US20140135337A1 - Induction of immune tolerance by using methotrexate - Google Patents

Induction of immune tolerance by using methotrexate Download PDF

Info

Publication number
US20140135337A1
US20140135337A1 US14/116,486 US201214116486A US2014135337A1 US 20140135337 A1 US20140135337 A1 US 20140135337A1 US 201214116486 A US201214116486 A US 201214116486A US 2014135337 A1 US2014135337 A1 US 2014135337A1
Authority
US
United States
Prior art keywords
methotrexate
cells
matg
treatment
alemtuzumab
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.)
Abandoned
Application number
US14/116,486
Other languages
English (en)
Inventor
Alexandra Joseph
Susan Richards
Melanie Ruzek
Richard Garman
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.)
Genzyme Corp
Original Assignee
Genzyme Corp
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=47177257&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20140135337(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Genzyme Corp filed Critical Genzyme Corp
Priority to US14/116,486 priority Critical patent/US20140135337A1/en
Publication of US20140135337A1 publication Critical patent/US20140135337A1/en
Assigned to GENZYME CORPORATION reassignment GENZYME CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: RUZEK, MELANIE, GARMAN, RICHARD, JOSEPH, Alexandra, RICHARDS, SUSAN
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0008Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • This invention relates generally to immunology, and more specifically to the use of methotrexate to reduce undesired immune responses in patients.
  • the therapeutic proteins may elicit antibody responses in the patient (Schellekens, Nat Rev Drug Discov, 1(6):457-62 (2002); Zinkernagel, Semin Immunol, 12(3):163-71 (2000), discussion 257-344; Thorland et al., Haemophilia, 5(2):101-5 (1999); Goodeve, Blood Coagul Fibrinolysis, 14 Suppl 1:S17-21 (2003)).
  • ADA anti-drug antibody
  • ADA can develop in the enzyme-replacement therapy (ERT) against recombinant human acid alpha-glucosidase.
  • ERT enzyme-replacement therapy
  • patients who do not express measurable amounts of the endogenous enzyme sustained levels of high antibody titer correlate with patient decline (CRIM-Pompe patients) (Kishnani et al., Mol Genet Metab., 99(1):26-33 (2010); Hunley et al., Pediatrics, 114(4):e532-5 (2004); Amalfitano et al., Genet Med, 3(2): 132-8 (2001)).
  • ADA can also induce autoimmune disease as in the case of recombinant human erythropoietin (Schellekens, Clin Ther, 24(11):1720-40 (2002), discussion 1719; Locatelli et al., Perit Dial Int, 27 Suppl 2:S303-7 (2007)).
  • ADA responses can also occur with antibody therapeutics regardless of whether the therapeutics are non-human derived, humanized or even fully human.
  • the immunogenicity of both monoclonal and polyclonal antibody therapeutics can influence patient safety and drug efficacy.
  • Antibodies that develop against therapeutic monoclonal antibodies such as infliximab, adalimumab, rituximab and natalizumab have been associated with decreased serum levels and efficacy of the therapeutic antibodies (Bendtzen et al., Arthritis Rheum, 54(12):3782-9 (2006); Schmidt et al., Clin Immunol, 132(3):334-41 (2009); Bartelds et al., Ann Rheum Dis, 66(7):921-6 (2007); Baert et al., N Engl J Med, 348(7):601-8 (2003); Tahir et al., Rheumatology (Oxford), 44(4):561-2 (2005); Maini et
  • Alemtuzumab is another antibody therapeutic that can generate ADA in relapsing-remitting multiple sclerosis patients (Coles et al., N Engl J Med, 2008. 359(17):1786-801 (2008)).
  • Alemtuzumab is a lymphocyte-depleting monoclonal antibody that interacts with CD52, a cell surface antigen expressed on immune cells.
  • Alemtuzumab is in late-stage clinical trials for treating relapsing-remitting multiple sclerosis and has also been evaluated in rheumatoid arthritis.
  • Thymoglobulin® is also associated with deleterious ADA in a small subset of patients. Serum sickness, acute renal failure and cardiovascular reactions have been observed in Thymoglobulin®-treated transplant recipients (Boothpur et al., Am J Kidney Dis., 55(1):141-3 (2009); Lundquist et al., Liver Transpl, 13(5):647-50 (2007); Busani et al., Minerva Anestesiol, 72(4):243-8 (2006); Tanriover et al., Transplantation, 80(2):279-81 (2005); Buehler et al., Clin Transplant, 17(6):539-45 (2003)).
  • the invention provides a method of inducing immune tolerance in a subject in need of treatment with a therapeutic.
  • this method one administers to the subject an effective amount of methotrexate in a single cycle, thereby inducing immune tolerance toward the therapeutic in the subject.
  • the invention also provides a method of inhibiting antibody responses to a therapeutic in a subject in need of treatment with the therapeutic.
  • this method one administers to the subject an effective amount of methotrexate in a single cycle, thereby inhibiting antibody responses to the therapeutic in the subject.
  • the invention also provides a method of alleviating an infusion reaction to a therapeutic in a subject in need of treatment with the therapeutic.
  • this method one administers to the subject an effective amount of methotrexate, thereby alleviating an infusion reaction to the protein therapeutic in the subject.
  • the invention also provides a method of reducing secondary autoimmunity in an autoimmune subject in need of treatment with a therapeutic.
  • this method one administers to the subject an effective amount of methotrexate, thereby reducing secondary autoimmunity in the subject.
  • the invention also provides a method of increasing the efficacy of a therapeutic in a subject in need of treatment with the therapeutic.
  • this method one administers to the subject an effective amount of methotrexate, thereby increasing the efficacy of the protein therapeutic in the subject.
  • the invention also provides a method of increasing the percentage of T regulatory cells in the T cell population in a subject treated with a lymphocyte-depleting therapy, e.g., an alemtuzumab therapy or a Thymoglobulin® therapy.
  • a lymphocyte-depleting therapy e.g., an alemtuzumab therapy or a Thymoglobulin® therapy.
  • the invention also provides a method of increasing the percentage of B regulatory cells in the B cell population in a subject in need of treatment with a protein therapeutic such as an antibody therapeutic.
  • a protein therapeutic such as an antibody therapeutic.
  • the effective amount of methotrexate is administered in a single cycle.
  • the invention provides a method of increasing TGF-beta-, IL-10-, and/or FoxP3-expressing B cells in a subject in need of treatment with a protein therapeutic, comprising administering an effective amount of methotrexate in a single cycle.
  • the invention also provides a method of prolonging the pharmacokinetics of a therapeutic agent in a subject.
  • one administers methotrexate to the subject, before or during or after administration of the therapeutic agent, in an amount effective to prolong the pharmacokinetics of the therapeutic agent.
  • the invention further provides a method of depleting lymphocytes in a human patient in need thereof.
  • a lymphocyte-depleting agent treats the patient with a lymphocyte-depleting agent and administers methotrexate to the patient, before or during or after the treatment with the lymphocyte-depleting agent, in an amount effective to induce immune tolerance in the patient toward the lymphocyte-depleting agent or to reduce secondary autoimmunity.
  • the effective amount of methotrexate is administered in a single cycle.
  • the lymphocyte-depleting agent may be a monoclonal antibody therapeutic (e.g., alemtuzumab or rituximab).
  • the patient may be a multiple sclerosis patient (e.g., a remitting-relapsing multiple sclerosis patient).
  • the lymphocyte-depleting agent may be a polyclonal antibody therapeutic (e.g., anti-thymocyte globulin polyclonal antibody).
  • the invention also provides a method of inducing immune tolerance in a subject in need of tissue transplantation.
  • the effective amount of methotrexate is administered in a single cycle.
  • the transplanted tissue is renal tissue or cardiac tissue.
  • the subject also receives an agent for immune-modulation (e.g., an immunosuppressant), such as polyclonal anti-thymocyte globulin antibody).
  • the invention also provides a method of inhibiting T cell responses in a subject in need of a therapeutic or tissue transplantation.
  • this method one administers to the subject an effective amount of methotrexate prior to, concurrently with, or after, treating the subject with the therapeutic or tissue transplantation, thereby inhibiting T cell responses in the subject.
  • the therapeutic is a protein therapeutic.
  • the therapeutic is an antibody therapeutic.
  • the antibody therapeutic may be a monoclonal antibody therapeutic and/or a lymphocyte-depleting agent (e.g., alemtuzumab), or a polyclonal antibody therapeutic (e.g., polyclonal rabbit anti-thymocyte globulin antibody).
  • the antibody therapeutic is alemtuzumab
  • the subject may be a multiple sclerosis patient.
  • the antibody therapeutic is polyclonal rabbit anti-thymocyte globulin antibody
  • the subject may be a human patient who is in need of organ transplantation, has aplastic anemia, and/or has or is at risk of having graft-versus-host disease.
  • the therapeutic is an enzyme.
  • the enzyme may be human alpha-galactosidase A or human acid alpha-glucosidase.
  • the subject is a human.
  • the effective amount of methotrexate may be 0.1 mg/kg to 5 mg/kg.
  • the single cycle of methotrexate may consist of 1 day of methotrexate administration, or 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11 consecutive days of methotrexate administration.
  • the single cycle of methotrexate may be administered between 48 hours prior to and 48 hours after the onset of the therapeutic treatment.
  • FIGS. 1A-B show anti-rabbit IgG responses to a single and multiple courses of mATG, a rabbit anti-murine thymocyte globulin polyclonal antibody. Average anti-rabbit IgG titers were measured, and mice treated with rabbit IgG (rbIgG) only were used as a control.
  • FIG. 1A shows responses to a single course of mATG over an 8 week period.
  • FIG. 1B shows responses to multiple courses of mATG over a 20 week period. Arrows indicate time points at which mATG or rbIgG were administered.
  • FIG. 2 shows average alemtuzumab-specific IgG titers following five monthly treatments with alemtuzumab. Arrows indicate time points at which alemtuzumab was administered.
  • FIG. 3 shows suppression of anti-mATG IgG responses by methotrexate (MTX) following a single course of mATG.
  • Mice were treated with mATG only, rbIgG only, or mATG and methotrexate.
  • FIGS. 4A-C show the effects of methotrexate on anti-rabbit IgG responses throughout a course of five monthly treatments with mATG. Arrows indicate time points at which mATG or methotrexate were administered.
  • FIG. 4A shows that three cycles of methotrexate significantly reduce average anti-rabbit IgG titers.
  • FIG. 4B shows that a single course of methotrexate significantly reduces average anti-rabbit IgG titers.
  • FIG. 4C compares the average anti-rabbit IgG titers of mice treated with mATG alone, mATG and a single cycle of methotrexate, or mATG and three cycles of methotrexate. A single cycle of methotrexate reduces anti-rabbit IgG titers more significantly than three cycles of methotrexate.
  • FIG. 5 shows that average anti-rabbit IgG titers are reduced after mATG re-challenge in methotrexate-treated mice (single and multiple methotrexate cycles), as compared to mice treated with mATG but not methotrexate. Arrows indicate time points at which mATG or methotrexate were administered.
  • FIG. 6 shows that the average anti-rabbit IgG titer in mice treated with mATG and a single cycle of methotrexate is 100 fold less than that in mice treated with mATG alone. Arrows indicate time points at which mATG or methotrexate were administered.
  • FIGS. 7A-C show that methotrexate can reduce average anti-alemtuzumab IgG titers in huCD52 Tg mice.
  • FIG. 7A Anti-alemtuzumab titers are lower in mice treated with a single cycle of 1 mg/kg or 5 mg/kg of methotrexate than in mice treated with 0.5 mg/kg or no methotrexate. Mice were treated with alemtuzumab alone, or alemtuzumab and a single cycle of methotrexate at 0.5 mg/kg, 1 mg/kg, or 5 mg/kg.
  • FIG. 7B Study design to test anti-alemtuzumab titers.
  • FIG. 7C Methotrexate reduced anti-alemtuzumab antibody titers in mice receiving alemtuzumab and methotrexate, compared to mice receiving only alemtuzumab, 24 hours after the fifth dose of alemtuzumab.
  • FIGS. 8A-D show the absolute cell number/ ⁇ l of whole blood of circulating T cells in mice treated with five daily doses of 0.5 mg/kg alemtuzumab or phosphate-buffered saline (PBS).
  • FIG. 8A shows that total T cells (CD3 + ) were reduced two, three, and four weeks post-treatment.
  • FIG. 8B shows that total B cells (CD19 + ) were reduced three weeks post-treatment.
  • FIG. 8C shows that T helper cells (CD4 + ) were reduced at two, three, and four weeks post-treatment
  • FIG. 8D shows that cytotoxic T cells (CD8 + ) were reduced at two, three, and four weeks post-treatment.
  • FIGS. 9A-B show alemtuzumab-specific IgG responses.
  • FIG. 9A shows responses following three cycles of treatment with alemtuzumab at 0.5 mg/kg, and one cycle of treatment with methotrexate at 0.5 mg/kg, 1 mg/kg, or 2 mg/kg.
  • the first cycle of alemtuzumab treatment consisted of five consecutive days of dosing, and the second and third cycles each consisted of three consecutive days of dosing.
  • FIG. 9B shows anti-alemtuzumab IgG titers of each animal per group at week 14.
  • FIG. 10 shows that a single cycle of 5 mg/kg of methotrexate restores circulating mATG levels in mice treated with five monthly doses of mATG.
  • FIG. 11 shows that mice treated with five monthly treatments of mATG and a single cycle of methotrexate have enhanced mATG-mediated CD4 + and CD8 + T cell depletion in blood after the fifth dose of mATG.
  • FIGS. 12A-B show that mice treated with a single cycle of methotrexate exhibit increased percentages of T regulatory (CD25 + Foxp3 + ) cells following a fifth monthly mATG treatment. Mice were treated with five monthly treatments of mATG alone, or five monthly treatment of mATG and a single cycle of methotrexate, or five monthly treatments of mATG and three cycles of methotrexate.
  • FIG. 12A shows T regulatory cell levels in the spleen.
  • FIG. 12B shows T regulatory cell levels in the blood.
  • FIG. 13A shows that an anti-rabbit IgG titer greater than 10,000 can interfere with pharmacokinetics of mATG.
  • FIG. 13B shows that an anti-rabbit titer greater than 100,000 can interfere with CD4 and CD8 cell depletion.
  • % of pretreatment control refers to the percent of CD4 and CD8 titers relative to their respective levels prior to mATG treatment.
  • FIG. 14 shows that mice treated with 5 mg/kg of methotrexate exhibit enhanced alemtuzumab depletion of circulating CD3 + T cells and CD19 + B cells after a fifth monthly dose of alemtuzumab.
  • Mice were treated with alemtuzumab alone, or alemtuzumab and a single cycle of methotrexate for the first three days of the study.
  • Asterisks indicate measurements with statistically significant differences (*, p ⁇ 0.05; ***, p ⁇ 0.0001).
  • FIG. 15A shows that a single cycle of methotrexate can reduce recombinant human acid alpha-glucosidase (rhGAA)-specific IgG titers throughout treatment, and even four weeks after the last rhGAA treatment, in a 16-week study.
  • Arrows indicate time points at which rhGAA and methotrexate were administered. Mice were treated with rhGAA alone, or rhGAA and a single cycle of methotrexate, or rhGAA and three cycles of methotrexate.
  • FIG. 15B shows that, in a six-week study, a single cycle of methotrexate reduces rhGAA-specific IgG titers.
  • FIG. 15C shows that a single cycle of methotrexate reduces rhGAA-specific IgG titers in an 18 week study.
  • FIG. 16A shows that methotrexate decreases anti-rabbit IgG titers in a murine allogeneic heart transplant model when administered with mATG, as compared to mATG alone.
  • FIG. 16B shows that methotrexate increases circulating levels of mATG in a murine allogeneic heart transplant model. Mice were treated with saline, mATG alone, or mATG and a single cycle of methotrexate. mATG was administered at 20 mg/kg at days 0 and 4 of the study, while 2 mg/kg of methotrexate was administered on days 0-6 of the study.
  • FIG. 17 shows a Kaplan-Meier plot indicating that a combined treatment of mATG and methotrexate prolongs the survival of allogeneic hearts transplanted into recipient mice.
  • Mice were left untreated, or were treated with 20 mg/kg of mATG alone at days 0 and 4 of the study, or with 2 mg/kg of methotrexate alone on days 0-6 of the study, or with both mATG and 2 mg/kg of methotrexate on days 0-6, or with both mATG and 0.5 mg/kg of methotrexate on days 0-6 or days 0-11.
  • FIG. 18 shows that mice with an allogeneic heart transplanted treated with either methotrexate alone or methotrexate in combination with mATG experience a reduction in anti-allograft antibody responses.
  • FIG. 18A shows recipient IgG binding to allogeneic fibroblasts on day 21.
  • FIG. 18B shows alloantibody levels on day 21 in mice given syngeneic transplants (Syn Tx) or an allogeneic transplant (Allo Tx) with the indicated treatments. Shown is the binding of individual recipient mouse serum IgG to allogeneic fibroblasts and is expressed as a ratio of the mean fluorescence intensity (MFI) to unstained fibroblasts.
  • MFI mean fluorescence intensity
  • FIG. 19 shows that B10 regulatory B cells are significantly increased following methotrexate treatment. Mice were treated with rhGAA alone or rhGAA and a single three-day cycle of methotrexate or saline. Cell numbers were counted on day 7 and day 8 of the study.
  • FIG. 20 shows that activated B cell subpopulations are significantly increased on day 6 following treatment with rhGAA and a single three-day cycle of methotrexate, as compared to treatment with rhGAA alone.
  • Absolute cell numbers of CD86+ transitional 2 B cells, CD86+ transitional 3 B cells, CD86+ follicular B cells, and CD86+ marginal zone B cells were counted on day 6 of the study.
  • Asterisks indicate measurements with statistically significant differences (*, p ⁇ 0.05; **, p ⁇ 0.001).
  • FIG. 21 shows that for activated splenic B cell subpopulations, such as activated marginal zone B cells, activated follicular B cells, and activated transitional 3 B cells, cell numbers remain enhanced even following treatment with rhGAA and methotrexate, as compared to treatment with rhGAA alone.
  • Arrows represent the treatment with rhGAA and methotrexate.
  • rhGAA was administered on day 1 and day 8.
  • Methotrexate was administered on days 1, 2, and 3, and days 8, 9, and 10.
  • Significant differences are represented by stars (*, p ⁇ 0.05). Data not shown for days 8 and 9 (indicated by vertical dotted lines).
  • FIG. 22 shows that activated splenic T cell populations, such as activated T helper cells, activated T cytotoxic cells, and activated T regulatory cells, remain largely unchanged following treatment with rhGAA and methotrexate, as compared to treatment with rhGAA alone.
  • Arrows represent the treatment with rhGAA and methotrexate.
  • rhGAA was administered on day 1 and day 8.
  • Methotrexate was administered on days 1, 2, and 3, and days 8, 9, and 10. Significant differences are represented by stars (*, p ⁇ 0.05). Data not shown for days 8 and 9 (indicated by vertical dotted lines).
  • FIG. 23 shows a six month-long study design for examining methotrexate treatment in combination with mATG.
  • Solid arrows represent 5 mg/kg mATG injections
  • dashed arrows represent 5 mg/kg methotrexate injections
  • dotted arrows represent terminal sacrifices.
  • FIGS. 24A-B show that a single cycle of methotrexate in connection with mATG enriches splenic B cells, as compared to mATG alone or three cycles of methotrexate.
  • FIG. 24A activated follicular B cells;
  • FIG. 24B activated transitional 3 B cells.
  • FIGS. 25A-B show the effects of methotrexate on alemtuzumab's pharmacodynamics in the blood. Mice were treated with 0.5 mg/kg of alemtuzumab for five months, either with or without three daily doses of 5 mg/kg/day of methotrexate in connection with the first administration of alemtuzumab.
  • FIG. 25A Methotrexate enhances depletion of total T cells (CD3 + ), T helper cells (CD4 + ), and T regulatory cells (CD4 + CD25 + Foxp3 + ) by alemtuzumab (AZM).
  • FIG. 25B Methotrexate enhances depletion of B cells by alemtuzumab. Stars indicate measurements with statistically significant differences (*, p ⁇ 0.05).
  • FIG. 26 shows the effects of methotrexate on alemtuzumab's pharmacodynamics in the spleen.
  • Mice were treated with 0.5 mg/kg of alemtuzumab for five months, either with or without three daily doses of 5 mg/kg/day of methotrexate in connection with the first administration of alemtuzumab.
  • T cells are depleted following the fifth treatment with alemtuzumab.
  • CD8 CEN CD8 + central memory T cells
  • CD8 EFF MEM CD8 + effector memory T cells
  • CD4 CEN MEM CD4 + central memory T cells
  • CD4 EFF MEM CD4 + effector memory T cells.
  • Stars indicate measurements with statistically significant differences (*, p ⁇ 0.05).
  • FIGS. 27A-B show the effects of methotrexate on B cell numbers in the spleen. Mice were treated with 0.5 mg/kg of alemtuzumab for five months, either with or without three daily doses of 5 mg/kg/day of methotrexate in connection with the first administration of alemtuzumab. Stars indicate measurements with statistically significant differences (*, p ⁇ 0.05).
  • FIGS. 28A-B show depletion of splenic lymphocytes three days after a single dose of alemtuzumab.
  • FIG. 28A T cells and B cells are significantly depleted. Small checks represent PBS-treated control mice, and large checks represent alemtuzumab-treated mice.
  • FIG. 28B B cells (CD 19 + ) are 92% depleted at 24 hours after treatment, and remain 36% depleted three days after treatment.
  • the three graphs represent different group of animals, each of which was bled at different time points. Stars indicate measurements with statistically significant differences (*, p ⁇ 0.05; **, p ⁇ 0.001; ***, p ⁇ 0.0001).
  • FIGS. 29A-B show the effects of methotrexate on cytokine levels.
  • FIG. 29A shows a study design for a six month study to assess cytokine levels in the spleen and lymph nodes. The star indicates that data were collected 24 hours after the fifth treatment with alemtuzumab. Arrows indicate time points at which alemtuzumab or methotrexate were administered, or terminal sacrifices were performed, as indicated.
  • FIG. 29B shows levels of B cell activating factor belonging to the TNF family (BAFF) in mice treated with alemtuzumab alone or with alemtuzumab and methotrexate.
  • BAFF B cell activating factor belonging to the TNF family
  • FIGS. 30A-B show the levels of cytokines after treatment with alemtuzumab and methotrexate.
  • FIG. 30A shows a study design for a four month study. The star indicates that data were collected one week after the second cycle of methotrexate. Arrows indicate time points at which alemtuzumab or methotrexate were administered, or terminal sacrifices were performed, as indicated.
  • FIG. 30B shows the levels of various cytokines in mice treated with alemtuzumab (AZM) alone or with 0.5 mg/kg, 1.0 mg/kg, or 2.0 mg/kg of methotrexate.
  • ZAM alemtuzumab
  • FIG. 31 shows the effects of methotrexate on anti-rhGAA titers in IL10 ⁇ / ⁇ (knockout) and C57BL/6 mice. 20 mg/kg of rhGAA was administered weekly for nine weeks in IL10 ⁇ / ⁇ knockout mice and C57BL/6 wild-type mice, with or without 5 mg/kg/day of methotrexate at 0, 24, and 48 hours after the first three weekly treatments of rhGAA.
  • FIG. 32 shows that some, but not all, splenic B cell populations are depleted at one and/or two weeks following treatment with alemtuzumab.
  • Small hatched bars represent phosphate buffered saline (PBS) treated huCD52 transgenic control mice; large hatched bars depict huCD52 transgenic mice treated with 0.5 mg/kg of alemtuzumab for five consecutive days.
  • PBS phosphate buffered saline
  • Asterisks indicate measurements with statistically significant differences (*, p ⁇ 0.05; **, p ⁇ 0.001; ***, p ⁇ 0.0001).
  • FIG. 33 shows a study design for examining the effects of methotrexate on B cell populations in the context of treatment with alemtuzumab.
  • Thyroid and lymph nodes (LN) were used for pathological evaluation. Arrows indicate time points at which alemtuzumab and methotrexate were administered, or terminal sacrifices as indicated.
  • FIG. 34 shows the effects of three daily doses of 5 mg/kg/day methotrexate alone, 0.5 mg/kg alemtuzumab alone, and 0.5 mg/kg alemtuzumab and 5 mg/kg/day methotrexate in combination on the depletion of B cell populations.
  • Asterisks indicate measurements with statistically significant differences (*, p ⁇ 0.05; **, p ⁇ 0.001; ***, p ⁇ 0.0001); ns, not significant).
  • FIG. 35 shows the effects of methotrexate on B cell depletion after five cycles of treatment with 0.5 mg/kg alemtuzumab alone or three daily doses of 5 mg/kg/day methotrexate alone or 0.5 mg/kg alemtuzumab and 5 mg/kg/day methotrexate in combination.
  • Asterisks indicate measurements with statistically significant differences (*, p ⁇ 0.05).
  • FIG. 36 shows that levels of the cytokines MCP-1, IL-13, IL-6, and IL-12 are decreased in mice treated with a single cycle of three days of 5 mg/kg of methotrexate administered on the first day of alemtuzumab treatment. Data were collected 24 hours after a fifth dose of alemtuzumab, four months after treatment with methotrexate.
  • FIGS. 37A-B show rhGAA titers at weeks 2, 6, and 12 in nu/nu nude mice.
  • FIG. 37A shows rhGAA titers at weeks 2 and 6. From left to right, week 2 measurements in control mice treated with saline, mice treated with rhGAA, and mice treated with rhGAA and methotrexate, followed by week 6 measurements in control mice treated with saline, mice treated with rhGAA, and mice treated with rhGAA and methotrexate.
  • FIG. 37A shows rhGAA titers at weeks 2, 6, and 12 in nu/nu nude mice.
  • FIG. 37A shows rhGAA titers at weeks 2 and 6. From left to right, week 2 measurements in control mice treated with saline, mice treated with rhGAA, and mice treated with rhGAA and methotrexate, followed by week 6 measurements in control mice treated with saline, mice treated with rhGAA
  • 37B shows week 12 measurements in, from left to right, nu/nu mice treated with rhGAA alone (Myo), nu/nu mice treated with methotrexate and rhGAA, BL6 mice treated with rhGAA alone, and BL6 treated with methotrexate and rhGAA.
  • FIGS. 38A-B show that the numbers and percentages of IL-10 expressing B10 B cells is increased in mice tolerized to rhGAA with methotrexate.
  • B 10 B cells isolated from animals treated with rhGAA or rhGAA and methotrexate were assessed for IL-10 protein expression by flow cytometry.
  • FIGS. 39A-B show that IL-10 is expressed in both activated (CD86+) and non-activated (CD86 ⁇ ) B10 B cells.
  • FIG. 39A depicts a FACS plot of B10 B cells stained with CD86
  • FIG. 39B depicts the numbers of CD86 + IL10 + B10 B cells and CD86 ⁇ IL10 + B10 B cells in response to treatment with rhGAA or rhGAA and methotrexate.
  • FIGS. 40A-B show that methotrexate treatment with rhGAA induces B10 B cells to increase their expression of TGF-beta.
  • the second and third panels of FIG. 40A are FACS plots showing B10 B cells stained with TGF-beta and CD86 from animals treated with rhGAA or rhGAA and methotrexate.
  • the first panel of FIG. 40A depicts the number of TGF-beta + B 10 B cells in animals treated with rhGAA or rhGAA and methotrexate.
  • FIG. 40B depicts CD86 + TGF-beta + B10 B cell and CD86 ⁇ TGF-beta + B10 B cell counts.
  • FIG. 41A depicts that B10 B cells appear to express FoxP3 in animals treated with rhGAA ( FIG. 41A ).
  • FIG. 41B depicts that the numbers of FoxP3+ B cells increase with treatment with both methotrexate and rhGAA.
  • FIG. 41C depicts that both activated (CD86+) and non-activated (CD86 ⁇ ) B10 B cells express FoxP3.
  • FIGS. 42A-C show that follicular, transitional 2, and transitional 3 B cells (top to bottom) express IL-10 and that the cell numbers of the IL-10-expressing B cell subsets increase with methotrexate as compared to mice treated with rhGAA alone.
  • FIGS. 43A-C show that follicular, transitional 2, and transitional 3 B cells (top to bottom) express TGF-beta and that the cell numbers of the TGF-beta-expressing B cell subsets increase with methotrexate as compared to mice treated with rhGAA alone.
  • FIGS. 44A-C show that follicular, transitional 2, and transitional 3 B cells (top to bottom) express FoxP3 and that the cell numbers of the FoxP3-expressing B cell subsets increase with methotrexate as compared to mice treated with rhGAA alone.
  • FIG. 45 shows anti-rhGAA titers at week 6 in animals treated with rhGAA or rhGAA and methotrexate, in the presence or absence of 5 mg/kg of anti-TGF-beta antibody (1D11, Genzyme) or the isotype control (13C4).
  • Antibody titers were assessed bi-weekly in the four different groups of animals.
  • FIGS. 46A-C show that 1D11 treatment interfered with methotrexate-induced expansion of B10 B cells expressing TGF-beta, IL-10, or FoxP3.
  • Spleens were isolated from animals treated with rhGAA or rhGAA and methotrexate that also were co-administered 1D11 or 13C4 seven days following a single rhGAA treatment or a single rhGAA and methotrexate treatment. Cells in each group were then pooled and cultured for two days and then counted using flow cytometry.
  • FIGS. 47A-C show that 1D11 treatment interfered with methotrexate-induced expansion of follicular B cells expressing TGF-beta or IL-10, although FoxP3+ Follicular B cells did not appear to experience 1D11 effects.
  • Spleens were isolated from animals treated with rhGAA or rhGAA and methotrexate that also were co-administered 1D11 or 13C4 seven days following a single rhGAA treatment or a single rhGAA and methotrexate treatment. Cells in each group were then pooled and cultured for two days and then counted using flow cytometry.
  • FIGS. 48A-C show that, in transitional 2 B cells, while 1D11 treatment interfered with methotrexate-induced expansion of TGF-beta-expressing transitional 2 B cells, no effects were seen on IL-10+ transitional 2 B cells.
  • Spleens were isolated from animals treated with rhGAA or rhGAA and methotrexate that also were co-administered 1D11 or 13C4 seven days following a single rhGAA treatment or a single rhGAA and methotrexate treatment. Cells in each group were then pooled and cultured for two days and then counted using flow cytometry.
  • FIGS. 49A-C show that in transitional 3 B cells there is detectable TGF-beta, IL-10 and FoxP3, but no apparent effect of 1D11 treatment on the cells.
  • Spleens were isolated from animals treated with rhGAA or rhGAA and methotrexate that also were co-administered 1D11 or 13C4 seven days following a single rhGAA treatment or a single rhGAA and methotrexate treatment, and cells were counted using flow cytometry.
  • FIGS. 50A-C show that 1D11 treatment does not affect basal levels of IL-10, TGF-beta, and FoxP3 in follicular B cells, transitional 2 B cells, and transitional 3 B cells (top to bottom).
  • FIG. 51A is a schematic showing the transfer of total splenic B cells from a mouse tolerized to rhGAA (Myozyme® or “MYO”) into an rhGAA-na ⁇ ve recipient mouse. After transfer, the recipients (along with non-transferred control animals treated with either rhGAA or rhGAA and methotrexate) were treated weekly with 20 mg/kg of rhGAA.
  • Myozyme® or “MYO” Myozyme® or “MYO”
  • FIG. 51B shows titer analysis demonstrating that total splenic B cells isolated from animals treated with rhGAA and single cycle of methotrexate can transfer immune tolerance to rhGAA in na ⁇ ve hosts.
  • FIG. 52 depicts cell counts from the blood or spleen of normal mice treated with rabbit IgG (rbIgG), mATG alone, rbIgG and methotrexate, or mATG and methotrexate. Methotrexate does not deplete CD4+, CD8+, T regulatory (CD4+CD25+FoxP3+) T cells, or total CD 19+ B cells in normal animals.
  • FIG. 53 depicts cell counts from the blood or spleen of transplant mice treated with rbIgG, mATG alone, rbIgG and methotrexate, or mATG and methotrexate 14 days after transplantation. Methotrexate does not deplete CD4+, CD8+, T regulatory (CD4+CD25+FoxP3+) T cells, and total CD19+ B cells in transplant animals.
  • FIG. 54 shows that methotrexate treatment induces statistically significant increases in IL-10 in multiple cell subsets as viewed by a shift in mean fluorescence intensity (MFI) of these proteins in animals treated with rhGAA or rhGAA and methotrexate.
  • MFI mean fluorescence intensity
  • FIG. 55 shows that methotrexate treatment induces statistically significant increases in TGF-beta in multiple cell subsets as viewed by a shift in mean fluorescence intensity (MFI) of these proteins in animals treated with rhGAA or rhGAA and methotrexate.
  • MFI mean fluorescence intensity
  • FIG. 56 shows that methotrexate treatment induces statistically significant increases in FoxP3 in multiple cell subsets as viewed by a shift in mean fluorescence intensity (MFI) of these proteins in animals treated with rhGAA or rhGAA and methotrexate.
  • MFI mean fluorescence intensity
  • the present invention is based on our surprising discovery that a single cycle or short course of methotrexate administration reduces undesired immune responses (such as ADA responses, and other undesired T- and/or B-cell mediated immune responses) in patients receiving protein therapeutics such as replacement enzymes or therapeutic antibodies, and anti-graft antibody responses in tissue transplantation.
  • This discovery leads to new methods for increasing both safety and efficacy of protein therapies and organ transplantation.
  • Thymoglobulin® is a rabbit anti-human thymocyte globulin polyclonal antibody used for immunosuppression in the setting of solid organ transplantation, aplastic anemia and in prevention of graft-versus-host disease.
  • mATG rabbit anti-murine thymocyte globulin polyclonal antibody
  • methotrexate is also useful in organ transplantation.
  • the survival of a heart allograft was significantly longer.
  • Methotrexate is classically known as a dihydrofolate reductase antagonist that is thought to kill proliferating cells by inhibiting purine metabolism and interfering with de novo DNA synthesis (Kremer, Arthritis Rheum, 50(5):1370-82 (2004)). It could be easily assumed that methotrexate may simply kill the reactive cells through this well-described mechanism, but this seems unlikely with the single cycle regimen that we have discovered. Methotrexate has a short half-life and is not likely to be in cells or circulation long enough to actively kill cells three to four months following treatment (Walling, Invest New Drugs, 24(1):37-77 (2006); Slavikova et al., Neoplasma, 25(2):211-6 (1978)).
  • methotrexate reduces undesired antibody responses by inducing an active mechanism of immune control, not by indiscriminately depleting lymphocytes.
  • methotrexate is working in a unique, previously unknown way.
  • Low, continual doses of methotrexate in infliximab-treated rheumatoid arthritis patients have been shown to reduce anti-infliximab antibody responses; yet as exposure is continuous, this regimen is more likely to involve constant immunosuppression, rather than tolerance induction.
  • Methotrexate treatment alone has been shown to reduce disease activity in rheumatoid arthritis when given weekly in low doses.
  • methotrexate upon continual administration of low dose methotrexate (every other day), autoantigen-specific T regulatory cells appear, which may help account for the efficacy of methotrexate treatment in rheumatoid arthritis (Xinqiang et al., Biomed Pharmacother, 64(7):463-471 (2010)).
  • methotrexate is truly unique in that it involves a short course of methotrexate treatment that can provide long-lasting control of undesired immunological responses.
  • methotrexate acts through a unique mechanism of action that involves the expansion of activated B cell subsets that may represent regulatory B cells active in suppressing immune responses.
  • methotrexate also may act through a mechanism of T regulatory cell expansion.
  • the methods of this invention can control undesired immunological responses (e.g., ADA responses, and other undesired T- and/or B-cell mediated immune responses) in a variety of biological therapies (e.g., therapy using a biologic such as proteins, nucleic acids, carbohydrates, lipids, and metabolites).
  • biological therapies e.g., therapy using a biologic such as proteins, nucleic acids, carbohydrates, lipids, and metabolites.
  • Protein therapy refers to therapy in which the therapeutic agent is a proteinaceous substance, including peptides and proteins.
  • Protein therapeutics can, for example, be enzymes, cytokines, growth factors, immunomodulators, thrombolytics, antibodies (including polyclonal and monoclonal antibodies), antibody fragments or modified antibodies (e.g., Fab, F(ab′) 2 , Fv, Fd, scFv, and dAb).
  • antibodies including polyclonal and monoclonal antibodies
  • antibody fragments or modified antibodies e.g., Fab, F(ab′) 2 , Fv, Fd, scFv, and dAb.
  • Fabrazyme® recombinant human alpha-galactosidase
  • Cerezyme® imiglucerase
  • Aldurazyme® laronidase
  • MFS I Mucopolysaccharidosis I
  • Myozyme® and Lumizyme® alglucosidase alpha
  • antibody therapeutics include Campath® (alemtuzumab), Thymoglobulin®, Avastin® (bevacizumab), Lucentis® (ranibizumab), Remicade® (infliximab), Humira® (adalimumab), Rituxan® (rituximab), Tysabri® (natalizumab), Simulect® (basiliximab), Zenapax® (daclizumab), OKT30 (muromonab-CD3), Erbitux® (Cetuximab), Mylotarg® (gemtuzumab), Herceptin® (trastuzumab), and Benlysta® (belimumab).
  • protein therapeutics include Enbrel® (etanercep), and other fusion proteins.
  • ADA undesired immune responses can be generated in a patient against the protein therapeutic, causing variable effects on patient outcome.
  • Such responses occur because biologic therapeutics often contain sequences and conformations that are foreign to a human patient.
  • ADA interferes with therapeutic efficacy and/or increases safety risks.
  • ADA may cause hypersensitivy reactions, anaphylaxis, serum sickness, immune complex disease, acute renal failure.
  • ADA can be monitored in patients receiving protein therapy by a clinician using well established methods, including ELISA and immunohistochemistry.
  • a “protein therapy” as used herein refers to a viral therapy where a viral vector is used to deliver a nucleic acid therapeutic.
  • viruses used in such therapies include, but are not limited to, adenoviruses, adeno-associated viruses, and retroviruses.
  • Antibodies may develop against the capsid proteins of the virus, reducing the efficacy and increasing the safety risks of such therapies.
  • the methods of this invention are useful to control undesired immunological responses (e.g., ADA responses, and other undesired T- and/or B-cell mediated immune responses) in viral therapies as well.
  • the methods of this invention also may control undesired immunological responses in non-protein biological therapies.
  • exemplary non-protein bio-therapies include, but are not limited to, nucleic acid therapies, (e.g., antisense therapies, siRNA therapies, and miRNA therapies).
  • the methods of this invention can also be used to induce immune tolerance in a patient receiving tissue transplantation such as renal transplantation, liver transplantation, cardiac transplantation, and stem cell transplantation. Host versus graft and graft versus host rejections often occur in tissue transplantation, especially allograft and xenograft transplantation.
  • a single cycle of methotrexate can be used alone or together with another immune-modulating agent, (e.g., an immunosuppressant such as Thymoglobulin®) to manage anti-graft antibody response.
  • an immunosuppressant such as Thymoglobulin®
  • the combination of Thymoglobulin® and methotrexate in transplantation may act to prolong graft survival.
  • methotrexate may allow for safer re-treatment of Thymoglobulin®, protecting the patient from developing significant anti-rabbit antibodies (such as IgG and/or IgM) and/or infusion-related reactions.
  • a single, short cycle of methotrexate can significantly reduce undesired immunological responses such as ADA in subjects receiving biologic therapeutics (e.g., protein therapeutics) and anti-allograft responses in patients receiving tissue transplantation.
  • ADA immunological responses
  • biologic therapeutics e.g., protein therapeutics
  • anti-allograft responses in patients receiving tissue transplantation.
  • Reducing undesired ADA may not only improve patient safety, but also may improve the efficacy of a protein therapeutic through improving the protein therapeutic's pharmacodynamics and/or pharmacokinetics.
  • Methotrexate a small molecule compound, has been used to treat patients with severe active rheumatoid arthritis, severe psoriasis, and certain types of cancer including cancers that begin in the tissues that form around a fertilized egg in the uterus, breast cancer, lung cancer, certain cancers of the head and neck, certain types of lymphoma, and leukemia (cancer that begins in the white blood cells).
  • Methotrexate treats cancer by slowing the growth of cancer cells.
  • Methotrexate treats psoriasis by slowing the growth of skin cells to stop scales from forming Methotrexate may treat rheumatoid arthritis by decreasing the activity of the immune system.
  • Methotrexate has been studied in the context of controlling ADA responses elicited against ⁇ -galactosidase A and ⁇ -glucosidase. However, those studies were done with multiple cycles of methotrexate treatment (Garman et al. Clin Exp Immunol, 137(3):496-502 (2004); Joseph et al., Clin Exp Immunol, 152(1):138-46 (2008); Mendelsohn et al., N Engl J Med, 360(2):194-5 (2009)), rather than a single cycle of methotrexate.
  • methotrexate can reduce ADA in antibody therapy.
  • a single cycle regimen refers to a treatment regimen, or a treatment unit, of consecutive or non-consecutive days and are started at preferably no more than five (e.g., no more than three) days following the dosing of the primary protein therapeutic or transplantation.
  • a single cycle of treatment of methotrexate preferably does not extend past the first period of protein therapeutic dosing.
  • a single cycle of methotrexate consists of three consecutive days of methotrexate intake (e.g., orally), starting on day 0, the day when the primary protein therapeutic is given to the patient for the first time, or when the patient receives a tissue transplant.
  • a single cycle of methotrexate may also consist of, for example, 2, 3, 4, 5, 6, 7, or 8 consecutive daily doses of methotrexate, starting on day 0.
  • Methotrexate also can be administered at other times as deemed appropriate, e.g., when managing secondary autoimmunity in e.g., a lymphocyte-depleting therapy.
  • a single cycle of methotrexate preferably does not last longer than 8 days.
  • a single cycle of methotrexate begins between 48 hours prior to and 48 hours after the onset of the primary therapeutic treatment (i.e., the treatment with the biologic therapeutic).
  • a single cycle of methotrexate may begin 48 hours prior to, 36 hours prior to, 24 hours prior to, 12 hours prior to, concurrently with, 12 hours after, 24 hours after, 36 hours after, or 48 hours after, the onset of the primary therapeutic treatment.
  • methotrexate may be administered in more than one cycle, but at a low total dosage.
  • the methotrexate can be administered in two or more (e.g., 3, 4, 5, 6, etc.) cycles, but with a total combined dosage of no more than 5 mg/kg in a patient.
  • the dosage of methotrexate will be an effective amount of methotrexate in reducing undesired immunological responses, such as antibody or cellular responses, when given in a single cycle.
  • An effective amount of methotrexate in human patients may be in the range of 0.05 mg/kg to 5 mg/kg. In some embodiments, the effective amount is 0.1 mg/kg to 1.5 mg/kg. In some embodiments, the effective amount is 0.12 mg/kg to 1.28 mg/kg. In certain embodiments, the effective amount is 0.12 mg/kg. In certain embodiments, the effective amount is 1.28 mg/kg.
  • methotrexate may pose minimal safety risks because the dosing regimen involves only a brief course of methotrexate at dose levels that are more similar to doses for rheumatoid arthritis than low neoplastic doses.
  • the total amount of methotrexate tested in each cycle in mice was 14 or 15 mg/kg. 14 mg/kg of methotrexate in mice is equivalent to approximately 68 mg or 5.92 mg/m 2 in an average adult weighing 60 kg. Rheumatoid arthritis patients can receive up to 25 mg of methotrexate per week without suffering from significant toxicities.
  • the low neoplastic dose of methotrexate is considered to be 30 mg/m 2 , significantly higher than 5.92 mg/m 2 .
  • methotrexate regimen is likely to be well-tolerated in adults.
  • the exact dosage and regimen of methotrexate should of course be established by a clinician, taking into account the patient's physical condition, age, weight, gender, other medications he/she is taking and their known side-effects, and any other relevant factors.
  • methotrexate on managing undesired antibody responses in the patient can be monitored by well known methods, including clinical examination of the patient, symptoms, blood tests assaying ADA or anti-allograft antibody titers, immunohistochemical assays (e.g., C4 deposition assays and other solid-phase antibody detection methods such as the enzyme-linked immnuosorbent assay (ELISA) and bead-based flurometric assays).
  • immunohistochemical assays e.g., C4 deposition assays and other solid-phase antibody detection methods such as the enzyme-linked immnuosorbent assay (ELISA) and bead-based flurometric assays.
  • ELISA enzyme-linked immnuosorbent assay
  • bead-based flurometric assays bead-based flurometric assays
  • the effect also can be monitored by measuring levels of biomarkers such as MCP-1, IL-13, IL-6, and IL-12, which we have shown to be reduced in level by methotrexate treatment, and transitional 2 B cells, transitional 3 B cells, follicular B cells, marginal zone B cells, B10 B cells, and B1 B cells, which we have shown to be increased in number by methotrexate treatment.
  • biomarkers such as MCP-1, IL-13, IL-6, and IL-12, which we have shown to be reduced in level by methotrexate treatment
  • transitional 2 B cells, transitional 3 B cells, follicular B cells, marginal zone B cells, B10 B cells, and B1 B cells which we have shown to be increased in number by methotrexate treatment.
  • TGF-beta, FoxP3, IL-5, IL-10, IL-15, and GM-CSF may be used as biomarkers to monitor the effects of methotrexate on undesired immune responses as needed.
  • the levels of biomarkers also may be used to monitor the effects of methotrexate on T cell responsiveness to a therapeutic (e.g., a protein therapeutic).
  • a therapeutic e.g., a protein therapeutic.
  • Biomarkers for T cell activation such as IL-2, interferon- ⁇ , and TNF- ⁇ , may also be monitored as readouts for methotrexate's effect on T cell responses.
  • the single cycle methotrexate regimen of this invention can expand the use of many protein therapeutics whose repeated uses in a given patient have been limited in the past due to safety and efficacy concerns.
  • the concomitant use of methotrexate with Thymoglobulin® may expand the utility of Thymoglobulin® to other disease settings where re-dosing is desired, such as T cell-mediated autoimmune diseases including, but not limited to, diabetes, lupus, scleroderma, rheumatoid arthritis, psoriasis and multiple sclerosis.
  • methotrexate may expand upon the efficacy and safety of alemtuzumab, for example, in autoimmune diseases such as multiple sclerosis, wherein alemtuzumab is usually administered in repeated annual cycles, or in chronic B-cell lymphocytic leukemia, wherein alemtuzumab is administered in a 12-week cycle, dosing starting at 3 mg/day (until the infusion reactions are equal to or less than grade 2), then scaling up to 10 mg/day (until the infusion reactions are equal to or less than grade 2), and finally moving up to 30 mg/day (on alternate days, 3 times weekly).
  • autoimmune diseases such as multiple sclerosis
  • alemtuzumab is usually administered in repeated annual cycles
  • chronic B-cell lymphocytic leukemia wherein alemtuzumab is administered in a 12-week cycle
  • dosing starting at 3 mg/day (until the infusion reactions are equal to or less than grade 2), then scaling up to 10 mg/day (until
  • lymphocyte depletion is a type of immunosuppression by reduction of circulating lymphocytes, e.g., T cells and/or B cells, resulting in lymphopenia.
  • lymphocyte depletion can be achieved by a protein therapeutic such as Thymoglobulin®, humanized anti-CD52 monoclonal antibody CAMPATH-1H® (alemtuzumab), and rituximab.
  • Lymphocyte depletion is desired in treatment of a number of autoimmune conditions, including multiple sclerosis (Coles et al., Ann. Neurol. 46, 296-304 (1999); Coles et al., 2008), rheumatoid arthritis, vasculitis, and lupus.
  • Lymphocyte depletion therapy may cause secondary autoimmunity.
  • Autoimmunity is referred to herein as “secondary autoimmunity” when it arises subsequent to the onset of a first (“primary”) disease, for example, a “primary” autoimmune disease.
  • Secondary autoimmunity sometimes arises in MS patients having, or having had, lymphopenia following, e.g., lymphocyte depleting therapy. In some individuals, secondary autoimmunity arises soon after lymphocyte depleting therapy (e.g., treatment with alemtuzumab).
  • lymphocyte depletion may not arise until months or years after lymphocyte depleting therapy; in some of those individuals, by the time they develop secondary immunity, substantial lymphocyte recovery (total lymphocyte count) may have occurred so that they may no longer be lymphopenic. Lymphocyte depletion may occur in the context of treatment with antibody therapeutics or small molecule therapeutics.
  • Secondary autoimmunity arising in lymphopenic MS patients can be any type of autoimmune condition other than MS, including but not limited to thyroid autoimmunity (e.g., Graves' disease), thrombocytopenic purpura, immune thrombocytopenia (ITP), Goodpasture's disease, autoimmune neutropenia, autoimmune hemolytic anemia, and autoimmune lymphopenia.
  • thyroid autoimmunity e.g., Graves' disease
  • thrombocytopenic purpura thrombocytopenic purpura
  • ITP immune thrombocytopenia
  • Goodpasture's disease e.g., autoimmune neutropenia
  • autoimmune hemolytic anemia e.g., autoimmune lymphopenia
  • the secondary autoimmunity is B cell mediated, i.e., B cell responses and auto-antibodies are directly linked with disease development and pathology.
  • autoantibody levels in a patient's body fluid can be determined as a means of detecting signs of autoimmunity.
  • anti-nuclear antibodies, anti-smooth muscle antibodies, and anti-mitochrondrial antibodies can be measured.
  • additional assays can be performed to measure anti-double-stranded DNA antibodies, anti-ribonucleoprotein antibodies, and anti-La antibodies.
  • Anti-thyroid peroxidase (TPO) and anti-thyroid stimulating hormone (TSH) receptor antibodies can be measured to detect autoimmune thyroid diseases; if anti-TPO or anti-TSH receptor antibodies are detected, one can measure whether thyroid function is affected by measuring free T3, free T4 and TSH levels.
  • Anti-platelet antibodies can be measured to detect autoimmune thrombocytopenia, and a measurement of blood platelet levels may serve to determine if the presence of anti-platelet antibodies is causing a reduction in platelet number. See also WO 2010/041149.
  • the single cycle methotrexate regimen of this invention can be used to improve the safety and efficacy of lymphocyte-depleting therapy by reducing ADA as well as minimizing secondary autoimmunity. Without wishing to be bound by theory, we believe that methotrexate may reduce secondary autoimmunity by tolerizing multiple autoantigens simultaneously.
  • mice normal female C57BL/6 mice between 6 and 12 weeks of age were used for the in vivo studies of rabbit anti-murine thymocyte globulin polyclonal antibody (mATG) and were obtained from Jackson Laboratories (Bar Harbor, Me.) or Taconic Laboratories (Hudson, N.Y.).
  • Alemtuzumab-related studies employed human CD52 (huCD52) transgenic (Tg) mice between 6-12 weeks of age that were obtained from Charles River Laboratories/Genzyme Corp. Mice were housed and maintained in accordance with the Guide for Care and Use of Laboratory Animals and under American Association for Accreditation of Laboratory Animals Care I accreditation and all animal protocols used in these studies were approved by the Institutional Animals Care and Use Committee.
  • the huCD52 Tg mouse used for nonclinical pharmacology studies were generated by Xenogen (Cranbury, N.J., USA).
  • a bacmid construct containing approximately 145 kilobases of genomic DNA from human chromosome 1 was randomly integrated into the mouse genome of CD-1 embryonic stem cells.
  • the construct included a total of 5 partial or full genes of unknown function in addition to human CD52.
  • the 5 partial or full gene segments contained in the bacmid were as follows: the human CD52 gene, the 3′ end of a novel gene (DKFZP434L0117), the SH3BGRL3 gene (SH3 domain binding glutamic acid-rich protein like 3), the gene for socius (SOC), the AIM1L (absent in melanoma 1-like) gene, and the zinc finger protein 683 gene.
  • Three founder lines were generated and line 107 was established at Genzyme.
  • Monoclonal antibody alemtuzumab was administered intravenously as either a single injection of 0.5 mg/kg or in either a three or five day cycle of 0.5 mg/kg/day.
  • rhGAA Recombinant human alglucosidase alfa
  • Myozyme® Recombinant human alglucosidase alfa
  • mice were treated weekly with 20 mg/kg of rhGAA by bolus tail vein injection unless stated differently. All mice were treated prophylactically with 5 to 30 mg/kg diphenhydramine (Baxter Healthcare Corporation, Deerfield, Ill.) intraperitoneally prior to rhGAA administration. Control animals were treated intravenously with either sterile 0.9% saline or rhGAA-formulation buffer.
  • Methotrexate (Calbiochem catalog #454125) was administered at 0.5, 1.0, 2.0 or 5 mg/kg by intraperitoneal injection for 1-3 cycles, where each cycle equals either three, six or seven consecutive days of injection depending upon the experiment.
  • methotrexate was administered intraperitoneally at 5 mg/kg at 0, 24, and 48 hours following either the initial mATG treatment or the first three mATG treatments.
  • methotrexate was given daily at 2 mg/kg from days 0 to 6, daily at 0.5 mg/kg from days 0 to 6, or daily at 0.5 mg/kg from days 0 to 11.
  • Polyclonal antibody mATG was administered as an intraperitoneal injection of 5 mg/kg every four weeks or as two 20 mg/kg doses given four days apart when in the transplant setting with the first dose given on the day of transplant (day 0).
  • splenocyte and lymph node cell preparations single-cell suspensions were generated from harvested mouse spleens or inguinal and mesenteric lymph nodes by homogenization between frosted glass slides into PBS containing 2% FCS.
  • red blood cells were lysed by 1-2 minute incubation with a red blood cell lysis solution (BD Biosciences, San Diego, Calif.). Blood was isolated by retro-orbital bleeding and cell preparations were performed by lysing red blood cells with red blood cell lysing solution (BD Biosciences) for 20-30 minutes.
  • live cells were enumerated using the ViCell automated counter (Beckman Coulter, Fullerton, Calif.). Following isolation, all cell preparations were washed with PBS/2% FCS prior to use in the assays described below.
  • CD4 na ⁇ ve cells CD4 + CD25 ⁇ CD62L + CD44 ⁇ ,
  • CD4 memory cells CD4 + CD25 ⁇ CD62L ⁇ CD44 + ,
  • CD8 memory cells CD8 + CD44 + CD62L ⁇ ,
  • B2/follicular B cells CD19 + CD21 hi CD23 lo ,
  • B1 B cells CD19 + CD43 + CD11b + ,
  • transitional 1 B cells CD19 + CD93 + CD23 ⁇ IgM hi ,
  • transitional 2 B cells CD19 + CD93 + CD23 + IgM hi ,
  • transitional 3 B cells CD19 + CD93 + CD23 + ⁇ m lo ,
  • marginal zone B cells CD19 + CD21 hi CD23 lo .
  • B10 B cells CD19 + CD5 + CD1d + .
  • ELISA enzyme linked immunosorbent assay
  • the plates were washed and horseradish peroxidase-conjugated goat anti-mouse IgG secondary antibody (Southern Biotechnology Associates, Birmingham, Ala., USA) was added and allowed to incubate for 1 h at 37° C. Following a final wash, 3,3′,5,5′-tetramethylbenzidine substrate (BioFx, Owings Mills, Md., USA) was added and allowed to develop for 15 min at room temperature. The reaction was stopped by the addition of 1 N HCl and absorbance values were read at 450/650 nm on an ELISA plate reader (Molecular Devices, Sunnyvale, Calif., USA). End-point titers were defined as the lowest dilution that averages above an absorbance of 0.100 using Softmax software (Molecular Devices, Sunnyvale, Calif., USA).
  • Mouse serum was determined by ELISA. Briefly, 96-well plates (Corning Inc., Corning, N.Y., USA) were coated overnight with 1 ⁇ g/ml of goat anti-rabbit IgG-Fc fragment antibody (Bethyl Laboratories, TX, USA). Following blocking with 0.5% BSA (high purity), standard controls and serum samples were diluted as necessary and added in duplicate to the wells of the coated plates and incubated at 36-38° C. for 1 hour with gentle shaking. The plates were washed and horseradish peroxidase-conjugated goat anti-rabbit IgG-Fc fragment antibody (Bethyl Laboratories, TX, USA) was added as appropriate and incubated for 1 hour at 36-38° C.
  • mice were bled 4-6 days following alemtuzumab treatment and specific anti-alemtuzumab IgG was measured by ELISA. Briefly, 96-well plates (Corning Inc., Corning, N.Y., USA) were coated overnight with 3 ⁇ g/ml of alemtuzumab in PBS (pH 7.2). Following blocking with 0.1% BSA in PBS, serial dilutions of serum were added in duplicate to alemtuzumab-coated plates and incubated at 37° C. for 1 h.
  • mice were bled 4-6 days following rhGAA treatment and specific IgG was measured by ELISA. Briefly, 96 well plates (Corning Inc., Corning, N.Y., USA) were coated overnight with 5 ⁇ g/ml of rhGAA in sodium acetate buffer (pH 5.0). Following blocking with 0.1% BSA in PBS, serial dilutions of serum were added in duplicate to rhGAA-coated plates and incubated at 37° C. for 1 hour. The plates were washed, and HRP-conjugated goat anti-mouse IgG secondary antibody (Southern Biotechnology Associates, Birmingham, Ala.) was added and allowed to incubate for 1 hour at 37° C.
  • TMB 3,3′,5,5′-tetramethylbenzidine substrate
  • 1N HCl 1,3′,5,5′-tetramethylbenzidine substrate
  • Endpoint titers were defined as the reciprocal of the sample dilution resulting in an absorbance value of 0.2 using Softmax software (Molecular Devices, Sunnyvale, Calif.).
  • mice 8-12 weeks old were given 5 mg/kg of methotrexate (Calbiochem catalog #454125) by intraperitoneal injection for 1-3 cycles, where 1 cycle equals 3 consecutive injection days.
  • 20 mg/kg of Myozyme® (Genzyme Corporation) was given by tail vein injection once or for 2-6 weekly doses, commencing with the first methotrexate dose Animals were sacrificed weekly or daily after initiation of treatment. Spleen, mesenteric, and inguinal lymph nodes were collected for flow cytometric analysis of T and B cell subsets and sera was collected for ELISA assays.
  • Spleens were processed between glass slides and red blood cells (RBC) was lysed with lysing buffer purchased from BD Biosciences (catalog#555899) according to the manufacturer's instructions. Lymph nodes were processed between glass slides and washed with phosphate buffered saline (PBS) containing 2% fetal calf serum (FCS). Cells were resuspended in 200 ⁇ L of PBS containing 4% fetal bovine serum and 25 ⁇ g/mL of total mouse IgG and blocked for 30 minutes at 4° C.
  • PBS phosphate buffered saline
  • FCS 2% fetal calf serum
  • lymph node cells Approximately 3 million spleens cells and 1 million lymph node cells were stained with different antibody cocktails and analyzed with a high through put sampler (HTS) on a Becton Dickinson CANTOII flow cytometer. At least 100,000 cell events were acquired within the lymphocyte gate.
  • HTS high through put sampler
  • Anti-mouse antibody cocktails consisted of PE-CD21/35 catalog #552957, FITC-catalog #553138, PE-CD138 catalog #553714, PE-CD127 catalog #552543, APC-Cy7-CD19 catalog #557655, FITC-CD43 catalog #553270, PE-Cy7-CD4 catalog #552775, FITC-CD3e catalog #553062, APC-CD11b catalog #553312, PE-Cy7-IgM catalog #552867, APC-Cy7-CD8 catalog #557654, PE-CD273 (PD-L2) catalog #557796, APC-CD138 catalog #558626, PE-Cy7-CD11b catalog #552850, PE-CD93 (early B lineage) catalog #558039, APC-CD69 catalog #560689, Pe-Cy7-CD24 catalog #560536, FITC-CD1d catalog #553845, APC-CD5 catalog #
  • FITC-FoxP3 intracellular staining kit was purchased from eBioscience. Pacific Blue (PB)-CD25 catalog #102022, PB-CD23 catalog #101616 and PB-CD86 catalog #105022 were purchased from BioLegend. Analysis of lymphocyte subsets was performed with FCS express version 3 software provided by De Novo Software. Percentages were generated with the batch processing option and absolute numbers were calculated according to the cell counts obtained. Spleen and lymph node cell counts were obtained with a Beckman Coulter Vi-cell XR cell viability analyzer according to the manufacturer's instructions.
  • mice 8-12 weeks old were given 5 mg/kg of methotrexate (Calbiochem catalog #454125) by intraperitoneal injection for 1 cycle (3 consecutive daily doses) commencing with treatment with 20 mg/kg of rhGAA.
  • methotrexate Calbiochem catalog #454125
  • animals were treated with intraperitoneal injections of 5 mg/kg of either 1D11 or 13C4 (Genzyme Corporation) 3 time per week, every other day, commencing with rhGAA and methotrexate treatment. Animals were sacrificed on day 6 or 7 post rhGAA initiation depending on the mouse strain.
  • Spleens were prepared in single cell suspension and loaded onto the RoboSep (STEMCELL technologies) instrument according to the manufacturer's instructions and subjected to B cell negative selection.
  • Purified B cells were seeded at 500,000 cells per well in 96 well round bottom plates (Costar catalog #3799) and either incubated with no stimulation or with 10 ⁇ g/mL of LPS (Sigma catalog #L5014) for 48 hours at 37° C. All wells received Monensin (BD Bioscience catalog #554724) according to the manufacturer's instructions. Cells were allowed to incubate for at least 4 hours at 37° C. Samples were transferred to V bottom wells (USA Scientific catalog #651201) and spun at 1200 rpm for 5 minutes at 4° C.
  • Samples could be resuspended in buffer for surface analysis of protein and immediate acquisition or resuspended in Fix/Perm (eBioscience catalog #11-5773) for intracellular staining of IL-10 (BioLegend catalog #505008), TGF-beta (BioLegend catalog #141404) and FoxP3 (eBioscience catalog #11-5773-82) according to the manufacturer's instructions. Additional surface staining included TGF-beta and Tim-1 (BioLegend catalog #119506) antibodies. All samples were acquired and analyzed as described above.
  • C57BL/6 and BALB/c mice were obtained from Charles River (Kingston, N.Y. or Raleigh, N.C.) and used in these experiments between 8 and 13 weeks of age.
  • the donor allogenic C57BL/6 mice were first anesthetized with an intraperitoneal injection of Ketamine (Fort Dodge Animal Health/Pfizer, Fort Dodge, Iowa) and Xylazine (Lloyd, Shenandoah, Iowa) and a median sternotomy was performed.
  • the donor heart was slowly perfused in situ with 1 ml of cold heparinized Ringer's lactate solution (Baxter Healthcare, Deerfield, Ill.) through the inferior vena cava and aorta before the superior vena cava and pulmonary veins were ligated and divided.
  • the ascending aorta and pulmonary artery were then transected, the graft removed from the donor and the heart was stored in ice-cold saline until engraftment.
  • a recipient mouse (Balb/c) was similarly anesthetized and prepped as described above for donor mice, except that the abdominal cavity was opened.
  • the abdominal aorta (AA) and the inferior vena cava (IVC) was isolated.
  • the donor heart was placed into the recipient abdomen (upside down) and the grafts revascularized with end-to-side anastomoses between the donor pulmonary artery and the recipient inferior vena cava, as well as the donor's aorta and the recipient abdominal aorta.
  • the abdominal muscle was closed with a running 5-O Vicryl suture (Ethicon, Johnson & Johnson, Somerville, N.J.), and the skin closed with running 5-0 Ethilon suture (Ethicon). Standard post-op pain assessment and management was performed. Grafts were assessed by palpation 5-7 times per week for the first 30 day, and then 3-4 times per week until the end of the study.
  • C57Bl/6 mice were treated with one 20 mg/kg intravenous dose of rhGAA (Genzyme Corporation), 3 consecutive intraperitoneal doses of 5 mg/kg methotrexate (APP Pharmaceuticals, LLC), and either 1D11 or 13C4 at 5 mg/kg (Genzyme Corporation) for 3 doses every other day.
  • Methotrexate, 1D11, and 13C4 treatment commenced with rhGAA injections. Spleens were collected 7 days after treatment initiation and were processed as described above for B cells, culture and flow analysis.
  • rhGAA titer data was obtained by treating animals as described above, with rhGAA dosed weekly over 12 weeks, methotrexate dosed for either 1 or 3 cycles, and 1D11 or 13C4 dosed 3 times a week every other day for 12 weeks. Serum samples were collected every 2 weeks for ELISA analysis.
  • Cardiac grafts were fixed in 10% neutral buffered formalin, bisected along the longitudinal axis to expose the right and left ventricles and the outflow tract, and routinely processed for paraffin embedding. Sections were cut at 5 microns and were stained with hematoxylin and eosin (H&E) or Masson's trichrome. Serial sections were also immunostained as described below. Each H&E-stained section was evaluated qualitatively for various features of allograft rejection pathology (e.g., vasculitis, myocardial degeneration and necrosis, myocarditis) using a histologic grading scheme.
  • H&E hematoxylin and eosin
  • Immunohistochemistry was performed using a Bond-Max automated immunostaining system (Leica Microsystems Inc., Buffalo Grove, Ill.). To detect CD3 and Foxp3 dual immunopositive cells, graft tissue sections were subjected to double immunostaining with anti-CD3 and anti-Foxp3 antibodies using Bond Polymer Refine Detection kit and Bond Polymer AP Red kit (Leica, Buffalo Grove, Ill.) following manufacturer's guidelines.
  • deparaffinized sections of paraffin-embedded grafts were subjected to heat-induced epitope retrieval (25 min at 99° C.), incubated with serum free protein block (Dako, Carpentaria, Calif.), rabbit monoclonal anti-CD3 antibody (Lab Vision/Neo Marker), peroxidase-conjugated polymer, peroxidase block, and diamino benizidene detection reagent followed by rat anti-mouse Foxp3 antibody (eBioscience Inc., San Diego, Calif.) and then a rabbit anti-rat antibody (Vector Laboratories, Inc., Burlingame, Calif.).
  • Serum alloantibody levels were determined by incubating serum from cardiac transplanted or normal mice at 1:50 dilutions with an SV40 transformed C57BL/6 fibroblast line (SVB6) followed by detection of fibrobast bound antibodies (alloantibodies) using FITC rabbit anti-mouse IgG (Dako, Carpinteria, Calif.) and by flow cytometric analysis. Geometric mean fluorescent intensities of serum stained fibroblasts were divided by isotype control stained fibroblasts to normalize alloantibody levels between experiments. The binding of serum antibodies to allogenic fibroblasts were specifically alloantibodies because the same serum samples did not bind to a SV40 transformed BALB/c fibroblast line (SVBalb) (data not shown).
  • SVBalb SV40 transformed BALB/c fibroblast line
  • mice C57BL/6 mice were obtained from Jackson Laboratories and were housed under specific pathogen free conditions. Control mice were given a single intravenous injection of rhGAA at 20 mg/kg. Tolerized mice were given a single intravenous injection of rhGAA at 20 mg/kg, in addition to three daily consecutive methotrexate intraperitoneal injections of 0.5 mg. Spleens were harvested on day six after the initial rhGAA injection, from both donor groups, and processed into pooled single cell suspensions. Cells were washed and filtered through a 0.22 ⁇ M filter.
  • Tolerized and non-tolerized recipient groups received either a high cell 10 ⁇ 10 6 or a low cell 5 ⁇ 10 6 concentration via intravenous injections.
  • Control groups were given rhGAA only, or rhGAA and methotrexate (single cycle of three consecutive daily MTX injections). All groups received weekly intravenous rhGAA 20 mg/kg injections, and were retro-orbitally bled biweekly for 16 weeks, into BD Vacutainer serum separator tubes. Serum was removed and stored below ⁇ 20° C. until used for ELISA.
  • Anti-rhGAA antibody titers were determined using ELISA, read on a SpectraMax M2, and calculated using Softmax to extrapolate titer value.
  • Raw data Softmax files and EXCEL spreadsheets containing control and titer information were stored on network servers. All graphs and statistics were generated using GraphPad Prism software.
  • Anti-Drug Antibodies are Produced in Response to Antibody Therapeutics
  • Polycloncal antibody mATG binds to a variety of immune cell types, including antigen-presenting cells.
  • Our data show that a single course of mATG (two doses of 25 mg/kg given three days apart, administered intraperitoneally) could generate anti-mATG IgG titers as high as 100,000 ( FIG. 1A ) in mice.
  • anti-mATG titers were further increased with titers up to 5 ⁇ 10 6 after five monthly injections ( FIG. 1B ).
  • rabbit IgG rbIgG was used as a control.
  • alemtuzumab does not cross-react with murine CD52
  • preclnical studies with alemtuzumab must be done in huCD52 Tg mice where the transgene expression pattern is similar to CD52 expression in humans. Similar to mATG, intravenous administration of alemtuzumab (0.5 mg/kg) elicited significant ADA responses in huCD52 Tg mice. These responses increased through the first four treatments and then declined such that following the fifth dose of alemtuzumab, huCD52 Tg mice no longer generated anti-alemtuzumab antibodies ( FIG. 2 ). This non-responsiveness suggests that natural tolerance had occurred (Rosenberg et al., Blood, 93(6):2081-8 (1999)).
  • Methotrexate Controls Anti-mATG IgG Responses with a Single Cycle of Administration
  • methotrexate could reduce anti-ATG responses, and thus, mitigate these safety concerns
  • a three-day regimen of methotrexate, given only as a single cycle was evaluated as a means of controlling anti-mATG IgG responses in mice. This is distinct from the regimen that was previously published in that only a single cycle of methotrexate was administered with mATG as opposed to at least three cycles that were given in the context of ERTs.
  • Methotrexate (Calbiochem catalog #454125) administered intraperitoneally at 5 mg/kg for six consecutive days starting on the first of two mATG administrations (25 mg/kg, 3 days apart) could suppress anti-mATG IgG responses through at least eight weeks following treatment by 95% when comparing area under the effect curves ( FIG. 3 ).
  • mATG may be the specific binding of mATG to antigen presenting cells (APCs) such as follicular dendritic cells, which when in the presence of complement may significantly enhance B cell responses.
  • APCs antigen presenting cells
  • follicular dendritic cells which when in the presence of complement may significantly enhance B cell responses.
  • Two treatment regimens of methotrexate also were evaluated.
  • ERT enzyme-replacement therapy
  • three cycles of methotrexate given during the first three doses of acid alpha-glucosidase provided a sustained reduction in antibody titer through at least eight months of weekly ERT dosing (Joseph et al., Clin Exp Immunol, 152(1):138-46 (2008)).
  • methotrexate was evaluated in the context of mATG where 5 mg/kg of methotrexate was given within 15 minutes of mATG administration as well as 24 and 48 hours following each of the first three monthly mATG treatments.
  • This regimen successfully decreased anti-mATG antibody responses from titers of approximately 4 million to titers of 816,000, yielding a reduction of 79% comparing area under the effect curves ( FIG. 4A ).
  • methotrexate had thus far been evaluated in the context of five consecutive monthly doses of mATG. To further evaluate whether an immune tolerance mechanism had been activated, animals that originally received five monthly mATG injections were withheld from treatment for eight weeks. Following this rest period, the animals were given a final injection of mATG. If a mechanism of immune tolerance was employed, anti-mATG IgG titers should not increase significantly following the sixth mATG treatment.
  • methotrexate-treated animals generated measurable titers that increase with successive mATG treatments, overall, titer levels in methotrexate-treated animals were consistently 100-fold lower than those observed in animals treated with mATG alone ( FIG. 6 ).
  • the lower level of antibody titers should significantly reduce the potential for safety risks and efficacy effects.
  • alemtuzumab In relapsing-remitting multiple sclerosis, alemtuzumab is dosed in annual cycles and patients can generate ADA (Coles et al., N Engl J Med, 359(17):1786-801 (2008)). As immunogenicity and pharmacokinetic testing is ongoing in multiple phase III studies, it is unclear whether anti-alemtuzumab antibodies will impact exposure, efficacy, and/or safety in a subset of patients. Thus, we evaluated whether a single cycle of methotrexate could control ADA titers following five monthly single injection cycles of alemtuzumab. HuCD52 Tg mice were given alemtuzumab (0.5 mg/kg) intravenously monthly for five consecutive months.
  • Methotrexate was given at 0.5, 1 or 5 mg/kg 15 minutes prior to the first monthly alemtzumab dose as well as 24 and 48 hours after the dose. 1 mg/kg of methotrexate provided some benefit, as it reduced anti-alemtuzumab responses by 88% ( FIG. 7A ). 5 mg/kg of methotrexate successfully reduced anti-alemtuzumab IgG responses by 99% ( FIG. 7A ). Methotrexate appeared to have no effect on natural tolerance.
  • huCD52 transgenic mice were treated with five monthly doses of 0.5 mg/kg alemtuzumab.
  • the mice also were treated with or without three daily doses of 5 mg/kg/day of methotrexate in connection with the first administration of alemtuzumab ( FIG. 7B ).
  • Serum samples were collected throughout the study to assess anti-alemtuzumab titers and confirm tolerance.
  • Titer data were obtained at 24 hours after the fifth monthly dose. The data demonstrated that methotrexate reduced anti-alemtuzumab antibody titers ( FIG. 7C ).
  • Methotrexate can Control Anti-Alemtuzumab Antibody Responses in the Context of a Clinically Relevant Alemtuzmab Dosing Regimen
  • alemtuzumab is dosed as an initial cycle of five daily treatments of 12 mg/day. Twelve months following the initial treatment cycle in patients, an additional cycle of three daily 12 mg doses of alemtuzumab is administered.
  • the levels of circulating CD 19 + B cells have recovered to baseline values; however, the levels of circulating CD4 + T helper cells and CD8 + T cytotoxic cells have not fully repopulated (Coles et al., N Engl J Med, 359(17):1786-801 (2008)).
  • alemtuzumab was re-administered in huCD52 Tg mice between 4 and 5 weeks following the first cycle. Since the initial course of alemtuzumab was a five-day cycle, methotrexate was administered 15 minutes prior to each daily alemtuzumab treatment and for two days afterwards.
  • the maximal cumulative cycle dose of methotrexate given in this regimen is 14 mg/kg (2 mg/kg/day), which is very similar to the cumulative dose of 15 mg/kg when methotrexate is given as a three-day course of 5 mg/kg/day.
  • Methotrexate can Improve the Pharmacokinetics and Pharmacodynamics of mATG
  • ADA can interfere with the pharmacokinetics and pharmacodynamics of protein therapeutics.
  • Methotrexate can Improve the Pharmacodynamics of Alemtuzumab
  • Methotrexate not only enhances the pharmacodynamics of mATG, but also restores alemtuzumab-mediated depletion of circulating T and B cells when anti-alemtuzumab responses appear to neutralize some of the depleting activity.
  • five monthly intravenous injections of alemtuzumab were given to huCD52 Tg mice with and without methotrexate.
  • 5 mg/kg of methotrexate was administered daily for the first three days of the 6 month study. Blood was harvested from animals of both treatment groups two days prior to the fifth dose and one day after the fifth dose of alemtuzumab. Cell populations were assessed by flow cytometry as described in Example 6.
  • T cell deficient Nu/Nu mice Studies also have been conducted in T cell deficient Nu/Nu mice to evaluate the role of T cells in generating anti-rhGAA titers. In these experiments, we have repeatedly observed that little to no ADA to rhGAA develop in these T cell deficient mice ( FIGS. 37A-B ). These data support the notion that T cells contribute to anti-rhGAA titers. Thus, as methotrexate can control ADA to rhGAA, it is also likely affecting T cell responses to rhGAA.
  • Methotrexate Enhances mATG-Mediated Survival of Heart Allogeneic Transplants
  • methotrexate could enhance the efficacy of mATG in normal mice
  • mATG function could be augmented by methotrexate in a transplantation setting. Since Thymoglobulin® is used clinically as an induction therapy to prolong transplant survival, we evaluated whether the addition of methotrexate could augment the efficacy of mATG in a murine allogeneic heart transplant model. 20 mg/kg of mATG was administered on days 0 and 4, while 2 mg/kg methotrexate was administered as a single cycle of treatment on days 0-6.
  • methotrexate 0.5 mg/kg
  • mice either received no treatment (saline control), mATG alone, or a combination of the mATG and methotrexate regimens.
  • methotrexate coadministered with mATG reduced anti-drug antibody titers to mATG regardless of regimen used ( FIG. 16A and Table 1).
  • coincidental to the reduction in antibody titers was an observed increase in mATG exposure in this transplant setting ( FIG. 16 ).
  • the anti-drug antibody titers increased even faster than in a normal mouse setting and resulted in mATG levels being near undetectable within 7 days of the first mATG administration ( FIG. 16B ).
  • anti-rabbit IgG antibody titers were significantly lower in mice treated with methotrexate, and circulating mATG levels were significantly higher in those mice. This emphasizes that under conditions of an ongoing inflammatory response, anti-drug antibody responses can be accelerated and perhaps have an even greater impact on pharmacodynamics and efficacy.
  • methotrexate had a profound inhibitory effect on mATG anti-drug antibodies and enhanced mATG exposure.
  • circulating mATG levels were still low to undetectable by 21 days with combination mATG and methotrexate treatment, additional tolerance mechanisms are likely at play given the >100 day graft survival.
  • methotrexate treatment alone was able to significantly reduce anti-allograft antibodies, further substantiating the effects of methotrexate on controlling antibody responses in general ( FIG. 18 ).
  • methotrexate appears to be able to control antibody responses against multiple antigens of the heart allograft simultaneously.
  • a further reduction was induced by combining mATG with methotrexate treatment ( FIG. 18C ).
  • methotrexate is a folate antagonist that is thought to mediate its suppressive effects by inducing the death of proliferating cells.
  • mATG data presented above demonstrates that antibody responses are induced but remain significantly decreased with each successive mATG treatment in methotrexate-treated animals. These data suggest that B cell responses are actively managed. Without wishing to be bound by theory, we hypothesize that methotrexate may induce a regulatory cell population(s) that controls these responses as they occur.
  • activated B cell subsets were significantly increased following methotrexate treatment ( FIG. 20 ). These populations included activated marginal zone B cells, activated follicular B cells and activated transitional 2 and 3 B cells.
  • Cell populations were defined as follows: B2/follicular B cells: CD19 + CD21 int CD23 hi ; transitional 2 B cells: CD19 + CD93 + CD23 + IgM hi ; and transitional 3 B cells: CD19 + CD93 + CD23 + IgM lo ; marginal zone B cells: CD19 + CD21 hi CD23 lo .
  • Methotrexate Increases Selected B Cell Populations in Combination with mATG
  • mice that received a single cycle of methotrexate were compared to mice that received either mATG alone or mATG with three cycles of methotrexate.
  • Two cell populations that unexpectedly demonstrated effects were activated follicular B cells and activated transitional 3 B cells ( FIGS. 24A-B , respectively).
  • mice treated with either mATG alone or in combination with three cycles of methotrexate decreases were observed in the absolute cell number of these cell populations.
  • mice that received only a single cycle of methotrexate no statistically significant decreases in these populations observed, suggesting that this dosing regimen of methotrexate induced some enrichment of these populations.
  • both of these cell populations were also shown to be enriched directly following methotrexate treatment in combination with in Myozyme ( FIG. 21 ). Similar subsets also have been identified in tolerant transplant patients. It is possible that a single cycle of methotrexate treatment can induce these B cell subsets which, upon antigen exposure, become activated and suppressive.
  • Methotrexate Increases Selected B Cell Populations in Combination with Alemtuzumab
  • huCD52 transgenic mice were treated with single monthly doses of 0.5 mg/kg alemtuzumab for five months, either with or without three daily doses of 5 mg/kg/day of methotrexate in connection with the first administration of alemtuzumab.
  • Cell populations were evaluated in the blood and spleen of the mice 2 days prior to, and 1, 7 and/or 28 days following, the fifth dose of alemtuzumab by flow cytometry.
  • alemtuzumab In blood, the pharmacodynamic effect of alemtuzumab was enhanced in methotrexate-treated animals 24 hours following the fifth dose of alemtuzumab. Statistically significant cell depletion was observed in both T cell and B cell subsets one day after the fifth dose, consistent with previous data indicating that anti-alemtuzumab titers are low in these animals and would not be likely to interfere with alemtuzumab-mediated depletion. By contrast, mice treated with alemtuzumab alone may have more alemtuzumab-neutralizing antibodies that interfere with alemtuzumab pharmacodynamics. As shown in FIG.
  • mice treated with alemtuzumab and methotrexate exhibit significant alemtuzumab-mediated depletion in total T cells, T helper cells and T regulatory cells one day following alemtuzumab dosing.
  • Similar findings were observed in circulating B cell subsets ( FIG. 25B ), although trends towards decreasing cell numbers were also observed in animals treated with alemtuzumab alone
  • splenic T cell populations were significantly depleted in methotrexate-treated animals one day following the fifth alemtuzumab treatment ( FIGS. 26A-B ).
  • methotrexate may enrich some or all of these B cell populations, which counters their alemtuzumab-mediated depletion. This enrichment is not expected to occur in the fast, fluid environment of the blood because immune cells do not differentiate in blood.
  • splenic cell subsets were significantly depleted ( FIG. 28A ). It is possible that at 24 hours, B cell depletion may be greater in alemtuzumab-treated animals than at three days following alemtuzumab treatment, as B cell repopulation may have begun by the three-day mark. This has been demonstrated in several studies assessing these populations in peripheral blood. Depletion was observed as early as three hours post-dosing in the peripheral blood.
  • methotrexate may have enriched B cell populations that potentially help mediate tolerance induction.
  • the populations that appear enriched are similar to those that are increased directly after methotrexate treatment ( FIG. 21 ). Taken together, this suggests that methotrexate may induce an environment that allows immune responses to be actively controlled at the time of antigen exposure, even long after the treatment with methotrexate.
  • Cytokines play a dual role in B cell responses. For instance, B-cell activating cytokines IL-6 and BAFF are also required for B cell differentiation. Since methotrexate seems to increase B cell populations, one may expect these cytokines to be increased. However, IL-6 is also pro-inflammatory, and therefore, elevated levels may interfere with methotrexate-induced effects. Like IL-6, IL-10 is involved in B cell differentiation into plasma cells and immunosuppression as well.
  • BAFF data were generated from serum samples taken 24 hours after the 5 th dose of alemtuzumab ( FIG. 29A ). This is a continuation of cellular data from this study presented above. At this time point no difference in BAFF levels were observed ( FIG. 29B ).
  • Cytokine levels were assessed one week after the second cycle of alemtuzumab ( FIG. 29B ). Generally, one week after the second cycle of alemtuzumab, cytokine levels appeared low. At this time point, a statistically significant increase was observed in TNF-alpha levels in animals treated with 2 mg/kg of methotrexate. This increase may reflect a change that is related to methotrexate-induced tolerance, or that is a sign of an inflammatory response. Trends observed in other cytokines, such as apparent increases in IL-6 and potentially slight decreases in IL-7, also were noted ( FIG. 30 ).
  • IL-10-secreting regulatory B cells have been associated with IL-10 secretion.
  • One way to assess whether IL-10-secreting regulatory B cells play a role in methotrexate-induced tolerance is to evaluate whether methotrexate can control antibody responses in IL-10 deficient animals. This type of assessment may be challenging in that, as mentioned above, IL-10 is necessary for plasma cell differentiation and therefore antibody responses may be lower in these animals. With this caveat in mind, we observed interesting trends suggesting that IL-10 may play a role in methotrexate-induced tolerance.
  • Anti-rhGAA responses in C57BL/6 wild-type animals treated with rhGAA and methotrexate were decreased at 4, 6 and 9 weeks.
  • anti-rhGAA titers at weeks 4 and 6 were not decreased in IL-10 knockout animals that were treated with rhGAA and methotrexate.
  • week 9 there was a slight decrease, which may indicate a delayed induction of tolerance in IL-10 deficient mice. This would be consistent with reports that IL-10 is not the only suppressive cytokine secreted by regulatory B cells (Sagoo et al., J. Clin. Investigation; 120(6):1848-1861 (2010)). TGF-beta also has been associated with the regulatory B cell response.
  • IL-10 may play a role in methotrexate-induced tolerance.
  • Alemtuzumab-treated multiple sclerosis patients can develop secondary autoimmunity.
  • the most common autoimmune disorders that develop following alemtuzumb treatment are those related to thyroid autoimmunity.
  • immune thrombocytopenic purpura and Good Pasture's syndrome also have been observed in multiple sclerosis patients treated with alemtuzumab.
  • All three types of autoimmunity are B cell mediated in that B cell responses and auto-antibodies are directly linked with disease development and pathology. The association of alemtuzumab treatment with the development of these secondary diseases is not well understood.
  • T cells and B cells are depleted. A large percentage of these depleted T and B cells are likely to be auto-reactive cells that interact with antigens expressed in the central nervous system. As a result, their subsequent depletion by alemtuzumab is thought to contribute to the therapeutic benefit of this monoclonal antibody therapy.
  • Patients suffering from autoimmune disease have been described to contain autoreactivities (i.e., autoreactive B cells and autoreactive antibodies) against multiple antigens that are associated with a variety of autoimmune diseases. Environmental, physiological, and genetic factors all contribute to the determination of whether autoimmune disease will likely ensue and influence which autoimmune disease will present in the patient. It is not uncommon for patients who suffer from one type of autoimmune disease to also develop another.
  • marginal zone B cells have been associated with thyroid autoimmunity (Segundo et al., Thyroid, 11(6):525-530 (2001).
  • the regulatory B cell subset, B10 B cells which have been shown to help quell autoimmunity in the murine model of multiple sclerosis, EAE (Matsushita et al., J. Clin. Investigation, 118:3420-3430 (2008)), and have been shown to exist in humans (Iwata et al., Blood, 117:530-541 (2011)) is depleted for longer periods of time than marginal zone B cells.
  • a short course of methotrexate treatment during the first cycle of alemtuzumab may help increase the representation of regulatory B10 B cells, and to restore the B cell balance such that marginal zone B cells are more equally represented in the B cell repertoire following alemtuzumab treatment and/or differentiate the marginal zone B cells into regulatory marginal zone B cells (CD1d+marginal zone cells).
  • Splenic B cell populations were studied to determine the effects of alemtuzumab on B cell depletion ( FIG. 32 ).
  • 0.5 mg/kg of alemtuzumab was administered intravenously for five consecutive days in huCD52 Tg mice.
  • populations of follicular B cells which are typically not autoreactive
  • B1 B cells and marginal zone B cells both of which are autoreactive
  • B10 B cells which are regulatory
  • transitional B cells and marginal zone B cells which are thought to be able to differentiate into B regulatory cells
  • mice treated with alemtuzumab alone were included in this study, and revealed that mice treated only with methotrexate in this short cycle regimen exhibited cellular effects in the absence of antigen stimulation, which can be different from the effects observed in control mice treated with methotrexate alone ( FIG. 34 ).
  • Methotrexate induces tolerance to protein therapies and transplanted cardiac tissue antigens, thereby abrogating B cell immune responses that relate to the production and secretion of ADA and anti-allograft antibodies.
  • methotrexate may not only help control the cellular environment following alemtuzumab treatment, but that it also may induce tolerance to self-proteins and mitigate the B cell immune responses that relate to the generation of auto-antibodies that contribute to the development and pathology of B cell-mediated autoimmune diseases.
  • Methotrexate Induces Immune Tolerance Through Specific Induction of Regulatory B Cell Populations
  • methotrexate induces immune tolerance not by the expected means of killing proliferating cells as suggested by others (Messinger et al., Genetics in Medicine 14:135-142 (2012) and Lacaná et al., Am J Med Genet Part C Semin Med Genet 160C:30-39 (2012)), but through the specific induction of regulatory B cells populations that express TGF-beta, IL-10 and FoxP3. B cells from mice tolerized to Myozyme® by a single cycle of methotrexate appeared to transfer immune tolerance to na ⁇ ve animals. Moreover, both IL-10 and TGF-beta appeared to be necessary for methotrexate-induced immune tolerance.
  • methotrexate induced TGF-beta, which in turn induced IL-10 and FoxP3.
  • This mechanism is novel and unexpected, and questions the value of a current clinical immune tolerance protocol that involves co-treatment with three cycles of methotrexate, Rituximab® (a B cell-depleting agent), and optionally, intravenous immunoglobulin (IVIG) (Messinger et al., supra).
  • IVIG intravenous immunoglobulin
  • rituximab-mediated B cell depletion is not thought to comprehensively deplete all B cells in the blood and tissues.
  • alemtuzumab actively depletes B10 B cells
  • treatment with methotrexate still is able to access those cells that seem to help maintain alemtuzumab tolerance for many months following the initial cycle of methotrexate.
  • rituximab treatment is rapidly followed by increased representation of transitional B cells, which, as shown herein, seem influenced by methotrexate to induce and mediate immune tolerance.
  • a single cycle of low dose methotrexate is a surprising and effective method of inducing immune tolerance to, inter alia, lymphocyte-depleting protein therapies.
  • B10 B cells One cell type associated with immune regulation is B10 B cells.
  • B10 B cells in both human and mouse are characterized by their expression of IL-10 (Matsushita et al., J. Clin. Invest. 118:3420-3430 (2008), Iwata et al., Blood 117:530-541 (2011)), and can only suppress immune responses in IL-10 competent mice.
  • B10 B cells were increased in methotrexate-tolerized mice ( FIG. 19 ), and IL-10 knockout animals were unresponsive to methotrexate-induced immune tolerance ( FIG. 31 ).
  • B10 B cells isolated from animals treated with Myozyme® or Myozyme® and methotrexate were assessed for IL-10 protein expression by flow cytometry.
  • IL-10 was expressed in B10 cells from both treatment groups, the number of B10 B cells expressing IL-10 was increased in animals treated with Myozyme® and methotrexate ( FIG. 38 ). IL-10 was expressed in both activated, CD86+ and non-activated, CD86 ⁇ B10 B cells following two days of culture ( FIG. 39 ). Previous studies appear to show that IL-10 expression is measured only following in vitro stimulation of cells by stimulants such as PMA/Ionomycin or LPS (Carter et al., J Immunol 186:5569-5579 (2011); Yanaba et al., J Immunol 182:7459-7472 (2009)).
  • TGF-beta expression is associated with immune regulation in T regulatory cells and is often linked with IL-10 expression in T regulatory cells. Moreover, some reports seem to indicate that regulatory B cells can express TGF-beta. Although B10 B cells have never been reported to express TGF-beta, we decided to assess TGF-beta expression in B10 B cells of mice treated with Myozyme® alone or with Myozyme® and methotrexate using flow cytometry. Unexpectedly, we found that B10 B cells express TGF-beta, and that the numbers of TGF-beta-expressing cells is increased in methotrexate-tolerized animals ( FIG. 40A ).
  • TGF-beta is expressed in both activated (CD86+) and non-activated (CD86 ⁇ ) B10 B cells ( FIG. 40B ). This is an additional novel observation that methotrexate treatment increases the numbers of cells that express TGF-beta. Additionally, methotrexate increases the expression level of TGF-beta ( FIG. 55 ).
  • FoxP3 is another protein associated with immune regulation. FoxP3 is a marker for T regulatory cells. FoxP3 has not been reported to be expressed in B10 B cells in mice. We investigated FoxP3 expression in B10 B cells in the presence and absence of methotrexate-induced immune tolerance by using flow cytometry. B10 B cells appear to express FoxP3, as seen in animals treated with Myozyme® alone ( FIG. 41A ). The numbers of FoxP3+ B cells appear to increase with treatment with both methotrexate and Myozyme® ( FIG. 41B ). Additionally, both cultured activated (CD86+) and non-activated (CD86 ⁇ ) B10 B cells appear to express FoxP3 ( FIG. 41 ). This was the first report that B10 B cells express FoxP3. We found that FoxP3 was expressed in both activated (CD86+) and unactivated (CD86 ⁇ ) B10 B cells and the expression of FoxP3 is increased with methotrexate treatment ( FIG. 56 ).
  • Methotrexate treatment also induces statistically significant increases in IL-10, TGF-beta and FoxP3 in multiple cell subsets as viewed by the shift in mean fluorescence intensity of these proteins in animals treated with Myozyme® alone or Myozyme® and methotrexate ( FIGS. 54-56 ).
  • TGF-beta expressing B cell populations enriched by treatment with methotrexate and Myozyme® we next looked to determine whether TGF-beta is required for methotrexate-induced immune tolerance.
  • Animals were treated with Myozyme® or Myozyme® and methotrexate with or without the presence of 5 mg/kg of anti-TGF-beta antibody (1D11, Genzyme) or the isotype control (13C4) given three times per week via intraperitoneal injection throughout the study.
  • Antibody titers were assessed bi-weekly in four different groups of animals.
  • TGF-beta was required for methotrexate-induced immune tolerance, then we would expect that animals treated with the anti-TGF-beta antibody should not exhibit reduced anti-Myozyme titers.
  • Week 6 titers are depicted in FIG. 45 , and suggest that TGF-beta may be necessary for methotrexate-induced immune tolerance. Because this is an early time point, only some of the animals have had time to generate anti-Myozyme responses. Importantly, at this time point titers appear similar to those shown in FIG. 15C at week 6, where two animals in the rhGAA alone and the rhGAA- and methotrexate- and 1D11-treated animals exhibited high titers. In comparison, none of the animals treated with rhGAA and methotrexate or rhGAA and methotrexate and 13C4 exhibited high titers.
  • spleens were isolated from animals treated with Myozyme® or Myozyme® and methotrexate that also were co-administered 1D11 or 13C4 seven days following a single Myozyme® treatment or a single Myozyme® and methotrexate treatment.
  • transitional 2, transitional 3, B10 and follicular B cells that expressed IL-10, TGF-beta, and FoxP3 were increased in animals treated with Myozyme® and methotrexate.
  • 1D11 treatment interfered with not only the methotrexate-induced expansion of cells expressing TGF-beta, but also with the expansion of some subsets expressing IL-10 and FoxP3. This was true specifically for B10 B cells ( FIG. 46 ) and follicular B cells ( FIG. 47 ), though FoxP3+ Follicular B cells did not appear to experience 1D11 effects. In transitional 2 B cells where 1D11 treatment interfered with TGF-beta-expressing transitional 2 B cells, no effects were seen with IL-10+ transitional 2 B cells (including activated transitional 2 B cells; FIG. 48 ).
  • interfering with TGF-beta by injecting a TGF-beta antibody during methotrexate-induced immune tolerance reduces the numbers of cells that are expressing TGF-beta in comparison to animals treated with the isotype control.
  • the typical increases observed with methotrexate-induced tolerance in IL-10 and FoxP3-expressing B10 B cells are inhibited by 1D11 treatment.
  • 1D11 treatment appears to influence methotrexate effects on TGF-beta, IL-10, and/or FoxP3 in certain B cell types, but not all B cell types.
  • methotrexate induces TGF-beta, which in turn induces IL-10 and potentially FoxP3 in certain cells, such as B10 B cells.
  • TGF-beta TGF-beta
  • FoxP3 FoxP3
  • methotrexate did not induce decreases in B cells as would have been expected if methotrexate was killing activated B cells to reduce antibody responses in either normal or transplanted animals ( FIGS. 52 and 53 ). These data suggest that methotrexate effects on anti-drug antibodies may not be mediated by anti-folate induced depletion of activated B cells. As expected, mATG treatment also did not impact total B cell numbers in this heterotopic cardiac allograft model. Overall, the combined treatment of mATG and methotrexate attenuated the severity of graft rejection pathology and was associated with decreased T cell infiltrate in the graft, but not with increases in cells with a regulatory T cell phenotype. This is consistent with the results generated in the context of Myozyme® and alemtuzumab.
  • cardiac grafts were collected and evaluated for pathology as well as cellular composition.
  • regulatory T cells have been associated with long-term graft survival in transplantation, are induced by Thymoglobulin® and mATG, and have been demonstrated to be responsible for delayed graft rejection following mATG treatment
  • CD3+Foxp3+ cells which bear a phenotype consistent with T regulatory cells, were evaluated.
  • cardiac grafts were collected from the mATG and methotrexate combination-treated group and the untreated syngeneic group at least 100 days after transplantation.
  • T cells within the inflammatory cell infiltrates in the epicardium appeared to have a T regulatory cell phenotype as indicated by dual CD3 and Foxp3 immunoreactivity, but this frequency appeared no greater within inflammatory infiltrates than in other groups. Therefore, reduced pathology also was observed early after treatment with mATG and methotrexate and was associated with both reduced and epicardium-restricted T cell infiltration.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Rheumatology (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Transplantation (AREA)
  • Obesity (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Materials For Medical Uses (AREA)
US14/116,486 2011-05-16 2012-05-03 Induction of immune tolerance by using methotrexate Abandoned US20140135337A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/116,486 US20140135337A1 (en) 2011-05-16 2012-05-03 Induction of immune tolerance by using methotrexate

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161486697P 2011-05-16 2011-05-16
US14/116,486 US20140135337A1 (en) 2011-05-16 2012-05-03 Induction of immune tolerance by using methotrexate
PCT/US2012/036405 WO2012158362A1 (en) 2011-05-16 2012-05-03 Induction of immune tolerance by using methotrexate

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/036405 A-371-Of-International WO2012158362A1 (en) 2011-05-16 2012-05-03 Induction of immune tolerance by using methotrexate

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/828,407 Continuation US11672802B2 (en) 2011-05-16 2020-03-24 Induction of immune tolerance by using methotrexate

Publications (1)

Publication Number Publication Date
US20140135337A1 true US20140135337A1 (en) 2014-05-15

Family

ID=47177257

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/116,486 Abandoned US20140135337A1 (en) 2011-05-16 2012-05-03 Induction of immune tolerance by using methotrexate
US16/828,407 Active US11672802B2 (en) 2011-05-16 2020-03-24 Induction of immune tolerance by using methotrexate
US18/141,128 Pending US20230372347A1 (en) 2011-05-16 2023-04-28 Induction of immune tolerance by using methotrexate

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/828,407 Active US11672802B2 (en) 2011-05-16 2020-03-24 Induction of immune tolerance by using methotrexate
US18/141,128 Pending US20230372347A1 (en) 2011-05-16 2023-04-28 Induction of immune tolerance by using methotrexate

Country Status (27)

Country Link
US (3) US20140135337A1 (ja)
EP (2) EP2709627B1 (ja)
JP (3) JP6174572B2 (ja)
KR (3) KR20140040744A (ja)
CN (4) CN113209288A (ja)
AU (1) AU2012256281B2 (ja)
BR (1) BR112013029501A2 (ja)
CA (1) CA2835819A1 (ja)
CL (1) CL2013003298A1 (ja)
CO (1) CO6841993A2 (ja)
DO (1) DOP2013000268A (ja)
EC (1) ECSP13013081A (ja)
ES (1) ES2836823T3 (ja)
GT (1) GT201300278A (ja)
IL (2) IL229245B (ja)
MA (1) MA35165B1 (ja)
MX (1) MX2013013445A (ja)
MY (1) MY173304A (ja)
NI (1) NI201300121A (ja)
PE (1) PE20140469A1 (ja)
PT (1) PT2709627T (ja)
RU (1) RU2674036C2 (ja)
SG (1) SG194802A1 (ja)
TN (1) TN2013000472A1 (ja)
UA (1) UA117556C2 (ja)
WO (1) WO2012158362A1 (ja)
ZA (1) ZA201308315B (ja)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160322251A1 (en) * 2015-04-30 2016-11-03 Nitto Denko Corporation Film for semiconductor device, method for manufacturing semiconductor device, and semiconductor device
WO2018209300A1 (en) * 2017-05-12 2018-11-15 Duke University Methods for the use of low-dose immune modulators transiently for treating patients undergoing protein replacement therapy
WO2019126647A1 (en) 2017-12-22 2019-06-27 Genzyme Corporation Biomarkers of methotrexate-induced immune tolerance
WO2021159092A1 (en) 2020-02-08 2021-08-12 Genzyme Corporation Compositions and methods for treating pompe disease

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107261154A (zh) 2011-04-29 2017-10-20 西莱克塔生物科技公司 致耐受性合成纳米载体
AU2012256281B2 (en) 2011-05-16 2017-06-15 Genzyme Corporation Induction of immune tolerance by using methotrexate
AU2014262163A1 (en) 2013-05-03 2015-11-19 Selecta Biosciences, Inc. Delivery of immunosuppressants having a specified pharmacodynamic effective-life and antigen for the inducation of immune tolerance
CN112933218A (zh) * 2013-06-04 2021-06-11 西莱克塔生物科技公司 非免疫抑制性抗原特异性免疫治疗剂的重复施用
EP3189138A4 (en) 2014-09-07 2018-04-18 Selecta Biosciences, Inc. Methods and compositions for attenuating gene editing anti-viral transfer vector immune responses
CA3055936A1 (en) 2017-03-11 2018-09-20 Selecta Biosciences, Inc. Methods and compositions related to combined treatment with anti-inflammatories and synthetic nanocarriers comprising an immunosuppressant

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4080325A (en) 1976-11-17 1978-03-21 The United States Of America As Represented By The Department Of Health, Education And Welfare Synthesis of methotrexate
US4558690A (en) 1982-01-26 1985-12-17 University Of Scranton Method of administration of chemotherapy to tumors
US5108987A (en) 1982-02-25 1992-04-28 Faulk Ward P Conjugates of proteins with anti-tumor agents
US5028697A (en) 1988-08-08 1991-07-02 Eli Lilly And Company Cytotoxic antibody conjugates of hydrazide derivatized methotrexate analogs via simple organic linkers
DE4000154A1 (de) 1990-01-04 1991-07-11 Symbiotec Gmbh Chemotherapeutisches mittel, insbesondere zur behandlung von krebserkrankungen
US6015557A (en) 1999-02-24 2000-01-18 Tobinick; Edward L. Tumor necrosis factor antagonists for the treatment of neurological disorders
US7485314B2 (en) * 2002-05-06 2009-02-03 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Induction of antigen specific immunologic tolerance
US8088916B2 (en) 2004-03-05 2012-01-03 Denki Kagaku Kogyo Kabushiki Kaisha Hyaluronic acid-methotrexate conjugate
US7884196B2 (en) 2004-10-06 2011-02-08 Oliver Lawless Vaccine composition comprising methylated DNA and immunomodulatory motifs
SI1988882T1 (sl) * 2006-03-02 2015-07-31 Alexion Pharmaceuticals, Inc. Podaljšanje preživetja alografta z inhibiranjem aktivnosti komplementa
WO2008094937A2 (en) 2007-01-29 2008-08-07 Joslin Diabetes Center, Inc. Methods of modulating angiogenesis
US20090155207A1 (en) * 2007-11-29 2009-06-18 Hariri Robert J Use of Immunomodulatory Compounds for the Treatment of Transverse Myelitis, Multiple Sclerosis, and Other Disorders
US7914785B2 (en) * 2008-01-02 2011-03-29 Bergen Teknologieverforing As B-cell depleting agents, like anti-CD20 antibodies or fragments thereof for the treatment of chronic fatigue syndrome
EP2077281A1 (en) * 2008-01-02 2009-07-08 Bergen Teknologioverforing AS Anti-CD20 antibodies or fragments thereof for the treatment of chronic fatigue syndrome
PL2344677T3 (pl) 2008-10-08 2017-11-30 Cambridge Enterprise Limited Sposoby i kompozycje do diagnozowania i leczenia choroby autoimmunologicznej wtórnej względem stwardnienia rozsianego
CN102271674A (zh) 2008-11-07 2011-12-07 4Sc股份公司 用于治疗自身免疫病的包含dhodh抑制剂和氨甲喋呤的联合治疗
AU2012256281B2 (en) 2011-05-16 2017-06-15 Genzyme Corporation Induction of immune tolerance by using methotrexate

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Currier (Low Dose oral methotrexate treatment of multiple sclerosis: a pilot study, Journal of Neurology, Neurosurgery, and Psychiatry, 1993; 56 pages 1217-1218) *
Jones (Improvement oin disability after alemtuzumab treatment of multiple sclerosis is associated with neuroprotective autoimmunity, Brain Advance Access, 2010, pages 1-16) *
Seland (Evaluation of antihymocyte globulin in acute relapses of multiple sclerosis, Neurology, 1974, vol 24, no 134, abstract only). *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160322251A1 (en) * 2015-04-30 2016-11-03 Nitto Denko Corporation Film for semiconductor device, method for manufacturing semiconductor device, and semiconductor device
WO2018209300A1 (en) * 2017-05-12 2018-11-15 Duke University Methods for the use of low-dose immune modulators transiently for treating patients undergoing protein replacement therapy
US20200405722A1 (en) * 2017-05-12 2020-12-31 Duke University Methods for the Use of Low-Dose Immune Modulators Transiently for Treating Patients Undergoing Protein Replacement Therapy
US11944628B2 (en) * 2017-05-12 2024-04-02 Duke University Methods for the use of low-dose immune modulators transiently for treating patients undergoing protein replacement therapy
WO2019126647A1 (en) 2017-12-22 2019-06-27 Genzyme Corporation Biomarkers of methotrexate-induced immune tolerance
WO2021159092A1 (en) 2020-02-08 2021-08-12 Genzyme Corporation Compositions and methods for treating pompe disease

Also Published As

Publication number Publication date
RU2674036C2 (ru) 2018-12-04
CL2013003298A1 (es) 2014-07-25
JP2018184466A (ja) 2018-11-22
CN110124039A (zh) 2019-08-16
GT201300278A (es) 2015-02-12
JP6174572B2 (ja) 2017-08-02
TN2013000472A1 (en) 2015-03-30
ES2836823T3 (es) 2021-06-28
CN113209288A (zh) 2021-08-06
PT2709627T (pt) 2020-12-24
MX2013013445A (es) 2014-07-28
CA2835819A1 (en) 2012-11-22
EP2709627B1 (en) 2020-09-23
CO6841993A2 (es) 2014-01-20
AU2012256281A1 (en) 2013-11-28
IL229245B (en) 2020-08-31
ZA201308315B (en) 2014-07-30
JP2014513722A (ja) 2014-06-05
KR20140040744A (ko) 2014-04-03
JP6381707B2 (ja) 2018-08-29
MA35165B1 (fr) 2014-06-02
NZ716716A (en) 2018-06-29
US20200360385A1 (en) 2020-11-19
IL276879B (en) 2022-06-01
BR112013029501A2 (pt) 2017-01-24
KR20200116543A (ko) 2020-10-12
ECSP13013081A (es) 2015-04-30
IL276879A (en) 2020-10-29
CN103648501A (zh) 2014-03-19
EP2709627A1 (en) 2014-03-26
US20230372347A1 (en) 2023-11-23
JP6694020B2 (ja) 2020-05-13
NI201300121A (es) 2014-02-28
KR102316283B1 (ko) 2021-10-21
CN109620830A (zh) 2019-04-16
RU2013155618A (ru) 2015-06-27
PE20140469A1 (es) 2014-04-23
EP2709627A4 (en) 2014-11-12
EP3824890A1 (en) 2021-05-26
IL229245A0 (en) 2014-01-30
KR20190132551A (ko) 2019-11-27
MY173304A (en) 2020-01-14
AU2012256281B2 (en) 2017-06-15
WO2012158362A1 (en) 2012-11-22
KR102163285B1 (ko) 2020-10-08
US11672802B2 (en) 2023-06-13
SG194802A1 (en) 2013-12-30
DOP2013000268A (es) 2014-01-31
JP2017141236A (ja) 2017-08-17
NZ742216A (en) 2018-10-26
UA117556C2 (uk) 2018-08-27
NZ617575A (en) 2016-10-28

Similar Documents

Publication Publication Date Title
US11672802B2 (en) Induction of immune tolerance by using methotrexate
Kishimoto Development of ImmTOR tolerogenic nanoparticles for the mitigation of anti-drug antibodies
CN104177464B (zh) Cd83在联合治疗中的用途
Rodrigo et al. Acute rejection following kidney transplantation: state-of-the-art and future perspectives
Miura et al. TNX-1500, a crystallizable fragment–modified anti-CD154 antibody, prolongs nonhuman primate cardiac allograft survival
US20210236512A1 (en) Clozapine for the treatment of a immunoglobulin driven b cell disease
NZ716716B2 (en) Induction of immune tolerance by using methotrexate
OA16781A (en) Induction of immune tolerance by using methotrexate.
NZ617575B2 (en) Induction of immune tolerance by using methotrexate
NZ742216B2 (en) Induction of immune tolerance by using methotrexate
Trambley Costimulation blockade in transplantation: Limitations and adjunct therapies

Legal Events

Date Code Title Description
AS Assignment

Owner name: GENZYME CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JOSEPH, ALEXANDRA;RICHARDS, SUSAN;RUZEK, MELANIE;AND OTHERS;SIGNING DATES FROM 20111012 TO 20111022;REEL/FRAME:033668/0964

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION