WO2003012432A1 - Erythropoietin and anti-tumor necrosis factor alpha combination therapy - Google Patents

Erythropoietin and anti-tumor necrosis factor alpha combination therapy Download PDF

Info

Publication number
WO2003012432A1
WO2003012432A1 PCT/US2001/024384 US0124384W WO03012432A1 WO 2003012432 A1 WO2003012432 A1 WO 2003012432A1 US 0124384 W US0124384 W US 0124384W WO 03012432 A1 WO03012432 A1 WO 03012432A1
Authority
WO
WIPO (PCT)
Prior art keywords
epo
tnfα
study
week
patients
Prior art date
Application number
PCT/US2001/024384
Other languages
French (fr)
Inventor
Loretta Itri
Original Assignee
Ortho-Mcneil Pharmaceutical, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ortho-Mcneil Pharmaceutical, Inc. filed Critical Ortho-Mcneil Pharmaceutical, Inc.
Priority to PCT/US2001/024384 priority Critical patent/WO2003012432A1/en
Priority to CA002456177A priority patent/CA2456177A1/en
Priority to JP2003517575A priority patent/JP2004537569A/en
Priority to EP01959479A priority patent/EP1423693A4/en
Publication of WO2003012432A1 publication Critical patent/WO2003012432A1/en

Links

Classifications

    • 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
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • 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
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1793Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1816Erythropoietin [EPO]
    • 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
    • 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
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • Erythropoietin is a glycoprotein hormone produced by the kidney in response to tissue hypoxia that stimulates red blood cell production in the bone marrow.
  • the gene for erythropoietin has been cloned and expressed in Chinese hamster ovary (CHO) cells as described in United States Patent No. 4,703,008.
  • Recombinant human erythropoietin r- HuEPO, Epoetin alfa
  • r- HuEPO Epoetin alfa
  • Recombinant human erythropoietin acts by increasing the number of cells capable of differentiating into mature erythrocytes, triggering their differentiation and augmenting hemoglobin synthesis in developing erythroblasts (SB. Krantz, Blood (1991) 77: 419-434; BS. Beckman, M. Mason-Garcia, The Faseb Journal (1991) 5: 2958-2964).
  • Epoetin alfa has been evaluated in normal subjects as well as in subjects with various ' anemic conditions. Epoetin alfa induces a brisk haematological response in normal human volunteers, provided that adequate supplies of iron are available to support increased hemoglobin synthesis. A majority of trials have investigated the safety and effectiveness of Epoetin alfa in the treatment of chronic renal failure and of anemia in cancer.
  • Epoetin alfa for the treatment of anemia associated with rheumatoid arthritis, prematurity, AIDS, bone marrow transplantation, myelofibrosis, sickle cell anemia, as a facilitator of presurgical autologous blood donation, and as a perisurgical adjuvant.
  • Epoetin alfa is approved for sale in many countries for the treatment of anemia in chronic renal failure (dialysis and predialysis), anemia in zidovudine treated HIN positive patients (US), anemia in cancer patients receiving platinum-based chemotherapy, as a facilitator of autologous blood predonation, and as a perisurgical adjuvant to reduce the likelihood of requiring allogeneic blood transfusions in patients undergoing orthopedic surgery.
  • Epoetin alfa can correct anemia in cancer patients, at doses several times higher than those shown to be effective in renal patients.
  • Anemia may result from the disease itself, the effect of concomitantly administered chemotherapeutic agents, or a combination of both.
  • the condition often takes on the characteristics of the anemia of chronic disease (ACD).
  • ACD is associated with erythroid hypoplasia of the bone marrow, a somewhat shortened circulating life of red cells and decreased bone marrow re-utilization of iron. If erythropoietin levels are measured, they are found to be within the normal range, but inappropriately low for the degree of anemia.
  • the patient has a blunted erythropoietin response.
  • About 50- 60% of anemic cancer patients receiving chemotherapy responded with a hemoglobin rise of at least 2 g/dL to Epoetin alfa therapy given three times weekly at a dose of 150 IU/kg over a period of 12 weeks (RI. Abels, KM. Larholt, KD. Krantz, and EC. Bryant, Proceedings of the Beijing Symposium, Alpha Medical Press, Dayton, Ohio, (1991) 121- 141).
  • doses up to 300 IU/kg were sometimes required, demonstrating the relative resistance to the effect of Epoetin alpha in these patients.
  • Tumor necrosis factor is part of a group of inflammatory cytokines that has been implicated in the pathologic changes seen in a number of diseases. TNF has been implicated as a contributing cause of fatigue, asthenia, anorexia and cachexia in a number of disorders. Elevated levels of TNF were associated with post-dialysis fatigue in one study (AW. Dreisbach, T. Hendrickson, LA. Beezhold, et al, Int. J. Artif. Organs (1998) 21:83-6) and were seen in patients with disorders of excessive daytime sleepiness in another. (AN. Vgontzas, DA. Papanicolaou, EO. Bixler, et al., J. Clin.
  • TNF may act as a negative modulator of erythropoiesis.
  • murine models there are conflicting data regarding the ability to abrogate TNF mediated anemia.
  • human studies there is an association between circulating TNF levels and the need for increased doses of epoetin alfa in hemodialysis patients (M. Goiechea, J. Nartin, P.
  • the unmet need of enhanced EPO treatment for patients who suffer from cancer and fatigue has been met by the present invention that describes a method to treat patients with a combination of EPO and an anti- Tumor Necrosis Factor alpha agent.
  • the present invention provides a method for the treatment of a patient, comprising the steps, in any order or concurrently of administering a therapeutic amount of at least one anti-Tumor Necrosis Factor compound and a therapeutic amount of erythropoietin.
  • the present invention further provides a method for the treatment of a patient having malignancies, comprising the steps, in any order or concurrently of administering a therapeutic amount of at least one anti-Tumor Necrosis Factor compound, a therapeutic amount of erythropoietin, and a therapeutic amount of an anti-tumor agent.
  • the methods of the present invention result in a reduced need for transfusion and a greater hemoglobin level in the patient. Further, the present invention provides that anemia may be successfully prevented or treated during the course of a chemotherapeutic regimen using the method of the present invention. The methods of the present invention result in patients that show greater quality of life, show improvement in physical performance and well being.
  • EPO Erythropoietin
  • EPO molecules may include small organic or inorganic molecules, synthetic or natural arnino acid peptides, purified protein from recombinant or natural expression systems, or synthetic or natural nucleic acid sequences, or any chemical derivatives of the aforementioned.
  • the generally preferred, form of EPO is purified, recombinant EPO, distributed under the trademarks of EPREX® or ERYPO®.
  • Epoetin alfa is a sterile, clear, colourless, aqueous solution for injection, that is provided in prefilled, single-use syringes containing either 4,000 or 10,000 IU epoetin alfa (a recombinant human erythropoietin) and 2.5 mg/mL human serum albumin in 0.4 mL (4,000 IU syringe) or 1.0 mL (10,000 IU syringe) of phosphate buffer.
  • Erythropoietin shall include those proteins that have the biological activity of human erythropoietin, as well as erythropoietin analogs, erythropoietin isoforms, erythropoietin mimetics, erythropoietin fragments, hybrid erythropoietin proteins, erythropoietin receptor agonists, renal erythropoietin, brain erythropoietin, oligomers and multimers of the above, homologues of the above, and muteins of the above, regardless of the biological activity of same, and further regardless of the method of synthesis or manufacture thereof including but not limited to naturally occurring, recombinant, synthetic, transgenic, and gene activated methods.
  • anti-Tumor Necrosis Factor Compound refers to drug products that decrease the amount of circulating, active TNF ⁇ .
  • the compound may achieve this by decreasing the amount of cellular TNF ⁇ mRNA transcription, by decreasing mRNA translation into TNF ⁇ protein, or by decreasing cellular secretion of TNF ⁇ .
  • Roy A. Black et al, from Immunex Corp. have discovered a compound that inhibits the enzyme that releases TNF from cell surfaces ⁇ Nature, 370, 218(1994)). This compound, called TNF- ⁇ protease enzyme inhibitor, curbs production of soluble TNF.
  • Other suitable anti-TNF ⁇ compounds could work by increasing the rate of clearance or decreasing the amount of functional TNF ⁇ in circulation.
  • Preferred anti-TNF ⁇ compounds are Thalidomide, Pentoxifylline, Infliximab, glucocorticoids, and Etanercept.
  • the anti-TNF ⁇ compounds may be administered as combinations in order to maximize modulation of TNF since these agents acts as TNF ⁇ inhibitors at a different points in TNF synthesis and pharmacokinetic activity.
  • Pentoxifylline inhibits TNF- ⁇ gene transcription (Doherty et al, Surgery (St. Louis), 110:192, 1991), while thalidomide enhances TNF- ⁇ m-RNA degradation (Moreira et al., 1993) and glucocorticoids such as dexamethasone inhibit TNF- ⁇ m-RNA translation (Han et al., J. Exp. Med. (1990) 172:391).
  • Infliximab and etanercept act by reducing the amount of circulating, active TNF ⁇ .
  • Pentoxifylline decreases circulating TNF ⁇ at the Standard dose of 400 mg 3 times daily. Pentoxifylline inhibits TNF- ⁇ gene transcription (Doherty et al., Surgery (St. Louis), 110:192, 1991).
  • Glucocorticoids such as dexamethasone inhibit TNF- ⁇ m-RNA translation.
  • Dexamethasone is administered orally, intramuscularly, or intravenously in the dose range of 8-40 mg (pediatric dose: 0.25-0.5 mg/kg). If given intravenously, dexamethasone should be given over 10-15 minutes, since rapid administration may cause sensations of generalized warmth, pharyngeal tingling or burning, or acute transient perianal and/or rectal pain.
  • Methylprednisolone is also administered orally, intramuscularly, or intravenously at doses and schedules that vary from 40-500 mg every 6-12 hours for up to 20 doses.
  • Thalidomide may act by enhancing TNF- ⁇ m-RNA degradation (Shannon et al. (1990) Amer. Society for Microbiology Ann. Mtg., Abs. U- 53). Thalidomide is given by oral administration in the range of about 30 mg to 1500 mg per 24 hours, preferably 200 to 500 mg per 24 hours for an adult human weighting 70 kg.
  • REMICADETM is a monoclonal antibody that blocks the biological activity of circulating TNF ⁇ . Infliximab does not neutralize TNF ⁇ (lymphotoxin ⁇ ), a related cytokine that utilizes the same receptors as TNF ⁇ . Remicade is supplied as a sterile, white, lyophilized powder for intravenous infusion. Following reconstitution with 10 mL of Sterile Water for Injection, USP, the resulting pH is approximately 7.2. Each single- use vial contains 100 mg Infliximab, 500 mg sucrose, 0.5 mg polysorbate 80, 2.2 mg monobasic sodium phosphate and 6.1 mg dibasic sodium phosphate. No preservatives are present.
  • E ⁇ BRELTM (etanercept) is a dimeric fusion protein consisting of the extracellular ligand- binding portion of the human 75 kilodalton (p75) tumor necrosis factor receptor (T ⁇ FR) linked to the Fc portion of human IgGl.
  • the Fc component of etanercept contains the C H domain, the C R 3 domain and hinge region, but not the C H I domain of IgGl.
  • Etanercept is produced by recombinant DNA technology in a Chinese hamster ovary (CHO) mammalian cell expression system. It consists of 934 amino acids and has an apparent molecular weight of approximately 150 kilodaltons.
  • ENBRELTM is supplied as a sterile, white, preservative-free, lyophilized powder for parenteral administration after reconstitution with 1 mL of the supplied Sterile Bacteriostatic Water for Injection, USP (containing 0.9% benzyl alcohol). Following reconstitution, the solution of ENBRELTM is clear and colorless, with a pH of 7.4 ⁇ 0.3. Each single-use vial of ENBRELTM contains 25 mg etanercept, 40 mg mannitol, 10 mg sucrose, and 1.2 mg tromethamine. ENBRELTM is administered as a single subcutaneous (SC) injection.
  • SC subcutaneous
  • Such tumor types include, but are not limited to, solid tumors, hematological tumors, sarcomas, carcinomas, neoplasms, as well as tumors of the breast, Non-Hodgkin's Lymphoma, myeloma, Hodgkin's lymphoma, leukemia, colon, rectal, colo-rectal, stomach, gastrointestinal, ovarian, lung, pancreas, and prostate.
  • Platinum containing anti-tumor agents include, but are not limited to, cisplatin, and cis-dichlorodiammineplatinum.
  • Non-platinum containing anti-tumor agents include, but are not limited to, cyclophosphamide, fluorouracil, epirubicin, methotrexate, vincristine, doxorubicin, bleomycin, and etoposide.
  • Quality of life is usually affected by malignancy, e.g., due to the underlying disease, effects of therapy and the psychological burden of coping with cancer.
  • Fatigue is the most frequently reported symptom in cancer patients and impairs significantly their quality of life (ML. Winningham, LM. Nail, M. Barton Burke, et al., Oncol Nursing Forum (1994) 21: 23-34)
  • Anemia contributes to fatigue and to the reduction in quality of life.
  • the effects on fatigue (FACT An) and quality of life (ECOG performance score, Cancer Linear Analogue Scale, SF36) of early intervention and/or treatment of anemia with the combination therapy of EPO + anti-TNF ⁇ can be alleviated with concurrent use of EPO + anti-TNF ⁇ during chemotherapy.
  • Randomization will be used to avoid bias in the assignment of patients to treatment, to increase the likelihood that subject attributes are evenly balanced across groups, and to enhance the validity of comparisons across treatment groups.
  • Investigators and patients will be blinded to the identity of etanercept and placebo to enhance the validity of comparisons that are subject to observer bias or the placebo effect (e.g. - QoL end points).
  • EPO will be given to all patients in an open label manner. Patients will be assigned randomly to each treatment regimen.
  • Serum TNF ⁇ and anti-TNF ⁇ levels • FACT- An, Linear Analog Scale Assessment (LAS A), CLOX and EXIT 25 within 3 days prior to the administration of chemotherapy
  • Subjects will be assigned to treatment groups based on a computer- generated randomization schedule. The randomization will be balanced by using permuted blocks and will be stratified by center and type of cancer. Based on this randomization code, the study drug will be packaged and labeled for each subject. [Subject numbers] [Medication code numbers] will be preprinted on the study drug labels and assigned sequentially as subjects qualify for the study and are randomized to treatment.
  • the etanercept or placebo drug container will have a two-part, tear-off label with directions for use and other information on each part.
  • the tear-off section of the label will contain a concealed area identifying the study drug (e.g., active or placebo) and will be removed and attached to the subject's CRF when the drug is dispensed.
  • the second part of the label will remain affixed to the study drug container and will contain all identifying information except for the identity of the drug contained.
  • the study drugs will be identical in appearance and will be packaged in identical containers.
  • the blind should not be broken.
  • the blind should be broken only if specific emergency treatment would be dictated by knowing the treatment status of the subject. In such cases, the investigator must contact the sponsor. If the investigator is unable to contact the sponsor, the investigator may in an emergency determine the identity of the treatment by exposing the concealed area of the label attached to the subject's CRF. Individual code breaks by the investigator will normally result in withdrawal of the subject from the trial. The date, time, and reason for the unblinding must be documented on the appropriate page of the CRF (Study Completion Information) and the sponsor must be informed as soon as possible.
  • the randomization schedule will not be revealed to study subjects, parents or guardians, investigators and clinical staff, or site managers until all subjects have completed the double-blind phase of the trial.
  • patients should remain blinded to study-related test results prior to the completion of each set of QoL and cognitive measures.
  • the treating physician may provide laboratory results as soon as the forms are completed for that visit.
  • evaluations For patients receiving every three week chemotherapy, evaluations should be completed at least 3 days following the end of the last dose of chemotherapy. For patients receiving every four week chemotherapy, evaluations should be completed within 3 days prior to the scheduled chemotherapy course.
  • EPO will be started at a dose of 40,000 U per week given SC, without regard to the timing of chemotherapy. If at the evaluation prior to the week 5 dose the hemoglobin has not increased by at least 1 g/dl over the baseline value, the dose of EPO will be increased to 60,000 U per week for weeks 5-8. If the hemoglobin increases by at least 2 g/dl over baseline at the time of the week 9 evaluation, the patient will be considered a responder. Responders will continue to receive the dose of EPO given during weeks 5-8 until week 16. Non-responders will be offered the opportunity to receive EPO + ETAN in an 8 week open label extension.
  • Placebo injections will be given at a dose of 1 ml SC, at a site separate from the EPO site, on days 1 and 4 of each week (day 1 is counted as the day the EPO and placebo are given together).
  • the patient should receive two doses of placebo during week 8, prior to the week 9 evaluation.
  • Responders will continue to receive placebo in a blinded manner through week 16.
  • Two placebo injections should be given in week 16, prior to the off study evaluation.
  • Non-responders will be offered the opportunity to receive EPO + ETAN in an 8 week open label extension.
  • EPO will be started at a dose of 40,000 U per week given SC, without regard to the timing of chemotherapy. If at the evaluation prior to the week 5 dose the hemoglobin has not increased by at least 1 g/dl over the baseline value, the dose of EPO will be increased to 60,000 U per week for weeks 5-8. If the hemoglobin increases by at least 2 g/dl over baseline at the time of the week 9 evaluation, the patient will be considered a responder. Responders will continue to receive the dose of EPO given during weeks 5-8 until week 16. Non-responders will proceed to the Off Study Evaluation at week 9.
  • ETAN injections will be given at a dose of 25 mg (1 ml) SC, at a site separate from the EPO site, on days 1 and 4 of each week (day 1 is counted as the day the EPO and ETAN are given together).
  • the patient should receive two doses of ETAN during week 8, prior to the week 9 evaluation. Responders will continue in ETAN through week 16. Two ETAN injections should be given during week 16 and prior to the off study evaluation. Non-responders will proceed to the off study evaluation at week 9.
  • ETAN ETAN
  • Week 9 placebo will be replaced with ETAN at week 9 for patients who are non-responders in Regimen A.
  • the following doses of EPO and ETAN will be given during weeks 9-16 in these patients: (1) Patients on an EPO dose of 60,000 U/week at week 8 will continue on that dose for weeks 9-12.
  • ETAN will be given at a dose of 25 mg SC, at a site separate from the EPO site, on days 1 and 4 of each week (day 1 is counted as the day the EPO and placebo are given together).
  • ETAN will be started on the day of the week 9 EPO dose and continue until 2 doses are given during week 12.
  • the patient will be considered a responder and continue on study through week 16. ETAN should continue until 2 doses are given in week 16. The patient then will proceed to the Off Study Evaluation. If the patient is a non-responder at week 13, then the patient will proceed to the Off Study Evaluation.
  • ETAN ETAN will be given at a dose of 25 mg SC, at a site separate from the EPO site, on days 1 and 4 of each week (day 1 is counted as the day the EPO and placebo are given together).
  • the first dose of ETAN will be given on the day of the week 9 EPO dose and continue until 2 doses are given during week 12. If the evaluation at week 13 indicates at least a 1 g/dl increase in hemoglobin over the week 9 value, or a 2 g/dl increase over the baseline value, then the patient will continue on study through week 16 at the same dose of EPO. ETAN should continue until 2 doses are given during week 16 EPO.
  • EPO should be withheld until the hemoglobin drops to 12 g/dl. EPO should then be resumed at 75% of the last dose given before discontinuation. If the hemoglobin increases by > 1.3 g/dl in a 2 week period, EPO should be continued at 75% of the previous dose.
  • Epoetin alfa 40,000 U/ml should be brought to room temperature and drawn up into a plastic syringe immediately prior to administration by SC injection according to standard techniques.
  • the EPO and ETAN/placebo may be given at the same time on day 1 of each week, but should be administered at separate sites.
  • Each vial of EPO should be used only once.
  • the maximum injection volume per site is 2 ml.
  • Study medication (ETAN/placebo) should be reconstituted with 1 ml of the supplied sterile bacteriostatic water for injection, USP (0.9% benzyl alcohol). The diluent should be slowly injected into the vial. Some foaming may occur. To avoid excessive foaming, do not shake or agitate vigorously. Swirl gently until dissolution occurs, usually over less than 5 minutes. The solution should be clear and colorless. The medication should then be drawn up into a plastic syringe and administered SC as soon as possible after reconstitution. New injections should be given at least 1 inch from an old site and never into areas where the skin is tender, bruised, hard or red. Prior to injection of the study medication, Part 2 of the two-part vial label is to be attached to the subject's case report form after entry of the subject's initials and number, and the date of study medication administration. Each vial of study medication should be used only once. Iron
  • Iron deficiency may develop during the use of EPO and may limit the efficacy of EPO if left untreated. If laboratory evidence of iron deficiency develops during the study, the patient should be given 150-200 mg of elemental iron per day. The appropriate formulation of the iron supplement is left to the discretion of the treating physician.
  • Patients may be transfused with packed red blood cells when judged to be necessary by the physician of record.
  • a hemoglobin level should be obtained at the time the type and cross-match specimen is drawn.
  • the pre-transfusion hemoglobin value, along with the number of units used and the total volume transfused, should be recorded in the case report form.
  • the sponsor must be notified in advance (or as soon as possible thereafter) of any instances in which prohibited therapies are administered.
  • ⁇ Response is defined as a 2g/dl or greater increase in the hemoglobin when compared to the Baseline value.
  • ⁇ Significant weight loss will be defined as Off Study Weight divided by Baseline Weight less than 0.90.
  • ⁇ Disease status will be defined according the current version of the RECIST Criteria issued by the National Cancer Institute.

Abstract

The present invention provides a method to treat anemic and non-anemic subjects having malignancies with a combination of EPO and an anti-Tumor Necrosis Factor compound such that there is a reduced need for transfusion and a greater hemoglobin level. The present invention provides a method to treat anemic and non-anemic subjects having malignancies with a combination of EPO and an anti-Tumor Necrosis Factor compound such that anemia may be successfully prevented or treated during the course of a chemotherapeutic regimen. The present invention provides a method to treat anemic and non-anemic subjects having malignancies with a combination of EPO and an anti-Tumor Necrosis Factor compound such that the patients show greater quality of life, show relief from fatigue symptoms, and show improvement in physical performance and well being.

Description

TITLE OF THE INVENTION
ERYTHROPOIETIN AND ANTI-TUMOR NECROSIS FACTOR ALPHA COMBINATION THERAPY
BACKGROUND OF THE INVENTION
Erythropoietin (EPO) is a glycoprotein hormone produced by the kidney in response to tissue hypoxia that stimulates red blood cell production in the bone marrow. The gene for erythropoietin has been cloned and expressed in Chinese hamster ovary (CHO) cells as described in United States Patent No. 4,703,008. Recombinant human erythropoietin (r- HuEPO, Epoetin alfa) has an amino acid sequence identical to that of human urinary erythropoietin, and the two are indistinguishable in chemical, physical and immunological tests.
Recombinant human erythropoietin acts by increasing the number of cells capable of differentiating into mature erythrocytes, triggering their differentiation and augmenting hemoglobin synthesis in developing erythroblasts (SB. Krantz, Blood (1991) 77: 419-434; BS. Beckman, M. Mason-Garcia, The Faseb Journal (1991) 5: 2958-2964).
In clinical trials to date, Epoetin alfa has been evaluated in normal subjects as well as in subjects with various ' anemic conditions. Epoetin alfa induces a brisk haematological response in normal human volunteers, provided that adequate supplies of iron are available to support increased hemoglobin synthesis. A majority of trials have investigated the safety and effectiveness of Epoetin alfa in the treatment of chronic renal failure and of anemia in cancer.
Other trials have evaluated Epoetin alfa for the treatment of anemia associated with rheumatoid arthritis, prematurity, AIDS, bone marrow transplantation, myelofibrosis, sickle cell anemia, as a facilitator of presurgical autologous blood donation, and as a perisurgical adjuvant. Epoetin alfa is approved for sale in many countries for the treatment of anemia in chronic renal failure (dialysis and predialysis), anemia in zidovudine treated HIN positive patients (US), anemia in cancer patients receiving platinum-based chemotherapy, as a facilitator of autologous blood predonation, and as a perisurgical adjuvant to reduce the likelihood of requiring allogeneic blood transfusions in patients undergoing orthopedic surgery.
Clinical experience has been collected over the past years to show that Epoetin alfa can correct anemia in cancer patients, at doses several times higher than those shown to be effective in renal patients. Anemia may result from the disease itself, the effect of concomitantly administered chemotherapeutic agents, or a combination of both. The condition often takes on the characteristics of the anemia of chronic disease (ACD). ACD is associated with erythroid hypoplasia of the bone marrow, a somewhat shortened circulating life of red cells and decreased bone marrow re-utilization of iron. If erythropoietin levels are measured, they are found to be within the normal range, but inappropriately low for the degree of anemia. Therefore the patient has a blunted erythropoietin response. About 50- 60% of anemic cancer patients receiving chemotherapy responded with a hemoglobin rise of at least 2 g/dL to Epoetin alfa therapy given three times weekly at a dose of 150 IU/kg over a period of 12 weeks (RI. Abels, KM. Larholt, KD. Krantz, and EC. Bryant, Proceedings of the Beijing Symposium, Alpha Medical Press, Dayton, Ohio, (1991) 121- 141). In a subsequent open-label dose titration study, doses up to 300 IU/kg, were sometimes required, demonstrating the relative resistance to the effect of Epoetin alpha in these patients. In three large community based studies, which enrolled over 7,000 patients with a wide range of non-myeloid malignancies, quality of life improvements correlated with the change in hemoglobin from baseline. (J. Glaspy, R. Bukowski, D. Steinberg, et al. /. Clin. Oncol. (1997) 15:1218-34; GD. Demetri, M. Kris, J. Wade, et al. J. Clin. Oncol. (1998) 16:3412-25; JL. Gabrilove, LH Einhorn, RB. Livingston, et al. American Society of Clinical Oncology Annual Meeting (1999) Abstract 2216). However, patients who did not experience an increase in hemoglobin over their baseline level did not show improvement in energy level, activity level, and overall quality of life. In each of the studies, approximately 35% of patients did not have an increase in their hemoglobin of 2 g/dl over baseline. Patients with progressive disease do not respond well to epoetin alfa, but the proportion (approximately 20%) of patients with progressive disease does not explain the overall rate of non-response of 35%. There is room to improve the overall response rate to erythropoietin in patients suffering from cancer.
Quality of life measures improve in association with increased hemoglobin levels in patients with cancer related anemia. Yet approximately one-third of patients fail to respond to erythropoietin treatment. Among the responders, quality of life measures improve significantly, but do not approach those seen in healthy subjects. TNF is implicated in many of the processes that complicate cancer therapy, including anemia, fatigue, and anorexia and cachexia. Cancer related fatigue has emerged as the most prevalent and bothersome symptom of cancer and its treatment. (RK. Portenoy, LM. Itri, The Oncologist (1999) 4:1-10). It is reported by 75%-100% of patients with cancer, depending on the patient's diagnosis, treatment and disease status. Cancer related fatigue is a multi-factorial problem. However, the treatment of cancer related anemia with erythropoietin has emerged as a safe and effective therapy to improve quality of life and relieve cancer related fatigue. Therefore, modulation of TNF simultaneously with EPO should increase the effectiveness of EPO for increasing hemoglobin levels, ameliorating anemia, and increasing the Quality of life measures in cancer patients.
Tumor necrosis factor (TNF) is part of a group of inflammatory cytokines that has been implicated in the pathologic changes seen in a number of diseases. TNF has been implicated as a contributing cause of fatigue, asthenia, anorexia and cachexia in a number of disorders. Elevated levels of TNF were associated with post-dialysis fatigue in one study (AW. Dreisbach, T. Hendrickson, LA. Beezhold, et al, Int. J. Artif. Organs (1998) 21:83-6) and were seen in patients with disorders of excessive daytime sleepiness in another. (AN. Vgontzas, DA. Papanicolaou, EO. Bixler, et al., J. Clin. Endrocrinol. Metab. (1997) 82:1313-6) TNF inhibition associated with the use of thalidomide has been exploited to treat the anorexia and cachexia of HIV disease (PAJ. Haslett. Semin. Oncol (1998) 25(6):53-7). The "procachetic" characteristics of some inflammatory cytokines, and TNF in particular, were recently reviewed (JM. Argiles, FJ. Lόpez- Soriano. Med Res Rev (1999) 19:223-48). The evidence of increased circulating TNF levels in cancer patients remains controversial; and, no single molecule has yet been identified as the mediator of cancer related cachexia. Still, TNF is an important candidate. An extensive review of asthenia echoes this sentiment. (DD. Von Hoff. Cancer Therapeutics (1998) 1:184-97).
A number of pre-clinical and clinical studies have demonstrated that TNF may act as a negative modulator of erythropoiesis. In murine models, there are conflicting data regarding the ability to abrogate TNF mediated anemia. (U. Clibon, L. Bonewald, J. Caro, et al. Exp Hematol. (1990) 18:458-41; CS. Johnson, CA. Cook, P. Furmanski, Exp Hematol. (1990) 18:109-13). In human studies, there is an association between circulating TNF levels and the need for increased doses of epoetin alfa in hemodialysis patients (M. Goiechea, J. Nartin, P. de Sequera, et al, Kidney Int (1998) 54:1337-43) and between local production of TNF in the bone marrow of patients with the anemia of chronic disease secondary to rheumatoid arthritis. (M. Jongen-Lavrencic, HR. Peeters, A. Wognum, et al, / Rheumatol. (1997) 24:1504-9). TNF blockade with a monoclonal antibody lead to improvement in the anemia of rheumatoid arthritis patients in one study. (D. Davis, PJ. Chrles, A. Potter, et al. BrJ Rheumatol (1997) 36:950-6).
The unmet need of enhanced EPO treatment for patients who suffer from cancer and fatigue has been met by the present invention that describes a method to treat patients with a combination of EPO and an anti- Tumor Necrosis Factor alpha agent.
SUMMARY OF THE INVENTION
The present invention provides a method for the treatment of a patient, comprising the steps, in any order or concurrently of administering a therapeutic amount of at least one anti-Tumor Necrosis Factor compound and a therapeutic amount of erythropoietin. The present invention further provides a method for the treatment of a patient having malignancies, comprising the steps, in any order or concurrently of administering a therapeutic amount of at least one anti-Tumor Necrosis Factor compound, a therapeutic amount of erythropoietin, and a therapeutic amount of an anti-tumor agent.
The methods of the present invention result in a reduced need for transfusion and a greater hemoglobin level in the patient. Further, the present invention provides that anemia may be successfully prevented or treated during the course of a chemotherapeutic regimen using the method of the present invention. The methods of the present invention result in patients that show greater quality of life, show improvement in physical performance and well being.
DETAILED DESCRIPTION OF THE INVENTION
The term Erythropoietin (EPO), as defined herein, refers to any molecule that specifically stimulates terminal differentiation of red blood cells from hematopoietic stem cells and stimulates production of hemoglobin. For example, but not to limit the scope of the current invention, EPO molecules may include small organic or inorganic molecules, synthetic or natural arnino acid peptides, purified protein from recombinant or natural expression systems, or synthetic or natural nucleic acid sequences, or any chemical derivatives of the aforementioned. The generally preferred, form of EPO is purified, recombinant EPO, distributed under the trademarks of EPREX® or ERYPO®. Epoetin alfa (EPREX®, ERYPO®) is a sterile, clear, colourless, aqueous solution for injection, that is provided in prefilled, single-use syringes containing either 4,000 or 10,000 IU epoetin alfa (a recombinant human erythropoietin) and 2.5 mg/mL human serum albumin in 0.4 mL (4,000 IU syringe) or 1.0 mL (10,000 IU syringe) of phosphate buffer.
The term "Erythropoietin" shall include those proteins that have the biological activity of human erythropoietin, as well as erythropoietin analogs, erythropoietin isoforms, erythropoietin mimetics, erythropoietin fragments, hybrid erythropoietin proteins, erythropoietin receptor agonists, renal erythropoietin, brain erythropoietin, oligomers and multimers of the above, homologues of the above, and muteins of the above, regardless of the biological activity of same, and further regardless of the method of synthesis or manufacture thereof including but not limited to naturally occurring, recombinant, synthetic, transgenic, and gene activated methods.
The term "anti-Tumor Necrosis Factor Compound" refers to drug products that decrease the amount of circulating, active TNFα. The compound may achieve this by decreasing the amount of cellular TNFα mRNA transcription, by decreasing mRNA translation into TNFα protein, or by decreasing cellular secretion of TNFα. Roy A. Black et al, from Immunex Corp., have discovered a compound that inhibits the enzyme that releases TNF from cell surfaces {Nature, 370, 218(1994)). This compound, called TNF-α protease enzyme inhibitor, curbs production of soluble TNF. Other suitable anti-TNFα compounds could work by increasing the rate of clearance or decreasing the amount of functional TNFα in circulation. Preferred anti-TNFα compounds are Thalidomide, Pentoxifylline, Infliximab, glucocorticoids, and Etanercept. The anti-TNFα compounds may be administered as combinations in order to maximize modulation of TNF since these agents acts as TNFα inhibitors at a different points in TNF synthesis and pharmacokinetic activity. Pentoxifylline inhibits TNF-α gene transcription (Doherty et al, Surgery (St. Louis), 110:192, 1991), while thalidomide enhances TNF-α m-RNA degradation (Moreira et al., 1993) and glucocorticoids such as dexamethasone inhibit TNF-α m-RNA translation (Han et al., J. Exp. Med. (1990) 172:391). Infliximab and etanercept act by reducing the amount of circulating, active TNFα.
Pentoxifylline (PENTOXIL™, Trental) decreases circulating TNFα at the Standard dose of 400 mg 3 times daily. Pentoxifylline inhibits TNF-α gene transcription (Doherty et al., Surgery (St. Louis), 110:192, 1991).
Glucocorticoids such as dexamethasone inhibit TNF-α m-RNA translation. Dexamethasone is administered orally, intramuscularly, or intravenously in the dose range of 8-40 mg (pediatric dose: 0.25-0.5 mg/kg). If given intravenously, dexamethasone should be given over 10-15 minutes, since rapid administration may cause sensations of generalized warmth, pharyngeal tingling or burning, or acute transient perianal and/or rectal pain. Methylprednisolone is also administered orally, intramuscularly, or intravenously at doses and schedules that vary from 40-500 mg every 6-12 hours for up to 20 doses.
Thalidomide (N-phthalidoglutarimide) may act by enhancing TNF-α m-RNA degradation (Shannon et al. (1990) Amer. Society for Microbiology Ann. Mtg., Abs. U- 53). Thalidomide is given by oral administration in the range of about 30 mg to 1500 mg per 24 hours, preferably 200 to 500 mg per 24 hours for an adult human weighting 70 kg.
REMICADE™ (Infliximab) is a monoclonal antibody that blocks the biological activity of circulating TNFα. Infliximab does not neutralize TNFβ (lymphotoxin α), a related cytokine that utilizes the same receptors as TNFα. Remicade is supplied as a sterile, white, lyophilized powder for intravenous infusion. Following reconstitution with 10 mL of Sterile Water for Injection, USP, the resulting pH is approximately 7.2. Each single- use vial contains 100 mg Infliximab, 500 mg sucrose, 0.5 mg polysorbate 80, 2.2 mg monobasic sodium phosphate and 6.1 mg dibasic sodium phosphate. No preservatives are present. Data from a study of single intravenous infusions of 1, 5, 10 or 20 mg/kg show a direct and linear relationship between the dose administered and the maximum serum concentration (Cmax) and area under the concentration-time curve. The volume of distribution at steady state (Vd), clearance and mean residence time are independent of the administered dose. Infliximab has a prolonged terminal half-life and is predominantly distributed within the vascular compartment. A single infusion of the recommended dose of 5 mg/kg resulted in a median Cmax of 118 μg/mL, a median Nd equal to 3.0 liters and a terminal half-life of 9.5 days.
EΝBREL™ (etanercept) is a dimeric fusion protein consisting of the extracellular ligand- binding portion of the human 75 kilodalton (p75) tumor necrosis factor receptor (TΝFR) linked to the Fc portion of human IgGl. The Fc component of etanercept contains the CH domain, the CR3 domain and hinge region, but not the CHI domain of IgGl. Etanercept is produced by recombinant DNA technology in a Chinese hamster ovary (CHO) mammalian cell expression system. It consists of 934 amino acids and has an apparent molecular weight of approximately 150 kilodaltons. ENBREL™ is supplied as a sterile, white, preservative-free, lyophilized powder for parenteral administration after reconstitution with 1 mL of the supplied Sterile Bacteriostatic Water for Injection, USP (containing 0.9% benzyl alcohol). Following reconstitution, the solution of ENBREL™ is clear and colorless, with a pH of 7.4 ± 0.3. Each single-use vial of ENBREL™ contains 25 mg etanercept, 40 mg mannitol, 10 mg sucrose, and 1.2 mg tromethamine. ENBREL™ is administered as a single subcutaneous (SC) injection.
It is readily apparent to those skilled in the art that a wide variety of malignancies are treatable according to the methods of the present invention.
Such tumor types include, but are not limited to, solid tumors, hematological tumors, sarcomas, carcinomas, neoplasms, as well as tumors of the breast, Non-Hodgkin's Lymphoma, myeloma, Hodgkin's lymphoma, leukemia, colon, rectal, colo-rectal, stomach, gastrointestinal, ovarian, lung, pancreas, and prostate.
It is readily apparent to those skilled in the art that a wide variety of non-platinum and platinum containing anti-tumor agents are suitable for use in the methods of the present invention. Platinum containing anti-tumor agents include, but are not limited to, cisplatin, and cis-dichlorodiammineplatinum. Non-platinum containing anti-tumor agents include, but are not limited to, cyclophosphamide, fluorouracil, epirubicin, methotrexate, vincristine, doxorubicin, bleomycin, and etoposide.
Quality of life is usually affected by malignancy, e.g., due to the underlying disease, effects of therapy and the psychological burden of coping with cancer. Fatigue is the most frequently reported symptom in cancer patients and impairs significantly their quality of life (ML. Winningham, LM. Nail, M. Barton Burke, et al., Oncol Nursing Forum (1994) 21: 23-34) Anemia contributes to fatigue and to the reduction in quality of life. The effects on fatigue (FACT An) and quality of life (ECOG performance score, Cancer Linear Analogue Scale, SF36) of early intervention and/or treatment of anemia with the combination therapy of EPO + anti-TNFα can be alleviated with concurrent use of EPO + anti-TNFα during chemotherapy.
The following examples illustrate the present invention without, however, limiting the same thereto.
EXAMPLE 1
A Randomized, Controlled Trial of Epoetin Alfa and Etanercept
OVERVIEW OF STUDY DESIGN
The study will be a double-blind, randomized, placebo controlled trial. After initial evaluation for eligibility, cancer patients with treatment related anemia will be assigned in a 1:1 randomization to receive either epoetin alfa plus placebo (Regimen A) or epoetin alfa plus etanercept (Regimen B). Patients who do not achieve an increase in hemoglobin of 1 g/dl without transfusion after four weeks of treatment will receive an increased dose epoetin alfa during weeks 5-8. Patients on Regimen A (epoetin alfa + placebo) who do not achieve an increase in hemoglobin of 2 g/dl over baseline by week 9 will be crossed over to Regimen B in an 8 week open label extension phase. Patients on Regimen B who do not achieve a 2 g/dl increase in hemoglobin over baseline by week 9 will be taken off study. Responding patients will continue on their assigned treatment and blinding will be maintained for 16 weeks. All patients must have received at least one course of chemotherapy and be scheduled to receive a minimum of 8 weeks of additional chemotherapy after study enrollment. Measures of Quality of Life (QoL), executive function, and weight will be obtained at study entry and at weeks 9 and 17 or at the time of withdrawal from the study. Hematologic parameters will be measured bi-weekly. All measurements should be made within 3 days prior to a subsequent course of chemotherapy. Assessments of tumor response will be made at enrollment, with subsequent assessments at weeks 9 and 17 or at the time of withdrawal from the study.
STUDY POPULATION
General Considerations
The specific inclusion and exclusion criteria for enrolling subjects in this study are described in the following sections. Exceptions to these inclusion/exclusion criteria should occur infrequently and should be discussed in advance with the Ortho Biotech medical monitor. If an exception is agreed upon and a subject is allowed to participate, the medical monitor will send confirmation to the site acknowledging the exception. This confirmation form or letter is to be kept with the case report forms (CRFs) both at the site and at the sponsor.
Inclusion Criteria
Subjects must satisfy the following criteria before entering the study:
• Histologically proven breast or lung cancer
• Prior chemotherapy < 1 course
• No evidence of progressive disease at study entry
• Treatment related anemia (must have started chemotherapy prior to study entry) with hemoglobin > 8 g/dl and < 12 g/dl
• Age > 18 years • Iron replete (transferrin saturation of > 20% and serum ferritin > 100 ng/ml)
• ECOG Performance status of 0-1 (see Appendix X) as determined by the investigator of designee,
• Life expectancy of at least 6 months
• Scheduled to receive at least an 8 weeks of chemotherapy
• Understands and signs informed consent
Exclusion Criteria
Subjects who meet any of the following criteria will be excluded from participating in the study:
• Prior chemotherapy > 1 course
• Use of biological response modifiers or cytokines within 30 days of study entry
• Diagnosis of rheumatoid arthritis, inflammatory bowel disease, or any disease for which etanercept may be primary therapy
• Chronic infection within 30 days of study entry
• Active infection at study entry
• Acute infection within 7 days of study entry
• Brain metastases
• Uncontrolled seizures
• Uncontrolled hypertension
• Transfusion within 2 months of study entry
• Iron deficiency or other nutritional anemia • Active hemolytic anemia
• Bleeding
• Known hypersensitivity to mammalian cell-derived products
• Known hypersensitivity to human albumin
• Prior treatment with epoetin alfa or any investigational erythropoietin within 8 weeks of study entry
• Significant medical disease other than cancer
• Second malignancy, other than basal cell carcinoma of the skin, within 5 years of study entry
• Use of corticosteroids other than for occasional use for anti-emesis or pre-treatment for a medication
• Advanced or poorly controlled diabetes mellitus
• Pregnant or lactating woman
RANDOMIZATION AND BLINDING
Overview
Randomization will be used to avoid bias in the assignment of patients to treatment, to increase the likelihood that subject attributes are evenly balanced across groups, and to enhance the validity of comparisons across treatment groups. Investigators and patients will be blinded to the identity of etanercept and placebo to enhance the validity of comparisons that are subject to observer bias or the placebo effect (e.g. - QoL end points). EPO will be given to all patients in an open label manner. Patients will be assigned randomly to each treatment regimen. PROCEDURES
Pre-Randomization Phase (Baseline)
The following evaluations and procedures are to be performed within 14 days prior to randomization unless otherwise specified. All laboratory tests and QoL and cognitive measures must be performed prior to the start of the first chemotherapy course after randomization.
• Patient demographics
• ECOG Performance Status as rated by the investigator or designee
• Tumor assessment using the NCI RECIST Criteria within 14 days prior to randomization
• Chemotherapy, radiation and surgery history
• Weight measured fully dressed except for shoes on the same scale to be used throughout the study
• Clinical laboratory tests
• Hemoglobin, hematocrit, MCV, absolute reticulocyte count, white blood cell count with differential and platelet count within 3 days prior to randomization
• Iron studies: ferritin, total iron binding capacity (TIBC), transferrin saturation, serum iron, transferrin receptor levels
• RBC ate and vitamin B 12 levels
• BUN, creatinine
• AST, ALT, total bilirubin
• Serum TNFα and anti-TNFα levels • FACT- An, Linear Analog Scale Assessment (LAS A), CLOX and EXIT 25 within 3 days prior to the administration of chemotherapy
• Transfusion history
• Signed informed consent
Subjects will be assigned to treatment groups based on a computer- generated randomization schedule. The randomization will be balanced by using permuted blocks and will be stratified by center and type of cancer. Based on this randomization code, the study drug will be packaged and labeled for each subject. [Subject numbers] [Medication code numbers] will be preprinted on the study drug labels and assigned sequentially as subjects qualify for the study and are randomized to treatment.
To maintain the blind, the etanercept or placebo drug container will have a two-part, tear-off label with directions for use and other information on each part. The tear-off section of the label will contain a concealed area identifying the study drug (e.g., active or placebo) and will be removed and attached to the subject's CRF when the drug is dispensed. The second part of the label will remain affixed to the study drug container and will contain all identifying information except for the identity of the drug contained. The study drugs will be identical in appearance and will be packaged in identical containers.
Under normal circumstances, the blind should not be broken. The blind should be broken only if specific emergency treatment would be dictated by knowing the treatment status of the subject. In such cases, the investigator must contact the sponsor. If the investigator is unable to contact the sponsor, the investigator may in an emergency determine the identity of the treatment by exposing the concealed area of the label attached to the subject's CRF. Individual code breaks by the investigator will normally result in withdrawal of the subject from the trial. The date, time, and reason for the unblinding must be documented on the appropriate page of the CRF (Study Completion Information) and the sponsor must be informed as soon as possible.
The randomization schedule will not be revealed to study subjects, parents or guardians, investigators and clinical staff, or site managers until all subjects have completed the double-blind phase of the trial.
To allow for the cross-over of non-responding patients randomized to EPO alone, the investigator will call the study sponsor with the week 9 hemoglobin results. If a patient is a non-responder, the study sponsor will inform the investigator of the patient's treatment assignment. Patients who do not respond to EPO + ETAN (Regimen B) will proceed to Off Study Evaluation procedures. Non-responding patients treated on EPO + placebo (Regimen A) will be treated with EPO + ETAN for an additional 8 weeks in an open label extension phase. Non- responding patients treated with EPO+ETAN who subsequently respond indicate synergistic action of the anti-tumor necrosis factor agent with EPO Comparison of the response of patients treated with EPO or with EPO+ETAN will further indicate synergistic action of the combination therapy.
DOUBLE-BLIND TREATMENT PHASE
During this phase patients should remain blinded to study-related test results prior to the completion of each set of QoL and cognitive measures. The treating physician may provide laboratory results as soon as the forms are completed for that visit.
For patients receiving every three week chemotherapy, evaluations should be completed at least 3 days following the end of the last dose of chemotherapy. For patients receiving every four week chemotherapy, evaluations should be completed within 3 days prior to the scheduled chemotherapy course.
Week 3
• ECOG Performance Status as rated by the investigator or designee
Weight measured fully dressed except for shoes on the same scale to be used throughout the study
Clinical laboratory tests
• Hemoglobin, hematocrit, MCV, absolute reticulocyte count, white blood cell count with differential and platelet count
• Iron studies: ferritin, total iron binding capacity (TIBC), transferrin saturation, serum iron, transferrin receptor levels
• Serum TNFα and anti-TNFα levels
Transfusion history
Week 5
• ECOG Performance Status as rated by the investigator or designee
• Chemotherapy, radiation and surgery history
• Weight measured fully dressed except for shoes on the same scale to be used throughout the study
Clinical laboratory tests • Hemoglobin, hematocrit, MCV, absolute reticulocyte count, white blood cell count with differential and platelet count Iron studies: ferritin, total iron binding capacity (TIBC), transferrin saturation, serum iron, transferrin receptor levels
• Serum TNFα and anti-TNFα levels
• Transfusion history
Week 9 This evaluation will serve as the Off Study Evaluation for Non-responders on Regimen B
• ECOG Performance Status as rated by the investigator or designee
Tumor assessment using the NCI RECIST Criteria (see Appendix X) within 14 days
Chemotherapy, radiation and surgery history
Weight measured fully dressed except for shoes on the same scale to be used throughout the study
Clinical laboratory tests
• Hemoglobin, hematocrit, MCV, absolute reticulocyte count, white blood cell count with differential and platelet count
• Iron studies: ferritin, total iron binding capacity (TIBC), transferrin saturation, serum iron, transferrin receptor levels
• RBC Mate and vitamin B 12 levels
• BUN, creatinine
• AST, ALT, total bilirubin • Serum TNFα and anti-TNFα levels
• FACT-An, Linear Analog Scale Assessment (LASA), CLOX and EXIT 25 within 3 days prior to the administration of chemotherapy
• Transfusion history
Responding patients on both regimes will continue on their assigned treatment and be evaluated through week 17 as follows.
Week 13
ECOG Performance Status as rated by the investigator or designee
Clinical laboratory tests
• Hemoglobin, hematocrit, MCV, absolute reticulocyte count, white blood cell count with differential and platelet count
• Iron studies: ferritin, total iron binding capacity (TIBC), transferrin saturation, serum iron, transferrin receptor levels
• Serum TNFα and anti-TNFα levels
Transfusion history
Determination of response: non-responders already on 60,000 U/week proceed to Off Study Evaluation
Week 17
Proceed to Off Study Evaluation OPEN LABEL EXTENSION PHASE FOR NON-RESPONDERS IN REGIMEN A
Week 13
• ECOG Performance Status as rated by the investigator or designee
• Clinical laboratory tests
• Hemoglobin, hematocrit, MCV, absolute reticulocyte count, white blood cell count with differential and platelet count
• Iron studies: ferritin, total iron binding capacity (TIBC), transferrin saturation, serum iron, transferrin receptor levels
• Serum TNFα and anti-TNFα levels
• Transfusion history
Week 17
• Proceed to Off Study Evaluation
OFF STUDY EVALUATION
• ECOG Performance Status as rated by the investigator or designee
• Tumor assessment using the NCI RECIST Criteria within 14 days
• Chemotherapy, radiation and surgery history
• Weight measured fully dressed except for shoes on the same scale to be used throughout the study
• Clinical laboratory tests • Hemoglobin, hematocrit, MCV, absolute reticulocyte count, white blood cell count with differential and platelet count
• Iron studies: ferritin, total iron binding capacity (TIBC), transferrin saturation, serum iron
• BUN, creatinine
• AST, ALT, total bilirubin
• Serum TNF and anti-TNF levels
• FACT-An, Linear Analog Scale Assessment (LASA), CLOX and EXIT 25 within 3 days prior to the administration of chemotherapy
• Transfusion history
DOSAGE AND ADMINISTRATION
Regimen A (EPO + Placebo)
EPO will be started at a dose of 40,000 U per week given SC, without regard to the timing of chemotherapy. If at the evaluation prior to the week 5 dose the hemoglobin has not increased by at least 1 g/dl over the baseline value, the dose of EPO will be increased to 60,000 U per week for weeks 5-8. If the hemoglobin increases by at least 2 g/dl over baseline at the time of the week 9 evaluation, the patient will be considered a responder. Responders will continue to receive the dose of EPO given during weeks 5-8 until week 16. Non-responders will be offered the opportunity to receive EPO + ETAN in an 8 week open label extension.
Placebo injections will be given at a dose of 1 ml SC, at a site separate from the EPO site, on days 1 and 4 of each week (day 1 is counted as the day the EPO and placebo are given together). The patient should receive two doses of placebo during week 8, prior to the week 9 evaluation. Responders will continue to receive placebo in a blinded manner through week 16. Two placebo injections should be given in week 16, prior to the off study evaluation. Non-responders will be offered the opportunity to receive EPO + ETAN in an 8 week open label extension.
Regimen B (EPO + ETAN)
EPO will be started at a dose of 40,000 U per week given SC, without regard to the timing of chemotherapy. If at the evaluation prior to the week 5 dose the hemoglobin has not increased by at least 1 g/dl over the baseline value, the dose of EPO will be increased to 60,000 U per week for weeks 5-8. If the hemoglobin increases by at least 2 g/dl over baseline at the time of the week 9 evaluation, the patient will be considered a responder. Responders will continue to receive the dose of EPO given during weeks 5-8 until week 16. Non-responders will proceed to the Off Study Evaluation at week 9.
ETAN injections will be given at a dose of 25 mg (1 ml) SC, at a site separate from the EPO site, on days 1 and 4 of each week (day 1 is counted as the day the EPO and ETAN are given together). The patient should receive two doses of ETAN during week 8, prior to the week 9 evaluation. Responders will continue in ETAN through week 16. Two ETAN injections should be given during week 16 and prior to the off study evaluation. Non-responders will proceed to the off study evaluation at week 9.
Open label phase for non-responders on Regimen A
To assess the effectiveness of ETAN in overcoming resistance to EPO, placebo will be replaced with ETAN at week 9 for patients who are non-responders in Regimen A. The following doses of EPO and ETAN will be given during weeks 9-16 in these patients: (1) Patients on an EPO dose of 60,000 U/week at week 8 will continue on that dose for weeks 9-12. ETAN will be given at a dose of 25 mg SC, at a site separate from the EPO site, on days 1 and 4 of each week (day 1 is counted as the day the EPO and placebo are given together). ETAN will be started on the day of the week 9 EPO dose and continue until 2 doses are given during week 12. If the evaluation at week 13 indicates at least a 1 g/dl increase in hemoglobin over the week 9 value or a 2 g/dl increase over the baseline value, then the patient will be considered a responder and continue on study through week 16. ETAN should continue until 2 doses are given in week 16. The patient then will proceed to the Off Study Evaluation. If the patient is a non-responder at week 13, then the patient will proceed to the Off Study Evaluation.
(2) Patients on an EPO dose of 40.000 U/week at week 8 will continue that dose through for weeks 9-12. ETAN will be given at a dose of 25 mg SC, at a site separate from the EPO site, on days 1 and 4 of each week (day 1 is counted as the day the EPO and placebo are given together). The first dose of ETAN will be given on the day of the week 9 EPO dose and continue until 2 doses are given during week 12. If the evaluation at week 13 indicates at least a 1 g/dl increase in hemoglobin over the week 9 value, or a 2 g/dl increase over the baseline value, then the patient will continue on study through week 16 at the same dose of EPO. ETAN should continue until 2 doses are given during week 16 EPO. The patient then will proceed to the Off Study Evaluation. If the patient is a non-responder at week 13, then the EPO dose will be increased to 60,000 U/week and the patient will continue on study through week 16. ETAN should continue until 2 doses are given during week 16 EPO. The patient then will proceed to the Off Study Evaluation.
Dose adjustment of EPO
If the hemoglobin is > 13 g/dl on 2 consecutive evaluations, EPO should be withheld until the hemoglobin drops to 12 g/dl. EPO should then be resumed at 75% of the last dose given before discontinuation. If the hemoglobin increases by > 1.3 g/dl in a 2 week period, EPO should be continued at 75% of the previous dose.
Patients who develop sepsis
All study medication should be discontinued in patients who develop sepsis while on study. The Study Completion/Early Withdrawal form should be completed and the study sponsor informed immediately.
Administration of EPO
Epoetin alfa 40,000 U/ml should be brought to room temperature and drawn up into a plastic syringe immediately prior to administration by SC injection according to standard techniques. The EPO and ETAN/placebo may be given at the same time on day 1 of each week, but should be administered at separate sites. Each vial of EPO should be used only once. The maximum injection volume per site is 2 ml.
Administration of ETAN
Study medication (ETAN/placebo) should be reconstituted with 1 ml of the supplied sterile bacteriostatic water for injection, USP (0.9% benzyl alcohol). The diluent should be slowly injected into the vial. Some foaming may occur. To avoid excessive foaming, do not shake or agitate vigorously. Swirl gently until dissolution occurs, usually over less than 5 minutes. The solution should be clear and colorless. The medication should then be drawn up into a plastic syringe and administered SC as soon as possible after reconstitution. New injections should be given at least 1 inch from an old site and never into areas where the skin is tender, bruised, hard or red. Prior to injection of the study medication, Part 2 of the two-part vial label is to be attached to the subject's case report form after entry of the subject's initials and number, and the date of study medication administration. Each vial of study medication should be used only once. Iron
Iron deficiency may develop during the use of EPO and may limit the efficacy of EPO if left untreated. If laboratory evidence of iron deficiency develops during the study, the patient should be given 150-200 mg of elemental iron per day. The appropriate formulation of the iron supplement is left to the discretion of the treating physician.
CONCOMITANT THERAPY
Patients may be transfused with packed red blood cells when judged to be necessary by the physician of record. A hemoglobin level should be obtained at the time the type and cross-match specimen is drawn. The pre-transfusion hemoglobin value, along with the number of units used and the total volume transfused, should be recorded in the case report form.
All concomitant therapy administered during the study will be recorded on the case report form. The use of androgens or hematinic agents (folate, B12) other than iron is prohibited during the study. The use of corticosteroids, other than occasional use as a pre-medication or an anti-emetic is prohibited during the study.
The sponsor must be notified in advance (or as soon as possible thereafter) of any instances in which prohibited therapies are administered.
EFFICACY EVALUATIONS
The primary endpoints to be evaluated in this study are as follows:
1. a comparison of the proportion of patients in each regimen who achieve a response by week 9
2. a comparison of the change in scores on QoL measures in each regimen between Baseline, week 9 and Off Study in responders, and using an intent to treat analysis of all patients 3. the proportion of non-responders in Regimen A who show a response at week 13 and week 17 when crossed over to the EPO + ETAN arm (Regimen B)
4. a comparison of the proportion of patients in each regimen who experience significant weight loss.
EFFICACY AND DISEASE STATUS EVALUATION CRITERIA
♦ Response is defined as a 2g/dl or greater increase in the hemoglobin when compared to the Baseline value.
♦ Significant weight loss will be defined as Off Study Weight divided by Baseline Weight less than 0.90.
♦ Disease status will be defined according the current version of the RECIST Criteria issued by the National Cancer Institute.
The following measurement tools will be used in this study:
• Linear Analog Scale Assessment
• Fact-An
• CLOX: An Executive Clock Drawing Task
• EXIT 25: The Executive Interview.

Claims

WHAT IS CLAIMED IS:
1. A method for treating a patient, comprising the steps, in any order or concurrently: a) administering a therapeutic amount of at least one anti-TNF alpha compound; and b) administering a therapeutic amount of erythropoietin.
2. The method of claim 1 wherein the patient has at least one malignancy.
3. The method of claim 1 wherein only one anti-TNF alpha compound is administered.
4. The method of claim 1 wherein two or more anti-TNF alpha compounds are administered.
5. The method of claim 1 wherein the anti-Tumor Necrosis Factor compound decreases circulating, active TNFα by a mechanism selected from a group consisting of decreasing TNFα mRNA transcription, increasing TNFα mRNA degradation, inhibiting TNFα mRNA translation, decreasing cellular secretion of TNFα protein, and reducing circulating TNFα protein.
6. The method of claim 1 wherein the anti-Tumor Necrosis Factor compound is selected from the group consisting of Thalidomide, Pentoxifylline, Infliximab, Glucocorticoids, and Etanercept.
7. A method for treating a patient having a malignancy, comprising the steps, in any order or concurrently: a) administering a therapeutic amount of at least one anti-TNF alpha compound; b) administering a therapeutic amount of erythropoietin; and c) administering a therapeutic amount of an anti-tumor agent.
8. The method of claim 7 wherein only one anti-TNF alpha compound is administered.
9. The method of claim 7 wherein two or more anti-TNF alpha compounds are administered.
10. The method of claim 7 wherein the anti-Tumor Necrosis Factor compound decreases circulating, active TNFα by a mechanism selected from a group consisting of decreasing TNFα mRNA transcription, increasing TNFα mRNA degradation, inhibiting TNFα mRNA translation, decreasing cellular secretion of TNFα protein, and reducing circulating TNFα protein.
11. The method of claim 7 wherein the anti-Tumor Necrosis Factor compound is selected from a group consisting of Thalidomide, Pentoxifylline, Infliximab, Glucocorticoids, and Etanercept.
PCT/US2001/024384 2001-08-02 2001-08-02 Erythropoietin and anti-tumor necrosis factor alpha combination therapy WO2003012432A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
PCT/US2001/024384 WO2003012432A1 (en) 2001-08-02 2001-08-02 Erythropoietin and anti-tumor necrosis factor alpha combination therapy
CA002456177A CA2456177A1 (en) 2001-08-02 2001-08-02 Erythropoietin and anti-tumor necrosis factor alpha combination therapy
JP2003517575A JP2004537569A (en) 2001-08-02 2001-08-02 Erythropoietin and anti-tumor necrosis factor alpha combination therapy
EP01959479A EP1423693A4 (en) 2001-08-02 2001-08-02 Erythropoietin and anti-tumor necrosis factor alpha combination therapy

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2001/024384 WO2003012432A1 (en) 2001-08-02 2001-08-02 Erythropoietin and anti-tumor necrosis factor alpha combination therapy

Publications (1)

Publication Number Publication Date
WO2003012432A1 true WO2003012432A1 (en) 2003-02-13

Family

ID=21742749

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/024384 WO2003012432A1 (en) 2001-08-02 2001-08-02 Erythropoietin and anti-tumor necrosis factor alpha combination therapy

Country Status (4)

Country Link
EP (1) EP1423693A4 (en)
JP (1) JP2004537569A (en)
CA (1) CA2456177A1 (en)
WO (1) WO2003012432A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007513164A (en) * 2003-12-02 2007-05-24 セルジーン・コーポレーション Methods and compositions for the treatment and management of abnormal hemoglobinosis and anemia
WO2008065387A2 (en) * 2006-11-27 2008-06-05 Bionature E.A. Limited Potentiation of erythropoietin (epo) action by membrane steroid receptor agonists

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007139150A1 (en) * 2006-05-30 2007-12-06 The University Of Tokushima ANTI-INFLUENZA VIRAL AGENT COMPRISING TNF-α INHIBITOR

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL365664A1 (en) * 2000-08-02 2005-01-10 Ortho-Mcneil Pharmaceutical, Inc. Improved anti-viral and anti-tumor chemotherapy by administration of erythropoeitin
US7087224B2 (en) * 2000-10-31 2006-08-08 Amgen Inc. Method of treating anemia by administering IL-1ra

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
ARRIETA O. ET AL.: "Protective effect of pentoxifylline plus thalidomide against septic shock in mice", INT. J. EXP. PATH., vol. 80, no. 1, February 1999 (1999-02-01), pages 11 - 16, XP002906600 *
BLADE J. ET AL.: "Treatment approaches for relapsing and refractory multiple myeloma", ACTA ONCOL., vol. 39, no. 7, 2000, pages 843 - 847, XP002906599 *
MAKONKAWKEYOON S. ET AL.: "Thalidomide inhibits the replication of human immunodeficiency virus type 1", PROC. NATL. ACAD. SCI. USA, vol. 90, July 1993 (1993-07-01), pages 5974 - 5978, XP000938703 *
MARROTT J.B. ET AL.: "A double-blind placebo-controlled phase II trial of thalidomide in asymptomatic HIV-positive patients: Clinical tolerance and effect on activation markers and cytokines", AIDS RES. HUM. RETROVIRUSES, vol. 13, no. 18, 1997, pages 1625 - 1631, XP002906601 *
PEUCKMANN V. ET AL.: "Potential novel uses of thalidomide focus on palliative care", DRUGS, vol. 60, no. 2, August 2000 (2000-08-01), pages 273 - 292, XP002906602 *
REGULIER E. ET AL.: "Continuous delivery of human and mouse erythropoietin in mice by genetically engineered polymer encapsulated myoblasts", GENE THERAPY, vol. 5, no. 8, August 1998 (1998-08-01), pages 1014 - 1022, XP001013508 *
See also references of EP1423693A4 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007513164A (en) * 2003-12-02 2007-05-24 セルジーン・コーポレーション Methods and compositions for the treatment and management of abnormal hemoglobinosis and anemia
WO2008065387A2 (en) * 2006-11-27 2008-06-05 Bionature E.A. Limited Potentiation of erythropoietin (epo) action by membrane steroid receptor agonists
WO2008065387A3 (en) * 2006-11-27 2008-07-31 Bionature E A Ltd Potentiation of erythropoietin (epo) action by membrane steroid receptor agonists

Also Published As

Publication number Publication date
EP1423693A1 (en) 2004-06-02
CA2456177A1 (en) 2003-02-13
EP1423693A4 (en) 2006-06-07
JP2004537569A (en) 2004-12-16

Similar Documents

Publication Publication Date Title
CA2418531C (en) Therapeutic methods for treating subjects with a recombinant erythropoietin having high activity and reduced side effects
MXPA06014421A (en) Method of administering and using vegf inhibitors for the treatment of human cancer.
RU2225221C2 (en) Pharmaceutical compositions of erythropoietin
JP5737826B2 (en) Treatment of anemia by inhibition of VEGF
KR100478573B1 (en) The use of erythropoietin and iron preparations for producing pharmaceutical combination preparations for treating rheumatic diseases
WO2000067769A1 (en) Enhanced survival of cancer patients treated with erythropoietin and antitumor agents
Hellström et al. Treatment of myelodysplastic syndromes with recombinant human erythropoietin
KR20030043924A (en) Improved anti-viral and anti-tumor chemotherapy by administration of erythropoietin
Morreale et al. Clinical and economic comparison of epoetin alfa and darbepoetin alfa
Kuan et al. A randomized double blind control trial comparing filgrastim and pegfilgrastim in cyclophosphamide peripheral blood hematopoietic stem cell mobilization
TW201517913A (en) Methods of treating iron deficiency
EP3868403A1 (en) Taci-fc fusion protein and use thereof
US20120082642A1 (en) Reversal of Adult Onset Disorders with Granulocyte-Colony Stimulating Factors
EP3845550A1 (en) Composition for accelerating cell proliferation comprising erythropoietin-derived peptide
IE61008B1 (en) Composition for increasing the hematocrit of a normal mammal
WO2003013568A1 (en) Cytokine modulation therapy
WO2003012432A1 (en) Erythropoietin and anti-tumor necrosis factor alpha combination therapy
KR19990066997A (en) How to move hematopoietic stem cells
Foa Erythropoietin: clinical applications
Hudson et al. Darbepoetin alfa, a new therapy for the management of anemia of chronic kidney disease
Vilan et al. Pure red cell aplasia: successful treatment with cyclophosphamide
EP4292603A1 (en) Method for treating iga nephropathy with taci-fc fusion protein
Nayak et al. Novel Approaches in Erythropoietin: A Review
Wick et al. Disturbances of Iron Distribution Diagnosis and Therapy
US20080249001A1 (en) Agents that alleviate side-effects caused by chemotherapy agents

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BY BZ CA CH CN CO CR CU CZ DE DM DZ EC EE ES FI GB GD GE GH HR HU ID IL IN IS JP KE KG KP KR LC LK LR LS LT LU LV MA MD MG MN MW MX MZ NO NZ PL PT RO RU SE SG SI SK SL TJ TM TR TT TZ UA UZ VN YU ZA

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ UG ZW AM AZ BY KG KZ MD TJ TM AT BE CH CY DE DK ES FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW MR NE SN TD TG

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

Ref document number: 2003517575

Country of ref document: JP

Ref document number: 2456177

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2001959479

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2001959479

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642