HK1084862B - Use of omega interferon in the manufacturing of a medicament for treating viral disease in a warm-blooded animal subject - Google Patents
Use of omega interferon in the manufacturing of a medicament for treating viral disease in a warm-blooded animal subject Download PDFInfo
- Publication number
- HK1084862B HK1084862B HK06100312.5A HK06100312A HK1084862B HK 1084862 B HK1084862 B HK 1084862B HK 06100312 A HK06100312 A HK 06100312A HK 1084862 B HK1084862 B HK 1084862B
- Authority
- HK
- Hong Kong
- Prior art keywords
- interferon
- omega
- administered
- omega interferon
- treatment
- Prior art date
Links
Description
Technical Field
The field of the invention is the use of omega interferon for the treatment of viral, infectious, immunological or proliferative diseases.
Background
Interferons are a group of endogenous peptides produced in response to a variety of infectious or immune diseases. Endogenous interferons have antiviral, infection immunomodulatory or antiproliferative activity. Alpha and beta interferons are known as type I interferons and have been shown to bind to a common receptor, the so-called alpha-beta receptor. Exogenous interferons, such as recombinant alpha interferon (of various subtypes) or recombinant consensus interferon (consensus) have been demonstrated to be useful in the treatment of, for example, viral hepatitis c and certain cancers. A small percentage of patients treated with alpha or consensus interferon for a period of months may no longer be positive for a hepatitis C virus ribonucleic acid blood test. Treatment with interferon may stabilize or reduce the volume of certain cancers.
Such treatment may include monotherapy with interferon alone, or interferon may be combined with an adjuvant. Exogenous interferon-beta (of various subtypes) has been shown to be useful as monotherapy in the treatment of multiple sclerosis. Exogenous recombinant gamma interferon has been shown to be useful as a monotherapy for the treatment of chronic granulomatous disease, and recently it has been proposed to be useful in the treatment of certain lung diseases. Some interferons are chemically modified by the addition of polyethylene polymers and as a result may have enhanced antiviral activity or patient acceptance.
Co-agents administered in combination with interferon may enhance the effectiveness of treatment with interferon. For example, ribavirin is a non-peptide small molecule that, among its activities, is known to inhibit inosine monophosphate dehydrogenase and has antiviral and immunomodulatory activities. For example, the addition of ribavirin (or other inosine monophosphate dehydrogenase inhibitor) to interferon-alpha may increase the long-term response rate in certain subpopulations of hepatitis c patients. Other co-agents of interferon-alpha may also be useful in certain clinical situations, interleukin-2 analogs or derivatives, histamine analogs or derivatives; a monoclonal antibody; a polyclonal antibody; or any combination thereof.
However, these currently available antiviral or immunomodulatory therapeutic agents are not without limitation. For example, the long-term success rate of hepatitis c treatment is estimated as follows: interferon-alpha alone (≈ 10-15%); consensus interferon alone (≈ 10-15%); pegylated alpha interferon alone (≈ 20-25%); interferon-alpha is associated with ribavirin (≈ 30-40%); and interferon alpha plus histamine related compounds (. apprxeq.30-40%). There is evidence that treatment with interferon-alpha in combination with ribavirin or a histamine analog can induce a response in patients who exhibit an incomplete response to interferon-alpha alone. Consensus interferon at certain dose levels has been reported to induce responses in patients who fail to achieve sustained results with lower doses of alpha interferon.
However, in a large percentage of patients, there is no significant response to either interferon-alpha or interferon-consensus, whether or not it is combined with another agent (primary viral resistance). In addition, a significant proportion of patients who do respond initially to the disease do not have a lasting response (secondary resistance) after cessation of drug therapy. Of those patients who do not respond to interferon alpha, most also do not respond to subsequent treatment with consensus interferon. The reasons for primary or secondary resistance are not completely understood, but may involve significant changes in blood levels of interferon, the production of antibodies to interferon, intracellular changes that limit interferon-induced responses, or genetic characteristics (or other changes) of the virus or the patient or both.
Moreover, due to adverse side effects, not all patients are resistant to interferon therapy (either alone or in combination with adjunctive agents). There are significant limitations in the administration of alpha, beta, consensus, gamma, leukocyte and tau interferon, as the effectiveness of interferon depends, for example, on the dose administered, and any limitations in administration due to adverse side effects have further negative clinical consequences: the medical utility of interferons is reduced by the inability to administer higher and more effective doses due to the adverse side effects of limiting the dose.
For example, the side effects of interferon-alpha include the following (as listed in the current FDA label for alpha-2 c): headache, fever, fatigue, myalgia, leukopenia, neutropenia, thrombocytopenia, arthralgia, rigidity, irritability, nausea, vomiting. Some of the side effects caused by interferons, even at low doses, can be serious, life-threatening or even fatal. These effects include, in particular, severe infections, seizures and depression. Suicidal ideation or actual suicide is also associated with prolonged application of currently marketed interferons.
The occurrence of such side effects can often result in reduced interferon administration or require complete cessation of treatment. In either case, medical utility is reduced or lost altogether. For example, in a recent study comparing pegylated interferon ALFA-2a to non-pegylated interferon ALFA-2a in 531 patients, more than 25% of patients in each treatment group had to be dose-reduced or discontinued (see PEGINTERFERON ALFA-2a IN PATIENTSWITH CHRONIC HEPTATIS C. Zeuzem S, Feinman SV, RasenackJ et al, New Engl J Med 2000; 343; 1666-72). It is noteworthy, however, that in this particular comparative test in hepatitis patients, due to the better pharmacokinetics, the viral response rate at the end of pegylated interferon alfa treatment was about 68% and for non-pegylated interferon alfa about 27%. However the response rate as judged by viral response and liver enzyme decline at the end of treatment was even lower, 42% and 25%, respectively.
Thus, while current interferon administration provides a useful therapeutic modality for certain diseases, significant problems with tolerance and overall therapeutic success persist. We have now found that omega interferon provides a solution to these problems.
Summary of The Invention
One aspect of the invention is a method of treating an immunological, proliferative, or infectious disease in a warm-blooded animal. The method comprises administering to the animal omega Interferon (IFN) at a dose and activity sufficient to induce a therapeutic response in the animal for the disease state being treated, the dose and activity for the disease state being treated being higher than would be well tolerated based on non-omega IFN data.
Another aspect is the method, wherein omega interferon is administered to such an animal, optionally in combination with a therapeutically effective amount of at least one adjunctive therapeutic agent, for a length of time subject to tolerance of the omega interferon by the animal, monitoring the level of a disease marker in the animal during the administration, and continuing administration of the omega interferon for a length of time subject to a continuing beneficial change in the level of the disease marker.
Another aspect of the invention is a preparation for use in the treatment of an immunological, proliferative, or infectious disease in a warm-blooded animal subject, the preparation comprising 1) omega interferon in a form suitable for administering to the subject a therapeutically effective amount of omega IFN to induce the desired therapeutic response, and 2) instructions for use of the omega IFN to administer the omega IFN at a dose and activity higher than would be well tolerated based on non-omega IFN data for the disease state being treated.
Another aspect of the invention is a method of preparing an omega interferon based preparation for use in the treatment of an immunological, proliferative or infectious disease in a warm-blooded animal subject, the method comprising providing omega IFN as a composition suitable for administration to the subject in a therapeutically effective dose, and combining the omega IFN so provided with instructions for administration of the omega IFN for use in such a disease.
Another aspect of the invention is the use of an omega Interferon (IFN) for the manufacture of a medicament for the treatment of an immunological, proliferative or infectious disease in a warm-blooded animal. The medicament is for administering to the animal a dose and activity sufficient to induce a therapeutic response in the animal for the disease being treated, the dose and activity for the disease state being treated being higher than would be well tolerated based on non-omega IFN data.
Drawings
FIG. 1: the graph shows the relationship between Hepatitis C Virus (HCV) ribonucleic acid (RNA) levels over time for patients with genotype 1 treated with omega interferon.
Detailed Description
Definition of
The term "interferon alpha" (sometimes referred to as "alpha") or "interferon alpha" refers to a family of highly homologous species-specific proteins (i.e., glycoproteins) that are known in the art to inhibit viral replication and cellular proliferation and to modulate immune responses. Exemplary suitable alpha interferons include recombinant Interferon alpha-2 b such as Intron-A Interferon available from Schering Corporation, Kenilworth, N.J., recombinant Interferon alpha-2 a such as Roferon Interferon available from Hoffmann-La Roche, Nutley N.J., recombinant Interferon alpha-2C such as Berofor alpha-2 Interferon available from Boehringer Ingelheim Pharmaceutical, Inc., Ridgefield, Conn., Interferon alpha-n 1, a purified mixture of natural alpha interferons such as Sufermion available from Sumitomo, Japan, or from Glaxo-Welle Ltd, London, Greatatain Brillon Interferon alpha-n 1(INS), or natural alpha-Interferon alpha-n 1(INS) such as those described in U.S. Pat. Nos. 32 and 32, U.S. Puff, and Califon 3936, and mixtures of natural alpha-n interferons such as those commercially available from Califon, Inc., and Califon 3, Inc., as blends of natural alpha-n Interferon alpha-n 2, such as Interferon alpha-n 2, and its derivatives, such as Interferon alpha-n 3, Interferon alpha-n, a purified mixture of natural Interferon alpha-n, such as described in U., norwalk, conn., under the trade name Alferon.
The term "interferon beta" or "beta IFN" refers to the known in the prior art can induce to viral antigen resistance protein (i.e. glycoprotein). Examples are described in U.S. Pat. Nos. 4,820,638 and 5,795,779, and include equivalents or derivatives thereof.
The term "interferon gamma" or "gamma interferon" or "gamma IFN" refers to a protein (i.e., glycoprotein) that induces resistance to certain viral antigens and is described in U.S. patent No.4,727,138; 4,762,791, respectively; 4,845,196, respectively; 4,929,554, respectively; 5,005,689, respectively; 5,574,137, respectively; 5,602,010, respectively; and 5,690,925, or equivalents or derivatives thereof.
The term "interferon tau" or "tau interferon" or "tau-IFN" refers to a protein (i.e., glycoprotein) that induces resistance to certain viral antigens and is described in U.S. Pat. Nos. 5,939,286 and 6,204,022, or equivalents or derivatives thereof.
[0053] The term omega interferon or omega-interferon as used herein refers to the species-specific proteins (i.e., glycoproteins) described in U.S. patent nos. 5,120,832 and 5,231,176. It can inhibit viral replication, cell proliferation and modulate immune response, even in cases or patients where interferon alpha is ineffective or of limited effectiveness. Omega interferon is a naturally occurring interferon with limited homology to alpha interferon (about 65%) and even lower homology to beta interferon (about 35%). Thus, omega interferon is structurally distinctive. Nonetheless, as noted above, omega interferon appears to bind to the "alpha-beta receptor", as judged by in vitro testing. Recombinant omega interferon is produced in mammalian cells using genetic engineering techniques. We have found that antibodies produced in animals exposed to interferon-alpha do not cross-react with interferon-omega, i.e. interferon-omega is immunologically distinctive.
Throughout the specification and claims, IFN and interferon are used interchangeably.
Non-omega interferon refers to an IFN that is not an omega IFN or a combination of IFNs that are not omega IFNs. Non-omega interferons will include alpha IFN, beta IFN, gamma IFN, tau IFN, leukocyte-derived IFN and the like.
Method of treatment
One aspect of the invention is a method of treating an immunological, proliferative, or infectious disease in a warm-blooded animal subject with an omega IFN. The dose and activity of omega IFN administered is sufficient to induce a therapeutic response in the animal for the disease state being treated. Surprisingly, the dose and activity administered for the disease state being treated will be well tolerated for omega IFNs over non-omega IFN-based data. Generally, the units of activity per microgram (μ g) of omega IFN exceed the units of activity per microgram (μ g) of non-omega IFN by more than a factor of 1 up to about 3, preferably by a factor of about 2. Thus, when the dose is administered to the animal for at least one month, the side effects resulting from administration of omega interferon are greater than would be expected from the use of other interferon products. The preferred dose of omega interferon is about 135-.
More specifically, the present invention is a method of treating interferon-responsive diseases with greater tolerability and greater efficacy, thereby increasing the therapeutic index for treating interferon-responsive diseases. Thus, omega interferon is administered at a dose and for a time sufficient to produce the desired therapeutic response, while advantageously limiting undesirable adverse side effects. Omega interferon may be administered alone or in combination with one or more adjunctive therapeutic agents.
The method is useful in any warm-blooded animal that has not previously been treated with an interferon, and is also useful in treating any warm-blooded animal that exhibits residual sensitivity or resistance to treatment with another interferon, wherein (1) the adverse side effect is unacceptably high; (2) the therapeutic response was unacceptably low; (3) or some combination of (1) and (2). The animal may be a domestic animal, a domestic pet, or preferably a human. Thus, the methods have both veterinary and human medical uses. Livestock that can be treated by this method include horses, cattle, pigs, sheep, goats, etc. Domestic pets include cats, dogs, rabbits, birds, and the like. Preferably, however, the method of the invention is primarily applied to the treatment of humans, whether male or female, young or old.
Diseases treatable by the methods of the invention include those of infectious (e.g., viral), immunological, or proliferative origin, which in some populations may be treated by administration of interferon. Infectious diseases are those diseases that result from the proliferation of parasitic or viral organisms and can be transmitted by infection with or without physical contact. The disease includes hepatitis C, hepatitis B, hepatitis D, hepatitis G, other viral hepatitis, condyloma acuminatum, human immunodeficiency virus, yellow fever, Ebola virus, hemorrhagic fever, etc. Other diseases of viral origin are those caused by viruses, such as those listed in Stedman's medical dictionary, 26 th edition. Immune diseases are those in which the patient's immune system is unbalanced or abnormal. These diseases include, for example, multiple sclerosis, myasthenia gravis, systemic lupus erythematosus, dermatomyositis, scleroderma, CREST syndrome, hashimoto's thyroiditis, kawasaki disease, vasculitis, and the like. Other immune diseases suitable for treatment are found in the latest edition of Merck ManualOr Harrison's The Principles and Practices of internal MedicineProliferative diseases are generally those that include many types of malignancies, most of which invade surrounding tissues and can metastasize to several sites. These diseases are often referred to as cancers and include, for example, hairy cell leukemia, malignant melanoma, multiple myeloma, follicular lymphoma, non-hodgkin's lymphoma, cutaneous T-cell lymphoma, chronic myelogenous leukemia, basal cell carcinoma, carcinoid syndrome, superficial bladder cancer, renal cell carcinoma, colorectal cancer, laryngeal papillomatosis, actinic keratosis, kaposi's sarcoma, or other interferon-sensitive cancers. Proliferative diseases may also include diseases in which non-cancerous cells produce tissue hyperplasia or hypertrophy, leading to fibrosis or scarring or hyperproliferation of normal tissue. Such conditions may include, inter alia: response to any physical, chemical, genetic, infectious, or traumatic injury; fibrosis of any organ or tissue, such as bone marrow, intestine, brain, endocrine glands, heart, kidney, liver, lung, smooth or striated muscle, central or peripheral nerves, skin, spinal cord or vascular system of any tissue, etc. These include mycosis fungoides, multiple sclerosis, chronic granulomatous disease, pulmonary fibrosis, liver fibrosis, cirrhosis of the liver or tuberculosis.
As described above, the method can be used to treat patients who have not been previously treated with IFN or who exhibit resistance to other non-omega interferons such as alpha IFN, consensus IFN, tau IFN, beta IFN, gamma IFN, leukocyte-derived IFN, and the like. Such resistance may be "primary resistance" to the therapeutic effect of the non-omega interferon, e.g., when such interferon is administered alone or when combined with at least one adjunctive therapeutic agent, whether or not the therapeutic agent is used before, during or after the interferon. Such resistance may also be "secondary" to a therapeutic intervention with the non-omega interferon, whether such an interferon is administered alone or in combination with one or more adjunctive therapeutic agents, whether or not the therapeutic agent is used before, during, or after the interferon And (4) element. The invention is particularly useful in preventing or eliminating this type of "underdose resistance".
As part of the present invention, it was found that the Maximum Tolerated Dose (MTD) is very limited for currently available interferons. For example, in patients with life-threatening cancer, higher MTDs are more acceptable than in patients with less aggressive cancer. In patients with chronic hepatitis c, the MTD is lower, sometimes significantly lower. We analyzed MTD data for multiple interferons in many different clinical situations (representative studies collected are listed in table 1 below).
TABLE 1 maximum tolerated dose regimen approved for interferon for the treatment of hepatitis C
| # | Interferon | Indications of | MTDScheme(s) | Reference to the literature |
| 1 | α-2a | Hepatitis C | 6MIUTIWsc | FDA Summary Basis of Approval(PLA94-0782,29Oct.1996) |
| 2 | α-2b | Hepatitis C | 3-5MIUTIWsc | FDA approved product label |
| 3 | PEGα-2b | Hepatitis C | 105μgQWsc(≈21 MIU QWsc) | FDA approved product label |
| 4 | PEGα-2c | Hepatitis C | 180μgQWsc(≈36MIU QW sc) | Product label |
| 5 | α-2a | Cancer treatment | 30MIU/wk(+etretrinate) | Roth et al, Acta Oncol 1999; 38(5): 613-7 |
| 6 | α-2a | Cancer treatment | 15.5MIU/wk7wks | Rajkumar SV et al, Int J Radiat Oncolobiol Phys 1998 Jan.15; 40(2): 297-302 |
| 7 | α-2a | Cancer treatment | 6MIU TIW sc (idarubicin, dexamethasone) | Hubel K et al, Leukemia 1997 Dec; 11Suppl 5: s47-51 |
| 8 | α-2a | Cancer treatment | 5MIU TIWsc (all-trans retinoic acid) | Adamson PC et al, J Clin Oncol 1997 Nov; 15(11): 3330- |
| 9 | α-2a | Cancer treatment | 3MIUQD×7d×4wks(+IL-2) | Gause BL et al, J Clin Oncol1996 Aug; 14(8): 2234-41 |
| 10 | α-2a | Cancer treatment | 3.4MIUTIW (+5-FU + cisplatin) | Trudeau M et al, Cancer ChemotherPharmacol 1995; 35(6): 496-500 |
| 11 | α-2a | Cancer treatment | 3.4 one week out of the week of MIUQD × 5d4 (+ RT, + cisplatin, + hydroxyurea) | Vokes EE et al, Cancer ChemotherPharmacol 1995; 35(4): 304-12 |
| 12 | α-2a | Cancer treatment | 3MIUTIW (+ cisplatin) | Gosland MP et al, Cancer ChemotherPharmacol 199537 (1-2): 39-46 |
| 13 | α-2b | Cancer treatment | 3.6MIU QD sc | Dorr RT et al, J Interferon Res 1988; 8: 717-25 |
| 14 | α-2b | Cancer treatment | 24MIU/mQDiv is limited to 7 days; maximum 12MIU/mQD2 Weekly (1 patient) | Iacoboeli S et al, Am J Clin Oncol 1995; 18: 27-33 |
| 15 | α-2b | Cancer treatment | 5MIU/mTIW sc | Kirkwood JM et al, J Clin Oncol 1996; 14: 7-17 |
| 16 | PEGα-2b | Cancer treatment | 75μg/kgQW(35MIU/wk) | TalpazM et al, Blood 2001; 98: 1708-13 |
| 17 | In common with | Hepatitis C | 3-9μgTIWsc(≈3-9MIUTIWsc) | FDA approved product labelamg100697S.pdf |
Footnotes of the tables:*assuming that each μ g of pegylated IFN is as potent as μ g of non-pegylated;**consensus IFN is assumed to be about 5 x alpha-2 a or alpha-2 b in efficacy.
Using the same study designation as above, the monthly maximum tolerated dose calculated as MIU for each dosing regimen is shown below. Adjustments are made relative to body weight or body surface area to normalize dosages.
It should be noted that in table 1, MIU represents millions of international units of antiviral activity; in some cases, there is a μ g equivalent. But the relative potency of the interferon also needs to be considered. For example, while the clinical activity profile of pegylated interferon alfa tends to outperform that of non-pegylated interferon alfa in some cases (because pegylation improves pharmacokinetics, e.g., by keeping the molecule in circulation for a longer period of time), there is a loss of antiviral activity on a μ g to μ g basis when pegylated interferon is compared directly to non-pegylated interferon alfa. Pegylation improves pharmacokinetics at the expense of (in vitro) antiviral activity. In general, we observed that for all pegylated or all non-pegylated interferons, the antiviral activity of MIU is also an excellent code or predictor of side effects. The higher the MIU, the greater the rate and severity of side effects and the lower the MTD and the lower the efficacy that can be achieved.
TABLE 2.4 maximum tolerated interferon dose for weeks
| # | Interferon | Indications of | Maximum tolerated 4 week dose (MIU) |
| 1 | α-2a | Hepatitis C | 72 |
| 2 | α-2b | Hepatitis C | 60 |
| 3 | PEGα-2b | Hepatitis C | 34 or 84 |
| 4 | PEGα-2a | Hepatitis C | 36 or 144 |
| 5 | α-2a | Cancer treatment | 120 |
| 6 | α-2a | Cancer treatment | 62 |
| 7 | α-2a | Cancer treatment | 72 |
| 8 | α-2a | Cancer treatment | 20 |
| 9 | α-2a | Cancer treatment | 84 |
| 10 | α-2a | Cancer treatment | 41 |
| 11 | α-2a | Cancer treatment | 68 |
| 12 | α-2a | Cancer treatment | 36 |
| 13 | α-2b | Cancer treatment | 100 |
| 14 | α-2b | Cancer treatment | 290 |
| 15 | α-2b | Cancer treatment | 60 |
| 16 | PEGα-2b | Cancer treatment | 140 |
| 17 | In common with | Hepatitis C | 36-108 |
*Depending on the presumed or calculated MIU/μ g ratio, as discussed previously herein.
MTDs derived from FDA approved product labels include multiple phase I, II, and/or III clinical trials for each interferon specified. Other references represent a single study.
We observed that overestimating MTD based on small sample or cohort sizes is a common error, especially in phase I studies. A good example is the recent experience with pegylated interferon alpha-2 a. A study of 27 patients was performed with 3-6 patients per dosing group, which initially concluded that 450 μ g was the appropriate weekly dose [ see Motzer RJ et al, J Clin Oncol 2001; 19: 1312-9]. (however, the estimated antiviral activity was only 7% of that of non-pegylated interferons, and 450. mu.g of pegylated alpha-2 a provided only 45. mu.g of 70% of that determined by in vitro assays for normal alpha-2 a (see Bailon p et al, Bioconjug Chem 2001; 12: 195- -although these optimistic estimates, subsequent larger studies were generally limited to doses of 180. mu.g/week or less (PEGINTERFERONANLFA-2 a IN PATIENTS WITH CHRONIC HAITITIIS C. Zeuzem S, Feinman SV, Rasenack J et al, New Engl J Med 2000; PAT343: 1666-72).
However, as can be seen from an examination of table 2, the maximum tolerated dose for any interferon approved for the treatment of hepatitis does not exceed 180MIU for 4 weeks. This is true even for the most favorable hypothesis regarding the number of antiviral MIUs per μ g of interferon administered. For alpha-2 a, alpha-2 b and consensus interferon, doses above the indicated maximum value produce unacceptable, severe, sometimes irreversible clinical toxicity or cause the patient to stop treatment.
The potency of a number of previously identified interferons was determined by different assay systems, resulting in a number of different MIU/μ g ratios. As a result, the cumulative and mean ± SD MIU values for various interferons may differ based on the presumed MIU/μ g for a given interferon. This false is accommodated in the calculations shown below.
TABLE 3 mean 4 week dose for hepatitis and cancer
| Scheme(s) | Indications of | Number of studies | Hypothesis of efficacy (MIU/. mu.g ratio) | Mean of maximum mean tolerated 4 week dose (MIU) + -SD |
| 1 | Hepatitis (HAV) | 1-4 | 3MIU ≈ 15 μ g for the same MIU/. mu.g of α -2a and α -2b, pegylated or unpegylated interferon | 90±37 |
| 2 | Hepatitis (HAV) | 1-4 | Pegylated Interferon ≈ 40% efficacy of non-Pegylated Interferon μ g/μ g in vitro | 51±19 |
| 3 | Hepatitis (HAV) | 1-4,17 | Same 1 and consensus interferon potency ≈ 5 × α -2a or α -2b potency | 108±52 |
| 4 | Hepatitis (HAV) | 1-4,17 | Same 2 and consensus interferon potency ≈ 1 MIU/. mu.g | 48±17 |
| 5 | Cancer treatment | 5-16 | 3MIU≈15μg | 94±33 |
| 6 |
An "optimally tolerated" average MIU continuous MIU exposure of at least 4 weeks would appear to be represented by the calculations shown in scheme 3 above, which for the consensus interferon study assumed a very high MIU/μ g ratio. Mean ± SD is 108 ± 52, while mean ± SEM would be 108 ± 26. Accordingly, any MIU/4 week value above the mean ± 3SEM or 186 would be unexpected for all interferons considered together. None of the reference protocols met the test even though it was assumed that the μ g/μ g potency of pegylated interferon was the same as that of the matched non-pegylated interferon. Overall, in vitro testing does not confirm this hypothesis (see Bailon P, above reference).
Once the data is presented in this manner, it is clear that interferons with more favorable potency/side effect profiles are highly desirable. It is desirable that the interferon be administered at higher doses to achieve greater antiviral effects while still being acceptable clinically.
In the case of treating hepatitis c or cancer, certain side effects may be exacerbated by the addition of ribavirin (hepatitis), interleukin-2 (cancer), or other adjunctive therapies (cellular pathway blockers such as tyrosine kinase inhibitors) now in use or under development.
Inadequate treatment of hepatitis, cancer or other interferon-responsive diseases can also occur due to highly variable interferon levels caused by inherent biological variability in the patient. Perhaps more importantly, however, we observed that interferon blood levels were quite variable for interferons such as alpha, consensus and possibly tau due to the short half-life in blood. This change in blood levels is important when administering interferon alpha dosing regimens such as daily (QD), every other day (QOD), thrice weekly (TIW), or once weekly (QW). Even when QW is administered to a modified interferon, such as pegylated alpha interferon, significant blood level variability occurs. This variability may further contribute to the occurrence of adverse side effects and their unpredictable nature in, for example, chronic hepatitis c patients.
There is a clear medical need for safer, better tolerated and more effective interferons with antiviral, immunomodulatory and/or antiproliferative properties. In particular, in the treatment of hepatitis c, there is a need for interferons that can be administered at higher doses with greater tolerability and lower incidence and severity of adverse side effects, i.e., interferons with superior therapeutic indices.
For example, in the field of treatment of hepatitis, there is also a need for interferons with improved pharmacokinetic profiles, active as monotherapy or as part of a combination therapy, in patients judged to be under-treated with alpha interferon alone or in combination with, for example, ribavirin, particularly patients infected with one or more hepatitis c viruses or virus subtypes that are partially or fully resistant to, for example, alpha or consensus interferon therapy.
Furthermore, there is a need for effective and safe interferon therapy that safely and tolerably suppresses viral replication to acceptable levels for months or even years if complete eradication is not achieved.
Surprisingly, we have now found that omega interferon is not only effective in treating patients with chronic viral infections, such as patients with hepatitis c virus infections, but is also particularly well tolerated. In addition, omega interferon may be tolerated at μ g-and MIU-dosage levels, which are much higher than those that can be safely used with other interferons, such as alpha or consensus interferon. This clinical tolerance can be achieved even if omega interferon binds to a receptor to which alpha interferon and consensus interferon also bind. Furthermore, this surprising effectiveness and tolerability occurs despite the fact that omega interferon has significantly greater potency (MIU/μ g) than alpha interferon and thus would be predicted to be unusable at higher doses.
Thus, we have found that in the disease state treated, the dose and activity of omega IFN is sufficient to induce the desired therapeutic response in animals without the predicted poor tolerance of such treatment with non-omega IFN. This gives the physician greater flexibility in treating different diseases. For example, in the treatment of hepatitis c, higher doses of omega IFN may be required than potentially lower doses required for hepatitis cirrhosis. In either case, however, the disease can be treated with higher and more efficacious while better tolerated doses of omega IFN than those skilled in the art would predict from existing data for non-omega IFNs.
As one of several favorable clinical outcomes for safety and tolerability profiles, we have also demonstrated that omega interferon can suppress hepatitis c virus replication in the viral subtype most resistant to interferon, HCV genotype 1. Also surprisingly, we have also found that non-pegylated omega interferon alone is more effective than other existing treatments in suppressing hepatitis c virus replication in genotype 1 at about selected dosage levels. This superiority for non-pegylated omega interferon is evident when comparing existing clinical results for omega interferon with historical clinical data from the following study:
1. other non-pegylated interferon administered alone;
2. pegylated interferon administered alone; and most surprisingly
3. Two pharmaceutical antiviral regimens of the expensive and toxic interferon-alpha plus ribavirin.
Furthermore, it has also been demonstrated in vitro in cells infected with immunodeficiency virus that the patterns of alpha and omega interferon-induced gene signaling are different, i.e., omega interferon is functionally distinctive. Omega interferon induces a more durable anti-HIV gene response, while the response to alpha interferon is transient. We have now also demonstrated that omega interferon is uniquely able to substantially suppress viral replication, for example of the yellow fever virus, whereas other interferons are not.
In the methods of the invention, omega IFNs can be administered alone or in combination with adjunctive therapeutic agents, i.e., physiologically or pharmacologically active substances that complement or complement the activity of omega IFNs. For the treatment of diseases such as hepatitis, inosine monophosphate dehydrogenase inhibitors (IMPDI) such as ribavirin or ribavirin analogs are often used. Other inosine monophosphate dehydrogenase inhibitors include mycophenolic acid, mycophenolate mofetil (mofetil), mycophenolate sodium, aminothiadiazole, thiophenfurin, thiazolidin, viramidine, VX-148, VX-497, and VX-944. Other non-IMPDI agents include interleukin-2 or interleukin-2 derivatives, histamine derivatives, monoclonal antibodies, polyclonal antibodies, or small molecule inhibitors of hepatitis c virus replication. Specific examples of such antibodies include HBV-Ab (XTL) -17 and-19.
Ribavirin is chemically known to be 1- β -D-ribofuranosyl-1H-1, 2, 4-triazole-3-carboxamide and is commercially available from ICN Pharmaceuticals, inc. It is in the Merck Index, 11thVersion of which is described on page 8199. Its preparation and formulation are described in U.S. Pat. No.4,211,771.
Chemically VX-497 is known to be (S) -N-3[3- (3-methoxy-4-oxazol-5-yl-phenyl) -ureido]-benzyl-carbanic acid tetrahydrofuran-3-yl ester and available from VertexPharmaceuticals, inc. It is further described in PharmaprojectsAnd U.S. Pat. No.5,807,876.
It is chemically known that mycophenolic acid is 6- (1, 3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzylfluranyl) -4-methyl-4-hexanoic acid and is produced by Penicillium brevi-compactum, Penicillium repens (P-stolonefrum) and related species. It is further described in Merck Index, 11thVersion 6238.
Sodium mycophenolate is the sodium salt of mycophenolic acid and is available from Novartis Corp, Basel, Switzerland.
Mycophenolate mofetil is 2-morpholinoethyl ester of mycophenolic acid and can be used as CellCeptPurchased from Roche Laboratories, inc, Nutley, New Jersey. It is further described in the Physicians Desk Reference, 53rdVersion, page 2657.
The aminothiadiazole is 1, 3, 4-thiadiazol-1-amine and has a CAS registry number 4005-51-0. The molecular weight is 101.004755, and the molecular formula is C2H3N 35. Further information may be obtained from the service Pharmaprojects, accession number 5433.
Thiophenfurin and thiazolfurin are compounds that are active in vivo in mice. See j.med.chem., 1995, 38, 3829 and Pharmaprojects.
Viramidine is a ribavirin derivative to be used for hepatitis C either as monotherapy or in combination with IFN. ICN Pharmaceuticals is focusing on this compound. See also Pharmaprojects.
VX-148 and VX-944 are being developed by Vertex Pharmaceuticals. For further information see Pharmaprojects.
CDN-4007 is a pioneering compound from Oncor, Inc. Further information can be found in Pharmaprojects, accession number 25549.
XTL-17 and XTL-19 are monoclonal antibodies against hepatitis C.
Once a patient with a suspected interferon-sensitive disease is identified, the patient is treated by administering an amount of omega interferon, alone or in combination with an adjunctive therapeutic agent, for a duration sufficient to effect a therapeutic response while mitigating any adverse side effects of the treatment. The amount of omega interferon will be determined by the physician administering the dose, on an individual patient basis, depending on factors such as age, weight and size, sex, concomitant medical condition, concomitant medication, known or suspected genetic characteristics, and the like.
To enhance the therapeutic response, the amount of omega interferon will preferably be greater (suitably judged by quality or potency) than the amount employed with a different interferon. A similar procedure is followed if a patient with a disease or condition resistant to prior treatment with another interferon is identified.
In a preferred embodiment, omega interferon is administered parenterally (i.e., by injection, not to the intestinal tract, such as intramuscularly, intraperitoneally, intravenously, or subcutaneously) to a human patient suffering from an interferon-responsive disease at a dose of about 135-700 μ g per week, or about 19-420MIU per week. The dose may be administered by a single injection of a small amount, for example 15-100 μ g per dose. Such amounts may be administered continuously, multiple times per day, QD, QOD, TIW or QW. Such an amount or greater may also be administered via a long acting formulation or a sustained release formulation, e.g., containing 270 and 10,000 μ g. Such long acting formulations or sustained release forms are administered less frequently than once weekly and are intended to remain in the body for at least 2 weeks or even more than 1 month. For example, a 12 week dose for controlled release at a relatively constant rate of 175 μ g per week (representing about 35-105MIU per week) would contain about 2100 μ g of omega IFN (12X 175), while a 24 week dose would contain 4200 μ g (24X 175), and so on. All suitable dosage forms and routes of administration may be employed.
The method is particularly useful for treating human patients suffering from chronic hepatitis c.
In another embodiment, omega interferon is administered enterally, particularly orally. Such administration may be as a single dose or multiple doses over a week period. Omega interferon may be administered in substantially pure form or mixed with one or more excipients, and may be chemically or physically modified to enhance bioavailability.
For example, omega interferon may be administered topically or by inhalation in addition to parenteral administration (e.g., intravenous, intramuscular, intraperitoneal). Effective administration of omega interferon can also be achieved by increasing endogenous omega interferon or fragments thereof, by using specific or non-specific inducers, or by administering genetic material (e.g., nucleic acids) that encode all or part of the genetic material required for expression of omega interferon.
The pharmaceutical formulations comprising omega interferon may further comprise at least one pharmaceutically acceptable carrier, which may include excipients such as stabilizers (to facilitate long-term storage), emulsifiers, binders, thickeners, salts, preservatives, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with omega interferon, its use in therapeutic compositions and formulations is contemplated. Alternatively, cells expressing omega interferon may also be administered, preferably producing omega interferon in an amount sufficient to increase effectiveness without substantially increasing adverse side effects as compared to other interferons. In addition, omega interferon may be administered by non-cellular delivery systems such as liposomes.
Since the treatment of proliferative, immunological or infectious diseases is usually carried out for a long time, it may be preferable in this case to implant or inject an article containing an omega IFN formulation which is biocompatible for the subject to be treated and releases IFN in a regulated manner over time, i.e. a controlled release formulation. The formulation may be bioerodible, such as a gel, non-gel polymer, or pill (pellet), or non-bioerodible, such as a mechanical device, e.g., a pump. The pump may also be external to the body, with only one catheter or tube or the like penetrating the skin into the subcutaneous or intramuscular space.
An example of a suitable non-bioerodible formulation or device is the use of DUROSSystem (ALZA Corporation), a miniature drug dispensing pump, is currently made primarily of titanium and can be as small as a match.
DUROSThe pump operates like a micro-syringe containing the drug in a drug reservoir. By osmosis, water from the body is slowly drawn through the semipermeable membrane into the pump by salt or other suitable osmotically active substance present in the engine compartment. This water is absorbed by the osmotic agent, which then swells and slowly and continuously pushes the piston, dispensing an accurate amount of drug out of the drug reservoir and into the body. The osmotic engine does not require a battery, switch or other electromechanical component to operate. The amount of drug delivered by the system is regulated by a number of factors, including, for example, the materials used in the preparation, the control of the amount of water entering the pump by the membrane, the strength of the osmotic agent, the frictional resistance to movement of the piston, the size and shape of the reservoir, the size, shape and length of the port through which the drug exits the pump, the formulation and type of drug, and whether the formulation is a liquid, suspension, or the likeOr a gel, and a pressure generated within the device to expel the drug or a counter pressure generated within the tissue to counter such expulsion.
Other useful long-term delivery formulations may use ALZET developed by ALZA CorporationIs prepared by the technology. These formulations can be delivered externally. Details of the ALZET technique can be found at www.alzet.com.
Patents that provide useful guidance regarding the preparation of long-term delivery devices useful in the methods and kits of the present invention include those assigned to Alkermes. Other patents include those assigned to ALZA Corporation (now a subsidiary of Johnson and Johnson, Inc.), specifically referred to as "DUROS"technique. Representative patents that may be used in various aspects of the present invention include the following U.S. patents: 5,529,914; 5,858,746; 6,113,938; 6,129,761; 5,985,305; 5,728,396; 5,660,847, respectively; 5,112,614; 5,543, 156; 5,443,459, respectively; 5,413,572; 5,368,863, respectively; 5,324,280; 5,318,558, respectively; 5,221,278, respectively; 4,976,966, respectively; 4,917,895, and 4,915,954. All patents are incorporated herein by reference.
When administering an adjunctive therapeutic agent such as ribavirin (or other IMPDI), ribavirin is administered to the patient in conjunction with omega interferon, i.e., the ribavirin dose is administered during some or all of the same time period that the patient receives omega interferon. Most interferon formulations are not effective when administered orally, except when chemically modified or protected in some other way from degradation by enteropeptidases as described above. Thus, a preferred method of administering omega interferon is parenterally, preferably by subcutaneous, intravenous, or intramuscular injection. More preferred is the administration of a long acting formulation, with or without the use of equipment such as a pump, or another long-term administration form suitable for multi-week or multi-month, continuous or sustained delivery of omega interferon. The pump may be of any suitable design, such as fixed or variable delivery osmotic, electrical, mechanical, hydraulic, pneumatic, inserted under the skin or worn outside.
IMPDI such as ribavirin can be administered orally in capsule or tablet form in combination with administration of omega interferon. Of course, other types of administration of the two drugs are also contemplated as may be utilized, such as by nasal spray, transdermally, by suppository, by sustained release dosage form, and the like. Any form of administration will work so long as the appropriate dosage is delivered without destroying the active ingredient, and appropriate considerations are given to the absorption, distribution, metabolism, and excretion of the individual in the combination of the various dosage levels.
Ribavirin is commonly referred to as the Physicians Desk ReferenceThe recommended rate of administration, but may be administered at a rate of about 400 to about 1200 mg/day.
Another aspect of the invention can be considered a method, wherein omega interferon is administered to a subject in need thereof, any combination with a therapeutically effective amount of at least one adjunctive therapeutic agent, for a length of time that the animal tolerates omega interferon, the level of a disease marker in the animal is monitored during the administration period, and omega interferon is continued to be administered for a length of time subject to a continued decrease in the level of the disease marker.
The disease progression and adverse side effects, if any, are then monitored by a physician attending the patient. This can be done by assessing signs and symptoms of the disease or by monitoring the presence of such disease markers in the patient's bodily fluids (e.g., blood, plasma, urine). For example, a patient with chronic hepatitis c virus ("HCV") infection may exhibit one or more of the following signs or symptoms:
(a) elevated alanine aminotransferase ("ALT");
(b) an increase in aspartate aminotransferase ("AST");
(c) bilirubin is elevated;
(d) positive anti-HCV antibody test;
(e) the presence of HCV, as confirmed by a positive test for the disease marker HCV-RNA;
(f) clinical features of chronic liver disease, such as abnormal liver size, ascites or esophageal varices; and
(g) hepatocyte injury or dysfunction as shown by histopathology, laboratory or radiographic methods;
(h) hepatocellular carcinoma.
In patients with severe HCV infection, the patient's HCV-RNA copy number/ml serum may exceed 2X 106And (6) copying. By successful treatment according to the methods of the present invention, the copy number of HCV-RNA can be reduced to a barely detectable level, i.e., less than about 100 and 1000 copies of HCV-RNA per ml of patient serum, as determined by quantitative, multi-cycle, reverse transcriptase PCR methods.
Thus, it can be seen that another aspect of the invention can be viewed as the use of an omega Interferon (IFN) in the manufacture of a medicament for the treatment of an immune, proliferative or infectious disease in a warm-blooded animal. The medicament is administered to the animal in accordance with the teachings above, i.e., at a dose and activity to the disease treated sufficient to induce a therapeutic response in the animal, which is higher than would be well tolerated by non-omega IFN-based data. Preferred aspects of the method of treatment are also applicable to this "use" aspect of the invention.
Article of manufacture
Another aspect of the invention is a product for use in the treatment of an immune, proliferative or infectious disease in a warm-blooded animal subject. The article of manufacture comprises an omega interferon suitable for administering a therapeutically effective amount of an omega IFN to the subject, in combination with instructions for administration, administering the omega interferon at a dose higher than a dose predicted based on non-omega IFN data, wherein the dose is preferably administered to an animal for at least 1 month, and wherein side effects from administration of the omega interferon are less than expected for use of other interferon products. The preparation is suitable for enteral, parenteral, inhalation or topical administration, as discussed above. Preferably, the article is designed for injection into a subject, in particular for subcutaneous injection. The preparation is particularly valuable designed to administer omega IFN to a subject in a controlled release manner, for example at a rate of about 135-. It is preferred to continue the controlled rate for at least 1 month. Of course, if the omega IFN is formulated for injection, it is preferably a sterile aqueous composition.
Another aspect of the invention may be considered a kit for delivering a relatively constant amount of a drug over time, wherein the amount of drug delivered to a subject patient is about 135-700 μ g/week. The kit comprises a long-term delivery formulation designed to deliver the drug at a relatively constant rate over time, typically for at least 1 month, preferably 3-12 months. The kit may also contain other equipment or drugs for administering the formulation. A physician or other health care provider can individualize the dosage rate of a patient over time according to the patient's characteristics including age, sex, size, health condition, etc., as well as the severity and type of the disease.
Preparation method
Another aspect of the invention results from the foregoing discussion, namely a process for the preparation of an omega IFN-based preparation for use in the treatment of an immune, proliferative or infectious disease in a warm-blooded animal subject. The method comprises providing omega IFN as a composition suitable for administering a therapeutically effective dose to a subject and administering the omega IFN so provided in combination with instructions for use, the omega IFN for use in such a disease at a higher dose than would be predicted based on non-omega IFN data, wherein the side effects from administration of the omega IFN are less than expected using other interferon products. Such a method would result in omega IFN suitable for enteral, parenteral or topical administration, preferably for injection into a subject. Preferred methods result in the production of omega IFN suitable for subcutaneous injection, particularly for controlled release of omega IFN into a subject, at a rate of about 135-. The rate-controlled release may last for 1 month or more. If the method is designed to formulate omega IFN as a composition for injection, it is important that it be sterile, preferably as a sterile aqueous solution.
Another aspect of the invention is a method of preparing a delivery system for delivering omega IFN in a controlled manner over time. The method includes preparing a long-term delivery device designed to deliver omega IFN over time at a relatively constant rate determined to be about 135-. Once the system is prepared, it is combined with appropriate written instructions for administration to a subject in need thereof, as discussed above. The system may also be combined with other devices or medicaments that may be used to administer or deliver the system. Written instructions for administration may be affixed directly to the container (e.g., by affixing the label directly to a vial containing the interferon with or without a carrier or excipient). Alternatively, the interferon-containing container closure system may be placed in a second container, such as a box, and the written material in the form of a package insert may be placed in the second container along with the interferon-containing first container closure system. Written instructions may describe the indication for which omega interferon is prescribed, either as monotherapy or as part of combination therapy with adjunctive therapeutic agents. Such indications may include interferon-responsive diseases (e.g., viral hepatitis c). The written material will preferably be provided in the form required by regulatory agencies, such as the U.S. food and drug administration, having authority to approve the marketing of such interferons, in the form of package inserts for the prescribed medication. The written material will indicate that the interferon is prescribed for patients with an infection, proliferative, or immunological disorder. The written material will preferably describe the technique of administering the drug, e.g., injection or implant formulation. The written material may also include instructions for using other devices or medications contained in the kit. In a preferred embodiment, the written material will indicate that the omega interferon is for use in the treatment of viral hepatitis, particularly viral hepatitis c, or cirrhosis or fibrosis of any organ, particularly the liver, when such cirrhosis or fibrosis is caused by viral hepatitis c. The written material will indicate that the interferon may be used as a primary or secondary treatment or in combination with other treatments. It will be further described that although the interferon has an effect on the infected liver of a viral hepatitis C patient, the interferon may also reach other tissues where it may have no therapeutic or adverse side effects.
Major toxicities may also be described if appropriate, and may include, for example, headache, flu-like symptoms, pain, fever, weakness, chills, infection, abdominal pain, chest pain, injection site reactions (as appropriate), malaise, hypersensitivity, syncope, vasodilation, hypotension, nausea, constipation, diarrhea, dyspepsia, anorexia, anemia, thrombocytopenia, leukopenia, other blood dyscrasias, myalgia, arthralgia, insomnia, dizziness, suicidal ideation, depression, impaired mental concentration, amnesia, confusion, irritability, anxiety, tension, decreased libido, urticaria, hair loss, and others.
It is also described in the written material that when symptoms such as fever, chills or flu-like manifestations are observed, these symptoms can be treated with TylenolAntihistamines such as BenadrylTo treat, hypotension may be responsive to administration of a fluid or booster, or if symptoms or signs are severe enough, the dose should be reduced or treatment terminated.
The written material may also describe that the delivery of the interferon formulation intended for short-term administration is by injection, infusion, inhalation, oral or transdermal administration. The preferred embodiment is by injection or infusion, most preferably by injection. Warnings, precautions and contraindications should be described.
Examples
The following examples are provided to further illustrate how to make and use the invention. In the examples, the major measure of antiviral efficacy in the context of chronic hepatitis c is the determination of viral load, for which hepatitis c viral rna (hcv rna) is the standard measure. This assay was used in two studies on omega interferon.
In the context of treating hepatitis, it is useful to determine changes in the following indices:
(a) elevated ALT;
(b) elevated AST;
(c) elevated bilirubin;
(d) HBsAg, anti-HBc antibody and anti-HBe antibody are tested positive;
(e) clinical features of chronic liver disease;
(f) hepatocyte injury or dysfunction as evidenced by histopathology, laboratory or radiographic methods;
(g) hepatocellular carcinoma.
In the context of combination therapy with interleukin-2 for the treatment of renal cell carcinoma, it is useful to determine the extent and variation of metastatic disease by:
(a) a positive computed tomography or magnetic resonance imaging scan;
(b) positive bone scan;
(c) positive signs of physical examination, such as the presence of palpable masses;
(d) positive hematuria test.
In the context of combination therapy with interleukin-2 for the treatment of renal cell carcinoma, it is useful to determine the extent and variation of metastatic disease by:
(a) a positive computed tomography or magnetic resonance imaging scan;
(b) positive bone scan;
(c) positive signs of physical examination, such as the presence of palpable masses.
Example 1
The safety, tolerability and antiviral effects of omega interferon were investigated in 90 previously untreated patients chronically infected with hepatitis c virus of genotype 1,2, 3 or 4. Except for other causes of hepatic dysfunction. The minimum HCV RNA level at enrollment was > 100,000U/ml, with elevated ALT levels.
The aim of this study was to evaluate the effect of different doses of omega interferon on HCV RNA levels, alanine Aminotransferase (ALT) levels. Other objectives are to evaluate the safety and tolerability of elevated doses of omega interferon, as judged by physical examination, adverse side effects and laboratory examinations.
The study was designed for multicenter, open label and booster dose, divided into 5 groups of 15 or more subjects per group: 15, 30, 45, 60 and 90 μ g were administered subcutaneously three times a week. Thus, the weekly doses of omega interferon in these 5 groups were 45, 90, 135, 180 and 270 μ g. The cumulative weekly antiviral activity of these same doses of omega interferon was 18, 36, 54, 72, 108 and 144MIU, respectively. The cumulative 4-week antiviral activity of these same doses of omega interferon was approximately 72, 144, 216, 288, and 432MIU, respectively. [ antiviral Activity was determined by measuring the antiproliferative effect of omega interferon, compared with the effect of alpha-2 c in human A549 cells infected with encephalomyocarditis virus. In this assay system, the antiviral activity of omega interferon is about 4X 108U/mg, 2X 10 compared to alpha-2 a8U/mg。]
The dose is administered by a visiting nurse or other medical practitioner to ensure that the dose is properly administered, that blood tests are properly drawn, and that adverse side effects are quickly identified, recorded and reported.
Omega interferon is prepared as a stable and lyophilized powder, which is then dissolved in sterile water for injection. A dose of 15 μ g was initially administered subcutaneously from the formulation on a 3-time weekly schedule, with a target administration duration of 3-12 months. The dose of omega interferon is then gradually increased.
Hepatitis C Virus RNA levels (HCV RNA) by quantitative multicycle reverse transcriptase polymerase chain reaction technology (Amplicor)Hoffmann La Roche). HCV RNA levels were determined 3 times prior to initiation of treatment, and at 1-5 days of treatment and 2, 4,8, 1,2 and 16 weeks of treatment, at least 2 weeks apart. If the patient responds to the treatment, treatment is continued at the same dosage level and thereafter HCV RNA levels are determined at intervals of every 3 months. ALT levels were determined by standard laboratory techniques before, on day 0 and at 2, 4,8, 12 and 16 weeks of treatment, and every 3 months thereafter if treatment continued.
Safety was determined by routine physical examination, routine interrogation of patients for adverse side effects, and standard laboratory tests including hematology, chemistry, liver function tests, and the like.
The results of the study are shown below. Baseline characteristics were similar in all dosing groups. Most patients are male, mostly between 20-50 years old. Most patients have genotype 1 infection with a very high baseline viral load as determined by hcv rna. Inflammation of the liver was present as judged by elevated ALT levels of approximately 3 x normal.
TABLE 4 Baseline characteristics
Treatment with omega interferon is surprisingly effective and very well tolerated. Changes in HCV RNA levels of viral genotype 1 in treatment groups 1-5 are shown below. At 45, 60 and 90 μ g TIW (135, 180, 270 μ g/week), there was a significant dose response at 12 weeks of treatment (FIG. 1). There was an excellent viral response to treatment and a clear dose response (figure 5). Very surprisingly, in genotype 1, complete viral clearance (below detectable limit in HCV RNA assay) was over 80% for the two highest dose groups. The response was even higher in genotypes 2, 3 and 4. Regression of the hepatitis changes as judged by biochemical responses to changes in ALT levels was also significant, even in patients infected with genotype 1 virus (table 6).
TABLE 5 genotype 1 viral response at 12 weeks
| Dosage (μ g/week) | Antiviral Activity (MIU/week) | Patient in whom HCV RNA was not detectable (%) |
| 45 | 18 | 20 |
| 90 | 36 | 20 |
| 135 | 54 | 60 |
| 180 | 72 | 82 |
| 270 | 108 | 100 |
Genotypes 2, 3 and 4 responded at an even higher overall rate, except in the 270 μ g/week group, where genotype 1 had achieved a maximum of 100%.
TABLE 6 Biochemical reaction of genotype 1 at 12 weeks
| Dosage (μ g/week) | Patients with Normal ALT (%) |
| 45 | 50 |
| 90 | 50 |
| 135 | 60 |
| 180 | 57 |
| 270 | 100 |
Genotypes 2, 3 and 4 reacted at an even higher overall rate (except, of course, in the 270. mu.g/week group, where the response rate was already at a maximum of 100%).
There were mild and temporary or reversible adverse side effects (table 7). Only 1 patient discontinued dosing due to adverse events.
TABLE 7 incidence of dose-induced adverse side effects
Comment on: historically, interferon-alpha alone has been the available treatment for hepatitis c patients. It is generally recognized that for long-term treatment, the chances of a response to the treatment increase. The development of a combination of interferon alpha plus oral ribavirin increases the response rate. Pegylated interferon alfa may provide advantages over non-pegylated interferon alfa as monotherapy. However, it is not clear that pegylated interferon alfa plus ribavirin does not increase the adverse side effects that outweigh the benefits of combination therapy.
In a recently completed study, results for interferon alpha alone were compared to results for pegylated interferon alpha or a combination of interferon alpha plus oral ribavirin. These results are shown below (table 8).
TABLE 8 virological and biochemical reactions to non-omega interferon regimens
VR ═ virological responses; BR is a biochemical reaction; not applicable to
Reference documents:
zeuzem S, New Engl J Med 2000; 343: 1666-72. study of non-cirrhosis patients with chronic hepatitis C;
heathcote EJ, New Engl J Med 2000; 343: 1673-80. study of cirrhosis patients with chronic hepatitis c;
McHutchison J, New Engl J Med 1998; 339: 1485-92. study of non-cirrhosis patients with chronic hepatitis C.
In Zeuzem et al, the incidence of dose discontinuation or reduction due to adverse effects in α -2a treated patients is 10% and 18%, respectively. For pegylated alpha-2 a, the corresponding ratios were 7% and 19%. In Heathcote et al, the ratios are 8% and 14% for α -2a, respectively, and 13% and 10% for pegylated α -2a, respectively. In McHutchison et al, the ratio is 9% and 12% and 8% and 13% for alpha-2 a alone and 21% and 26% for the combination of alpha-2 a and ribavirin.
Given the measure of omega interferon administration in the study of example 1 (much higher than well tolerated doses of other interferons), the duration of omega interferon treatment (sufficient to detect time-dependent serious adverse events), the virological response rate (surprisingly high, even compared to multi-drug treatment with alpha interferon plus ribavirin), it is evident that the virological response rate, biochemical response rate and tolerability of omega interferon are all surprisingly good.
Thus, monotherapy with omega interferon can simplify treatment of hepatitis: increased effectiveness, reduced adverse side effects, reduced costs associated with diagnosis and treatment of side effects, and reduced overall treatment costs. Combination therapy with, for example, ribavirin and higher doses of omega interferon will produce even better therapeutic results.
Example 2
Omega interferon is uniquely active against yellow fever virus. An CPE (virus induced cytopathic effect) -inhibition assay using live dye uptake was used to evaluate the antiviral activity of compounds against yellow fever virus strain 17/D in Vero cells, an african green monkey kidney cell line. The antiviral assay was designed to test 6 concentrations of each compound in triplicate against the challenge virus, here Yellow Fever Virus (YFV). Cell controls containing medium alone, virus-infected cell controls containing medium and virus, drug cytotoxicity controls containing medium and each drug concentration, reagent controls containing medium only (no cells), and drug colorimetric controls containing drug and medium (no cells) were run simultaneously with the test samples.
Plates were incubated at 37 ℃ in the presence of 5% CO2Until maximum CPE was observed in untreated virus control cultures (day 6). Through Cell Titer 96The AQueous One Solution Cell Proliferation assay determines the CPE-inhibiting effect of a compound. This assay is a colorimetric method that measures the number of viable cells. The reagent contains a new tetrazole compound MTS [ (3- (4, 5-dimethylthiazole-2-yl) -5- (3-carboxymethylphenyl) -2- (4-sulfophenyl) -2H-tetrazole, inner salt)]And an electron coupling agent PMS [ phenazine methosulfate ]]. When combined, MTS and PMS form a stable solution. The MTS tetrazole compound is then bioreduced to formazan by NADPH or NADH produced by dehydrogenases in metabolically active cellsAnd (3) obtaining the product. Measured nailThe amount of product is directly proportional to the number of viable cells in the culture.
Typical arrangements of cells, variable concentrations of drug and controls for standard 8 × 12 96 well plates are shown in tables 9A and 9B. Each table shows half of a standard 96-well plate. The form is presented in this way only to comply with the format requirements of the PCT application. By placing table 9A to the left of table 9B so that there are 12 columns in the page horizontally and 8 rows in the page vertically, a complete standard 96-well plate will be better viewed. The contents of the wells are shown in the tables. In the table, the different terms have the following meanings:
medium-only reagent control, cell-free
Cell control ═ cells and culture media
Viral control-Vero grown in the presence of virus but in the absence of drug
Conc ═ concentration
TABLE 9A
TABLE 9B
The percentage of CPE reduction for virus-infected wells and the percentage of cell survival for uninfected drug control wells were calculated. The minimum inhibitory drug concentration that reduces CPE by 50% (IC50) and the minimum toxic drug concentration that causes a 50% reduction in viable cells (TC50) were calculated using a regression analysis program fitted with a semilogarithmic curve. The therapeutic index (TI50) for each active compound can be determined by dividing TC50 by IC 50.
The results of the study comparing interferon-alpha (alpha-2 b) and interferon-omega are shown below (Table 9)
TABLE 9 omega interferon abrogation of yellow fever virus replication
| Medicine | IC50(IU/ml) | TC50(IU/ml) | TI(TC50/IC50) |
| Alpha-2 b interferon | Has not reached | >200 | NA |
| Omega interferon | 0.8 | >5000 | >6300 |
Interferon alpha is completely ineffective against yellow fever virus. There was no concentration with measurable antiviral effect. Concentrations above 200IU/ml produced significant alpha-2 b-induced direct cell damage. As a result, it is impossible to calculate the therapeutic index. In contrast, omega interferon produced significant antiviral effects in the absence of drug-induced cytotoxicity, with TI50 exceeding 6300.
It will be apparent to those skilled in the art that many modifications and variations of the present invention can be made without departing from its spirit and scope. The specific embodiments described herein are offered by way of example only, and the invention is to be limited only by the terms of the appended claims, along with the full scope of equivalents to which such claims are entitled.
All articles, patents, and other information cited herein are incorporated by reference for all purposes.
Claims (24)
1. Use of omega interferon in the manufacture of a medicament for the treatment of a viral disease in a warm-blooded animal subject, wherein an adjunctive therapeutic agent which is an inosine monophosphate dehydrogenase inhibitor is also administered to the subject.
2. The use of claim 1, wherein the viral disease comprises a hepatitis c virus infection.
3. The use of claim 2, wherein the subject exhibits primary or secondary resistance to non-omega interferon treatment.
4. The use of claim 1, wherein the viral disease comprises a yellow fever virus infection.
5. The use of any one of claims 1-4 wherein omega interferon is administered in a therapeutically effective amount of about 135 and 700 μ g/week.
6. The use of claim 5 wherein omega interferon activity is between about 27 and 420 million international units.
7. The use of any one of claims 1-6, wherein the inosine monophosphate dehydrogenase inhibitor is ribavirin or a ribavirin analog.
8. The use of any one of claims 1-6, wherein the inosine monophosphate dehydrogenase inhibitor is selected from the group consisting of mycophenolic acid, mycophenolate mofetil, mycophenolate sodium, aminothiadiazole, thiophofurin, thiazoluralin, viramidine, VX-148, VX-497, and VX-944.
9. The use of claim 7 or 8, wherein the inosine monophosphate dehydrogenase inhibitor is administered to the human subject at a therapeutically effective dose of about 200 to 4800 mg/day.
10. The use of any one of claims 1-9, wherein the omega interferon is administered enterally, parenterally or topically.
11. The use of claim 10 wherein the omega interferon is administered parenterally.
12. The use of claim 11, wherein the omega interferon is administered intramuscularly, intraperitoneally, intravenously, or subcutaneously.
13. The use of claim 12 wherein omega interferon is administered subcutaneously 3 times per week.
14. The use of claim 11 wherein the omega interferon is administered by injection.
15. The use of claim 14, wherein the omega interferon is administered by one or more daily injections.
16. The use of any one of claims 1-9, wherein the omega interferon is administered by infusion.
17. The use of any one of claims 1-9, wherein omega interferon is administered at a controlled rate over time.
18. The use of claim 17, wherein omega interferon is administered using a device.
19. The use of claim 18, wherein the device comprises a pump.
20. The use of claim 19, wherein the device is implanted in the subject or external to the subject.
21. The use of claim 20, wherein the device is implanted in a subject.
22. The use of claim 21, wherein the device comprises an osmotic pump.
23. The use of any one of claims 1-22, wherein the omega interferon is a recombinant omega interferon.
24. The use of any one of claims 1-23, wherein the subject is a human.
Applications Claiming Priority (3)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US33794801P | 2001-11-09 | 2001-11-09 | |
| US60/337,948 | 2001-11-09 | ||
| PCT/US2002/036025 WO2003063573A2 (en) | 2001-11-09 | 2002-11-08 | Method for treating diseases with omega interferon |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| HK1084862A1 HK1084862A1 (en) | 2006-08-11 |
| HK1084862B true HK1084862B (en) | 2009-08-07 |
Family
ID=
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US20070248572A1 (en) | Method for treating diseases with omega interferon | |
| US7727519B2 (en) | Method for treating hepatitis C virus with omega interferon | |
| AU2002365436A1 (en) | Method for treating diseases with omega interferon | |
| AU2002225870A1 (en) | Method for short-term and long-term drug dosimetry | |
| US20110270212A1 (en) | Pharmacokinetic control for optimized interferon delivery | |
| US6362162B1 (en) | CML Therapy | |
| PT1535623E (en) | Renal cell carcinoma treatment | |
| HK1084862B (en) | Use of omega interferon in the manufacturing of a medicament for treating viral disease in a warm-blooded animal subject | |
| Moritz et al. | Tumor necrosis factor α modifies resistance to interferon α in vivo: First clinical data | |
| AU2004242456B2 (en) | CML therapy | |
| JP2015512900A (en) | Combination therapy to treat HCV infection in a special patient genotype subpopulation | |
| HK1068548B (en) | Use of omega interferon in the manufacture of a medicament for treating hepatitis c | |
| JP2015509980A (en) | Combination therapy to treat HCV infection in a population of HCV-HIV co-infected patients | |
| HK1075418A (en) | Chronic myeloid leukemia (cml) therapy | |
| HK1032348B (en) | Chronic myeloid leukemia (cml) therapy |