CA2515366C - Compositions and methods to modulate immune and inflammatory responses - Google Patents

Compositions and methods to modulate immune and inflammatory responses Download PDF

Info

Publication number
CA2515366C
CA2515366C CA2515366A CA2515366A CA2515366C CA 2515366 C CA2515366 C CA 2515366C CA 2515366 A CA2515366 A CA 2515366A CA 2515366 A CA2515366 A CA 2515366A CA 2515366 C CA2515366 C CA 2515366C
Authority
CA
Canada
Prior art keywords
rtf
atp
cells
expression
activity
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CA2515366A
Other languages
French (fr)
Other versions
CA2515366A1 (en
Inventor
Kenneth Beaman
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Rosalind Franklin University of Medicine and Science
Original Assignee
Rosalind Franklin University of Medicine and Science
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 Rosalind Franklin University of Medicine and Science filed Critical Rosalind Franklin University of Medicine and Science
Publication of CA2515366A1 publication Critical patent/CA2515366A1/en
Application granted granted Critical
Publication of CA2515366C publication Critical patent/CA2515366C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Abstract

The present invention is directed to compositions and methods to modulate inflammatory and immune responses in a subject, which includes immune activation and inhibiting inflammatory responses, by modulating the regeneration and tolerance factor (RTF).

Description

SPECIFICATION
TITLE OF THE INVENTION
"COMPOSITIONS AND METHODS TO MODULATE IMMUNE AND
INFLAMMATORY RESPONSES"

BACKGROUND OF THE INVENTION
The inflaminatory process initiates a localized defense against foreign antigens.
The process is characterized by immune activation as well as destruction of nearby tissues secondary to involved release of destructive agents. Iiiitially involved tissue assumes a significant role in provoking the inflammatory response at large (Matzinger, P., Tlae danger inodel: a renewed sense of self. Science, 2002. 296(5566): p.
301-5;
Medzhitov, R. and C.A. Janeway, Jr., Decoding the patterns of self and nonself by the innate im.naune system. Science, 2002. 296(5566): p. 298-300). One of the primary immunogenic mechanisms triggered by the inflammatory response includes the release of ATP into the extracellular space. ATP is liberated from injured tissue itself-as well as from nearby macrophages, neutropliils, platelets and dying cells-following incidents of stress, infection or other forms of tissue insult (Fredholm, B.B., Purines and neutrophil leukocytes. Gen Pharmacol, 1997. 28(3): p. 345-50; Beigi, R., et al., Detectioit. of local ATP release fi om activated platelets using cell surface-attached firefly luciferase. Am J Physiol, 1999. 276(1 Pt 1): p. C267-78; Schwiebert, E.M., ABC
transporter-facilitated ATP conductive transport. Am J Pliysiol, 1999. 276(1 Pt 1): p.
Cl-8; Mizumoto, N., et al., CD39 is the dominant Langerhans cell-associated ecto-NTPDase: modulatory roles in infammation and imrnune responsiveness. Nat Med, 2002. 8(4): p. 358-65; Dubyalc, G.R. and C. el-Moatassiin, Signal transduction via P2-purinergic receptors for extracellular ATP and other nucleotides. An7 J
Physiol, 1993.
265(3 Pt 1): p. C577-606; Gordon, J.L., Extracellular ATP: effects, sources and fate.
Biochem J, 1986. 233(2): p. 309-19). Extracellular ATP can then exert a wide variety of effects on surrounding cells, most notably the induction of apoptotic events leading ultimately to cell death in both lymphocytes and macrophages (Di Virgilio, F., The P2Z
purinoceptor: an. intriguing role in - immunity, inflammation and cell death.
Immunol Today, 1995. 16(11): p. 524-8; Gargett, C.E., J.E. Cornish, and J.S. Wiley, ATP, a partial agonist for the P2Z receptor of human lymphocytes. Br J Pharmacol, 1997.
122(5): p. 911-7; Buisman, H.P., et al., Extracellular ATP induces a large nonselective conductance in macrophage plasma membYanes. Proc Natl Acad Sci U S A, 1988.
85(21): p. 7988-92; Ferrari, D., et al., ATP-mediated cytotoxicity in microglial cells.
Neuropharinacology, 1997. 36(9): p. 1295-301; Zheng, L.M., et al., Extracellular ATP
as a trigger for apoptosis or programmed cell death. J Cell Biol, 1991.
112(2): p. 279-88). Diminution of cell integrity then leads to further ATP release, propagating the inflaininatory response and continued tissue destruction. Recent data have shown an attenuated effect of extracellular ATP in the presence of the regeneration and tolerance factor (RTF) protein. The present invention discloses compositions and methods to modulate inflammatory and immune responses by regulating the activities of RTF. RTF
prevents ATP activation of P2Z, thus preventing the cell from undergoing pro-inflammatory activation and apoptosis.

SUMMARY OF THE INVENTION
The present invention is directed to compositions and methods to modulate inflamnzatory and immune responses in a subject, which includes immune activation and inhibiting inflammatory responses, by modulating the regeneration and tolerance factor (RTF). Immune activation can be accomplished by inhibiting the RTF
activity.
In one embodiment, the RTF activity can be inhibited by administering a RTF
antagonist, which can be a small molecule, a peptide, or an antibody or a fragment thereof. Inhibiting the RTF activity may result in cell apotosis, especially cancer cells such as ovarian carcinoma cells.
Inflammatory responses can be inhibited by increasing the level of activity of RTF. In an embodiment, the method of increasing the level of activity of RTF
is by administering an effective amount of RTF or a fragment of RTF. The RTF or its fragment can be isolated or purified from mammalian cells such as the T-lymphocyte, the B-lymphocyte, the macrophage, thyinus, or the fetalplacental tissue.
Alternatively, the RTF can be synthetic or a recombinant RTF.
The present invention is also directed to a method of treating cancer, such as ovarian cancer, by iiihibiting the RTF activity. In one embodiment, the method to inhibit RTF activity is to administer an effective dose of a RTF antagonist.
The present invention is further directed to a method of treating or ameliorating an inflammatory disorder or an autoimmune disorder or one or more symptoms thereof by administering an effective amount of RTF or a fragment of RTF. The RTF or its fragment can be isolated or purified from mammalian cells such as the T-lymphocyte, the B-lymphocyte, the macrophage, thymus, or the fetalplacental tissue.
Altematively, the RTF can be synthetic or a recombinant RTF. The RTF can be formulated with one or more suitable pharmaceutical excipients for administration by parenteral, intramuscular, topical, nasal, puhnonary, or oral route. The formulation can also be in the form of micropariicles or nanoparticles. In an embodiment, the method of treating or ameliorating an inflammatory disorder or an autoimmune disorder or one or more symptoms thereof includes an additional step of administering a TNF-a antagonist, which includes but is not limited to infliximab, etanercept, D2E7, CDP571, CDP870, thalidomide and its analogs, and phosphodiesterase type N inhibitors. .
Additional features and advantages of the present invention are described in, and will be apparent from, the following Detailed Description of the Invention and the figures.
BRIEF DESCRIPTION OF THE FIGURES
FIG. I shows the expression of RTF and P2Z on PBMC and macrophages. Flow cytometric analysis of the expression of RTF and P2Z. Dotted line indicates isotype control antibody. Solid line represents either RTF expression (column 1) or expression (column 2). The first row represents unstimulated PBMC. The second, row represents PBMC stimulated for 72 hours with PHA. The third row represents macrophages.
FIG. 2 illustrates that RTF expression is concurrent with P2Z expression on lymphocytes during stimulation. Lymphocytes were stimulated with PHA for 0-72 hours. The expression on RTF and P2Z was measured at time points 0, 24hrs, and 72 hrs as indicated by flow cytometry. The Y axis represent the percent maximal expression of each molecule. The black bars represent RTF expression and the gray bars represent P2Z expression. Error bars represent the standard error of the mean. .
FIG. 3 shows that RTF moves to the surface of the membrane in a 50 kDa form during activation. PBMC were stimulated for 0-72 hours and RTF size was examined by westem blot and RTF location was examined by confocal microscopy. Panel A:
Western Blot: PBMC were stimulated with plate bound CD3 and CD28 and harvested at the time points indicated (in hours), western blotted and probed by 2C1. Row 1 represents proteins of 70 kDa size, and row 2 represents proteins of 50 kDa size. Each lane represents a stimulation time of 0, 18, 48, and 72 hours respectively.
Panel B:
Confocal Microscopy: Confocal microscopy images of PBMC taken before activation (left panel) and at 72 hrs of activation (right panel), permeabilized and stained with anti-RTF-FITC and counterstained with anti-CD8-PE.
FIG. 4 demonstrates that surface ATPase activity is inhibited by anti-RTF.
PBMC and J774A1 macrophages with anti-RTF antibody (black columns) or isotype control antibody (gray columns) were examined for surface ATPase activity, by adding [32]yP-ATP and measuring the release of [32]-Pi. The Y axis represents surface ATPase activity as measured in counts per minute [32]-Pi. Error bars represent the standard error of the mean.
FIG. 5 shows that anti-RTF asid ATP generate more apoptosis together than either alone. Top row represent flow cytometry histograms of J774A1 macrophages incubated with ATP, anti-RTF, or both together. Bottom row represents J774A1 cells incubated with ATP, anti-RTF, and different concentrations of ATPase as indicated. X
axis represents annexin-V staining. Means of four independent experiments with standard error are given for each.

FIG. 6 demonstrates the enhancement of secretion of IL-1(3 in THP-1 Macrophages by anti-RTF antibody.
FIG. 7 illustrates the cytotoxic effect of anti-RTF antibody to ovarian carcinoma cells.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is directed to compositions and methods to modulate inflammatory and immune responses in a subject by regulating the level of activity of regeneration and tolerance factor (RTF). Modulating inflammatory and immune responses includes both immune activation and the inhibition of the inflammatory responses. In a preferred embodiment, the subject is a mammal. In a more preferred einbodiment, the subject is a human.
A principle means by which RTF controls the extent of inflamination is via its interaction with P2Z, a cell surface receptor expressed primarily on cells of hematopoetic origin such as lymphocytes and macrophages. P2Z is one of a family of receptors known as purinoceptors which are activated by nucleotides.
Extracellular ATP binds P2Z, which triggers the opening of specific calcium channels as well as non-specific transmembrane channels (Dubyalc, G.R. and C. el-Moatassim, Signal transduction via P2-purinergic receptors for extracellular ATP and other nucleotides.
Am J Physiol, 1993. 265(3 Pt 1): p. C577-606; Gargett, C.E., J.E. Comish, and J.S.
Wiley, ATP, a partial agonist for the P2Z receptor of human lynzphocytes. Br J
Pharmacol, 1997. 122(5): p. 911-7; Zheng, L.M., et al., Extracellular ATP as a trigger for apoptosis or pnogranzmed cell death. J Cell Biol, 1991. 112(2): p. 279-88). These channels then allow for rapid influx of calcium ions into the cytosolic space.
This calcium influx in turn drives forward many specific activation pathways. The activation of P2Z can cause specific activation of NF-xB, and the maturation and secretion of IL-1 R, a potent inflammatory cytokine. This will subsequently increase both cellular activation and drive the inflainmatory response, making P2Z a key mediator in the inflammatory process. It can also cause apoptosis and an inflammatory counterpart to apoptosis, oncosis, inducing the release of further ATP. Recent reports have shown that the absence of the P2Z purinoceptor can attenuate the inflammatory response (Labasi JM, P.N., Donovan C, McCurdy S, Lira P, Payette MM, Brissette W, Wicks JR, Audoly L, Gabel CA., Absence of the P2X7 receptor alters leukocyte function and attenuates an inflammatory response. J Immunol, 2002. 168(12)(Jun 15): p. 6436-45).
Therefore it follows that extracellular ATP up-regulates the inflammatory response through its instigation of P2Z activation at the site of inflammation and also the stimulated release of pro-inflammatory cytokines. A key regulatory point at which to reduce the inflammatory response, then, is in the concentration of extracellular ATP..
It has been suggested that ecto-ATPases hold important regulatory surface ATPase activity, which attenuates inflainmatory as well as iininune responses (Labasi JM, P.N., Donovan C, McCurdy S, Lira P, Payette MM, Brissette W, Wiclcs JR, Audoly L, Gabel CA., Absence of the P2X7 receptor alters leukocyte function and attenuates an inflamnzatoi-y response. J Immunol, 2002. 168(12)(Jun 15): p. 6436-45;
Filippini, A., et al., Ecto-ATPase activity in cytolytic T-lym.plzocytes. Protection from the cytolytic effects of extracellular ATP. J Biol Chem, 1990. 265(1): p. 334-40). The present application discloses that RTF is the primary surface ATPase with this function.
RTF is the a-subunit of a2-isoform of vacuolar ATPase, and that RTF exists as a 70 kDa protein (Toyomura, T., et al., Three subunit a isofornzs of mouse vacuolar H(+)-ATPase. Preferential expression of the a3 isofoYm during osteoclast diferentiation. J
Biol Chem, 2000. 275(12): p. 8760-5; Nichols, T.C., et al., Expression of a membYane forrn of the pregnancy-associated protein TJ6 on lynzphocytes. Cell Immunol, 1994.
155(1): p. 219-29; Boomer, J.S., et al., Regeneration and tolerance factor's potential role in T-cell activation and apoptosis. Hum Immunol, 2000. 61(10): p. 959-71). A 20 kDa fragment can be cleaved, leaving the 50 kDa external membrane form (Nichols, T.C., et al., Expression of a meinbrane forin of the pyegnancy-associated protein TJ6 on lymphocytes. Cell Iinmunol, 1994. 155(1): p. 219-29). Current data support that RTF is known to have a role in certain forms of apoptosis (Boomer, J.S., et al., Regeneration and tolerance factor's potential role in T-cell activation and apoptosis. Hum Immunol, 2000. 61(10): p. 959-71; Boomer, J.S., et al., Regeneration and tolerance factor is expressed during T-lymphocyte activation an.d plays a role in apoptosis. Hum Immunol, 2001. 62(6): p. 577-88). It has also been shown equally that anti-RTF antibody can bloclc RTF's ATPase activity, subsequently allowing ATP to bind P2Z and induce apoptosis. It has additionally been shown that RTF works in an antagonistic fashion versus the P2Z surface purinoceptor. United States Patent No. 6,133,434 and International Patent Application (PCT) No. WO 95/33048 disclose that antibodies against P2Z receptors can be useful in treating a variety of diseases and conditions, including epilepsy, cognition, emesis, pain (especially migraine), asthma, peripheral vascular disease, hypertension, diseases of innnune system, irritable bowel syndrome and premature ejaculation.
One possible mechanism for RTF to modulate inflammatory and immune responses is by RTF acting as an ATPase to reduce the level of extracellular ATP. ATP, released during injury or stress from tissues, infiltrating cells, and dying cells at the site of inflainmation, interacts with the P2Z receptor. This interaction exacerbates the inflammatory response which leads to cell activation and the maturation and secretion of IL-1(3. By controlling the degradation of extracellular ATP, RTF down regulates immune responses, including immune activation and inflamination. Thus, inhibiting the RTF activity leads to irmnune activation, which may lead to cell death, while enhancing the RTF activity leads to inhibition of inflammatory responses.
Any metliod that inhibits RTF activity can be employed in the present invention for immune activation. In a preferred embodiment, the method to inhibit RTF is by administering a RTF antagonist. The RTF antagonist can be a small molecule, a peptide, an antibody or a fragment thereof, and the like. The antibody can be either polyclonal or monoclonal. In one embodiment, the antagonist acts by binding to RTF to inhibit its activity. A preferred RTF antagonist is an anti-RTF antibody or a fragment thereof, and a preferred anti-RTF antibody is a monoclonal RTF antibody or a fragment thereof. A method to produce and purify a monoclonal RTF antibody is disclosed by Boomer et al. (Boomer, J.S., et al., Regeneration and tolerance factor's potential role in T-cell actfvation and apoptosis. Hum Immunol, 2000. 61(10): p. 959-71).
In another embodiment, the antagonist acts by inln'biting cellular expression of RTF. An example of such an antagonist includes, but is not limited to, an antisense nucleic acid which binds to the DNA codi.ng for the RTF molecule. Using antisense nucleic acid to bind to DNA
to inhibit cellular expression of proteins is well known in the art. Another example of antagonizing RTF is by inducing RNA interference (RNAi). The RNAi process induces the degradation of the target gene mRNA so as to silence the target gene expression. A
typical RNAi process includes inttoducing into cells a double-stranded interfering RNA
that comprises a sense RNA having the sequence homologous to the target gene mRNA
and antisense RNA having the sequence complementary to the sense RNA. The double-stranded interfering RNA can be a small interfering double stranded RNA (also known as siRNA). The double-stranded interfering RNA can be introduced to the cells exogenously, or it can be expressed intracellular by incorporating into the cells an expression system to express the double-stranded interfering RNA.
Intracellular expression is preferred for siRNA.
The RTF autagonist can be prepared as a pharmaceutical formulation by including one or more suitable or appropriate excipients. In an embodiment, the pharmaceutical formulation is a nanoparticle or a microparticle. The antagonist can be administered to the subject by any route including but not limited to parenteral, topical, nasal, pulmonary, ophthalmic, bucal, vaginal, transdermal, intrathecal, and oral One of the results of inhibiting RTF activity is induction of cell apotosis, especially cancer cells, such as ovarian carcinoma cells. Thus, inhibiting RTF
can be used as a method to treat cancer. Any of the methods described above which inhibit RTF can then be used as a method to treat cancer. An example of cancer that can be treated in the present invention is ovarian cancer.
Since inflammatory responses are the result of immune activation by the injured tissue, it is anticipated that enhancing RTF activity will result in inhibiting the inflammatory responses. Inflammation is known in many disorders which include, but are not limited to, arthritis (including but not limited to rheumatoid arthritis, 7.

spondyloarthopathies, gouty arthritis, osteoarthritis, systemic lupus erythematosus and juvenile arthritis), asthina, bronclzitis, menstrual cramps, tendonitis, bursitis, and skin related conditions (such as psoriasis, eczema, bums and dermatitis), certain gastrointestinal conditions (such as inflammatory bowel disease, Crohn's disease, gastritis, irritable bowel syndrome and ulcerative colitis), vascular diseases, migraine headaches, periarteritis nodosa, thyroiditis, aplastic anemia, Hodgkin's disease, sclerodoma, rheumatic fever, type I diabetes, myasthenia gravis, multiple sclerosis, sarcoidosis, nephrotic syndrome, Behcet's syndrome, polymyositis, gingivitis, hypersensitivity, conjunctivitis, swelling occurring after injury, myocardial ischemia, certain central nervous system disorders (such as Alzheimer's disease and dementia), allergic rhinitis, respiratory distress syndrome, endotoxin shock syndrome, atherosclerosis, central nervous system dainage resulting from stroke, ischemia, trauma, and the like. Many of the above inflammatory disorders are autoimmune disorders in which the body develops iminune responses to its own protein. Thus, enhancing the activity of RTF can be used to treat or prevent inflaminatory disorders, including autoiminune disorders, or their sylnptoms.
Any method that enhances the level of activity of RTF can be used to inhibit inflammatory responses and to treat or prevent inflammation in the present invention.
In a preferred embodiment, the method is to administer to the subject an effective amount of RTF or a fragment thereof. In an embodiment, the RTF is an intracellular form which has a molecular weight of about 70 kDa. In another embodiment, the RTF
is extemal meinbrane form having a molecule weight of about 50 kDa.
In one embodiment, the RTF can be isolated or purified from a mammalian source, such as murine or human, which includes but is not limited to, lymphocyte, macrophage, thymus, fetalplacental tissue, and the lilce. The lymphocyte can be a T-lymphocyte or a B-lymphocyte. The T- or B-lymphocyte preferably is activated.
In another embodiment, the RTF or a fragment thereof can be synthetic or a recombinant RTF, which can be derived, for example, from a nucleic acid sequence (e.g. cDNA) encoding the amino acid sequence of the RTF or the fragment. The nucleic acid sequence can be incorporated by recombinant techniques into an appropriate host organism for the expression of the RTF or the fragment. Examples of host organisms include, but are not limited to, bacteria, fitngi, yeasts, protozoa, transgenic animals, transgenic plants, and the like. Recombinant techniques are well known to those skilled in the art and many biopharmaceuticals have been successfully produced by such techniques. A method for producing a recombinant RTF in E. coli is disclosed by Lee et al. (Lee, G. W., Bommer, J,S., Gilman Sachs, A., Chedid A., Gudelj, L., Rukavina, D.
and Beaman, K. D. Regeneration and tolerance factor of the human placenta induces IL-10 itzduction. Eur J Immunol, 2001. 31: p. 687-691) , In a preferred embodiment of the present invention, the RTF or the fragment is formulated with one or more acceptable pharmaceutical excipients for administration to the subject. The RTF or the fragment can also be formulated as microparticles or nanoparticles. The microparticles or nanoparticles can further include ligands to deliver the microparticles or the nanoparticles to a target tissue such as the T- or B-lymphocyte or the macrophage. The RTF or the fragment can be administered to the subject by any acceptable route including, but not limited to, parenteral, topical, nasal, pulmonary, ophthalmic, bucal, vaginal, transderm.al, intrathecal, oral and the like.
In another embodiment, the method to enhance RTF activity can be a method which enhances the expression of cellular RTF.
In yet another embodiment of the present invention, the method of treating inflam.mation by enhancing the RTF activity may include an additional step of administering to the subject a TNF-a antagonist. Since both IUl P and TNF-a are known to cause inflammation, inhibiting the secretion of IL-1 P by RTF and bloclQng, the TNF-a activity by a TNF-a antagonist should have synergistic effects in treating inflammation. There are many known TNF-a antagonists. Examples include, but are not limited to, infliximab, etanercept, D2E7, CDP571, CDP870, thalidomide and its analogs, and phosphodiesterase type IV inhibitors.
EXAMPLES
Example 1: Cell Culture J774A1 cells were maintained by allowing them to adhere to the bottom of a T75 flask with 25 mis of RPMI 1640 complete media at 37 C in a 5% COZ atmosphere.
Cells were split into a new flask every 4 days using trypsin and a gentle wash with RPMI.
Example 2: ATPase Assay PBMC or J774A1 macrophages were suspended in 50 1 of RPMI at a density of 2.5x105 in a 96 well flat bottom plate and incubated with 10 g of either 2C1 (anti-RTF) or an isotype matched control antibody for 45 mins at 37 C in a 5% CO2 atmosphere.

Cells were washed with PBS and re-suspended in 100 1 of assay buffer (5mM KC1, 2mM CaC12, 2mM MgC12, and 1.5 C of [y-32P]ATP) in 1.5m1 eppendorf tubes. The ATPase reaction was allowed to continue for 10 mins at room teinperature. 500 1 of 20% w/v activated charcoal (in distilled water) was added to scavenge any unused [7-32P]ATP and allowed to sit for 10 mins on ice. The cells were centrifuged at 3200 x g for 5 mins, pelleting the charcoal. 500 1 of supernatant containing released y-32P was removed and added to scintillation vials wit113mis of scintillation fluid and read in the scintillation counter for 1 min/tube (Filippini, A., et al., Ecto-ATPase activity in cytolytic T-lymphocytes. Protection ftom the cytolytic effects of extracellular ATP. J
Biol Chem, 1990. 265(1): p. 334-40; Doinbrowski, K.E., et al., Identification and partial characterization of an ectoATPase expressed by human natural killer cells.
Biochemistry, 1993. 32(26): p. 6515-22; Dombrowski, K.E., et al., Antigen f ecognition by CTL is dependent upon ectoATPase activity. J Imtnunol, 1995. 154(12):
p..6227-37).
Example 3: Apoptosis Induction Approximately 2.5x105 PBMC were suspended in 100 1 of RPMI in a 96 well round bottom plate and a final concentration of either 3mM ATP, 0.75mM ATP, 10 g anti-RTF, or 0.751nM ATP + 10 g anti-RTF together was added. Cells were incubated in these conditions at 37 C in 5% CO2 for 6 hours. Cells were washed twice in 0.01%
BSA/PBS, and re-suspended in 100 1 binding buffer (10mM HEPES pH 7.4, 140mM

NaCI, 2.5mM CaC12) with 5 l of annexin-V/FITC (BD/Pharmingen) for 15 mins.
Cells were washed in binding buffer and re-suspended in 300 1 of binding buffer with 3 l of lmg/ml propidium iodide. Apoptosis was measured by quantifying annexin-V
binding by flow cytometry by gating on PI-/annexin-V+ cells (Vermes, L, et al., A
novel assay for apoptosis. Flow cytonaetr-ic detection of phosphatidylserine expression on eaf-ly apoptotic cells using fluorescein labeled Annexin V. J Im.munol Methods, 1995.
184(1):
p. 39-51).

Example 4: P2Z and RTF Staining A total of 2.5x105 cells were washed twice with 0.01 %BSA/PBS for 5 mins and re-suspended in 100nl of staining buffer. 5 1 of rabbit anti-P2X7 (P2Z) (Chemicon) antibody, 5 1 anti-RTF-FITC, or 51il of isotype matched control antibody was added to the appropriate tubes and allowed in react in the dark for 15 mins. Cell were washed again and re-suspended in 100 1 of staining buffer (1X PBS, 5% FBS) with 5 I
of anti-Rabbit biotinylated antibody (Pharmingen) and allowed to react for 15 mins. A
third wash was performed and 20 of Streptavidin-PE (Pharmingen) was added for 15 mins.
After a final wash, the cells were re-suspended in 300 1 of staining buffer and run on a BD Facscalibur flow cytometer.

Example 5: Propidium Iodide and Lucifer Yellow Incflraoration J774 cells were suspended in 100 .1 of RPMI in a 96 well round bottom plate at a density of 2.5x105 in on of the following conditions: 3mM ATP, 0.75mM ATP, 50 g anti-RTF, or 0.75mM ATP + 10 g anti-RTF. Cells were incubated at 37 C in 5%

for 30 mins in the presence of l g PI or LY, washed in PBS, re-suspended in PBS and analyzed in the BD FACSCALIBURTM flow cytometer.

For slides, J774 cells were grown in 4 well chamber slides for 2 days. The supematant was removed and each chamber was incubated in one of the conditions described above for 15 mins in the presence of 1 g PI or LY. The slides were washed in PBS, and incubated with l0 Trypan blue for 5 mins, washed again and examined under a fluorescent microscope.

Example 6: Confocal Microscopy PBMC were obtained by FICOLLTM separation and 2.5x105 cells per well were incubated in a 96 we11 plate in RPMI 1640 medium in the presence of 2 g/ml PHA, or RPMI alone. After 72 hours both unstimulated, and stimulated cells were washed twice in PBS, permeabilized with ice cold 70% ethanol for 15 mins, and reacted with 7 l anti-RTF-FITC, anti-CD8-PE (Pharmingen), or isotype control antibody for 15 mins.
Cells were washed again, fixed with Coulter fixative solution, and a CYTOSPINTM was used to transfer them to a positively charged slide which was mounted with Dapco medium.
Slides were then exanmined by confocal microscopy.

Exainple 7: RTF Expression Follows P2Z Expression on Lymphocytes It has been previously shown that RTF expression in lymphocytes is dependent on activation (Boomer, J.S., et al., Regeneration and tolerance factor's potential role in T-cell activation and apoptosis. Hum Immunol, 2000. 61(10): p. 959-7). To characterize RTF expression in relation to P2Z expression during the immune response, the surface expression of RTF and P2Z on lymphocytes was measured following stimulation with PHA for 24-72 hours. In resting lymphocytes, RTF expression was 14.42 0.06% that of maximal expression while P2Z expression was 35.23 8.5%.
RTF and P2Z expression increased togetller on the surface of the cells as stimulation time increased, with RTF and P2Z expression at 88.64 8.8% and 81.34 13.2%
respectively at 72 hours (Figure 1). Both molecules matched the others percent maximal expression at 0 and 24 hours, reaching maximal expression at 72 hours (Figure 2)..
The expression of RTF and P2Z on the J774A1 macrophage cell line has also been examined. Figure 1 shows that each molecule to be expressed in a constitutive manner.

Example 8: RTF Exists as a 50 kDa Protein on the Surface of Activated Cells, and as a 70 kDa Protein Inside of Resting Cells When lymphocytes examined by western blot at the time points stimulated above (Figure 3a), unstimulated lymphocytes expressed a 70 kDa protein of RTF as their major form of RTF. Upon stimulation, expression of RTF fell below detectable levels at a time period of 16-24 hours. At a time period of 24-72 hours after stimulation RTF
expression returned to detectable levels, however predominantly in a 50 kDa sized form (Figure 3a).
It is hypothesized that the 70 kDa form represents the intracellular form of RTF, while the 50 kDa form represents the external membrane fonn. To test this, the activated lymphocytes from Figure 3a were further examined by confocal microscopy at 0 and 72 hours of stimulation (Figure 3b). In this figure RTF (green) is present in the cytoplasm of un-stimulated cells. In cells stimulated with PHA for 72 hrs, RTF
has re-localized to the surface of the membrane. Consistent with the western blot shown in Figure 3a, these findings suggest to us that the 70 kDa protein is intracellular in resting lymphocytes, whereas the 50 lcDa protein is found on the surface of activated lymphocytes.

Example 9: RTF Regulates Surface ATPase Activity in Lymphocytes and Macrophages It is hypothesized that RTF regulates surface ATPase activity and anti-RTF
blocks it. To demonstrate that RTF has surface ATPase activity, PBMC and macrophages were incubated with either anti-RTF or isotype control antibody for 45 mins, and their surface ATPase activity was measured by [y32-P]ATP degradation (Filippini, A., et al., Ecto-ATPase activity in cytolytic T-lynaphocytes.
Protection fy-onz the cytolytic effects of extracellular ATP. J Biol Chem, 1990. 265(1): p. 334-40;
Dombrowski, K.E., et al., Identification and partial characterization of an ectoATPase expressed by hufnan natural killer cells. Biochemistry, 1993. 32(26): p. 6515-22;
Dombrowski, K.E., et al., Antigen recognition by CTL is dependent upon ectoATPase activity. J Iinmunol, 1995. 154(12): p. 6227-37). The PBMC incubated with anti-RTF
had a 10 fold decrease in surface ATPase activity with 11,758 3,394 cpm compared to PBMC incubated witli isotype antibody with a cpm of 115,592 23,891.
(P<0.001;
Figure 4). The J774A1 cells incubated with anti-RTF had a complete iiiliibition of surface ATPase activity compared with the isotype control (45,752 16,175 cpm;
P<0.001; fig 4). Thin layer chromatography confinned that anti-RTF completely iiiliibited the surface ATPase activity (data not shown).

Example 10: RTF Prevents ATP from Inducing Apoptosis Since RTF moves to the surface of the membrane as a 50 kDa size protein after stimulation in PBMC, and RTF regulates ATPase activity, it is desirable to demonstrate that RTF can prevent ATP induced apoptosis. For this purpose the macrophage cell line J774A1 is chosen. J774A1 cells were to be used as a model to demonstrate the interaction of RTF with P2Z because this cell line expresses both molecules constitutively and is known to undergo P2Z mediated, ATP induced apoptosis (Di Virgilio, F., The P2Z purinoceptor: an intriguing role in ifnnaunity, inf anaination and cell death. Immunol Today, 1995. 16(11): p. 524-8; Coutinho-Silva, R. and P.M.
Persechini, P2Z purinoceptor-associated pores induced by extracellular ATP in niacrophages and J774 cells. Ain J Physiol, 1997. 273(6 Pt 1): p. C1793-800).
If the theory that RTF regulates surface ATPase activity and therefore regulates ATP
binding to P2Z is correct, then it follows that anti-RTF and ATP would work together to generate more apoptosis than either alone by blocking the ATPase activity of RTF.
When added together, at the same concentration as both alone, a near maximal amount of apoptosis occurred as measured by annexin-V (Figure 5). When 3mM ATP was added, 84.14 1.63 % of the cells underwent apoptosis, but when the amount of ATP

was decreased to 0.75mM, only 13.26 3.6% apoptosis was seen. When 10 g of anti-RTF was added alone, 15.49. 4.05% of the cells were apoptotic. However, anti-RTF
and 0.75mM ATP were added together, apoptosis was 83.05 5.09%. When ATPase was added back, no apoptosis occurred showing that RTF's apoptosis inducing ability is dependent on the presence of ATP (Figure 5). Previously, in initial experiments we demonstrated that anti-RTF had no effect on the ATPase used, negating the possibility of their interaction (data not shown).

Example 11: RTF Prevents P2Z Activation A standard teclmique for detecting P2Z activity is used to demonstrate that the ultimate cause of the apoptosis inducing effect of RTF and ATP was due to P2Z
activation (Di Virgilio, F., The P2Z purinoceptor: an intriguing role in imnzunity, inflammation and cell death. Immunol Today, 1995. 16(11): p. 524-8; Coutinho-Silva, R. and P.M. Persechini, P2Z pur-inoceptor-associated pores induced by extracellular ATP in macrophages and J774 cells. Am J Physiol, 1997. 273(6 Pt 1): p. C1793-800;
Steinberg, T.H., et al., ATP4- permeabilizes the plasma menzbrane of mouse macrophages to fluorescen.t dyes. J Biol Chem, 1987. 262(18): p. 8884-8). When ATP
interacts with P2Z, non-specific channels of about 0.9 kDa in size will open in the membrane (Dubyak, G.R. and C. el-Moatassim, Signal transduction via P2-purinergic receptors for extracellular ATP and other nucleotides. Atn J Physiol, 1993.
265(3 Pt 1):
p. C577-606; Di Virgilio, F., The P2Z purinoceptor: an intriguing role in immunity, inflammation and cell death. Immunol Today, 1995. 16(11): p. 524-8; Hickman, S.E., et al., P2Z adenosine triphosphate receptor activity in cultured human monocyte-derived macrophages. Blood, 1994. 84(8): p. 2452-6; Greenberg, S., et al., Extracellular nucleotides nzediate Ca2+ fluxes in. J774 macrophages by two distinct n2echanisnzs. J
Biol Chem, 1988. 263(21): p. 10337-43). This allows the testing for P2Z's influence by measuring the influx of molecules smaller than 0.9 kDa, to which the cell is normally not permeable. Propidium iodide (PI) (0.414 kDa) and lucifer yellow (LY) (0.46 kDa) should be allowed through if P2Z is activated, and trypan blue (TB) (0.96 kDa) should be excluded under all circumstances unless necrosis has occurred (Di Virgilio, F., The P2Z purinoceptor: an intriguing role in immunity, inflammation and cell death.
Imn.iunol Today, 1995. 16(11): p. 524-8; Coutinho-Silva, R. and P.M.
Persechini, P2Z
purinoceptor-associated pores induced by extracellular ATP in inacrophages and cells. Am J Physiol, 1997. 273(6 Pt 1): p. C1793-800; Steinberg, T.H., et al., pe>^meabilizes the plasma zzzeznbrane of mouse macrophages to fluorescent dyes. J Biol Chem, 1987. 262(18): p. 8884-8). The same conditions used in the previous experiment were repeated to observe and measure the entry of propidium iodide or trypan blue in J774A1 cells. Thus P2Z influence would be characterized by cells that were PI
and LY
positive but TB negative.
Using flow cytometry, cells incubated with anti-RTF + 0.75mM ATP had pronounced channel formation judging from their incorporation of PI (Figure 6). This was confirmed by immunofluorescence microscopy (Figure 6). Cells reacted with anti-RTF and 0.75mM ATP together were found to be permeable to PI, but not to trypan blue (Figure 6). In contrast, control cells permeabilized with 70% ETOH were positive for both dyes (data not shown). Cells that were incubated with anti-RTF alone or 0.75mM ATP alone (Figure 6) showed no PI incorporation, while those incubated with both did incorporate PI. These results were confirmed using lucifer yellow (not shown).
This demonstrates that anti-RTF and ATP togetlier can induce the opening of channels >0.9 kDa, and yet shows membrane integrity is still intact.

Example 12: RTF regulates IL-1(3 secretion To show that RTF regulates IL-1p secretion, THP-1 macrophages were incubated with both 1mM and 3mM ATP, or anti-RTF and assayed for IL-1(3 secretion by ELISA. Since anti-RTF abrogates RTF function, our hypothesis was that anti-RTF
would induce IL-(3 secretion, showing that RTF is a regulator for this molecule.
5x105 THP macrophage cells were stimulated with 5nghnl of LPS and 1x105 PBMC for 2.5 hours. These cells were then washed and iiicubated in a final concentration of either 0 ATP, 3mM ATP, 1mM ATP, or anti-RTF for a period of 3 hours. The supernatants were assayed for IL-1(3 by ELISA (R&D systems). pg IL-1(3 per well was calculated from the optical densities. Results are expressed as the mean of 3 independent experiments.

As shown in Figure 7, cells incubated with media alone, secreted 61+1-14 pg IL-1 j3, forming the baseli.ne. Cells incubated with 3mM ATP secreted 285+/-25 pg (78+/-5.1% above baseline). Cells incubated with 1.mM ATP secreted 130+/-44 pg IL-1(3 (49+/-5.6 above baseline). Ce1Ls incubated with anti-RTF alone secreted 114 +/-22 pg IIrl (3 (an increase 47+1-3.5%), about the equivalent of cells incubated with 1mM ATP.
This shows that anti-RTF alone can induce secretion, suggesting that RTF is a regulator oflL-1(i.

Example 13: Anti-RTF is-cytotoxic to ovarian carcinoma cell A2780 ovarian carcinoma cells were grown in 96 well plates until 100%
confluent. Wells were identified, and cells were then incubated with 3mM ATP, 6mM
ATP, 20 g of anti-RTF and 20 ug of anti-RTF combined with 3mM ATP and 6mM
ATP. Cells were incubated for 18 hours at 37 C in a 5% CO 2 environment. Cells were then harvested and assayed using flow cytometry for Caspase 3 activation as an indicator for cell death. As shown in Figure 8, anti-RTF is significantly more cytotoxic then extracellular ATP at the indicated concentrations. Addition of extracellular ATP
did not significantly enhance cytotoxicity.

Example 14. In vivo effect of anti-RTF
Female athymic nude mice purchased from Harlan Sprague Dawley approximately 11 weeks old. All mice were injected with 200 1 of a 1:1 slurry of a MATRIGELTM mix containing 5x 106 A2780 ovarian carcinoma cells. Tumor cells were transplanted subcutaneously in the upper flank of the right leg. Two mice were untreated and kept as a control to measure tumor growth. The remaining six mice were injected with anti RTF antibody. All mice were left untreated for I week in order for a tumor and its vascular bed to become established. On the eighth day all mice that were treated were injected with 100ug of anti RTF antibody in a volume of 100 1 of PBS.
Injections were given in the tumor bearing flank of the animal. Antibody was not injected directly into the tumor, but was injected into the area of the tumor bed. Mice received 300 g of antibody per week using a 28 gage needle. Mice were injected every Sunday, Tuesday and Friday. Injections took place over 5 weeks. The experiment was terminated when the control mice tumor burden became too obstructive for mice to ambulate.
The median tumor volume of the control animals after 5 weeks without anti-RTF
treatment was 8448 mm3 . The median tumor volume for the anti-RTF treated animals(n=6) was 364 mm3. In two of the treated animals no measurable tuinor was observed (tumor volume range 0-1670). These results clearly indicate the possible usefiilness of the antibody and or the inhibition of the RTF associated ATPase complex in tumor therapy It should be understood that various changes and modifications to the presently preferred embodiments described herein will be apparent to those skilled in the art.
Such changes and modifications can be made witliout departing fiom the spirit and scope of the present invention and without diminishing its intended advantages. It is therefore intended that such changes and modifications be covered by the appended claims.

Claims (2)

1. The use of an anti-Regeneration and Tolerance Factor (RTF) antibody for inducing apoptosis in an ovarian carcinoma cell.
2. The use of claim 1 wherein the antibody is a monoclonal antibody or a polyclonal antibody.
CA2515366A 2003-02-10 2004-02-10 Compositions and methods to modulate immune and inflammatory responses Expired - Fee Related CA2515366C (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US44649903P 2003-02-10 2003-02-10
US60/446,499 2003-02-10
US10/774,708 2004-02-09
US10/774,708 US7211257B2 (en) 2003-02-10 2004-02-09 Methods for inducing apoptosis in ovarian carcinoma cells using an anti-regeneration and tolerance factor antibody
PCT/US2004/003969 WO2004071441A2 (en) 2003-02-10 2004-02-10 Compositions and methods to modulate immune and inflammatory responses

Publications (2)

Publication Number Publication Date
CA2515366A1 CA2515366A1 (en) 2004-08-26
CA2515366C true CA2515366C (en) 2010-04-20

Family

ID=32871987

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2515366A Expired - Fee Related CA2515366C (en) 2003-02-10 2004-02-10 Compositions and methods to modulate immune and inflammatory responses

Country Status (4)

Country Link
US (1) US7211257B2 (en)
EP (1) EP1596811A4 (en)
CA (1) CA2515366C (en)
WO (1) WO2004071441A2 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8138160B2 (en) 2006-08-03 2012-03-20 Warsaw Orthopedic, Inc. Reagents, methods and systems to suppress pro-inflammatory cytokines

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5196526A (en) 1989-11-28 1993-03-23 University Of Health Sciences/The Chicago Medical School CDNA clone for T-cell suppressor inducer factor
EP0760850A1 (en) 1994-05-27 1997-03-12 Glaxo Group Limited P2x RECEPTORS (PURINOCEPTOR FAMILY)
US6133434A (en) 1997-04-28 2000-10-17 Glaxo Group Limited Purinergic receptor
NZ502568A (en) 1997-08-05 2003-01-31 Stressgen Biotechnologies Corp Compositions to enhance immune responses against HPV (human papilloma virus) protein antigens and against tumours exhibiting an HPV protein antigen

Also Published As

Publication number Publication date
WO2004071441A3 (en) 2006-01-05
CA2515366A1 (en) 2004-08-26
US7211257B2 (en) 2007-05-01
EP1596811A4 (en) 2007-10-03
WO2004071441A2 (en) 2004-08-26
EP1596811A2 (en) 2005-11-23
US20040219145A1 (en) 2004-11-04

Similar Documents

Publication Publication Date Title
Li et al. Inflammation: a novel therapeutic target/direction in atherosclerosis
JP2022081543A (en) Anti-trem2 antibodies and methods of use thereof
JP5164167B2 (en) Agonists, antagonists and immunotoxins of immunoregulatory tumor necrosis factor receptor 25 (TNFR25)
ES2328597T3 (en) MODULATION OF NKG2D.
JP5569946B2 (en) Immunosuppressants and preventive and therapeutic agents for autoimmune diseases
US11485787B2 (en) Agents that modulate RGMb-neogenin-BMP signaling and methods of use thereof
TW201934552A (en) Diaryl substituted 5,5-fused ring compounds as C5aR inhibitors
US11352400B2 (en) PI3K gamma inhibitor peptide for treatment of respiratory system diseases
CN111032658B (en) 5-5 fused rings as C5a inhibitors
WO2014058915A2 (en) Therapies based on control of regulatory t cell stability and function via a neuropilin-1:semaphorin axis
WO2020227159A2 (en) Methods of modulating immune activity
TW201900212A (en) Method for treating immune-related adverse events in cancer therapy using soluble CD24
US20130011835A1 (en) Compositions and methods for treating inflammation
US10975144B2 (en) CDNA encoding a monoclonal anti-IL-32 antibody and methods for the production of the antibody
Ko et al. A novel modified RANKL variant can prevent osteoporosis by acting as a vaccine and an inhibitor
KR20210053306A (en) Combination therapy with a C-C chemokine receptor 4 (CCR4) antagonist and one or more immune checkpoint inhibitors
US9428568B2 (en) Compositions and methods for modulating cell-cell fusion via intermediate-conductance calcium-activated potassium channels
JP6151692B2 (en) Methods and compositions for modulating voltage-gated calcium channel function
CA2515366C (en) Compositions and methods to modulate immune and inflammatory responses
WO2011000384A1 (en) Method for suppressing tumor growth by blocking fibroblast growth factor receptor, and method for diagnosing malignant neoplasms
WO2008055889A1 (en) Compositions and methods of treating oncological, inflammatory and autoimmune diseases mediated by sema4a
US7943130B2 (en) Methods and compositions relating to CCR5 antagonist, IFN-γ and IL-13 induced inflammation
Park et al. Activation of formyl peptide receptor 1 elicits therapeutic effects against collagen‐induced arthritis
EP3538550A1 (en) Or10h1 modulators and uses thereof
US20230128075A1 (en) Monoclonal antibodies targeting hsp70 and therapeutic uses thereof

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20130211