able to not only target apoptosing cells such as those present in tumor tissue, but also to target the surrounding cells and damage and/or kill them.
  • anti-annexin antibodies will be very effective as anti-tumor agents, or they can be used to support or complement existing tumor and cancer therapies.
  • Labelled anti-annexin antibodies are very useful as diagnostic agents.
  • One possible application of such antibodies is in the diagnosis of cancer. After an initial conventional cancer therapy, such as chemotherapy or radiation therapy, labelled anti-annexin antibodies can be administered to a patient.
  • Such antibodies will bind annexins present on or in association with cells that have started to undergo apoptosis. Since apoptosis is likely to increase after cancer therapy, the antibodies will be able to detect tumor tissue and even micrometastases and other tumor cells which are otherwise very difficult to detect. Thus, the antibodies will allow a more specific detection and thus a directed and specific treatment of tumor cells and tumor tissue.
  • anti- annexin antibodies could then be used to target those tumor cells and tumor tissues containing apoptosing cells to delete or damage the tumor cells and tissues by using antibodies coupled to effector molecules.
  • antibodies coupled to effector molecules can be used to supplement and/or improve existing tumor therapies.
  • the antibodies need not necessarily be labeled, whereas for other therapeutic or diagnostic methods it is preferred to use a labeled antibody.
  • This label can be a molecule which allows for detection of the bound antibodies in vitro or in vivo or a substance that is able to destroy cells, like for example radioactivity or a toxic substance.
  • substances which might be coupled to the antibodies according to the present invention are known to a person skilled in the art.
  • autoimmune diseases which are also associated with cells undergoing apoptosis.
  • diseases include, but are not limited to rheumatoid arthritis, lupus erythematosus, multiple sclerosis and diabetes and many other autoimmune diseases.
  • rheumatoid arthritis for example, it is possible to use anti-annexin antibodies to specifically target synovial cells and induce an inflammatory response to such cells and thereby delete them more efficiently.
  • Other diseases than can be diagnosed and/or treated include, but are not limited to vascular and cardiovascular diseases
  • anti-annexins function as anti-inflammatory signals, they might also be involved in the development of tolerance against tumor cells and other cells undergoing apoptosis.
  • anti-annexin antibodies as described above can also be used for blocking the development of tolerance against tumor cells and/or tumor tissue.
  • the use of the present antibody for treating tumors may lead to complete eradication of cancer cells with no remaining resistance.
  • the apoptotic cells showing annexin I on their surface are bound specifically and destroyed by the effector agent, the cells surrounding such apoptotic cells, however, are also destroyed due to the effect of the effector molecule.
  • the effector molecules can be selected according to their efficiency and their range of effectiveness.
  • annexin I on the vessel walls of tumor tissue might target anti-annexin I antibodies directly to tumors, even if the tumors do not express annexin I. Accordingly, anti-annexin I antibodies and/or anti-annexin II, anti-annexin IV and anti-annexin V antibodies as well as antibodies against other members of the annexin family might be useful in therapy of any vascularized tumor, including small cell lung carcinoma.
  • annexin I antibodies include all annexin I expressing tumors like T cell lymphoma, Melanoma and tumors of epithelial and endothelial origin and neuronal tumors. 12,15
  • antibodies against annexins II, IV and V could be used to monitor malignancies that express the respective annexins, including tumors of the hepato-pancreatic system, of the intestinal tract and of muscle tissues. 12
  • the use of the present antibody can also support and/or complement a cancer therapy.
  • Common tumor therapies often are able to destroy the majority of tumor cells, but for largely unknown reasons some cells remain resistant to tumor therapy and cannot be destroyed. Such cells might be the cause for further tumor growth after the first therapy has been terminated.
  • the use of the present antibody for tumor therapy can prevent such later growth of tumors from resistant tumor cells.
  • such antibodies can be used for a diagnosis of diseases with concomitant increased apoptosis rate of cells, as for example diabetes and auto-immune diseases.
  • diseases include those mentioned above, including cancer but also diseases such as diabetes, autoimmune diseases and also cardiovascular and vascular diseases, such as those mentioned above.
  • the invention relates to a method of detecting apoptosis comprising: (i) providing a sample to be analysed, comprising cells; (ii) detecting an annexin present on the surface of said cells by adding a substance capable of specifically binding to an annexin present on a cell that is undergoing apoptosis.
  • the substance of step (ii) is an antibody or fragment thereof capable of specifically binding to an annexin present on a cell that is undergoing apoptosis.
  • the annexin to be detected is preferably annexin I, annexin II, annexin IV or annexin V, in particular full-length annexin I having a molecular weight of about 38 kDa.
  • the antibody of fragment thereof is preferably specific for annexin I, in particular the 38 kDa annexin I (see Fig. 4).
  • annexin I is used as well as other members of the annexin family for certain indications. From the studies connected to the present invention, it could be gathered that annexin I, when presented on the cell surface, gives an anti-inflammation signal. Such anti-inflammation signals also occur when foreign tissues are transplanted into a patient.
  • the use of annexin I can avoid such problem when administered to a patient or to tissue to be transplanted. Presentation of annexin l on such tissues suppresses the immune response that otherwise might be initiated due to immune recognition of the transplanted tissue.
  • annexin I II, IV or V or fragments thereof leads to a marking of such transplantation tissue that avoids an inflammatory effect.
  • annexin I and other annexins are related to specific receptors, which again can be stimulated or blocked by either binding of one of the annexins or fragments thereof or an antibody against this receptor.
  • Such binding of receptors by an annexin or an antibody against the annexin-receptor can also influence the immune response and lead to the same possibilities for therapy as described above for the annexin antibodies or the use of annexins in transplantation.
  • annexins and/or functional fragments thereof and/or fusion proteins comprising an annexin or functional fragments thereof can be used to modulate the immune system.
  • annexin proteins When annexin proteins are isolated or recombinantly produced and contacted with cells and/or tissues containing cells undergoing apoptosis, they will bind to the surface of apoptotic cells. In some cases where there is increased cell death, the naturally occurring levels of annexin may not be sufficient to inhibit or suppress an inflammatory response, and thus the administration of annexins is useful for the prevention or reduction of such inflammation, thereby exerting a therapeutic or beneficial effect. Thus, they can be used to produce the same signals that annexins expressed by the apoptotic cells themselves will elicit.
  • annexins can be used to block inflammation or an inflammatory response.
  • Diseases which are linked to inflammation and/or cell death include, but are not limited to ischaemia- reperfusion damage, stroke, chronic heart failure, myocardial infarction, spinal cord injury, acute liver failure, renal ischaemia, neurodegenerative diseases such as Alzheimer and Parkinson's disease, sepsis and HIV-infection.
  • the annexins could be used to inhibit inflammatory responses to tissues such as transplantation tissues and the like.
  • Other diseases that can also be treated by the administration of annexins or functional fragments thereof are autoimmune diseases such as multiple sclerosis, lupus erythenatosus, rheumatoid arthritis, diabetes and others.
  • FIGURE 1 DAC5 binds to the surface of apoptotic cells a) Jurkat T cells were induced to undergo apoptosis by addition of 1 ⁇ M staurosporine (sts). At different time points, 2x10 5 cells per sample were incubated with 100 ⁇ l DAC5 supernatant or 1 ⁇ g/ml lgG2a isotype control Ab (iso), followed by incubation with FITC-labelled anti-mouse IgG antibodies. To discriminate between early and late apoptosis, aliquots were incubated in parallel with FITC-labelled annexin V (AxV) or 2 ⁇ g/ml propidium iodide (PI).
  • AxV FITC-labelled annexin V
  • PI propidium iodide
  • the kinetic is representative for 3 experiments.
  • Inset shows a representative DAC5 staining after 4 hours incubation with staurosporine (shaded histogram, in comparison to DAC5 staining on live cells.
  • Jurkat T cells were irradiated with 150 Gy and cultured for additional 6 hours. Alternatively, cells were incubated for 6 hours with 1 ⁇ M staurosporine (sts) or 1 ⁇ g/ml leucine zipper CD95 ligand (LZ-CD95L). Subsequently, cells were stained with FITC-labeled annexin V or FITC-labeled DAC5 in annexin Binding buffer.
  • FIGURE 2 DAC5 precipitates annexin I a)
  • Postnuclear lysates of 10 7 CEM cells were immmunoprecipitated with Protein A sepharose and purified DAC5 or lgG2a isotype control (iso), or protein A sepharose alone (-). Precipitates were resolved on a 12% SDS PAGE and silver stained. The protein bands labeled "annexin I" were cut from the gel and subjected to tryptic digest, followed by analysis via mass spectroscopy. By comparison with the NCBInr database peptides were identified as derived from annexin I. he - heavy chain; lc - light chain.
  • FIGURE 3 annexin I is bound to the outside of apoptotic cells
  • FIGURE 4 annexin I on the surface of cultured neutrophils
  • annexin I protein levels were monitored on Westernblot, using DAC5 and a peroxidase coupled, secondary anti mouse IgG antibody.
  • FIGURE 5 annexin I suppresses DC cytokine secretion
  • 2x10 5 d7 immature DCs were incubated for 4h with or without 50 ⁇ g/ml DAC5-F (ab') 2 -Fragments and 1x10 6 d1 neutrophils.
  • d1 neutrophils were added in a transwell chamber and d4 neutrophils were used, respectively.
  • 50ng/ml LPS were added to the wells for another 48h.
  • Cells were then harvested, stained with antibodies against CD83, CD86 and HLA-DR and analyzed by flowcytometry (a). Aliquots of cell culture supematants were collected 24 h after addition of LPS and analyzed with ELISAs for TNF (b).
  • FIGURE 6 Jurkat tumor model 5x10 6 apoptosis sensitive Jurkat J16 cells or apoptosis resistant Rapo cells were injected s.c. into NOD/SCID-mice. When tumors of ca. 8 mm in diameter were established, the mice received an injection of 250 ⁇ g anti-APO-1. After 24 h the tumors were explanted. Cells were separated and analyzed for apoptosis using flowcytometric analysis of DNA content after Nicoletti et al (ref).
  • FIGURE 7 lodinated DAC5 binds to apoptotic cells in vitro 1x10 6 CEM cells were irradiated with 200mJ/cm 2 UV-C light and cultured for another 4h. Apoptotic cells were then incubated with 131 I-DAC5 or 127 I-DAC5 for 20 in at 4°C. Cells incubated with the radioactively labeled DAC5 were centrifuged through oil and analyzed for bound radioactivity in a beta-counter. Cells stained with 127 l- DAC5 were analyzed by flowcytometry, using a secondary FITC-coupled anti mouse IgG antibody.
  • Example 1 Immunization and fusion C57BL/6 mice were kept under certified pathogen free conditions and immunized nine times i.p. with 2x 10 7 apoptotic J16 Jurkat T cells. Apoptosis was induced by incubation with 1 ⁇ M staurosporine (Sigma, Kunststoff, Germany) for
  • Example 2 CeW lines and hybridomas The human T-ALL cell line CEM and the human Jurkat T cell lines J16 and Rapo were cultured in RPMI 1640 (Gibco, Düsseldorf, Germany) supplemented with 10% fetal calf serum (Gibco, Düsseldorf, Germany) and 10 mM HEPES (Sigma, Kunststoff, Germany). Supernatant of the hybridoma DAC 5 was affinity purified using a Protein A-Sepharose column (Sigma, Kunststoff, Germany), followed by dialysis against PBS. F(ab') 2 fragments of DAC5 were generated using Immunopure kits (Pierce, Rockford, USA) according to manufacturers instructions.
  • DC primary human dendritic cells
  • adherent monocytes were washed thoroughly with PBS and seeded into 6 well cell culture plates at a density of 3x10 6 /well and cultured in X-Vivo 15 medium (Cambrex, Verviers, Belgium) supplemented with 2% inactivated donor serum and containing 1000 U/ml hu GM-CSF and 500 U/ml hu IL-4 (Immunotools, Osnabr ⁇ ck, Germany).
  • Fresh cytokines were supplemented again on day 3. Immature DCs were used on day 7, and >98% were positive for the DC marker CD11c.
  • Example 4 Preparation of primary human neutrophils Human neutrophils were obtained as described previously, 30 using density centrifugation of heparinized whole in PolymorphPrep (Axis Shield, Oslo, Norway). After preparation, neutrophils were cultured in X-Vivo 15 Medium
  • Example 5 Immunoprecipitation and silver staining
  • 1x10 7 J16 cells were lysed in Triton X-100 Lysisbuffer (20 mM Tris/HCI, pH 7.4, 1 % Triton X-100, 10 % glycerol, 150 mM NaCI, 1 mM PMSF and 1 ⁇ g/ml of Leupeptin, Antipain, Chymostatin and Pepstatin A) for 15 min on ice and centrifuged (15 min, 14000xg, 4°C). Supernatants were then subjected to immunoprecipitation with 10 ⁇ g DAC5 or isotype control antibody and Protein A Sepharose (Sigma, Kunststoff, Germany) at 4°C overnight. Precipitates were prepared in SDS sample buffer, resolved on a 12% SDS PAGE and stained with the Silverquest silver staining kit (Invitrogen, Paisley, UK), according to manufacturers instructions.
  • Example 6 Protein Sequencing Protein bands were excised from the silver stained gel, repeatedly washed with water, 40 mM ammonium bicarbonate/ ethanol (1:1 v/v), reduced with 10 mM DTT at 56 °C for 1 h and alkylated with 55 mM iodoacetamide at 25°C for 30 min in the dark. After alkylation, gel bands were repeatedly washed with 40mM ammonium bicarbonate and ethanol, dehydrated with 100% acetonitrile and air- dried for 15 min. Digestion with trypsin (10ng/ ⁇ l, Promega) was performed in 40 mM ammonium bicarbonate at 37°C overnight.
  • Example 7 MALDI-TOF mass spectrometry (MALDI-TOF-MS) Sample preparation was achieved by cocrystallization of ZipTip C18 (Millipore) concentrated samples with matrix. Peptides in the supernatant of the in-gel digest were absorbed to a prewashed (50% acetonitrile/water) and equilibrated (0.1% trifluoroacetic acid/water) ZipTip C18 by repetitive pipetting steps. Following washing of the ZipTip C18 by equilibration buffer the peptides were eluted from the ZipTip with 1 ⁇ l of matrix ( ⁇ -cyano-4-hydroxycinnamic acid saturated in 50% acetonitrile/water).
  • MALDI-TOF mass spectra were recorded in the positive ion reflector mode with delayed extraction on a Reflex II time-of- flight instrument (Bruker-Daltonik). Ion acceleration was set to 26.5 kV, the reflector was set to 30.0 kV and the first extraction plate was set to 20.6 kV. Mass spectra were obtained by averaging 50 to 200 individual laser shots. Calibration of the spectra was performed internally by a two-point linear fit using the autolysis products of trypsin at m/z 842.50 and m/z 2211.10.
  • Post source decay (PSD) analysis was performed in the positive ion reflector mode with delayed extraction by setting an ion gate width of 40 Da around the ion of interest.
  • Data were acquired in 14 segments by decreasing the reflector voltage in a stepwise fashion. For each segment 100 to 200 individual laser shots were accumulated. The fragment ion spectrum was obtained by pasting together all segments to a single spectrum using the FAST software provided by Bruker. Fragment ion calibration was performed externally with the fragment masses of the adrenocorticotropic hormone 18 - 39 clip.
  • Example 8 Database search Singly charged monoisotopic peptide masses were used as inputs for database searching. Searches were performed against the NCBInr database using the ProFound search algorithm (http://129.85.19.192/prowl- cgi/ProFound.exe). Isoelectric points were allowed to range from 0 to 14, carbamidomethylation was included as fixed modification and the oxidation of methionine was allowed as possible modification. Up to one missed tryptic cleavage was considered, and the mass tolerance for the monoisotopic peptide masses was set to +/-100 ppm.
  • Example 9 Surface staining 5 x 10 5 cells were incubated with 100 ⁇ l of hybridoma culture supernatant or with 10 ⁇ g/ml purified DAC5 in PBS/10% FCS for 30 min at 4°C, washed with 500 ⁇ l PBS/10% FCS, and incubated another 30 min with 5 ⁇ g/ml FITC-labeled goat anti mouse IgG antibodies (Dianova, Hamburg, Germany) and 2 ⁇ g/ml propidium iodide (Sigma, Kunststoff, Germany) in PBS/10% FCS. The addition of 10% FCS provided for calcium, necessary for annexin I to bind to the membrane of apoptotic cells.
  • annexin V-FITC Molecular Probes, Leiden, Netherlands
  • Example 10 EDTA-washes and Western Blot analysis 5x10 6 CEM cells were incubated in 500 ⁇ l medium with 1 ⁇ M staurosporine for different time periods or left untreated. Subsequently, the cells were centrifuged, the supernatant was collected and pellets were washed with 500 ⁇ l PBS/1 OmM EDTA. After centrifugation the washing solution was collected and the pellets were lysed in 500 ⁇ l Triton X-100 Lysis buffer.
  • Example 11 Cocultures of immature DCs and neutrophils Immature d6 DCs were plated at a density of 2x10 5 cells/well into 24-well plates and incubated over night. The next day, d1 neutrophils were incubated for 15 min at 37° with 50 ⁇ g/ml DAC5 F(ab') 2 fragments or left untreated, and overlaid onto the DCs at a ratio of 1:1.
  • Haslett.C Savill.J.S. & Meagher.L. The neutrophil. Curr. Opin. Immunol. 2, 10-18 (1989).

Abstract

The present invention relates to anti-annexin antibodies and their uses as well as uses of theirs ligands, the annexins. Such annexins and anti-annexin antibodies are useful for detecting apoptosis and for the production of pharmaceutical compositions for the diagnosis and/or treatment of cancer, autoimmune diseases, cardiovascular and/or vascular diseases.

Description

ANTIBODIES AGAINST NNEXINS , USE THEREOF FOR THERAPY ND DIAGNOSIS , USE OF ANNEXINS FOR THERAPY AND DIAGNOSIS .
Description
The present invention relates to anti-annexin antibodies and their uses as well as uses of theirs ligands. Such annexins and anti-annexin antibodies are useful for detecting apoptosis and for the production of pharmaceutical compositions for the diagnosis and/or treatment of diseases linked to apoptosis, such as cancer, autoimmune diseases, cardiovascular and/or vascular diseases.
Apoptosis constitutes the most common form of cell death throughout the lifespan of an organism. During development and adulthood the removal of excess cells by apoptosis ensures tissue homeostasis.1,2
Upon apoptotic stimuli such as cytotoxic agents e.g. staurosporine, γ-irradiation or the engagement of the CD95 (APO-1/Fas) receptor, cell death proceeds via activation of caspases, finally leading to the formation of membrane enclosed apoptotic bodies.3,4
To preclude the release of noxious cellular contents, the remnants of dying cells are rapidly cleared by neighboring cells and phagocytes like macrophages or dendritic cells.5,6 This efficient and safe uptake is mediated via specific signals or ligands, which must provide for engulfment as well as prevention of immune responses against self antigens.7,8
Accordingly, apoptotic cells have been reported to mediate anti-inflammatory signals to phagocytosing cells, thereby actively down-regulating the immune response.9,10 Thus, in contrast to necrotic cell death, apoptosis causes little, if any, immune response.
Annexin I belongs to a well conserved family of lipid and calcium binding proteins with tissue- and development-specific expression.11 Expression of annexin I can be detected in a variety of tissues including all cells of hematopoietic origin except erythrocytes, neuronal cells and epithelial as well as endothelial cells.12 In contrast to other members of the annexin family like annexin IV and V, annexin I expression is absent in ceils of the hepato- pancreatic system in the adult organism. However, expression of annexin I in hepatocytes can be detected during early stages of embryogenesis. Moreover, annexin I expression can also be induced in adult hepatocytes after treatment with IL-6 and HGF and was reported to be strongly expressed in human hepatocellular carcinoma cells13"15. Therefore, expression of different members of the annexin family by a specific cell is well regulated and shows considerable plasticity. As determined so far, most if not all tissues express members of the annexin family to varying degrees. Notably, tissues in which expression of annexin I, II and IV cannot be detected show expression of annexin V instead, as for example astrocytes and skeletal muscle as well as cardiac muscle cells.16,17
Sharing a conserved lipid binding core, mammalian annexins differ considerably in their N-terminus.11 While annexin V possesses an N-terminus of only 14 amino acids 18,19, the N-terminus of annexin I consists of 40 amino acids and contains several target sequences for phosphorylation.20 In a number of studies, annexin I has been implicated in the suppression of inflammation. While originally described as an intracellular mediator of glucocorticoid actions,21,22 an extracellular form of annexin I has been shown to regulate neutrophil extravasation through interaction with the formyl peptide receptor.23 In addition, mice deficient for annexin I show increased and aberrant inflammation,24 adding further evidence to a role for annexin I in the control of inflammation. Reported anti-inflammatory properties of annexin I have been linked to its unique N- terminus as well as to a sequence of its core domain, which shows homology to a class of anti inflammatory peptides, known as antiflammins.25 Neutrophils represent the largest entity of lymphocytes in the blood. Due to their short half-life, neutrophils also constitute the major load of apoptotic cells to be disposed of under physiological conditions.17 Neutrophils also express high levels of endogenous annexin I,18 which is translocated to the surface upon prolonged culture in vitro. Simultaneously, the N-terminus of annexin I is cleaved (Fig. 4).
Dendritic cells (DCs) are the most potent antigen presenting cells and their activation and maturation are critical for the outcome of an immune response.19 Residing in an immature state in peripheral tissues, DCs can be activated upon encountering danger signals like the bacterial cell wall component lipopoly- saccharides (LPS).20,2 Following activation, DCs mature and migrate towards lymphatic tissues. DC maturation induces the expression of costimulatory surface markers like MHC class II molecules and members of the B7 family as well as the secretion of pro-inflammatory cytokines like TNFα and IL-12.22,23 Under steady state conditions, DCs have been shown to contain engulfed apoptotic material.24 Under these conditions, molecules on the surface of these engulfed apoptotic cells might prevent subsequent activation of the respective DCs and, thus, preclude an autoimmune response.25
USP 5,658,877 discloses pharmaceutical compositions containing safe and effective amounts of annexin I and methods for treating lung disease by alleviating the effects of endotoxin.
USP 5,632,986 discloses a conjugate comprising an annexin covalently bound to urokinase. The preferred annexin is annexin V.
USP 6,312,694 discloses methods for killing tumor vascular endothelial cells by administering a binding ligand comprising a cytotoxic agent operatively linked to a targeting agent. One of the targeting agents mentioned is annexin V. There is no hint of a connection between annexin V and apoptosis. US 5,296,467 discloses pharmaceutical compositions consisting essentially of a vascular anticoagulant annexin, Ca2+ and Zn ι2+
Apoptosis is a mechanism which could be used to eliminate cells without the deleterious effects of necrosis. It would be highly desirable if this mechanism could be used for cancer therapy.
It was therefore an object of the present invention to provide substances which allow the detection and/or monitoring apoptosis.
This object is solved by providing an anti-annexin antibody or a fragment thereof which is capable of binding specifically to an annexin present on a cell that is undergoing apoptosis.
It was surprisingly found that an antibody generated to be specific for an annexin, in particular anti-annexin I and further anti-annexin II, anti-annexin IV and anti-annexin V antibodies as well as antibodies against other members of the annexin family, can be used to detect apoptosis and to diagnose, monitor and/or treat a variety of conditions including cancer.
Such an anti-annexin antibody or fragment thereof preferably binds to annexin I or annexin V. Further preferred antibodies of the present invention include anti- annexin I and further anti-annexin II, anti-annexin IV and anti-annexin V antibodies as well as antibodies against other members of the annexin family. Such antibodies can be used for example and without being limited thereto, for the detection of apoptosis in vitro, ex vivo or in vivo.
A preferred such antibody of the present invention is DAC5 or a fragment thereof, which is described in the experimental part. In order to discover new molecules involved in the signaling of apoptotic cells towards phagocytes, mice were immunized with apoptotic Jurkat cells and monoclonal antibodies were generated. One of these antibodies, termed DAC5 for "Detector of Apoptotic Cells Nr.5", recognized a protein on the surface of early apoptotic but not live Jurkat cells (Fig 1a). Binding kinetics of DAC5 resembled the kinetics of the widely used early apoptosis marker annexin V.
Notably, DAC5 binding occurred before membrane integrity of the cells was lost, as measured by propidium iodide (PI) uptake. This indicates that the antigen was localized on the cell surface. Binding was not restricted to the apoptotic stimulus nor to the Jurkat cell line, as binding occurred also after treatment with leucine zipper CD95 (LZ-CD95), which triggers the CD95 (APO-1/Fas) pathway, as well as upon γ-irradiation (Fig 1b). Cells binding DAC5 upon apoptosis included primary human T cells, HeLa cells and the melanoma cell line A375 (data not shown). Using immunoprecipitation of Jurkat lysates resolved on a silverstained SDS-PAGE and subsequent mass spectroscopy, the antigen of DAC5 was identified as annexin I (Fig 2).
It is shown here that upon induction of apoptosis, annexin I is translocated to the outer cell membrane, where it binds to negatively charged phospholipids in a calcium dependent fashion. Consequently, we could detect increasing amounts of annexins I, II and IV in EDTA membrane-washes of early apoptotic cells (Fig. 3).
Thus, the antibody of the present invention allows the detection and also the monitoring of all stages of apoptosis, including early stages.
To investigate the role of surface annexin I on apoptotic cells in the generation of immune responses, we tested the effects of annexin I positive d1 neutrophils on the maturation of dendritic cells (DC).
We therefore measured the TNFα release and DC surface markers after coincubation of immature DCs with annexin l-positive d1 neutrophils and subsequent LPS incubation. As a control we also incubated late apoptotic d4 neutrophils with DCs, in which the full length annexin I is almost completely degraded (Fig 4). In contrast to d4 neutrophils, d1 neutrophils inhibited the LPS- induced maturation of DCs as measured by TNFα secretion and upregulation of the maturation markers CD83, CD86 and HLA-DR (Fig.δa). When surface annexin I was blocked by the addition of DAC 5 F(ab)2-fragments, however, the suppression exerted by d1 neutrophils was remarkably released (Fig 5b).
Surprisingly, it was found that annexin I probably serves as an endogenous "anti-inflammation" signal on the surface of apoptotic cells. In the course of physiological tissue turnover, annexin I prevents the activation of antigen presenting cells that have engulfed apoptotic cells. Thereby, annexin I helps to maintain tolerance towards self antigens derived from apoptotic cells and might serve as a powerful tool to control immune responses during transplantation and autoimmunity. In addition, anti-annexin I antibodies such as DAC5 can be used to block the development of tolerance against tumors.
Being a marker specific for apoptotic cells, DAC5 might also facilitate the detection of apoptotic cells in vivo. To this aim, tumors were established in immune-compromised NOD/SCI D mice, using apoptosis sensitive (J16) and apoptosis resistant (Rapo) Jurkat clones, respectively. Apoptosis in the sensitive tumor cells was induced through i.v. injection of the apoptosis inducing antibody anti APO-1.26 24 h after injection tumors were isolated and tested for apoptosis by flowcytometry. Indeed, the sensitive tumor cell showed 30% apoptosis, while while the resistant tumor showed only background apoptosis levels (Fig 6).
To monitor DAC5-binding to the sensitive tumor in vivo, the antibody was labeled with radioactive 131l and tested for binding on apoptotic cells. As can be seen in Fig 7, 131l DAC5 retained its capability to bind to apoptotic cells. To verify the potential of DAC5 to detect apoptotic cells in vivo, further studies are ongoing. In addition, a recent report from Oh ei al. showed the presence of annexin I on the outer surface of endothelial cells lining the vessels in the vicinity of tumors. Tumor therapy using an anti-annexin I antibody was successful in reducing tumor burden and survival of tumor bearing rats.27 Therefore, anti-annexin I antibodies might as well be suitable for targeting therapeutic agents directly to tumors, even if tumor cells themselves do not express annexin I.
The antibody of the invention is preferably a monoclonal antibody. The antibody of the invention can be a whole antibody, or it can be a fragment, such as a Fab or F(ab')2 fragment.
The antibody or fragment according to a preferred embodiment of this invention is labeled. The label should enable detection in vitro or in vivo or ex vivo, preferably in such a way that a diagnosis in vivo can be performed. Labels which are suitable therefore are known to the man in the art as well as methods for detecting this label. Examples for such labels include radioactive labels, luminescent labels. An example is FITC.
The label can also be an effector molecule or a toxic substance, e.g. a cytotoxic effector molecule which is able to destroy cells, e.g. tumor cells. Examples are radioactive or toxic substances. Such substances can be very specifically directed to the tumor locus by using the antibody according to the present invention, and such antibodies bind to tumor cells which due to apoptosis show annexin I or other annexins on their surface. The effector substance which is coupled to the antibody can destroy not only the cell to which the antibody is coupled via annexin I, but also destroys cells in the environment of such apoptotic cells, which themselves are not yet apoptotic or do not surrender to apoptosis for whatever reason.
From above-described studies and the studies which are presented in the following examples, several applications and uses can be deduced for annexins in general, especially annexin I and further also annexin II, IV and V, as well as anti-annexin antibodies, such as anti-annexin I and further anti-annexin II, anti- annexin IV and anti-annexin V antibodies as well as antibodies against other members of the annexin family.
As mentioned above, annexins are expressed on cells that are undergoing apoptosis. Such cells are often found in tissue affected by diseases such as cancer or tumor tissue, and tissue affected by autoimmune diseases such as rheumatoid arthritis, lupus erythematosus and multiple sclerosis, as well as diabetes and cardiovascular and vascular diseases.
Surprisingly it was found that anti-annexin antibodies can actually be used to modulate immune responses. The use of the antibody according to the present invention in therapy and especially in tumor therapy as well as in support of commonly used tumor therapies can be mediated by the following mechanism: Cells showing annexin I on their surface block inflammatory signals within the immune system. Therefore, such cells are not destroyed efficiently by the immune apparatus. When antibodies bind to the annexin I molecules, they block the anti-inflammatory signals that are presented to the immune effector cell. As a result, an inflammation can take place which itself supports the immune response in for example cancer therapy as well as support of an immune response against self-antigens. When such antibodies are brought in contact with cells or tissue containing cells that are undergoing apoptosis, the anti- annexin antibodies will bind to the annexins expressed on the apoptosing cells. This allows not only the use of such antibodies for closely monitoring and detecting apoptosis in vivo, in vitro and ex vivo, but also the specific targeting of such cells and/or tissues for diagnostic and/or therapeutic purposes. By way of such targeting, it is possible to modulate the immune system with regard to its responses to such cells. Since annexins expressed on apoptotic cells appear to function as anti-inflammatory signals, anti-annexin antibodies directed to such cells will block such anti-inflammatory signals and thus elicit and/or increase the inflammatory response to the cells and also to the surrounding cells. This way, it is possible to use such anti-annexin antibodies to induce and/or increae an inflammatory response to tumor tissue.
In addition, it is also possible to use anti-annexin antibodies which are coupled to an effector molecule which can be a toxic substance or a radioactive substance or a marker. When a toxic or cytotoxic substance is used as the effector molecule, the anti-annexin antibody will be able to not only target apoptosing cells such as those present in tumor tissue, but also to target the surrounding cells and damage and/or kill them. Thus, such anti-annexin antibodies will be very effective as anti-tumor agents, or they can be used to support or complement existing tumor and cancer therapies.
Labelled anti-annexin antibodies are very useful as diagnostic agents. One possible application of such antibodies is in the diagnosis of cancer. After an initial conventional cancer therapy, such as chemotherapy or radiation therapy, labelled anti-annexin antibodies can be administered to a patient. Such antibodies will bind annexins present on or in association with cells that have started to undergo apoptosis. Since apoptosis is likely to increase after cancer therapy, the antibodies will be able to detect tumor tissue and even micrometastases and other tumor cells which are otherwise very difficult to detect. Thus, the antibodies will allow a more specific detection and thus a directed and specific treatment of tumor cells and tumor tissue. In addition, anti- annexin antibodies could then be used to target those tumor cells and tumor tissues containing apoptosing cells to delete or damage the tumor cells and tissues by using antibodies coupled to effector molecules. Thus, such antibodies can be used to supplement and/or improve existing tumor therapies.
For some purposes the antibodies need not necessarily be labeled, whereas for other therapeutic or diagnostic methods it is preferred to use a labeled antibody.
This label can be a molecule which allows for detection of the bound antibodies in vitro or in vivo or a substance that is able to destroy cells, like for example radioactivity or a toxic substance. As mentioned above, such substances, which might be coupled to the antibodies according to the present invention are known to a person skilled in the art.
The same mechanism can also be used to treat and/or diagnose other diseases such as autoimmune diseases, which are also associated with cells undergoing apoptosis. Such diseases include, but are not limited to rheumatoid arthritis, lupus erythematosus, multiple sclerosis and diabetes and many other autoimmune diseases. In rheumatoid arthritis for example, it is possible to use anti-annexin antibodies to specifically target synovial cells and induce an inflammatory response to such cells and thereby delete them more efficiently. Other diseases than can be diagnosed and/or treated include, but are not limited to vascular and cardiovascular diseases
Because the anti-annexins function as anti-inflammatory signals, they might also be involved in the development of tolerance against tumor cells and other cells undergoing apoptosis. Thus, anti-annexin antibodies as described above can also be used for blocking the development of tolerance against tumor cells and/or tumor tissue.
Therefore, the use of the present antibody for treating tumors, either alone or in combination with common tumor therapies, may lead to complete eradication of cancer cells with no remaining resistance. In such treatment the apoptotic cells showing annexin I on their surface are bound specifically and destroyed by the effector agent, the cells surrounding such apoptotic cells, however, are also destroyed due to the effect of the effector molecule. The effector molecules can be selected according to their efficiency and their range of effectiveness.
Furthermore, the presence of annexin I on the vessel walls of tumor tissue might target anti-annexin I antibodies directly to tumors, even if the tumors do not express annexin I. Accordingly, anti-annexin I antibodies and/or anti-annexin II, anti-annexin IV and anti-annexin V antibodies as well as antibodies against other members of the annexin family might be useful in therapy of any vascularized tumor, including small cell lung carcinoma.
This might be very important in cancer therapy, for example for monitoring the progress of an anti-tumoral therapy and further detection whether a tumor is amenable to such therapy. Malignancies whose therapy could be monitored using anti-annexin I antibodies include all annexin I expressing tumors like T cell lymphoma, Melanoma and tumors of epithelial and endothelial origin and neuronal tumors.12,15 In addition, antibodies against annexins II, IV and V could be used to monitor malignancies that express the respective annexins, including tumors of the hepato-pancreatic system, of the intestinal tract and of muscle tissues.12
In the field of cancer therapy, the use of the present antibody can also support and/or complement a cancer therapy. Common tumor therapies often are able to destroy the majority of tumor cells, but for largely unknown reasons some cells remain resistant to tumor therapy and cannot be destroyed. Such cells might be the cause for further tumor growth after the first therapy has been terminated. The use of the present antibody for tumor therapy can prevent such later growth of tumors from resistant tumor cells.
Also, such antibodies can be used for a diagnosis of diseases with concomitant increased apoptosis rate of cells, as for example diabetes and auto-immune diseases. Especially preferred is the use of the anti-annexin I antibody for the detection of apoptosis in vivo and for the monitoring of therapy. Such diseases include those mentioned above, including cancer but also diseases such as diabetes, autoimmune diseases and also cardiovascular and vascular diseases, such as those mentioned above.
Further, the invention relates to a method of detecting apoptosis comprising: (i) providing a sample to be analysed, comprising cells; (ii) detecting an annexin present on the surface of said cells by adding a substance capable of specifically binding to an annexin present on a cell that is undergoing apoptosis. Preferably, the substance of step (ii) is an antibody or fragment thereof capable of specifically binding to an annexin present on a cell that is undergoing apoptosis. The annexin to be detected is preferably annexin I, annexin II, annexin IV or annexin V, in particular full-length annexin I having a molecular weight of about 38 kDa. Thus, the antibody of fragment thereof is preferably specific for annexin I, in particular the 38 kDa annexin I (see Fig. 4).
Another subject matter of the present invention is the use of annexin I and/or further annexin II, annexin IV and annexin V as well as other members of the annexin family for certain indications. From the studies connected to the present invention, it could be gathered that annexin I, when presented on the cell surface, gives an anti-inflammation signal. Such anti-inflammation signals also occur when foreign tissues are transplanted into a patient. The use of annexin I can avoid such problem when administered to a patient or to tissue to be transplanted. Presentation of annexin l on such tissues suppresses the immune response that otherwise might be initiated due to immune recognition of the transplanted tissue.
Thus, administration of annexin I II, IV or V or fragments thereof leads to a marking of such transplantation tissue that avoids an inflammatory effect.
This effect obviously is true also for other members of the annexin family, wherefore use and/or application of other annexins to transplantation patients or tissue to be transplanted is encompassed by the present invention, especially annexin I, II, IV or V, as another means according to the present invention to avoid inflammatory immune reaction against the transplanted tissues.
A further possibility of using the results of the present invention is that annexin I and other annexins are related to specific receptors, which again can be stimulated or blocked by either binding of one of the annexins or fragments thereof or an antibody against this receptor. Such binding of receptors by an annexin or an antibody against the annexin-receptor can also influence the immune response and lead to the same possibilities for therapy as described above for the annexin antibodies or the use of annexins in transplantation.
Thus, annexins and/or functional fragments thereof and/or fusion proteins comprising an annexin or functional fragments thereof can be used to modulate the immune system. When annexin proteins are isolated or recombinantly produced and contacted with cells and/or tissues containing cells undergoing apoptosis, they will bind to the surface of apoptotic cells. In some cases where there is increased cell death, the naturally occurring levels of annexin may not be sufficient to inhibit or suppress an inflammatory response, and thus the administration of annexins is useful for the prevention or reduction of such inflammation, thereby exerting a therapeutic or beneficial effect. Thus, they can be used to produce the same signals that annexins expressed by the apoptotic cells themselves will elicit. Thus, contacting cells or tissues with annexin proteins will lead to the production of anti-inflammatory signals. This can be used for the diagnosis and/or treatment of diseases which are linked with cell death in general, apoptosis and/or inflammation. Thus, annexins can be used to block inflammation or an inflammatory response. Diseases which are linked to inflammation and/or cell death include, but are not limited to ischaemia- reperfusion damage, stroke, chronic heart failure, myocardial infarction, spinal cord injury, acute liver failure, renal ischaemia, neurodegenerative diseases such as Alzheimer and Parkinson's disease, sepsis and HIV-infection. In the same way, the annexins could be used to inhibit inflammatory responses to tissues such as transplantation tissues and the like. Other diseases that can also be treated by the administration of annexins or functional fragments thereof are autoimmune diseases such as multiple sclerosis, lupus erythenatosus, rheumatoid arthritis, diabetes and others.
In the acute phases of multiple sclerosis, large numbers of neurons and other cells of the nervous system, e.g. oligo dendrocytes, undergo apoptosis. Often, this massive cell death is accompanied by inflammation. The treatment of patients of MS, before, after or during such an acute phase, will suppress this inflammation and thus reduce the damage to the tissue.
The following example and figures are meant to further elucidate the present invention.
Description of the Figures
FIGURE 1: DAC5 binds to the surface of apoptotic cells a) Jurkat T cells were induced to undergo apoptosis by addition of 1 μM staurosporine (sts). At different time points, 2x105 cells per sample were incubated with 100μl DAC5 supernatant or 1 μg/ml lgG2a isotype control Ab (iso), followed by incubation with FITC-labelled anti-mouse IgG antibodies. To discriminate between early and late apoptosis, aliquots were incubated in parallel with FITC-labelled annexin V (AxV) or 2 μg/ml propidium iodide (PI). The kinetic is representative for 3 experiments. Inset shows a representative DAC5 staining after 4 hours incubation with staurosporine (shaded histogram, in comparison to DAC5 staining on live cells. b) Jurkat T cells were irradiated with 150 Gy and cultured for additional 6 hours. Alternatively, cells were incubated for 6 hours with 1 μM staurosporine (sts) or 1 μg/ml leucine zipper CD95 ligand (LZ-CD95L). Subsequently, cells were stained with FITC-labeled annexin V or FITC-labeled DAC5 in annexin Binding buffer.
FIGURE 2: DAC5 precipitates annexin I a)
Postnuclear lysates of 107 CEM cells were immmunoprecipitated with Protein A sepharose and purified DAC5 or lgG2a isotype control (iso), or protein A sepharose alone (-). Precipitates were resolved on a 12% SDS PAGE and silver stained. The protein bands labeled "annexin I" were cut from the gel and subjected to tryptic digest, followed by analysis via mass spectroscopy. By comparison with the NCBInr database peptides were identified as derived from annexin I. he - heavy chain; lc - light chain.
FIGURE 3: annexin I is bound to the outside of apoptotic cells
5x106 CEM cells were treated with 1 μM sts for different time points and centrifuged to obtain the supernatant. The pellets were washed with PBS/EDTA to yield the membrane bound annexin I (membrane). Finally, cell pellets were lysed and aliquots of all fractions were resolved on a 12% SDS PAGE followed by detection with monoclonal anti-annexin I Abs. Western Blot shown is representative for 3 experiments.
FIGURE 4: annexin I on the surface of cultured neutrophils
Primary human neutrophils were cultured for 1-4 days. Aliquots were analyzed for annexin I and phosphatidylserine-exposure by flowcytometry, using annexin V-FITC or DAC5-FITC, respectively, annexin I protein levels were monitored on Westernblot, using DAC5 and a peroxidase coupled, secondary anti mouse IgG antibody.
FIGURE 5: annexin I suppresses DC cytokine secretion
2x105 d7 immature DCs were incubated for 4h with or without 50μg/ml DAC5-F (ab')2-Fragments and 1x106 d1 neutrophils. As controls, d1 neutrophils were added in a transwell chamber and d4 neutrophils were used, respectively. Following 4h of coculture, 50ng/ml LPS were added to the wells for another 48h. Cells were then harvested, stained with antibodies against CD83, CD86 and HLA-DR and analyzed by flowcytometry (a). Aliquots of cell culture supematants were collected 24 h after addition of LPS and analyzed with ELISAs for TNF (b).
FIGURE 6: Jurkat tumor model 5x106 apoptosis sensitive Jurkat J16 cells or apoptosis resistant Rapo cells were injected s.c. into NOD/SCID-mice. When tumors of ca. 8 mm in diameter were established, the mice received an injection of 250 μg anti-APO-1. After 24 h the tumors were explanted. Cells were separated and analyzed for apoptosis using flowcytometric analysis of DNA content after Nicoletti et al (ref).
FIGURE 7: lodinated DAC5 binds to apoptotic cells in vitro 1x106 CEM cells were irradiated with 200mJ/cm2 UV-C light and cultured for another 4h. Apoptotic cells were then incubated with 131I-DAC5 or 127I-DAC5 for 20 in at 4°C. Cells incubated with the radioactively labeled DAC5 were centrifuged through oil and analyzed for bound radioactivity in a beta-counter. Cells stained with 127l- DAC5 were analyzed by flowcytometry, using a secondary FITC-coupled anti mouse IgG antibody.
EXAMPLES
Example 1 Immunization and fusion C57BL/6 mice were kept under certified pathogen free conditions and immunized nine times i.p. with 2x 107 apoptotic J16 Jurkat T cells. Apoptosis was induced by incubation with 1 μM staurosporine (Sigma, Munich, Germany) for
0.5 h. Cells were thoroughly washed in sterile PBS before injection. During the first 3 immunizations, the apoptotic J16 cells were coated with 1,2 μg/ml recombinant human annexin V (Sigma, Munich, Germany) to promote generation of high affinity antibodies28. Hybridomas were generated from spleens of immunized mice by Polyethylene glycol-induced fusion with the mouse plasmacytoma line Ag8.
Example 2 CeW lines and hybridomas The human T-ALL cell line CEM and the human Jurkat T cell lines J16 and Rapo were cultured in RPMI 1640 (Gibco, Karlsruhe, Germany) supplemented with 10% fetal calf serum (Gibco, Karlsruhe, Germany) and 10 mM HEPES (Sigma, Munich, Germany). Supernatant of the hybridoma DAC 5 was affinity purified using a Protein A-Sepharose column (Sigma, Munich, Germany), followed by dialysis against PBS. F(ab')2 fragments of DAC5 were generated using Immunopure kits (Pierce, Rockford, USA) according to manufacturers instructions.
Example 3 Preparation of primary human dendritic cells (DC) Human immature DCs were prepared from PBMC of 500 ml heparinized blood of healthy donors. PBMC were prepared as described previously29 using density centrifugation on a Ficoll gradient (Biochrom, Berlin, Germany) and allowed to adhere to plastic tissue culture flasks for 1h. Subsequently, adherent monocytes were washed thoroughly with PBS and seeded into 6 well cell culture plates at a density of 3x106/well and cultured in X-Vivo 15 medium (Cambrex, Verviers, Belgium) supplemented with 2% inactivated donor serum and containing 1000 U/ml hu GM-CSF and 500 U/ml hu IL-4 (Immunotools, Osnabrϋck, Germany). Fresh cytokines were supplemented again on day 3. Immature DCs were used on day 7, and >98% were positive for the DC marker CD11c. Example 4 Preparation of primary human neutrophils Human neutrophils were obtained as described previously,30 using density centrifugation of heparinized whole in PolymorphPrep (Axis Shield, Oslo, Norway). After preparation, neutrophils were cultured in X-Vivo 15 Medium
(Cambrex, Verviers, Belgium), supplemented with 2% donor serum for up to 4 days (d1-d4 neutrophils).
Example 5 Immunoprecipitation and silver staining For immunoprecipitation, 1x107 J16 cells were lysed in Triton X-100 Lysisbuffer (20 mM Tris/HCI, pH 7.4, 1 % Triton X-100, 10 % glycerol, 150 mM NaCI, 1 mM PMSF and 1 μg/ml of Leupeptin, Antipain, Chymostatin and Pepstatin A) for 15 min on ice and centrifuged (15 min, 14000xg, 4°C). Supernatants were then subjected to immunoprecipitation with 10 μg DAC5 or isotype control antibody and Protein A Sepharose (Sigma, Munich, Germany) at 4°C overnight. Precipitates were prepared in SDS sample buffer, resolved on a 12% SDS PAGE and stained with the Silverquest silver staining kit (Invitrogen, Paisley, UK), according to manufacturers instructions.
Example 6 Protein Sequencing Protein bands were excised from the silver stained gel, repeatedly washed with water, 40 mM ammonium bicarbonate/ ethanol (1:1 v/v), reduced with 10 mM DTT at 56 °C for 1 h and alkylated with 55 mM iodoacetamide at 25°C for 30 min in the dark. After alkylation, gel bands were repeatedly washed with 40mM ammonium bicarbonate and ethanol, dehydrated with 100% acetonitrile and air- dried for 15 min. Digestion with trypsin (10ng/μl, Promega) was performed in 40 mM ammonium bicarbonate at 37°C overnight.
Example 7 MALDI-TOF mass spectrometry (MALDI-TOF-MS) Sample preparation was achieved by cocrystallization of ZipTip C18 (Millipore) concentrated samples with matrix. Peptides in the supernatant of the in-gel digest were absorbed to a prewashed (50% acetonitrile/water) and equilibrated (0.1% trifluoroacetic acid/water) ZipTip C18 by repetitive pipetting steps. Following washing of the ZipTip C18 by equilibration buffer the peptides were eluted from the ZipTip with 1 μl of matrix (α-cyano-4-hydroxycinnamic acid saturated in 50% acetonitrile/water). MALDI-TOF mass spectra were recorded in the positive ion reflector mode with delayed extraction on a Reflex II time-of- flight instrument (Bruker-Daltonik). Ion acceleration was set to 26.5 kV, the reflector was set to 30.0 kV and the first extraction plate was set to 20.6 kV. Mass spectra were obtained by averaging 50 to 200 individual laser shots. Calibration of the spectra was performed internally by a two-point linear fit using the autolysis products of trypsin at m/z 842.50 and m/z 2211.10. Post source decay (PSD) analysis was performed in the positive ion reflector mode with delayed extraction by setting an ion gate width of 40 Da around the ion of interest. Data were acquired in 14 segments by decreasing the reflector voltage in a stepwise fashion. For each segment 100 to 200 individual laser shots were accumulated. The fragment ion spectrum was obtained by pasting together all segments to a single spectrum using the FAST software provided by Bruker. Fragment ion calibration was performed externally with the fragment masses of the adrenocorticotropic hormone 18 - 39 clip.
Example 8 Database search Singly charged monoisotopic peptide masses were used as inputs for database searching. Searches were performed against the NCBInr database using the ProFound search algorithm (http://129.85.19.192/prowl- cgi/ProFound.exe). Isoelectric points were allowed to range from 0 to 14, carbamidomethylation was included as fixed modification and the oxidation of methionine was allowed as possible modification. Up to one missed tryptic cleavage was considered, and the mass tolerance for the monoisotopic peptide masses was set to +/-100 ppm. Searches with fragment masses from PSD experiments were performed against the NCBInr database using the MS-Tag search algorithm provided by the Protein Prospector software package (http://prospector.ucsf.edu). Parent mass tolerance was set to +/- 0.1 Da and fragment ion tolerance was set to +/- 0.9 Da).
Example 9 Surface staining 5 x 105 cells were incubated with 100 μl of hybridoma culture supernatant or with 10 μg/ml purified DAC5 in PBS/10% FCS for 30 min at 4°C, washed with 500 μl PBS/10% FCS, and incubated another 30 min with 5 μg/ml FITC-labeled goat anti mouse IgG antibodies (Dianova, Hamburg, Germany) and 2 μg/ml propidium iodide (Sigma, Munich, Germany) in PBS/10% FCS. The addition of 10% FCS provided for calcium, necessary for annexin I to bind to the membrane of apoptotic cells. After further washing, cells were analyzed on a FACScan cytometer (Becton Dickinson, San Jose, USA). Staining with annexin V-FITC (Molecular Probes, Leiden, Netherlands) was performed according to manufactures instructions in annexin Binding buffer (10 mM HEPES, 140 mM NaCI, 2,5 mM CaCI2, pH 7,4).
Example 10 EDTA-washes and Western Blot analysis 5x106 CEM cells were incubated in 500 μl medium with 1 μM staurosporine for different time periods or left untreated. Subsequently, the cells were centrifuged, the supernatant was collected and pellets were washed with 500μl PBS/1 OmM EDTA. After centrifugation the washing solution was collected and the pellets were lysed in 500 μl Triton X-100 Lysis buffer. A 200 μl aliquot of each fraction was separated on a 12 % SDS-PAGE, blotted onto nitrocellulose membrane (Amersham, Freiburg, Germany) and detected with monoclonal antibodies against annexin I (Pharmingen, San Diego, USA). Example 11 Cocultures of immature DCs and neutrophils Immature d6 DCs were plated at a density of 2x105 cells/well into 24-well plates and incubated over night. The next day, d1 neutrophils were incubated for 15 min at 37° with 50μg/ml DAC5 F(ab')2 fragments or left untreated, and overlaid onto the DCs at a ratio of 1:1. After 4 hours, 50 ng/ml LPS (Sigma, Munich, Germany; E.coli strain 026:B6) was added to a part of the cocultures. As controls, 50 μg/ml DAC5 F(ab')2 fragments alone and d4 neutrophils were cocultured with DCs as well. After 48h the cells were harvested, centrifuged and stained with antibodies against HLA-DR, CD86 and CD83 (CaJtag) and in PBS containing 10% mouse serum to block Fc-Receptor interactions. Cells were analyzed on a FACScan cytometer (Becton Dickinson, San Jose, USA). Supernatants were collected after 24 h and stored at - 20 °C for further cytokine analysis with ELISAs for TNFα (Pharmingen, San Diego, USA).
REFERENCES
1. Vaux.D.L. & Korsmeyer,S.J. Cell death in development. Cell 96 ; 245-254 (1999). 2. Krammer,P.H. CD95's deadly mission in the immune system. Nature 407, 789-795 (2000).
3. Kerr.J.F., Wyllie.A.H. & Currie.A.R. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br. J. Cancer 26, 239-257 (1972). 4. Peter.M.E. & Krammer,P.H. The CD95(APO-1/Fas) DISC and beyond. Cell Death. Differ. 10, 26-35 (2003).
5. Savill ., Fadok,V, Henson.P. & HaslettC. Phagocyte recognition of cells undergoing apoptosis. Immunol. Today 14, 131-136 (1993).
6. Lauber.K., Blumenthal.S.G., Waibel.M. & Wesselborg.S. Clearance of apoptotic cells: getting rid of the corpses. Mol. Cell 14, 277-287 (2004).
7. Savill,J., Dransfield,!., Gregory.C. & HaslettC. A blast from the past: clearance of apoptotic cells regulates immune responses. Nat. Rev. Immunol. 2, 965-975 (2002).
8. Steinman.R.M. et al. Dendritic cell function in vivo during the steady state: a role in peripheral tolerance. Ann. N. Y. Acad. Sci. 987, 15-25 (2003).
9. Liu.K. et al. Immune tolerance after delivery of dying cells to dendritic cells in situ. J. Exp. Med. 196, 1091-1097 (2002).
10. Fadok.V.A. et al. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAR J. Clin. Invest 101, 890- 898 (1998). 11. Gerke. & Moss.S.E. annexins: from structure to function. Physiol Rev. 82, 331-371 (2002).
12. Wallner.B.P. et al. Cloning and expression of human lipocortin, a phospholipase A2 inhibitor with potential anti-inflammatory activity. Nature 320, 77-81 (1986).
13. Solito.E., Raguenes-Nicol.C, de Coupade.C, Bisagni-Faure.A. & Russo- Marie,F. U937 cells deprived of endogenous annexin 1 demonstrate an increased PLA2 activity. Br. J. Pharmacol. 124, 1675-1683 (1998).
14. Cover.P.O., Baanah-Jones,F., John, CD. & Buckiηgham.J.C. annexin 1 (lipocortin 1) mimics inhibitory effects of glucocorticoids on testosterone secretion and enhances effects of interleukin-1beta. Endocrine. 18, 33-39 (2002).
15. Walther.A., Riehemann,K. & Gerke,V. A novel ligand of the formyl peptide receptor: annexin I regulates neutrophil extravasation by interacting with the FPR. Mol. Cell 5, 831-840 (2000).
16. Hannon,R. et al. Aberrant inflammation and resistance to glucocorticoids in annexin 1-/- mouse. FASEB J. 17, 253-255 (2003).
17. Haslett.C, Savill.J.S. & Meagher.L. The neutrophil. Curr. Opin. Immunol. 2, 10-18 (1989).
18. Perretti,M. & Flower.R.J. annexin 1 and the biology of the neutrophil. J. Leukoc. Biol. 76, 25-29 (2004).
19. Banchereau,J. & Steinman.R.M. Dendritic cells and the control of immunity. Nature 392, 245-252 (1998).
20. Schnare.M. et al. Toll-like receptors control activation of adaptive immune responses. Nat Immunol. 2, 947-950 (2001). 21. Weighardt.H. et al. Identification of a. Eur. J. Immunol. 34, 558-564 (2004).
22. Inaba.K., Metlay.J.R, Crowley.M.T, Witmer-Pack.M. & Steinman.R.M. Dendritic cells as antigen presenting cells in vivo. Int. Rev. Immunol. 6, 197-206 (1990).
23. O'Doherty.U. et al. Dendritic cells freshly isolated from human blood express CD4 and mature into typical immunostimulatory dendritic cells after culture in monocyte-conditioned medium. J. Exp. Med. 178, 1067-1076 (1993).
24. Huang, F.P. et al. A discrete subpopulation of dendritic cells transports apoptotic intestinal epithelial cells to T cell areas of mesenteric lymph nodes. J. Exp. Med. 191, 435-444 (2000).
25. Steinman,R.M., Turley.S., Mellman . & Inaba.K. The induction of tolerance by dendritic cells that have captured apoptotic cells. J. Exp. Med. 191, 411- 416 (2000).
26. Trauth.B.C. et al. Monoclonal antibody-mediated tumor regression by induction of apoptosis. Science 245, 301-305 (1989).
27. Oh, P. et al. Subtractive proteomic mapping of the endothelial surface in lung and solid tumours for tissue-specific therapy. Nature 429, 629-635 (2004).
28. Stach.C.M. et al. Treatment with annexin V increases immunogenicity of apoptotic human T-cells in Balb/c mice. Cell Death. Differ. 7, 911-915 (2000).
29. Klas.C, Debatin.K.M., Jonker.R.R. & Krammer.P.H. Activation interferes with the APO-1 pathway in mature human T cells. Int. Immunol. 5, 625-630 (1993).
30. Ferrante.A. & Thong.Y.H. A rapid one-step procedure for purification of mononuclear and polymorphonuclear leukocytes from human blood using a modification of the Hypaque-Ficoll technique. J. Immunol. Methods 24, 389-393 (1978).

Claims

Claims
1. An anti-annexin antibody or fragment thereof, wherein the antibody or fragment is capable of specifically binding to an annexin present on a cell that is undergoing apoptosis.
2. The antibody fragment according to claim 1 wherein it is a Fab or F(ab')2 fragment.
3. The antibody or fragment according to any one of the preceding claims, wherein it is a monoclonal antibody or fragment thereof.
4. The antibody or fragment according to any one of the preceding claims, wherein it is labeled.
5. The antibody or fragment according to claim 4, wherein the label is an effector molecule, a toxic substance or a radioactive substance.
6. A pharmaceutical composition, comprising as the active ingredient an antibody or fragment thereof capable of specifically binding to an annexin present on a cell that is undergoing apoptosis .
7. Use of an anti-annexin antibody or fragment thereof for the detection and/or monitoring of apoptosis, in vitro, ex vivo or in vivo.
8. Use of an anti-annexin antibody or fragment thereof for modulating an immune response.
9. Use of an anti-annexin antibody or fragment thereof for inducing and/or increasing an inflammatory response.
10. Use of an anti-annexin antibody or fragment thereof for targeting tumor cells and/or tumor tissue.
11. Use of an anti-annexin antibody or fragment thereof for inducing an inflammatory response to tumor cells.
12. Use of an anti-annexin antibody or fragment thereof for blocking the development of tolerance against tumor cells.
13. Use of an anti-annexin antibody or fragment thereof for the production of an agent for the diagnosis and/or treatment of diseases linked to apoptosis.
14. Use of claim 13, wherein the diseases linked to apoptosis are selected from the group consisting of cancer, diabetes, autoimmune diseases and cardiovascular and vascular diseases.
15. Use of claim 14, wherein the autoimmune disease is diabetes, rheumatoid arthritis, lupus erythematosus or multiple sclerosis.
16. Use of any one of claims 7 to 15, wherein the anti-annexin antibody is labelled.
17. Use of claim 16, wherein a labelled anti-annexin antibody is used for the production of a diagnostic agent for the detection of tumor cells after or during conventional cancer therapy.
18. Use according to anyone of claims 1-17, wherein the anti-annexin antibody is specific for annexin I, annexin II, annexin IV, or annexin V.
19. A method for detecting and/or monitoring apoptosis comprising: (i) providing a sample to be analysed, comprising cells; (ii) detecting an annexin present on the surface of said cells by adding a substance capable of specifially binding to an annexin present on a cell that is undergoing apoptosis.
20. The method of claim 19, wherein the substance of step (ii) is an antibody or fragment thereof capable of specifically binding to an annexin present on a cell that is undergoing apoptosis.
21. Use of an annexin and/or functional fragments thereof and/or fusion protein comprising an annexin or functional fragments thereof, for the production of an agent for inhibiting an inflammatory response.
22. Use of an annexin and/or functional fragments thereof and/or fusion protein comprising an annexin or functional fragments thereof, for the production of an agent for the diagnosis and/or the treatment of a disease linked with apoptosis and/or cell death and/or inflammation.
23. Use according to claim 22, wherein the disease is selected from the group consisting of a ischaemic reperfusion damage, stroke, chronic heart failure, myocardial infarction, spinal cord injury, acute liver failure, renal ischaemia, neurodegenerative diseases such as Alzheimer, Parkinson's diease, sepsis, HIV-infeetion and autoimmune diseases, in particular multiple sclerosis.
24. Use of an annexin and/or functional fragments thereof and/or fusion protein comprising an annexin or functional fragments thereof, for the production of an agent to inhibit an inflammatory response to tissue, in particular transplantation tissue.
5. Use according to anyone of claims 21-24, wherein the annexin used is annexin I, annexin II, annexin IV or annexin V.
PCT/EP2004/010756 2003-09-24 2004-09-24 Antibodies against annexins, use thereof for therapy and diagnosis. use of annexins for therapy and diagnosis. WO2005027965A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP04765595A EP1670510A1 (en) 2003-09-24 2004-09-24 Antibodies against annexins, use thereof for therapy and diagnosis. use of annexins for therapy and diagnosis.

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP03021581 2003-09-24
EP03021581.8 2003-09-24

Publications (1)

Publication Number Publication Date
WO2005027965A1 true WO2005027965A1 (en) 2005-03-31

Family

ID=34354425

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2004/010756 WO2005027965A1 (en) 2003-09-24 2004-09-24 Antibodies against annexins, use thereof for therapy and diagnosis. use of annexins for therapy and diagnosis.

Country Status (2)

Country Link
EP (1) EP1670510A1 (en)
WO (1) WO2005027965A1 (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005117999A2 (en) * 2004-06-02 2005-12-15 Sidney Kimmel Cancer Center Imaging and therapeutic agents targeting proteins expressed on endothelial cell surface
WO2005117848A2 (en) * 2004-06-02 2005-12-15 Sidney Kimmel Cancer Center Vascular targets for detecting, imaging and treating neoplasia or neovasculature
FR2929517A1 (en) * 2008-04-04 2009-10-09 Oreal Cosmetic use of polypeptide of amino acid sequence encoded by nucleic acid sequence represented by a sequence, nucleic acid sequence encoding for polypeptide or modulating agent, to prevent and/or treat e.g. signs of skin aging
US7645739B2 (en) 2001-02-21 2010-01-12 Alavita Pharmaceuticals, Inc. Modified annexin compositions and methods of using same
WO2010064012A2 (en) 2008-12-02 2010-06-10 Queen Mary & Westfield College Treatment
WO2011154705A1 (en) 2010-06-09 2011-12-15 Queen Mary & Westfield College Annexin 1 antibody
US8497072B2 (en) 2005-11-30 2013-07-30 Abbott Laboratories Amyloid-beta globulomer antibodies
US8691224B2 (en) 2005-11-30 2014-04-08 Abbvie Inc. Anti-Aβ globulomer 5F7 antibodies
US8877190B2 (en) 2006-11-30 2014-11-04 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US8895004B2 (en) 2007-02-27 2014-11-25 AbbVie Deutschland GmbH & Co. KG Method for the treatment of amyloidoses
US8987419B2 (en) 2010-04-15 2015-03-24 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9176150B2 (en) 2003-01-31 2015-11-03 AbbVie Deutschland GmbH & Co. KG Amyloid beta(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
EP3045470A1 (en) 2015-01-15 2016-07-20 Deutsches Krebsforschungszentrum Stiftung des Öffentlichen Rechts Modulators of the function of the core domain of annexins, and uses thereof in autoimmune and/or cancer therapy
US10233235B2 (en) 2008-09-22 2019-03-19 The Regents Of The University Of Colorado, A Body Corporate Modulating the alternative complement pathway
WO2020030827A1 (en) 2018-08-10 2020-02-13 Medannex Ltd. Cancer treatment with an antibody
WO2020161247A1 (en) 2019-02-08 2020-08-13 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Annexin-coated particles
US10752677B2 (en) 2011-12-14 2020-08-25 Queen Mary & Westfield College, University Of London Methods of treating obsessive compulsive disorder (OCD) or anxiety using an antibody that binds to annexin-1
US10947283B2 (en) 2016-06-10 2021-03-16 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Core domain of annexins and uses thereof in antigen delivery and vaccination
US11041019B2 (en) 2017-02-08 2021-06-22 Medannex Ltd. Anti human annexin A1 antibody
US11053305B2 (en) 2018-12-11 2021-07-06 Q32 Bio Inc. Fusion protein constructs comprising anti-C3d antibody and CR1 polypeptide

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH0772147A (en) * 1993-07-02 1995-03-17 Noboru Kaneko Antianexin v antibody as well as preparation and utilization of same antibody
DE19541284A1 (en) * 1995-11-06 1996-05-30 Kalden Joachim Robert Prof Dr Immunomodulation method
EP0741144A1 (en) * 1994-11-11 1996-11-06 International Reagents Corporation Antiannexin-v monoclonal antibody, process for producing the same, and use therof
WO1999021980A1 (en) * 1997-10-24 1999-05-06 Mcgill University P-40/annexin i and related proteins and their role in multidrug resistance
WO2002017857A2 (en) * 2000-09-01 2002-03-07 Philadelphia, Health And Education Corporation Methods and compositions for inhibiting angiogenesis

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH0772147A (en) * 1993-07-02 1995-03-17 Noboru Kaneko Antianexin v antibody as well as preparation and utilization of same antibody
EP0741144A1 (en) * 1994-11-11 1996-11-06 International Reagents Corporation Antiannexin-v monoclonal antibody, process for producing the same, and use therof
DE19541284A1 (en) * 1995-11-06 1996-05-30 Kalden Joachim Robert Prof Dr Immunomodulation method
WO1999021980A1 (en) * 1997-10-24 1999-05-06 Mcgill University P-40/annexin i and related proteins and their role in multidrug resistance
WO2002017857A2 (en) * 2000-09-01 2002-03-07 Philadelphia, Health And Education Corporation Methods and compositions for inhibiting angiogenesis

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
PATENT ABSTRACTS OF JAPAN vol. 1995, no. 06 31 July 1995 (1995-07-31) *
VERMES I ET AL: "A novel assay for apoptosis Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V", JOURNAL OF IMMUNOLOGICAL METHODS, ELSEVIER SCIENCE PUBLISHERS B.V.,AMSTERDAM, NL, vol. 184, no. 1, 17 July 1995 (1995-07-17), pages 39 - 51, XP004021001, ISSN: 0022-1759 *

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7645739B2 (en) 2001-02-21 2010-01-12 Alavita Pharmaceuticals, Inc. Modified annexin compositions and methods of using same
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US9176150B2 (en) 2003-01-31 2015-11-03 AbbVie Deutschland GmbH & Co. KG Amyloid beta(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
WO2005117999A2 (en) * 2004-06-02 2005-12-15 Sidney Kimmel Cancer Center Imaging and therapeutic agents targeting proteins expressed on endothelial cell surface
WO2005117848A2 (en) * 2004-06-02 2005-12-15 Sidney Kimmel Cancer Center Vascular targets for detecting, imaging and treating neoplasia or neovasculature
WO2005117848A3 (en) * 2004-06-02 2006-06-29 Sidney Kimmel Cancer Ct Vascular targets for detecting, imaging and treating neoplasia or neovasculature
WO2005117999A3 (en) * 2004-06-02 2008-02-07 Sidney Kimmel Cancer Ct Imaging and therapeutic agents targeting proteins expressed on endothelial cell surface
US8715675B2 (en) 2004-06-02 2014-05-06 Jan E. Schnitzer Vascular targets for detecting, imaging and treating neoplasia or neovasculature
US8497072B2 (en) 2005-11-30 2013-07-30 Abbott Laboratories Amyloid-beta globulomer antibodies
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US9540432B2 (en) 2005-11-30 2017-01-10 AbbVie Deutschland GmbH & Co. KG Anti-Aβ globulomer 7C6 antibodies
US10323084B2 (en) 2005-11-30 2019-06-18 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US8691224B2 (en) 2005-11-30 2014-04-08 Abbvie Inc. Anti-Aβ globulomer 5F7 antibodies
US10538581B2 (en) 2005-11-30 2020-01-21 Abbvie Inc. Anti-Aβ globulomer 4D10 antibodies
US8877190B2 (en) 2006-11-30 2014-11-04 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US9359430B2 (en) 2006-11-30 2016-06-07 Abbvie Inc. Abeta conformer selective anti-Abeta globulomer monoclonal antibodies
US9951125B2 (en) 2006-11-30 2018-04-24 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US9394360B2 (en) 2006-11-30 2016-07-19 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US8895004B2 (en) 2007-02-27 2014-11-25 AbbVie Deutschland GmbH & Co. KG Method for the treatment of amyloidoses
FR2929517A1 (en) * 2008-04-04 2009-10-09 Oreal Cosmetic use of polypeptide of amino acid sequence encoded by nucleic acid sequence represented by a sequence, nucleic acid sequence encoding for polypeptide or modulating agent, to prevent and/or treat e.g. signs of skin aging
WO2009136033A2 (en) * 2008-04-04 2009-11-12 L'oreal Cosmetic use of annexin ii-type proteins for treating dryness of the skin
WO2009136033A3 (en) * 2008-04-04 2010-04-29 L'oreal Cosmetic use of annexin ii-type proteins for treating dryness of the skin
US11407820B2 (en) 2008-09-22 2022-08-09 The Regents Of The University Of Colorado Modulating the alternative complement pathway
US10233235B2 (en) 2008-09-22 2019-03-19 The Regents Of The University Of Colorado, A Body Corporate Modulating the alternative complement pathway
EP2889312A3 (en) * 2008-12-02 2015-10-21 Queen Mary & Westfield College Treatment of autoimmune disease by modulating Annexin-1 (lipocortin 1)
WO2010064012A3 (en) * 2008-12-02 2010-07-29 Queen Mary & Westfield College Treatment of autoimmune disease by modulating annexin-i (lipocortin 1 )
WO2010064012A2 (en) 2008-12-02 2010-06-10 Queen Mary & Westfield College Treatment
US8980255B2 (en) 2008-12-02 2015-03-17 Queen Mary & Westfield College Treatment of autoimmune disease by modulating annexin-1 (lipocortin 1)
US8987419B2 (en) 2010-04-15 2015-03-24 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9822171B2 (en) 2010-04-15 2017-11-21 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
USRE47982E1 (en) 2010-06-09 2020-05-12 Queen Mary & Westfield College, University Of London Annexin 1 antibody
US9127051B2 (en) 2010-06-09 2015-09-08 Queen Mary & Westfield College, University Of London Annexin 1 antibody
WO2011154705A1 (en) 2010-06-09 2011-12-15 Queen Mary & Westfield College Annexin 1 antibody
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US10047121B2 (en) 2010-08-14 2018-08-14 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US10752677B2 (en) 2011-12-14 2020-08-25 Queen Mary & Westfield College, University Of London Methods of treating obsessive compulsive disorder (OCD) or anxiety using an antibody that binds to annexin-1
WO2016113022A1 (en) * 2015-01-15 2016-07-21 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Modulators of the function of the core domain of annexins, and uses thereof in autoimmune and/or cancer therapy
EP3045470A1 (en) 2015-01-15 2016-07-20 Deutsches Krebsforschungszentrum Stiftung des Öffentlichen Rechts Modulators of the function of the core domain of annexins, and uses thereof in autoimmune and/or cancer therapy
US10947283B2 (en) 2016-06-10 2021-03-16 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Core domain of annexins and uses thereof in antigen delivery and vaccination
US11851463B2 (en) 2016-06-10 2023-12-26 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Core domain of annexins and uses thereof in antigen delivery and vaccination
US11041019B2 (en) 2017-02-08 2021-06-22 Medannex Ltd. Anti human annexin A1 antibody
WO2020030827A1 (en) 2018-08-10 2020-02-13 Medannex Ltd. Cancer treatment with an antibody
US11858982B2 (en) 2018-08-10 2024-01-02 Medannex Ltd. Cancer treatment with an antibody
US11053305B2 (en) 2018-12-11 2021-07-06 Q32 Bio Inc. Fusion protein constructs comprising anti-C3d antibody and CR1 polypeptide
US11053306B2 (en) 2018-12-11 2021-07-06 Q32 Bio Inc. Fusion protein constructs comprising anti-C3d antibody and factor H
US11879008B2 (en) 2018-12-11 2024-01-23 Q32 Bio Inc. Methods of treating complement mediated diseases with fusion protein constructs comprising anti-C3d antibody and a complement modulator
WO2020161247A1 (en) 2019-02-08 2020-08-13 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Annexin-coated particles

Also Published As

Publication number Publication date
EP1670510A1 (en) 2006-06-21

Similar Documents

Publication Publication Date Title
WO2005027965A1 (en) Antibodies against annexins, use thereof for therapy and diagnosis. use of annexins for therapy and diagnosis.
Oldenborg CD47: a cell surface glycoprotein which regulates multiple functions of hematopoietic cells in health and disease
US20200030377A1 (en) Antigen-specific immune modulation
US20190177396A1 (en) Use of cd31 peptides in the treatment of thrombotic and autoimmune disorders
JPH09509826A (en) Ligands (ACT-4-L) for the receptor on the surface layer of activated CD4 (+) T cells
US20030026803A1 (en) Compositions for inhibiting macrophage activity
EP2453906A1 (en) Methods of stimulating liver regeneration
JP6769948B2 (en) An isolated interleukin-34 polypeptide for use in the prevention of transplant rejection and the treatment of autoimmune diseases
EP3679070A1 (en) Antibodies to programmed cell death protein 1
JPH10504039A (en) Compositions and treatments for multiple sclerosis
Zarantonello et al. C3‐dependent effector functions of complement
AU2017252344A1 (en) Diagnosis of immune_activation using CLEVER-1, TNF-alpha and HLA-DR binding agents
JP2000511417A (en) Immunomodulatory compounds containing D-isomer amino acids
EP3205347A1 (en) White blood cell extracellular trap formation inhibitor
CA2391530A1 (en) Tr3-specific binding agents and methods for their use
Park et al. Ionizing radiation and nitric oxide donor sensitize Fas-induced apoptosis via up-regulation of Fas in human cervical cancer cells
CN111936512A (en) Anticoagulant proteins and their use to treat diseases associated with neutrophil activation
EP1562632B1 (en) Inducible ligand for alpha1 beta1 integrin and uses
WO2000071147A1 (en) Novel therapeutic use of viral inflammation modulatory protein in blocking xenograft rejection
Pratt et al. Influence of complement on the allospecific antibody response to a primary vascularized organ graft
EP1367393A1 (en) Methods for using the CD 163 pathway for modulating an immune response
CA2341558A1 (en) Compositions and methods for protecting organs, tissue and cells from immune system-mediated damage
US20220401536A1 (en) Use of dd1alpha (vista) modulators in cancer treatment: immunotherapy based on the disruption of dd1alpha/pd-1 signaling
Kumamoto et al. TIRC7 is induced in rejected human kidneys and anti-TIRC7 mAb with FK506 prolongs survival of kidney allografts in rats
US20230203107A1 (en) Peptide for treating sepsis derived from rv3364c protein of mycobacterium tuberculosis

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 BW BY BZ CA CH CN CO CR CU CZ DK DM DZ EC EE EG ES FI GB GD GE GM HR HU ID IL IN IS JP KE KG KP KZ LC LK LR LS LT LU LV MA MD MK MN MW MX MZ NA NI NO NZ PG PH PL PT RO RU SC SD SE SG SK SY TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GM KE LS MW MZ NA SD SZ TZ UG ZM ZW AM AZ BY KG MD RU TJ TM AT BE BG CH CY DE DK EE ES FI FR GB GR HU IE IT MC NL PL PT RO SE SI SK TR BF CF CG CI CM GA GN GQ GW ML MR 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: 2004765595

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2004765595

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 2004765595

Country of ref document: EP