![]() Silibinin component for hepatitis treatment
专利摘要:
公开号:ES2662700T9 申请号:ES11005445.9T 申请日:2008-11-14 公开日:2018-08-27 发明作者:Peter Dr. Ferenci;Ulrich Dr. Mengs;Ralf-Torsten Dr. Pohl;Lucio Claudio Rovati;Massimo Maria D'amato 申请人:Madaus GmbH; IPC主号:
专利说明:
5 10 fifteen twenty 25 30 35 40 DESCRIPTION Silibinin component for hepatitis treatment This invention relates to the use of a silibinin component for the production of a medicament for the treatment of hepatitis C, in particular, for the reduction of the virus load. Preferably, the medicament is adapted for parenteral administration. Preferably, the silibinin component is a silybinin ester. Silibinin, {3,5,7-trihydroxy-2- (3- (3-hydroxy-4-methoxy-phenyl) -2- (hydroxymethyl) -2,3-dihydrobenzo [b] [1,4] -dioxin -6-yl) chroman-4-one; or according to Ph. Eur. {(2R, 3R) -3,5,7-trihydroxy-2 - [(2R, 3R) -3- (4-hydroxy-3-methoxyphenyl) -2- (hydroxymethyl) -2 , 3-dihydro-1,4-benzo-dioxin-6-yl] -2,3-dihydro-4H-1-benzopyran-4-one} is the main constituent of silymarin and the main flavonoid extracted from milk thistle ( Silybum marianum Gaertneri). Silibinin has the following structure: image 1 silybinin The diastereomers silibinin A and silibinin B are distinguished in the literature: image2 silybinin A silybinin B Silybinin is the main constituent of silymarin (in a 50:50 mixture of silybinin A and silybinin B). Other constituents include isosilibinin (isosilibinin A and isosilibinin B), silidianine (silydianine), silicristine (silycristine), isosylcycline, taxifoline and others. Methods for isolating silibinin are known from the prior art (for example, US 4,871,763). Silybinin and silymarin have been investigated and described in detail. In this regard, reference may be made, for example, to the publication of N-C Kim et al., Org. Biomol Chem. 2003, 1, 1684-9; DYW Lee et al., J. Nat. Prod. 2003, 66, 1171-4; DJ Kroll et al., Integrative Cancer Therapies, 2007, 6, 110-9; Z Wen et al., DMD Fast Forward, doi: 10.1124 / dmd.107.017566; and US 4,871,763. Silybum marianum has a history as a medical plant for almost 2 millennia. Silymarin, the extract of milk thistle seeds, is an ancient herbal remedy used to treat a range of disorders of the liver and gallbladder, including hepatitis, cirrhosis and as a liver protector against poisoning from wild mushrooms, alcohol , chemicals and environmental toxins The mode of action of silymarin is diverse. The largest randomized controlled trial conducted in the 1970s indicated that long-term treatment with silymarin may decrease mortality in cirrhosis patients (P Ferenci et al., J Hepatol 1989, 9, 105-13). However, the role of the drug for the treatment of liver diseases remains controversial (S Verma et al., Clinical Gastroenterology and Hepatology 2007, 5, 408-16; F Rainone, Am Fam Phys 2005, 72 (7), 1285 -8). Part of this uncertainty is due to limited data on their pharmacokinetic regimens and optimal dosages. Silymarin is poorly soluble in water and oral preparations have limited bioavailability. The pharmaceutical applications of silybinin are also known. Silibinin has strong antioxidant properties (see publications by A Pietrangelo et al., Gastroenterology 1995, 109, 1941-49; MI Lucena et al., Int J Clin Pharmacol Ther 2002, 40, 2-8; and L Mira et al ., Biochem Pharmacol 1994, 48, 753-9) and antifibrotic properties (see the publications of G Boigk et al., Hepatology 1987, 26, 643-9; and C Dehmlow et al., Hepatology 1996, 23, 749-54 ) that make it a potentially useful drug for the treatment of chronic liver diseases. Silibinin as a pure substance is administered intravenously, for example, in the case of liver poisoning by the white cup of death (amanitin, phalloidin) with the 5 10 fifteen twenty 25 30 35 40 Four. Five fifty 55 in order to preserve the liver from further deterioration (see the publication of K Hruby et al., Hum Toxicol 1983, 2, 138-195). The effect on mushroom poisoning is explained in part by the stimulation of nucleolar polymerase A that increases the synthesis of ribosomal proteins and inhibits lipid peroxidation (see the publication of J Sonnenbichler et al., Prog Clin Biol Res. 1986, 213 , 319-31). Clinical trials also show success in the prevention and treatment of certain types of cancer (L Varghese et al. Clin Cancer Res 2005, 11 (23), 8441-7; K Letschert et al., Toxicological Sciences 2006, 91, 140 -9). The silybinin ester is marketed as an infusion solution, for example, under the name Legalon® SIL in Germany. Viral hepatitis refers to infections that affect the liver and are caused by viruses. It is a major public health issue worldwide. Viral hepatitis does not only imply high mortality, it also affects medical resources and can have serious economic consequences. Most cases of viral hepatitis can be prevented. Viral hepatitis includes five types of disease, which can be caused by at least five different viruses. Hepatitis A and hepatitis B (viral and serum hepatitis, respectively) are separate diseases and both can be diagnosed by a specific serological test. Hepatitis C and E comprise a third category, each of a different type, hepatitis C being transmitted parenterally and hepatitis E is transmitted enterally. Hepatitis D, or hepatitis delta, is another distinct virus and depends on hepatitis B infection. This form of hepatitis can occur as a superinfection in a hepatitis B carrier or as a coinfection in an individual with acute hepatitis B. Hepatitis C is an infectious disease in humans, which is caused by the hepatitis C virus (HCV). Infection with HCV can lead to severe liver damage, for example, inflammation of the liver parenchyma, liver fibrosis, liver cirrhosis and liver carcinoma. In more than 80% of infected patients, HCV infection becomes chronic. HCV transmission usually takes place parenterally through the blood. It is estimated that approximately 170 million people worldwide are infected with the hepatitis C virus (HCV). Infected patients may be asymptomatic for decades until finally the development of cirrhosis of the liver and / or hepatocellular carcinomas. Approximately 40-50% of liver transplants in the United States are based on HCV infections. Six HCV genotypes (HCV1-HCV6) have been identified that differ in their geographic extent and in their response to medicinal therapies. HCV proteins have been shown to induce STAT-3 activation through oxidative stress and Ca2 + signaling (K Koike et al., Hepatol Res 2006; 34: 65-73; G Waris et al., J Virol 2005 , 79, 1569-80) as well as lipid peroxidation products and antioxidant gene expression (M Okuda et al., Gastroenterology 2002, 122, 366-375). It seems that the balance of potential oxidative and reductive capacities in the cell (cellular redox state) has profound consequences on signal transduction trajectories (YM Janssen et al., Am J Physiol 1997, 273: 789-96) that include signaling IFN-alpha hindered (D Di Bona et al., J Hepatol. 2006, 45, 271-9). HCV infection is divided according to ICD10 (WHO, 2007 version) into acute (B17.1) and chronic (B18.2) hepatitis C. HCV is one of the most important causes of acute and chronic hepatitis developments. The clinical course of the disease, however, can be very different and subject to wide variability. Therefore, it is not possible to talk about a typical course of the disease, since HCV infection is essentially manifested by a broad clinical spectrum, that is, by variable symptoms, different clinical scenarios and secondary hepatic and extrahepatic secondary diseases. In approximately 20% of patients with acute hepatitis, liver inflammation is attributed to an HCV infection. In the acute phase, however, hepatitis C usually proceeds asymptomatically and, therefore, is not diagnosed in approximately 85% of cases. In some cases, only non-specific symptoms of a falsely flu-like syndrome occur. Usually, the infection manifests during the acute phase. Hepatitis C becomes chronic in approximately 85% of patients with an acute HCV infection. This high chronification rate seems to be the result of the wide variability of HCV virus; that is, the gene encoding the HCV layer is subjected to a high mutation rate. Due to the high variability of the virus and, in particular, the high variability of the HCV antigenic epitope, the mutated HCV escapes recognition by the human immune system. Approximately 25% of patients, as a result of chronic inflammation of the liver, cirrhosis of the liver occurs with an increased risk of liver carcinoma development (see, for example, the publications of JH Hoofnagle, Hepatology 1997, 26, Suppl. 1, 15S-20S; MI Memon et al., Journal of Viral Hepatitis 2002, 9, 84-100; SL Tan et al., Nature Reviews, Drug Discovery 2002, 1, 867-81). Patients who are infected with HCV routinely receive a standard medicinal combination therapy 5 10 fifteen twenty 25 30 35 40 Four. Five fifty 55 consisting of pegylated interferon-a2a or interferon-a2b and ribavirin. In HCV infections due to genotype 2 or 3 (HCV2 or HCV3 infections), this combination therapy is carried out for 24 hours. In HCV1 positive patients of HCV infections due to genotype 1 (HCV1) combination therapy is carried out for 48 hours. However, many of the patients infected with HCV, however, discontinue treatment due to side effects that occur and / or low compliance as a result of parenteral administration and prolonged treatment period. In addition, only about 50% of patients who have HCV1 infection achieve a long-term treatment outcome, that is, the rest do not respond (see, for example, the publication of RET Smith, Nature Reviews, Drug Discovery, 2006 , 5, 715). Pegylated interferon plus ribavirin therapy for hepatitis C virus fails in approximately half of genotype 1 patients. Treatment failure is caused by an absence of response (minimal decrease in viral titration) or relapse (initial responses considerable followed by bounces of viral titers during therapy or afterwards). These different models could be affected by many factors that include the host's genetic characteristics, immune response and viral genetic differences (see publications by MW Fried et al., New England Journal of Medicine 2002, 347, 975-82; HS Conjeevaram et al., Gastroenterology 2006, 131, 470-7; MP Manns et al., Lancet 2001, 358, 95865; DB Strader et al., Hepatology 2004, 39, 114771; SJ Hadziyannis et al., Ann Intern Med, 2004, 140, 346-55). Viral genetic differences could include pre-therapy differences or differences that arise during treatment due to viral evolution in response to the pressures applied by the therapy. New treatments are being developed, including the optimization of the current standard treatment with peginterferon plus ribavirin, antiviral therapy specifically targeted for HCV, new immunomodulatory agents and treatments aimed at reducing fibrosis (see RE Stauber et al., Drugs 2008, 68 (10), 1347). To this day, no vaccine against HCV can be obtained. Standard medicinal therapies are very expensive, show only reduced success in the control of HCV infection and sometimes cause considerable side effects (SL Tan et al., Nature Reviews, Drug Discovery 2002, 1, 867; R. Bartenschlager, ibid. 911). There is a need for medications for the treatment of viral hepatitis, in particular hepatitis B and C. It is an object of the invention to make available a medicament for the treatment of viral hepatitis, in particular hepatitis C, which has advantages compared to prior art medications. The drug, if possible, should have no or only slight side effects and be effective, for example, in hepatitis C patients who do not respond sufficiently to conventional combination therapy with PEG interferon / ribavirin. In addition, the drug must have considerable antiviral properties and, therefore, decrease the virus load in a lasting way. This objective is achieved by the content object of the patent claims. Surprisingly it has been found that silibinin, its pharmaceutically tolerable salts and / or derivatives are suitable in the treatment of inflammatory viral liver diseases, in particular hepatitis C. Therefore, hepatitis C patients who do not respond (i.e., are so-called "non-sensitive") to an immunomodulatory / antiviral combination therapy such as PEG interferon / ribavirin, which currently represents the standard treatment for hepatitis C, a significant reduction in viral load can be achieved by administration, preferably by administration. parenteral of a silibinin component. Additionally, it seems that pretreatment with the silibinin component improves the patients' response to the subsequent administration of interferon and ribavirin. Research on the treatment of HCV infections, particularly for the inhibition of HCV infections by administration of silymarin, has been described in the prior art (see, for example, the publications of R. Saller et al., Drugs 2001, 61 (14), 2035-63; KE Mayer et al., Journal of Viral Hepatitis, 2005, 12, 559-67; US2005 / 0123628; SJ Polyak et al., Gastro-enterology 2007, 132, 1925-1936). R. Saller et al. They report that, although silymarin is not known to affect viral replication, from a pharmacological perspective it can be expected to inhibit the inflammatory and cytotoxic cascade of events triggered by viral infection. Oral administration of a silibinin-phosphatidylcholine complex (IdB1016, 240 mg of silibinin twice daily) in a short-term, placebo-controlled pilot study in 20 patients with chronic active hepatitis revealed that the evolution of AST levels it was significantly reduced in the silibinin group, without congruent differences in the other tests of viral functions (see the publications of A. Vailati et al., Phytotherapy, Volume LXIV, No. 3, 1993; G. Buzzelli et al., Int J. Clin. Pharmacol. Ther. Toxicol. 1993, 31, 456-60). K. E. Mayer et al. describe that oral silymarin treatment resulted in a decrease in serum transaminases compared to the reference in four studies and compared to placebo in only one study. However, there is no evidence that silymarin affects viral load or improves liver histology in 5 10 fifteen twenty 25 30 35 40 Four. Five fifty 55 hepatitis B or C (see publications by M.L. Chavez, J. Herb. Pharmacother. 2001, 1 (3), 79-90; L.B. Seeff et al., Hepatology, 2001, 34 (3), 595-603). The authors conclude that silymarin compounds probably decrease serum transaminases in patients with chronic viral hepatitis, but they do not appear to affect viral load or liver histology. US2005 / 0123628 relates, among other things, to the preparation and oral administration of compositions comprising glycyrrhizin, schisandra, ascorbic acid, L-glutathione, silymarin, lipoic acid and D-alpha-tocopherol. These compositions are said to be useful for reducing oxidative stress and lipid peroxidation and for treating chronic liver diseases, chronic hepatitis C virus infection and non-alcoholic steatohepatitis. Numerous studies have reported the hepatoprotective effects that silymarin has against a wide variety of toxins including acetaminophen, ethanol, carbon tetrachloride and D-galactosamine and against ischemic injury, radiation and iron toxicity. During the first twenty weeks of a trial in an open and randomized clinical center, subjects were given twice daily oral administration of a total of 1,000 mg of glycyrrhizin; three times a day of a total of 1,500 mg of schisandra extract; three times a day of a total of 6,000 mg of a = corbic acid; twice a day of a total of 300 mg of L-glutathione; three times a day of a total of 750 mg of milk thistle extract; twice a day of a total of 300 mg of lipoic acid and once a day of a total of 800 IU of D-alpha-tocopherol. During the first ten weeks of the study, the subjects were also given an intravenous (iv) injection twice a week of four different parenteral compositions, none of which contained silymarin. After 10 weeks, 12.0% of the subjects and after 20 weeks 24.0% of the subjects showed a reduction of 1 viral load record. There is no evidence in US2005 / 0123628 that silymarin, let alone silibinin, may be responsible for this comparatively slight reduction in viral load. S.J. Polyak et al. they compare in vitro a standardized silymarin extract (MK-001) with commercial silymarin preparations. Both preparations are said to show an antiviral activity in models based on cell cultures used, although the effects of commercial preparations were not as potent as MK-001. MK-001 inhibits the expression of tumor necrosis alpha factor in human peripheral blood mononuclear cells stimulated with anti-CD3 and nuclear kappa B factor dependent transcription in human hepatoma Huh7 cells. In addition, the dose of MK-001 dependently inhibits the infection of Huh7 and Huh7.5.1 cells by the JFH-1 virus. MK-001 shows effects against HCV infection of isolated cells and, when combined with interferon-a, inhibited HCV replication more than interferon-a alone. To compare the anti-HCV action of MK-001 with commercial silymarin preparations, Ultrathistle® (Natural Wellness, Montgomery, NY) and Silybinin® (Indena SpA, Milan) were also tested. However, MK-001 is said to cause a more potent viral action than Ultrathistle® and Silybinin®. The authors conclude from these in vitro tests that, as regards anti-HCV activity, the standardized silymarin extract MK-001 is superior to two commercial products. S.J. Polyak says nothing about parenteral administration of purified silibinin, not least about the treatment of non-sensitive people. In addition, the findings of Polyak et al. in disagreement with clinical studies that found no effect of silymarin on HCV in patients with chronic hepatitis C (MD Tanamly et al., Dig Liver Dis. 2004, 36, 752-9; E Gabbay et al., World J Gastroenterol. 2007 , 13, 5317-23). Berkson (MEDLINE, 1999) describes a conservative triple antioxidant proposal for the treatment of hepatitis C. WO 00/32177 describes the use of 17-ketosteroid compounds, as well as the derivatives, metabolites and precursors of these compounds and pharmaceutically acceptable salts of any of these compounds in the treatment or prevention of hepatitis C virus and / or virus Hepatitis G type in patients who need this treatment. WO 02/067853 describes the combination of a selective COX-2 inhibitor and cystine first, for the treatment of antiviral diseases, including HIV, immunosuppressed individuals, AIDS and hepatitis C. Ciriaci et al. (Gastroenterology, vol. 112, 1997, A1246) describes a randomized trial concerning interferon alfa versus silibinin plus interferon in chronic hepatitis C previously resists interferon. It has now been surprisingly found that administration, particularly parenteral administration, of a preferably pure silybinin component reduces viral load in viral hepatitis patients in vivo. Therefore, the silibinin component is able to reduce the viral load. This discovery allows optimizing the dose of silibinin in the absence of other silymarin constituents that may cause unwanted side effects. The reduction of viral load by parenteral administration of a silibinin component is particularly surprising, as clinical studies found an absence of the effect of silimama on HCV in patients with chronic hepatitis C (M Torres et al., PR Health Sci J 2004, 23 (2), 69-74; MD 5 10 fifteen twenty 25 30 35 40 Four. Five fifty Tanamly et al., Dig Liver Dis., 2004, 36: 752-9; A Gordon at al., J Gastroenterol Hepatol. 2006, 21, 275-80; E Gabbay et al., World J Gastroenterol. 2007, 13, 5317-23; and LB Seeff et al., Hepatology, 2008, 80 (11), 19006). M Torres et al. They report on a clinical trial in which the participation of patients aged 21-65 years with a diagnosis of chronic hepatitis C, who were not using antiviral therapy, was requested. 34 patients were randomized to a treatment with S. marianum, 160 mg orally, three times a week for four weeks or no treatment (control). The trial showed that S. marianum has no function as an antiviral agent. MD Tanamly et al. report on a clinical trial in which 177 patients with chronic hepatitis C virus were randomly assigned to receive silymarin or multivitamin supplements. The trial showed that the recommended dose of silymarin had no effect on hepatitis C virus viremia. To Gordon et al. report on a clinical trial in which 24 subjects with chronic hepatitis C were recruited for a double-blind, randomized, cross-controlled, placebo-controlled study. Subjects received 12 weeks of S. marianum (600 mg or 1,200 mg / day) and placebo. Reference biochemical, virological, psychological and quality of life trials were performed. Seventeen patients completed the trial. The trial showed that the mean changes in HCV aRn titers were not significantly different from those on S. marianum compared to placebo. E. Gabbay et al. report on a clinical trial in which 100 patients with chronic HCV infection who had failed in interferon treatment were recruited and randomly assigned to receive seven different antioxidants, including silymarin capsules, 250 mg, tid. The primary endpoints were liver enzymes, HCV-RNA levels and histology. The trial showed that antioxidant therapy had no treatment effect on viral load. LB Seeff et al. report on the trial of the long-term antiviral treatment of hepatitis C against cirrhosis (HALT-C), which involved people with advanced chronic hepatitis C, not sensitive to a previous antiviral therapy but who were still eager to participate in a treatment with pegylated interferon long-term. No beneficial effect of silymarin was found on hepatis C virus (HCV) RNA levels. In conclusion, silymarin users had HCV levels similar to non-users. In addition, it has been surprisingly found that administration, particularly parenteral administration of a silibinin component supports conventional treatment by peginterferon / ribavirin. It was found that the silibinin component (re) activates the susceptibility of patients to a conventional treatment by peginterferon / ribavirin and / or increases the antiviral effect of a conventional treatment by peginterferon / ribavirin. Brief description of the figures: Fig. 1: Example 1, study 1: Oxidative tension parameters during and after an infusion of 10 mg / kg of silibinin component for 4 hours. (Test of d-ROMs = compounds derived from reactive oxygen metabolites, BAP test = biological antioxidant potential). Fig. 2: Example 1, study 1: HCV-RNA (IU / ml record; mean ± SD) before (day 1) and after 7 days of 10 mg / kg iv of silibinin component / day. Fig. 3: Example 1, study 1: Changes in HCV-RNA after iv administration of 10 mg / kg / day of silibinin component for 7 days, followed by combination therapy with peginterferon alfa 2a / ribavirin and 140 mg of Silymarin TID. Fig. 4: Example 1, study 2: Changes in HCV-RNA during iv administration of silibinin component at various doses for 14 days, followed by combination therapy with peginterferon alfa 2a / ribavirin that was started on day 8. Fig. 5: Example 1, study 2: Mean decrease (± SD) of HCV-RNA during 7 days of iv administration of silibinin component monotherapy and 7 days of iv silibinin component in combination with peginterferon alfa 2a / ribavirin to various dose. Fig. 6: Example 1, study 2: Changes in HCV-RNA after completion of silibinin iv (week 2) in the 14 patients who received 15 or 20 mg / kg / silibinin / d. Combination therapy with peginterferon alfa 2a / ribavirin was started on day 8 and 280 mg of silymarin TID on day 15. Fig. 7: Example 2, individual patient, changes in HCV-RNA after iv administration of 20 mg / kg / day of silibinin component during two administration intervals comprising 14 consecutive days, beginning the first iv administration interval in week 24 and beginning the second interval of 5 10 fifteen twenty 25 30 35 40 Four. Five fifty administration at week 35, during a continuous combination therapy with 180 pg of peginterferon alfa 2a / ribavirin for 60 weeks. Fig. 8: Example 2, individual patient, changes in HCV-RNA after an iv administration of 20 mg / kg / day of silibinin component during an administration interval comprising 14 consecutive days and starting at week 32 during a Continuous combination therapy with 180 pg of peginterferon alfa 2a / ribavirin for 60 weeks. Fig. 9: Example 2, individual patient, changes in HCV-RNA after an iv administration of 20 mg / kg / day of silibinin component during an administration interval comprising 14 consecutive days and starting at week 72 during a Continuous combination therapy with 180 pg of peginterferon alfa 2A / ribavirin for 80 weeks. Fig. 10 schematically shows various ways of co-administration of ribavirin and / or pegylated interferon alpha and the drug containing the silibinin component. Fig. 11 shows data generated from an in vitro NS5B inhibition study for six purified silymarin constituents. Fig. 12 shows data generated from an in vitro NS5B inhibition study for silibinin bis (hydrogensuccinate). The invention relates to the use of a silibinin component for the production of a medicament, preferably virustatic or antiviral, more preferably reducing the viral load, for the treatment of viral hepatitis, in particular hepatitis C, preferably of virus infections. Chronic or acute hepatitis C, Preferably by parenteral administration. For the purposes of the specification, the term "medication" is preferably synonymous with the term "medication." The invention relates to the use of a silibinin component for the production of a medicament that essentially does not contain silidianine and / or does not contain silicristine and / or does not contain isosilibinin, for the treatment of hepatitis C. In a preferred embodiment, according to the invention, the treatment of viral hepatitis, in particular hepatitis C, is carried out by decreasing the virus load (viral load). It has been found that silybinin components are able to reduce the viral load of hepatitis B or C patients. This is particularly surprising, since in the prior art there is no evidence that silymarin, which is a mixture that contains a certain amount of silibinin, affects viral load or improves liver histology in hepatitis B or C (see the publication of KE Mayer et al., Journal of Viral Hepatitis, 2005, 12, 559-67). In another preferred embodiment according to the invention, the treatment of viral hepatitis, in particular hepatitis C, is carried out in patients who will experience or have undergone a liver transplant. Patients who have undergone a liver transplant due to viral hepatitis are at risk of restoring viral hepatitis in the recently transplanted liver. Usually, the virus is incompletely suppressed from the body when the infected liver is removed after surgery and the rest of the viruses retained in the body can infect the newly transplanted liver again. In patients infected with chronic hepatitis C, re-infection after liver transplantation occurs in 100% of cases. As it has surprisingly been found that silibinin is capable of decreasing the burden of the virus, the risk of reinfection after liver transplantation can be substantially reduced by the administration, preferably parenteral administration, of a silibinin component. Forms of viral hepatitis are known to those skilled in the art. In viral hepatitis, at least six different forms are definitely known: hepatitis A, B, C, D, E and G. The organisms that cause these infections are hepatotropic viruses. They belong to different virus families in each case and have a DNA or RNA genome. Transmission takes place through food or through the exchange of body fluids such as sperm and blood. Differences have also been observed between the different forms with respect to the course of the disease and the severity of the disease. Although hepatitis A and E occur basically in acute form, hepatitis B, C and D can lead to chronic courses, in some with serious complications. For the purposes of the description, the term "viral hepatitis" preferably comprises hepatitis B and C. In a preferred embodiment, the treatment is carried out by reducing the virus load of one or more viruses selected from the group consisting, but not limited to, of the HCV1, HCV2, HCV3, HCV4, HCV5 and HCV6 genotypes, preferably HCV1. 5 10 fifteen twenty 25 30 35 40 Four. Five fifty 55 If the genotype involved is HCV1, subtypes 1a, 1b, 1c, 1d, 1e, 1f, 1g, 1h, 1i, 1j, 1k and 1l are preferred. If the genotype involved is HCV2, subtypes 2a, 2b, 2c, 2d, 2e, 2f, 2g, 2h, 2i, 2j, 2k, 2, 2m, 2n, 2o, 2p and 2q are preferred. If the genotype involved is HCV3, subtypes 3a, 3b, 3c, 3d, 3e, 3f, 3g, 3h, 3i, 3j and 3k are preferred. If the genotype involved is HCV4, subtypes 4a, 4b, 4c, 4d are preferred. , 4e, 4f, 4g, 4h, 4i, 4j, 4k, 4l, 4m, 4n, 4o, 4p, 4q, 4r and 4t. If the genotype involved is HCV5, subtype 5a is preferred. If the genotype involved is HCV6, subtypes 6a, 6b, 6c, 6d, 6e, 6f, 6g, 6h, 6i, 6j, 6k, 6, 6m, 6n, 6o, 6p and 6q are preferred. With regard to the nomenclature of the genotypes and subtypes of the hepatitis C virus, reference may be made, for example, to the publication of P. Simmonds et al., Hepatology, 42, 2005, 962-73. The invention relates to the use of a silybinin component for the production of a medicament, which is preferably adapted for parenteral administration, for the treatment of hepatitis C in patients who do not respond to a conventional immunomodulatory / antiviral combination therapy as a therapy. of ribavirin / interferon ("non-sensitive") and / or patients who partially respond to a conventional combination immunomodulatory / antiviral therapy such as a ribavirin / interferon ("partially sensitive") therapy and / or patients who show a considerable initial response followed relapses of viral titers during therapy or later ("with relapse"). The invention also relates to the treatment of viral hepatitis C by means of a silibinin component, treatment that is subsequent to a conventional combination therapy by means of ribavirin / interferon. Preferably, therapy by administration of a silibinin component begins after ribavirin / interferon therapy has failed (either initially or after a certain period of treatment). In relation to conventional hepatitis C therapy by administration of ribavirin / interferon, the terms "non-sensitive", "partially sensitive" and "relapse" are known to those skilled in the art. Currently, pegylated interferon plus ribavirin therapy for hepatitis C virus fails in approximately half of genotype 1 patients. Treatment failure occurs through absence of response (minimal decrease in viral titer) or relapse (responses considerable initials followed by relapses of viral titers during therapy or afterwards). For descriptive memory purposes, a non-sensitive patient is preferably considered as a patient who does not show a decrease in viral load by <2 log10 IU / ml (i.e. factor 100) when ribavirin / interferon is administered (usually peg- interferon a), preferably for 12 weeks. In a preferred embodiment, non-sensitive patients have decreases in viral titers of <2.1 log10 IU / ml and absolute titers> 4.62 log10 IU / ml at the lowest point. For descriptive memory purposes, a partially sensitive patient is preferably considered as a patient who does not show a decrease in viral load <2 log10 IU / ml at week 12 with HCV RNA detectable at week 24. For the purposes of descriptive memory, a patient with relapse is preferably considered as a patient who has a decrease in viral titres of> 2.8 log10 and his absolute titre falls temporarily below the detection limit (2.78 log10 IU / ml) For the purposes of the description, the term "medication" is preferably synonymous with "method of administration" or "dose unit." For example, a medicament for oral administration is related, for example, in the form of a tablet, and this tablet is preferably the unit of dose to be administered, which contains the dose of silybinin component intended for the time of respective administration in a treatment scheme. If the dose unit comprises a single tablet, the dose unit corresponds to the method of administration. However, it is also possible that the dose unit is divided into a certain number of administration forms, for example, a certain number of tablets, in which case each contains only a partial dose, but in the whole the total dose of the Silibinin component, which is intended for the respective time of administration in a treatment scheme (these dose unit tablets are then intended for essentially simultaneous administration). For the purposes of the description, the term "silybinin component" preferably refers to silybinin, including all its stereoisomers, for example, silybinin A and silybinin B, their pharmaceutically tolerable salts and / or derivatives, in particular esters. Preferred esters are derivatives of inorganic acids such as phosphoric acid or sulfuric acid; or of organic acids such as formic acid, acetic acid, propionic acid, citric acid, malic acid, mandelic acid and the like. Particularly preferred are hemiesters of dicarboxylic acids, for example malonic acid, glutaric acid, succinic acid, adipic acid, subteric acid, azelaic acid, sebacic acid, fumaric acid, maleic acid, itaconic acid, phthalic acid, terephthalic acid, isophthalic acid, etc. . Preferred hemiesters are dihemisuccinates, which may be present as free acids or as salts, for example, such as sodium, potassium or ammonium salts. One or more of the silybinin hydroxyl groups may be esterified. Preferably, 1, 2, 3, 4 or all of the hydroxy groups of the silybinin are esterified. In a preferred embodiment, the component the silibinin component is C-2 ', 3-bis (hydrogensuccinate) of 5 10 fifteen twenty 25 30 35 silybinin or a pharmaceutically acceptable salt thereof, such as sodium salts, potassium salts, ammonium salts and the like, as well as mixtures thereof. Disodium salt is particularly preferred. Suitable esters are also esters of gluconic acid. Preferably, the silibinin component is a compound of general formula (I) image3 in which R1, R2, R3, R4 and R5 are independently selected from each other from the group consisting of -H, - SO3H, -PO3H2, -CO-C1-C8 alkylene-alkylene-OH, -CO-alkylene-C1C8-CO2H , -CO-C1-C8-SO3H alkylene, -CO- C1-C8-OPO3H2 alkylene, -CO-C1-C8-PO3H2 alkylene, - (C2-C3-O alkylene) nH where n = 1 to 20, - CO-C1-C8-N alkylene (C1-C3 alkyl) 3 + X ', wherein X- is a pharmaceutically tolerable anion, or its pharmaceutically tolerable salts. Preferably, R1, R2 and R5 are -H. More preferably, the silibinin component of the general formula (I) has the stereochemistry of the general formula (I-A) or (I-B): image4 In a preferred embodiment, the compound of the general formula (I-A) is mixed with the compound of the general formula (I-B) in any relative weight ratio, for example, 50 ± 5: 50 ± 5. However, in a preferred embodiment, the diastereomeric excess of the compound of the general formula (IA) is at least 50% of, more preferably, at least 75% of, even more preferably at least 90% of, even more preferably at least 95% of, most preferably at least 98% of and in particular at least 99% of. In another preferred embodiment, the diastereomeric excess of the compound of general formula (IB) is at least 50% of, more preferably, at least 75% of, even more preferably at least 90% of, even more preferably at least 95% of , most preferably at least 98% of and, in particular, at least 99% of. Other preferred silybinin components are described in WO 03/090741, to which reference is made in its entirety. Preferably, the silibinin component in pure water at room temperature has a better solubility than silibinin as such. In a preferred embodiment, the invention relates to the use of a silybinin ester for the production of a medicament, which is preferably formulated for parenteral or oral administration, for the treatment of viral hepatitis, in particular hepatitis C. The medicament essentially does not. It contains silidianine and / or does not contain silicristine and / or does not contain isosilibinin. In a preferred embodiment, the medicament is formulated for parenteral administration. Parenteral administration can be carried out, for example, by subcutaneous, intravenous, intramuscular, intraarterial, intraperitoneal, intracutaneous, intraarticular, intrathecal, intracardial, intravitreal, retrobulbar, intrapulmonary and intraosseous routes. Particularly preferably, the medicament is formulated for an injection or infusion, in particular 5 10 fifteen twenty 25 30 35 40 Four. Five fifty for intravenous or intraarterial administration. Suitable medications that are appropriate for an injection or infusion are known to those skilled in the art. In this regard, for example, reference may be made in its entirety to the publication of K. H. Bauer et al., Lehrbuch der Pharmazeutischen Technologie [Textbook of Pharmaceutical Technology], WVG Stuttgart 1999. Medications that are suitable for an injection are usually sterilized solutions, emulsions or suspensions, which are prepared by dissolving, emulsifying or suspending the active substance and, optionally, additional excipients in water, in a suitable non-aqueous liquid that does not have to be sterilized if warranted, or a mixture of these vehicles. Medications that are suitable for an infusion are usually aqueous solutions or emulsions sterilized with water as a continuous phase. Medications for an injection or infusion may optionally contain additional excipients. The type excipients are preferably solubilizers such as, for example, lecithin and poloxamer 188, substances for isotonicity such as, for example, sodium chloride, glucose and mannitol, buffers such as, for example, acetate, phosphate and citratate buffers, antioxidants such as, for example, ascorbic acid, sodium metahydrogen sulphite, sodium sulfite and sodium hydrogen sulphite, chelating agents such as, for example, disodium edetate, preservatives such as, for example, esters of p-hydroxybenzoic acid, benzyl alcohol and chlorocresol and emulsifiers such as, for example, lecithin, fatty acids, sterols, sorbitan fatty acid esters, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene fatty acid glycerides, polyoxyethylene fatty acid esters, polyoxyethylene fatty alcohol ethers, acid esters fatty acids of glycerol and poloxamers. A particularly preferred medicament is a powder for the preparation of an infusion solution comprising C-2 ', 3-bis (hydrogensuccinate) of silybinin, preferably as the disodium salt and optionally inulin as an excipient. Containers containing 598.5 mg of C-2 ', 3-bis (hydrogensuccinate) disodium salt powder of silibinin and inulin are adapted for the preparation of an infusion solution and are marketed in Germany under the trademark Legalon ® SIL. In a preferred embodiment, the medicament according to the invention is bioequivalent for this formulation. In another preferred embodiment, the medicament is formulated for oral administration. Preferably, the medicament is a form of oral administration selected from the group consisting of tablets, capsules, sugar-coated tablets, granules and sachets. When a silibinin component is administered through the oral route, it should be ensured that the bioavailability of the silibinin component from the oral dosage form is sufficiently high. In this regard, the limiting factor is considerable lipophilicity of silibinin. In a particularly preferred embodiment, the invention relates to the use of a silibinin component for the production of a medicament that is formulated for oral administration and essentially does not contain silidianine and / or does not contain silicristine and / or does not contain isosilibinin, for the treatment of hepatitis C. It seems that these additional constituents of silymarin also have a physiological effect (for example, they may cause side effects), but that with respect to the treatment of viral hepatitis, silibinin (or its analogues) is the most effective, particularly to reduce the burden viral. Therefore, when silymarin is administered, that is, a mixture of silibinin, silidianine, silicristine, isosilibinin and other constituents, the overall dose of silymarin has to be comparatively high in order to provide a particular amount of silybinin. For example, when silymarin contains, for example, 42% p of silibinin, the administration of 125 mg of silymarin only provides approximately 52 mg of silybinin and approximately 73 mg of other compounds that also have a physiological effect (but not the desired effect ). The risk of unwanted side effects increases with the dose of a physiologically active substance. Therefore, as regards the profile of unwanted side effects, the administration of 52 mg of substantially pure silibinin is superior to the administration of 125 mg of silymarin having a silibinin content of 42% p (see T. Ding et al., "Determination of active component in silymarin by RP-LC and LC / MS", J. Pharm. Biomed. Anal. 2001, 26 (1), 155-161). The structures of silibinin (silybin), silidianine (silydianin), silicristine (silychristin) and isosilibinin (isosilybin) are set out below, see D.Y.-W. Lee et al., J. Nat. Prod. 2003, 66, 1171-4; N.- C. Kim et al., Org. Biomol Chem., 2003, 1, 1684-9): 5 10 fifteen twenty 25 30 35 40 image5 silybinin silicristina image6 Isosilibinin Silidianine Suitable forms of administration that are appropriate for oral administration (oral medications) are known to those skilled in the art. In this regard, reference may be made in its entirety, for example, to the publication of K. H. Bauer et al., Lehrbuch der Pharmazeutischen Technologie [Textbook of Pharmaceutical Technology], WVG Stuttgart 1999. The oral administration form is preferably selected from the group consisting of tablets, powders, granules, granules, sugar-coated tablets, syrups, juices, solutions, effervescent powders, effervescent granules, effervescent tablets, lyophilized tablets and capsules. Particularly preferably, the oral administration form is a tablet, a tablet coated with sugars, granules, granules or powder, particularly a tablet. Suitable excipients for the formulation of oral administration forms are known to those skilled in the art. Reference may be made in this regard, for example, to the publication of H. P. Fiedler, Lexikon der Hilfstoffe für Pharmazie, Kosmetik und angrenzende Gebiete [Encyclopedia of excipients for pharmacy, cosmetics and related areas], Editio Cantor Aulendorf, 2001. The tablets may be obtained, for example, by mixing the silibinin component with known excipients, for example, inert diluents such as calcium carbonate, calcium phosphate or lactose, disintegrants such as starch or alginic acid, binders such as starch or gelatin, lubricants such as stearate. of magnesium or talc and / or agents to achieve the deposition effect such as carboxymethyl cellulose, cellulose acetate phthalate or polyvinyl acetate. The tablets may also consist of a certain number of layers. Apart from the mentioned vehicles, the tablets may also contain additives such as, for example, sodium citrate, calcium carbonate and dicalcium phosphate, together with various additional substances such as starch, preferably potato starch, gelatin and the like. Additionally, flow adjuvants such as magnesium stearate, sodium lauri sulfate and talc can be used for tabletting. Sugar-coated tablets can be produced, for example, by coating the cores produced analogously to the tablets with agents commonly used in coatings of sugar-coated tablets, for example, colidone or lacquer range, gum arabic, talcum, titanium dioxide or sugar. To avoid a deposit effect or to avoid incompatibilities, the core may also consist of a certain number of layers. The sugar coated tablet layer may also consist of a certain number of layers to achieve the deposition effect, it being possible to use the aforementioned excipients in the case of tablets. Juices, syrups, emulsions, suspensions and solutions for oral administration may additionally contain a sweetener such as saccharin, cyclamate, glycerol or sugar and an agent for improving flavor, for example, flavors such as vanillin or orange extract. They may also contain auxiliary components for suspension or thickeners such as sodium carboxymethyl cellulose, wetting agents, for example, condensation products of fatty acids with ethylene oxide or preservatives such as esters of p-hydroxybenzoic acid. 5 10 fifteen twenty 25 30 35 40 Four. Five fifty 55 The capsules can be produced, for example, by mixing the silibinin component with inert vehicles such as lactose or sorbitol and encapsulating in gelatin capsules. As excipients there may be mentioned, for example, water, pharmaceutically acceptable organic solvents such as paraffins (for example, petroleum fractions), oils of vegetable origin (for example, peanut oil or sesame), mono- or polyfunctional alcohols (for example, ethanol or glycerol), vehicles such as, for example, crushed natural minerals (for example, kaolins, clays, talcum, limestone), crushed synthetic minerals (for example, silicic acid and highly dispersed silicates), sugars (for example, sucrose, lactose and dextrose), emulsifiers (for example, lignin, residual sulphite liquids, methyl cellulose, starch and polyvinyl pyrrolidone) and flow aids (for example, magnesium stearate, talc, stearic acid and sodium lauri sulfate). The medicine can release the silibinin component immediately or in a controlled way. If the release takes place in a controlled manner, the release preferably takes place in a delayed manner. Delayed release is understood according to the invention Preferably directed to a release profile in which the silibinin component is released for a relatively long period of time with a reduced rate of absorption for the purpose of prolonged therapeutic action. This is achieved, in particular, in the case of oral administration. The term "with at least partially delayed release" according to the invention comprises any medicament that guarantees a modified release of the silibinin component contained therein. Medications are preferably coated or uncoated forms of administration that are produced using special excipients, according to the particular procedures or by combination of both possibilities, in order to selectively modify the release rate or release site. With respect to the course of the release time, in the case of the medicaments according to the invention the following types are included: delayed release (extended release), repeated action release, prolonged release and sustained release. With regard to other details, reference may be made, for example, to the publication of K. H. Bauer et al., Lehrbuch der Pharmazeutischen Technologie [Textbook of Pharmaceutical Technology], 6th edition, WVG Stuttgart, 1999. Suitable measurements for the controlled release of active compound are known to those skilled in the art. If the medicament is a form of oral administration, for example, a tablet, a delayed release can be achieved, for example, by incorporating the silibinin component in a polymer matrix and / or by coating the oral administration form with a film by means of a membrane According to the invention, solid, semi-solid or liquid medicaments with controlled release behavior can be used. Solid medicines are preferred, such as oral osmotic systems (OROS), coated tablets, matrix tablets, multilayer tablets, jacketed tablets, tablets coated with jacketed sugars, diffusion granules, adsorbates and soft gelatin capsules. The oral medicament with controlled release of active compound is particularly preferably a coated tablet, jacketed tablet or matrix tablet, particularly preferably a matrix tablet. Medications with controlled release of active compound may contain the silibinin component in dissolved, suspended and / or solid, amorphous or crystalline form. For the production of the medicaments according to the invention with controlled release of active compound, the silibinin component can be used in various particle sizes, for example, without crushing, crushing or in micronized form. In drugs with controlled release of active compound, the silibinin component is preferably present in the form of particles containing active substance such as, for example, granules, granules, microcapsules, tablets, extrudates or crystals, which are coated with a membrane of controlled diffusion. The controlled diffusion medicaments are preferably multiparticles, that is, they preferably consist of a multiplicity of coated cores such as neutral granules, to which a mixture of the silybin component with a usual binder and thickener is applied, optionally together with usual excipients and vehicles and are sequentially coated with a diffusion lacquer, plasticizer and other excipients. The controlled diffusion medicaments according to the invention may also consist of homogeneous cores comprising the silibinin component, which are produced, for example, by granulation, rotor granulation, fluidized bed agglomeration, tablet formation, wet extrusion or extrusion in optionally molten state with spheronization and are coated with a diffusion lacquer that may contain plasticizers and other excipients. The particles containing the silibinin component may contain excipients such as, for example, acids or buffering substances, which modify the pH and thus contribute to reducing the dependence of the release of the silibinin component on the pH of the release medium. 5 10 fifteen twenty 25 30 35 40 Four. Five fifty 55 The controlled diffusion membrane may also contain other excipients which, due to their pH-dependent solubility, exert an influence on the permeability of the membrane at various pH values and thus contribute to minimizing the pH dependence of the release of the silybinin component. The binders and expectants used in the production of coated neutral granules are preferably hydroxypropylmethylcelluloses (HPMC) and polyvinylpyrrolidone (PVP). Similarly, other natural, synthetic or partially synthetic polymers such as, for example, methylcellulose (MC), hydroxypropylcellulose (HPC), other hydroxyalkylcelluloses and hydroxyalkylmethylcelluloses, carboxymethylcelluloses and their salts, poly (acrylic acids), polymethacrylates, gelatin, starch or other may be used. starch derivatives. For the production of granules, particles and (mini) tablets containing the component silybinin, cellulose, microcrystalline cellulose, cellulose derivatives such as, for example, HMPc, HPC and low substituted hydroxypropylcellulose (L-HPC), dicalcium phosphate, Lactose, PVP and sucrose are preferably used as binders and fillers by means of granulation, fluidized bed agglomeration, wet extrusion and tabletting. Melt extrusion granules are produced by incorporating the silibinin component into thermoplastic excipients. Suitable thermoplastic excipients are preferably HPC, HPMC, ethyl cellulose, hydroxypropylmethylcellulose acetate succinate (HPMCAS), PVP, vinyl pyrrolidone / vinyl acetate copolymer, polyethylene glycol, poly (ethylene oxide), polymethacrylates, polyvinyl alcohols (PVA alcohols) ( , partially hydrolyzed polyvinyl acetate (PVA), polysaccharides, for example, alginic acid, alginates, galactomannans, waxes, fats and fatty acid derivatives. In particles containing the silybinin component, it is also possible to incorporate pH modifying substances such as acids, bases and buffering substances. By adding these substances, it is possible to considerably reduce the pH dependence of the release of the silybinin component and its salts, hydrates and sorbates. The excipients used that modify the pH in the nuclei that contain the silibinin component are, for example, adipic acid, malic acid, L-arginine, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, succinic acid, citric acid, acid ethanesulfonic acid, 2-hydroxyethanesulfonic acid, fumaric acid, gluconic acid, glucuronic acid, glutamic acid, potassium hydrogen hydrogen carbonate, maleic acid, malonic acid, methanesulfonic acid, toluenesulfonic acid, tromethamine or tartaric acid. Preferably, citric acid, succinic acid, tartaric acid and potassium hydrogen hydrogen peroxide are used. For the production of the diffusion lacquer, ethyl celluloses (for example, Aquacoat® or Surelease®) and polymethacrylates (for example Eudragit® NE, Eudragit® RS and RL) are preferably suitable. However, other materials such as cellulose acetate and cellulose acetate butyrate can also be used as film-forming diffusion control polymers. In addition to the diffusion-controlling polymer, the diffusion lacquer may also contain other excipients with pH-dependent solubility such as enteric polymers such as cellulose phthalate, in particular cellulose acetate phthalate and hydroxypropyl methylcellulose phthalate, cellulose succinates, in particular cellulose acetate succinate and hydroxypropylmethylcellulose acetate succinate or polymethacrylates (for example, Eudragit® L). By adding these substances, it is possible to reduce the pH dependence of the release of the silybinin component. The plasticizers used are, for example, citric acid derivatives, phthalic acid derivatives, benzoic acid and benzoic acid esters, other esters of aromatic carboxylic acids, esters of dicarboxylic acids, aliphatic, mono-, di- or triacetate glycerol, polyols , fatty acids and their derivatives, acetylated fatty acid glycerides, castor oil and other virgin oils, miglyol and fatty acid alcohols. In order to avoid adhesion of the coated particles during production and in the finished product, non-stick lacquers such as talcum, magnesium stearate, glycerol monostearate and Aerosil can be added to the lacquer. The release rate is controlled by the composition of the lacquer and the thickness of the lacquer layer. Additives that increase the permeability of the film that are pore-forming agents or to the particles to be coated containing the silybinin component can be added to the lacquer. The pore-forming agents employed are soluble polymers such as, for example, polyethylene glycols, PVP, PVA, HPMC, HPC, hydroxyethylcelluloses (HEC), MC, carboxymethylcelluloses or their salts, dextrins, maltodextrins, cyclodextrins, dextrans or other soluble substances such as, for example, urea, sodium chloride, potassium chloride, ammonium chloride, sucrose, lactose, glucose, fructose, maltose, mannitol, sorbitol, xylitol and lactitol. Excipients with pH-dependent solubility that may be constituents of the diffusion film are, for example, enteric polymers such as cellulose phthalates, in particular cellulose acetate phthalate and hydroxypropyl methylcellulose phthalate, cellulose succinates, in particular acetate succinate Cellulose and hydroxypropylmethyl cellulose acetate succinate and polymethacrylates (eg, Eudragit® L). 5 10 fifteen twenty 25 30 35 40 Four. Five fifty 55 In addition, the drug with controlled release of the silibinin component may be a coated administration form containing one or more inflatable excipients that swell strongly by penetration of liquid through the membrane and cause the coating to tear open as a consequence. of swelling and volume expansion. As a consequence of the tearing opening of the coating, the release of pharmaceutical product from the drug (impulse release) is made possible. As inflatable excipients, these medicaments preferably contain polyvinylpyrrolidones, crospovidones, cross-linked sodium carboxymethylcellulose, cross-linked sodium carboxymethyl starch, poly (ethylene oxides), polymethacrylates, hydroxypropyl methylcellulose with low substitution (L-HPC). Suitable coating materials are preferably cellulose acetate, ethyl cellulose and polymethacrylates. The coated, diffusion or impulse controlled medications described can be used directly and unchanged in a pharmaceutical form. However, they can be further treated, optionally by the addition of excipients, to provide the final form of administration (eg, capsule, tablet, sachet). In order to achieve a desired release profile, several coated particles can also be combined with each other in a pharmaceutical form and administration of a dose can be initiated, for example, by the combination of quick-release particles, for example , granules, granules or uncoated powders. Medications with controlled release that can be used are also formulations comprising the silibinin component in a matrix. These matrix formulations release the silibinin component by diffusion and / or erosion. Preferably, these medicaments are present in the form of a tablet or in the form of a certain number of tablets that may be, for example, encapsulated. The tablets may be coated or lacquered. These medications are produced, for example, by mixing the constituents and producing tablets directly or by dry or wet granulation with subsequent tablet formation. The matrix forming agents used can be water soluble, water swellable or water insoluble substances. Preferably, the medicaments contain one or more water swellable polymers. The water-soluble or water-swellable matrix-forming polymers used are preferably hydroxypropylmethylcelluloses (HPMC), hydroxyethylmethylcelluloses, hydroxypropylcelluloses (HPC), hydroxyethylcelluloses, methylcelluloses (MC), ethylcelluloses, other alkylcelluloses, hydroxyalkylcelluloses alginates, galactomannans such as guar gum and locust bean flour, xanthan, poly (ethylene oxides), poly (acrylic acids), poly (methacrylic acids), derivatives of poly (methacrylic acid), poly (vinyl alcohols) ( PVA), partially hydrolyzed polyvinyl acetate (PVAc), polyvinylpyrrolidone (PVP), agar, pectin, gum arabic, tragacanth, gelatin, starch or starch derivatives and mixtures of this substance. The use of HPMC is particularly preferred. In addition, water-insoluble substances can be used as structure-forming agents, for example, unsaturated or saturated (hydrogenated) fatty acids and their salts, esters or amides, mono-, di- or triglycerides of fatty acids, waxes, ceramides, cholesterol derivatives and mixtures of these substances. The medicaments may also contain usual tablet-forming excipients, preferably highly dispersed silica (Aerosil®), magnesium stearate, talc, PVP, lactose or microcrystalline cellulose. In addition, the substances can be incorporated into the matrix that controls the pH in the matrix. By adding these pH modifying excipients and / or by adding substances that dissolve with an increasing pH or that dissolve the matrix and thus increase the porosity or permeability of the matrix and / or favor the erosion of the matrix, the it is possible that these preferred embodiments of the present invention achieve an almost independent release of pH. The matrix containing the silibinin component may also be present in special geometric shapes in which the release is influenced by the special geometry and the surface of the matrix. The matrix surface and the release surface can be controlled, for example, by compression to provide special formats (for example, annular tablets) and / or by sub-area coating or application of barrier layers by means of a multilayer press. Formulations with different release profiles may preferably be combined to provide a pharmaceutical form in multilayer or core-encapsulated tablets. For example, by means of multilayered tablets comprising a quick-release layer, or core-encapsulated tablets having a quick-release jacket, controlled releases according to the invention are achieved with a high initial release of the silybinin component, while by means of core-encapsulated tablets with a quick-release core, an accelerated final release can be achieved. An additional drug with controlled release of the silibinin component is one in which the silibinin component is incorporated into a matrix consisting of one or more physiologically excipients 5 10 fifteen twenty 25 30 35 40 Four. Five fifty 55 60 acceptable through a melt process. The release of the silybinin component from these “molten extruded materials” takes place through diffusion and / or erosion. Preferably, these formulations with controlled release of the silybinin component are present in the form of granules, granules or tablets. The forms obtained by extrusion in the molten state, in particular the granules and granules, can be treated to provide other pharmaceutical forms such as, for example, by encapsulation or tablet formation, optionally with the addition of pharmaceutically customary excipients. In addition, the molten extrudates according to the invention can be crushed and subsequently used in this way fractured for the production of other medicaments such as matrix tablets. The additional treatment also comprises the combination of formulations having a different pharmaceutical release, such as delayed and rapid release particles, to provide a medicament. The extrudates in the molten state and / or the pharmaceutical forms that are produced from the extrudates in the molten state can be coated or lacquered. The extrudates in the molten state are preferably produced by mixing the silibinin component with at least one pharmaceutically acceptable fusible excipient (vehicle) and optionally other usual additional pharmaceutical substances, which melt at a temperature in the range of 50 to 250 ° C, preferably of 60 to 200 ° C, injection or extrusion molding and forming. In the course of this, the mixing of the components may take place before melting or during melting, or some of the components are melted and the other constituents are added to this molten material. The mixture of the vehicle, the silibinin component and the additional substances optionally present are thermoplastically deformable and, therefore, can be extruded. Numerous methods suggest for themselves the conformation of the mixture, for example, hot granulation, cold granulation, calendering, extrusion and deformation of the still plastic or rounding thread. Thermoplastic vehicles used that are preferably swellable or soluble in physiological media are preferably: polyvinylpyrrolidone (PVP), N-vinyl pyrrolidone (NVP) copolymers and vinyl esters, in particular vinyl acetate, vinyl acetate and crotonic acid copolymers, poly ( partially hydrolyzed vinyl acetate, polyvinyl alcohol, cellulose esters, cellulose ethers, in particular methylcellulose and ethylcellulose, hydroxyalkylcelluloses, in particular hydroxypropylcellulose, hydroxyalkylmethylcelluloses, in particular hydroxypropylmethylcellulose and hydroxyethylmethylcellulose, carboxymethylcelluloses Cellulose phthalate and hydroxypropylmethylcellulose phthalate, cellulose succinate, in particular cellulose acetate succinate and hydroxypropylmethylcellulose acetate succinate, poly (hydroxyalkyl acrylates), poly (hydroxyalkyl methacrylates), polyacrylates (polymethacrylate® types) ), copolymers of methyl methacrylate and acrylic acid, polylactides, polyethylene glycols, polyethylene oxides and polysaccharides such as galactomannans and alginic acid and their alkali metal and ammonium salts. The preferred thermoplastic excipients for the production of drugs with controlled release of the silybinin component are HPC, PVP, vinyl pyrrolidone / vinyl acetate copolymers, polymethacrylates, in particular Eudragit® L, HPMCAS, polyethylene glycols, poly (ethylene oxides) and their mixtures The plasticizing excipients that can be used for reducing the glass transition temperature of the mixture are, for example, propylene glycol, glycerol, triethylene glycol, butanediols, pentanoles such as pentaerythritol, hexanols, long chain alcohols, polyethylene glycols, polypropylene glycols, polyethylene / polyethylene. , silicones, derivatives of phthalic acid (for example, dimethyl phthalate, diethyl phthalate or dibutyl phthalate), benzoic acid and benzoic acid esters, other esters of aromatic carboxylic acids (for example, trimellitic acid esters), derivatives of citric acid (for example, triethyl citrate, tributyl citrate, acetyltriethyl citrate), esters of dicarboxylic acids (for example, dedialkyl adipates, sebacic acid esters, in particular diethyl sebacate, tartaric acid esters), mono-, glycerol di- or triacetate, fatty acids and derivatives (for example, glycerol monostearates ol, acetylated fatty acid glycerides, castor oil and other virgin oils, miglyol), fatty acid alcohols (for example, cetyl alcohol or cetyl stearyl alcohol), sugars, sugar alcohols and sugar derivatives (for example, erythritol, isomaltitol , lactitol, mannitol, maltitol, maltodextrin or xylitol). In addition to the silybinin component, the vehicle (s) and optionally plasticizer (s), the extrudable mixture may still contain other additional pharmaceutically customary substances, for example, lubricants and mold release agents, flow aids and flow agents , fillers and adsorbents, stabilizers, free radical scavengers, complexing agents, antioxidants, photostabilizers, propellants, surfactants, preservatives, colorants, sweeteners and flavors. Lubricants and mold release agents may contain, for example, stearic acid and stearates, in particular aluminum, calcium and magnesium stearates, calcium behenate, sodium stearyl fumarate, talc, silicones, waxes, and mono-, di- and triglycerides such as, for example, glycerol monostearate, glycerol distearate, glycerol dibehenate, glycerol monooleate, glycerol palmostearate. The flow agents are preferably animal and vegetable fats, preferably in hydrogenated form and with a melting point of at least 50 ° C, waxes (for example, carnauba wax), mono-, di- and triglycerides (for example, monostearate of glycerol, glycerol distearates, glycerol di-behenate, glycerol monooleate, 5 10 fifteen twenty 25 30 35 40 Four. Five fifty 55 glyceryl palmito stearate), phosphatides, in particular lecithin. The fillers used are preferably substances such as titanium dioxide, aluminum oxide, magnesium oxide, silicic acid and silicates, stearic acid and stearates, cellulose derivatives (for example, methyl cellulose), starch and starch derivatives, sugars, alcohols of sugars and sugar derivatives. Medications with controlled release of the silybinin component may also be molten extrudates containing excipients with pH modifying properties and / or a pH dependent solubility. By means of these excipients (for example, acids, bases, buffer substances and enteric polymers already described above), it is possible to minimize the pH dependence of the release of the silybinin component. In the production of the extrudates in the molten state, the formation of "solid solutions" can occur, in which the silibinin component is present in the matrix in a molecularly dispersed form. Medications with controlled release of the silibinin component may also be osmotic pharmaceutical release systems. In principle, osmotic systems of this type are known in the prior art. In this case, pharmaceutical release from the pharmaceutical form is generally based on an osmotic pressure in the form of a driving force. The osmotic system preferably consists of a core containing the silybinin component, optionally a hydrophilic swelling agent and optionally a water soluble substance to induce osmosis and optionally other pharmaceutically acceptable excipients and a layer consisting of a water permeable material that It is impermeable to the core components and has at least one opening, through which the constituents present in the core can be released. The material from which these medicaments according to the invention are formed with a controlled release of the silybinin component is semi-impermeable, that is, permeable to water, aqueous media and biological fluids and nothing or very narrowly permeable to the core components, and Suitable for film formation. The selectively semi-impervious wrapping material is insoluble in body fluids, does not erode, does not degrade in the gastrointestinal tract and is excreted unchanged or shows bioerosion only at the end of the release period. Typical materials for the production of the layers of the osmotic system are preferably acylated cellulose derivatives (cellulose esters), which are mono- to trisubstituted with acetyl groups or mono- to disubstituted with acetyl groups and an additional acyl radical other than acetyl, for example, cellulose acetate, cellulose triacetate, cellulose acetate / ethyl carbamate, cellulose acetate phthalate, cellulose acetate methylcarbamate, cellulose acetate succinate, cellulose acetate dimethylaminoacetate, cellulose acetate diethylaminoacetate, acetate cellulose ethyl carbonate, cellulose acetate chloroacetate, cellulose acetate ethoxylate, cellulose acetate methylsulphonate, cellulose acetate butyl sulphonate, cellulose acetate propionate, cellulose acetate octoate, cellulose acetate laurate, acetate-p - cellulose toluenesulfonate, cellulose acetate butyrate and other derivatives of cellulose acetates and also agar acetate and amylose acetate. A suitable semi-impervious membrane material of the osmotic system is additionally ethyl cellulose, alkylene oxide and alkyl glyceryl ether copolymers, polymeric epoxides, polyglycols and poly (lactic acid) derivatives. In addition, it is possible to use water soluble acrylates by themselves, for example, a copolymer of ethyl acrylate and methyl methacrylate. If necessary, the layer of the osmotic system may also contain plasticizers such as, for example, the plasticizers already mentioned above and other additional substances such as, for example, pore-forming agents. If necessary, a photoprotective lacquer can be applied to the semi-impermeable layer which may consist, for example, of HPMC or HPC and a suitable plasticizer (for example, polyethylene glycol) and pigments (for example, titanium dioxide or iron oxides). In order to make possible the administration of an initial dose of the silybinin component, the osmotic system may also be provided with a layer containing the silybinin component, from which the silybinin component is preferably rapidly released by contact with the release medium before the osmotically controlled release of the silibinin component from the nucleus begins. Suitable water swellable polymers that may be present in the core of the osmotic system are preferably poly (ethylene oxides) (eg, Polyox®), xanthan gum, vinylpyrrolidone and vinyl acetate copolymers, polyvinylpyrrolidones, crospovidones, carboxymethylcellulose cross-linked sodium, cross-linked sodium carboxymethyl starch, low-substituted hydroxypropylmethylcellulose (L-HPC), poly (hydroxyalkyl methacrylate), alginates and galactomannans and also hydrophilic polymer swelling agents additions and mixtures thereof. 5 10 fifteen twenty 25 30 35 40 Suitable osmotically active substances that can be added to the core for the induction of osmosis water soluble salts of inorganic or organic acids or non-ionic organic substances with a high degree of water solubility such as, for example, carbohydrates, in particular Sugars or amino acids, by way of example, can be mentioned a few substances that can be incorporated into the core of the osmotic system individually or as a mixture for the induction of osmosis: inorganic salts such as chlorides, sulfates, sulphites, carbonates, bicarbonates, phosphates, hydrogen phosphates and dihydrogen phosphates of alkali metals and alkaline earth metals such as sodium, lithium, potassium, calcium or magnesium, organic acids such as adipic acid, ascorbic acid, succinic acid, citric acid, fumaric acid, maleic acid, tartaric acid , benzoic acid and other alkali metal or alkaline earth metal salts s, acetates, pentoses such as, for example, arabinose, ribose or xylose, hexoses such as, for example, glucose, fructose, galactose or mannose, disaccharides such as, for example, sucrose, maltose or lactose, trisaccharides such as, for example, raffinose, sugar alcohols such as mannitol, sorbitol, maltitol, xylitol or inositol, and urea. Sodium chloride and sodium carbonate are used particularly preferably. In addition, the osmotic system may contain other additional pharmaceutically common substances such as, for example, lubricants and mold release agents, flow aids, binders, colored pigments, thickeners, protective colloids, stabilizers and surfactants. The production of the osmotic release system is preferably carried out by means of standard techniques such as, for example, wet granulation or dry compaction, tabletting and subsequent organic coating. The osmotic system layer has at least one outlet opening, through which the silibinin component is released, optionally together with other constituents of the core. The opening can be introduced into the layers in various ways, for example, by puncturing, mechanical drilling or by means of a laser drill. The term "opening" also comprises bioerodible materials that dissolve the layer after the administration of this medicament according to the invention and, therefore, leads to the formation of outlet openings in situ. In a further embodiment for the controlled release of the silibinin component, the silibinin component may also be present in the form of an ion exchange complex (adsorbate). Preferably, the medicament is formulated for administration once a day (c.d.), twice a day (b.i.d.), three times a day (t.i.d.) or four times a day. In a preferred embodiment, 0.5 to 75% by weight of the originally contained silibinin component has been released from the medicament after 1 h under in vitro conditions. Suitable conditions for the determination of the in vitro release of active substance are known to those skilled in the art. In this regard, reference can be made, for example, to the European Pharmacopoeia. Preferably, the determination of the release is carried out by means of a paddle shaker in artificial gastric juice (buffer pH 1.2) or artificial intestinal juice (buffer 7.6). The amount of silibinin component released can be analyzed, for example, by means of HPLC and UV detection. Preferred release profiles A1 to A8 are summarized in the following table: after [h] A1 A2 A3 A4 A5 A6 A7 As % in P. % in P. % in P. % in P. % in P. % in P. % in P. % in P. 0.5 5.0-34 6.0-33 7.0-32 9.0-31 11-30 13-30 15-29 17-28 one 12-53 15-52 18-50 20-48 22-46 24-44 27-42 30-40 2 25-74 27-71 29-68 31-65 33-62 36-60 39-58 42-56 3 33-85 36-82 39-79 42-76 45-73 48-71 50-69 52-67 4 41-92 44-89 47-86 50-83 53-81 55-79 58-77 60-75 6 52-98 55-97 58-96 60-94 63-92 66-90 69-88 72-86 8 > 62> 65> 68 71-99 74-98 76-98 78-97 80-97 12 > 70> 73> 76> 79> 82> 84> 86> 88 5 10 fifteen twenty 25 30 35 40 Four. Five fifty 55 Pharmaceutical formulations containing silibinin and cyclodextrin are known in the prior art (see, for example, EP 422 497). Preferably, silibinin forms an inclusion complex with cyclodextrin. Preferred cyclodextrins are a-, p- and Y-cyclodextrins or their derivatives of O-C1-C4 alkyl and hydroxy C1-C4 alkyl. Pharmaceutical formulations containing silibinin and phospholipids are analogously known in the prior art (see US 4,764,508). Preferably, the silibinin complexes with the phospholipid. Preferred phospholipids are phosphatidylcholine, phosphatidylethanolamine and phosphatidylserine. Preferred silibinin phospholipid complexes are tertiary complexes that additionally contain vitamin E (atocopherol). Complexes of this type are known in the prior art as "SPV complexes" (A Federico, Gut. 2006, 55 (6), 901-2). In addition to the silibinin component, the medication may contain one or more terpenes. Through the action of terpene, both the absorption requirements and the absorption procedures and therefore the absorption can be improved globally. Terpenes can be ethereal, natural or synthetic oils and / or their terpenoid constituents in the form of pure substances or mixtures of derivatives of these pure substances. Among the ethereal oils, mention may be made in particular of thyme oil, eucalyptus oil, pinocha oil, tea tree oil, cajeput oil, cardamom oil, peppermint oil, sage oil and rosemary oil, preferably thyme oil For terpenes as substances that are also intended to include terpenoid substances, mention may be made, in particular, of hemiterpenes, such as, for example, isoprene, tigeric acid, angelic acid, isovaleric acid; monoterpenes, which include acyclic monoterpenes such as 2,6-dimethyloctane, a-myrcene, (E) -p-ocimeno, perylene, linalool, geranial, (S) - (+) citronelal and monocyclic monoterpenes such as, for example, cyclopropane-monoterpenes and cyclobutane-monoterpenes such as chrysanthemic acid or junionone, cyclopentane-monoterpenes such as, for example, iridoids or nepetalactones or (-) -secologanine and (-) - oleuropein, cyclohexane-monoterpenes such as o-menthane, cis- or trans-p-methane, (R) - (+) - limonene, terpinoles, (-) - menthol, (+) - perilaldehyde, (-) - chin or (+) - carvone, bicyclic monoterpenes such as bridged terpenes oxygen 1,4-cineole, 1,8-cineole, or ascaridol; the carano and tujano cyclopropane bicycles, the pinano cyclobutane bicycles and the canfano and fenchano bicycloheptans; sesqui-terpenes like famesano, bisabolano, germacrano, elemano and humulano. Particularly preferred terpenes are thymol, menthol, cineole, borneol, carvone, limonene and pinene, usually thymol. The medicine contains a component of silibinin. Silybinin is a constituent of silymarin. Preferably, in addition to the silybinin or the silybinin components, the medicament does not contain other silymarin constituents. If the silibinin component is silibinin as such, the medicament does not preferably contain other silybinin constituents. If the silybinin component is not silybinin as such, for example, a silybinin ester, the drug preferably contains no silymarin constituents at all, that is, neither silybinin. One or more of the substances selected from the group consisting of isosilibinin, silidianine, silicristine, taxifoline, isosylcycline, silimonin, silandrin, silihermine and neosilihermine are not contained in the medication, that is, the medication is essentially free of at least one of the above mentioned substances. In this respect, "essentially exempt" means that the residual content of the referred substance is less than 2.0% by weight, more preferably less than 1.0% by weight, even more preferably less than 0.5% by weight. , most preferably less than 0.1% by weight and, in particular, less than 0.05% by weight, based on the total weight of the medicament. Analytical methods for the determination of the residual content of these substances are known to those skilled in the art, for example, HPLC. It has been found that the individual constituents of silymarin differ in their chemical and physical properties and contribute to the pharmacological activity of silymarin to a very different extent, so that it is advantageous to administer silybinin or its derivatives and / or salts as the sole constituent of silymarin, that is, uniquely. It seems that in this way both efficiency and patient compliance can be improved. In addition, it has been surprisingly found that tolerance to various silymarin constituents differs from one to another and that silybinin is more tolerable, particularly less toxic, than silymarin (i.e., the mixture containing compounds other than silybinin). In a preferred embodiment, the invention relates to the use of a silybinin component for the production of a medicament that is preferably formulated for parenteral or oral administration and, apart from silybinin component, does not contain other silymarin constituents, for the treatment of viral hepatitis, particularly hepatitis C. Particularly preferred medications that are adapted for oral administration of the silibinin component are described below. All of these oral dosage forms have in common that they preferably contain the silybinin component in substantially pure form, that is, preferably in the absence of other silymarin constituents, particularly in the absence of isosilibinin and / or silicristine and / or silidianine. Preferably, oral dosage forms are immediate release dosage forms, that is, the 5 10 fifteen twenty 25 30 35 Silibinin component is rapidly released from them, thus leading to a rapid onset of the drug in the gastrointestinal tract. In a preferred embodiment, 30 minutes of administration of the oral dosage form, at least 75% p, more preferably at least 80% p, even more preferably at least 85% p, most preferably at least 90 % py, in particular, at least 95% p of the originally contained silibinin component has been released from oral dosing. In a preferred embodiment, the medicament is provided in the form of a solid solution, the solid solution is preferably carried out by incorporating the silibinin component in molecular dispersed form into a highly soluble, preferably amorphous, polymer matrix having a large specific surface area. The silibinin component must be present in molecular dispersed form, that is, in no microcrystalline or fine form. A highly soluble amorphous state may have already been achieved using highly soluble solid polymer solvents when silybinin or the silybinin component is extracted from silymarin extract. This technical formulation of the drug increases the solubility of the silybinin component and its dissolution rate. An example of this solid solution comprises the silibinin component, a suitable polymer (for example, polyvinylpyrrolidone (PVP), or a polyvinylpyrrolidone copolymer, such as Kollidon® 25) and, optionally, a dextrin (for example, maltodextrin). The formulation may contain other excipients such as aerosol and / or talkum. Preferred embodiments B1 to B6 of the solid solution are shown in the following table: % p B1 B2 B3 B4 B5 B6 Silibinin component 1.0-50 2.5-20 8.0 ± 5.0 8.0 ± 4.0 8.0 ± 3.0 8.0 ± 2.0 PVP 1.0-97 10-80 64 ± 15 64 ± 12 64 ± 10 64 ± 7.0 Dextrin 1.0-70 5.0-50 22.8 ± 20 22.8 ± 15 22.8 ± 10 22.8 ± 7.0 aerosil 0-10 0-7.5 4.0 ± 3.0 4.0 ± 2.5 4.0 ± 2.0 4.0 ± 1.5 talcum powder 0-5.0 0-2.5 1.2 ± 1.0 1.2 ± 0.7 1.2 ± 0.5 1.2 ± 0.3 The formulation can be provided, for example, in a hard gelatin capsule. In another preferred embodiment, the medicament is provided in the form of a self-emulsifying microemulsion. Self-emulsifying lipid systems can be used as carriers and can lead to a high bioavailability of the drug contained therein. The lipid system is colloidal in nature and this allows the resorption of microparticles, especially colloidal size, also through the lymphatic system in the gastrointestinal tract. Normally, the dissolved drug is saturated but recrystallization does not occur. After oral administration of lipophilic drugs, for example, of the silibinin component, the microemulsion primarily serves as an optimized vehicle that increases the dissolution rate of the dissolved or highly dispersed drug at the site of absorption. In other words, the lipid system acts as an absorption enhancer. An example of this lipid system comprises the silibinin component, a suitable first emulsifier (for example, lauroyl-macrogolglyceride, such as Gelucire® 44/14) and, optionally, a suitable second emulsifier (for example, caprilocapril-macrogolglyceride, such as Labrasol® ). The formulation may contain other excipients, such as polysorbate. Preferred embodiments C1 to C6 of the solid solution are shown in the following table: % p C1 C2 C3 C4 C5 C6 Silibinin component 0.1-50 0.5-20 4.0 ± 3.5 4.0 ± 3.0 4.0 ± 2.5 4.0 ± 2.0 First emulsifier 1.0-99 5-97 54 ± 15 54 ± 12 54 ± 10 54 ± 7.0 Second emulsifier 0-70 0-70 41 ± 20 41 ± 15 41 ± 10 41 ± 7.0 Polysorbate 0-10 0-7.5 1.5 ± 1.0 1.5 ± 0.7 1.5 ± 0.5 1.5 ± 0.3 The formulation, which may be solid or, preferably, semi-liquid, may be provided, for example, in a hard gelatin capsule or as a soft gelatin capsule. 5 10 fifteen twenty 25 30 35 40 nanotechnology The particle size of the nanoparticles is preferably below 1 | jm. The nanoparticles are capable of transferring biological membranes of cellular structures. The silibinin component is preferably adsorbed to the surface of said nanoparticles. The nanoparticles are preferably selected from the group consisting of inorganic nanoparticles and organic nanoparticles. Inorganic nanoparticles comprise crystalline silicates, for example, of mineral origin or artificial silicates such as metalosilicates, for example, alumosilicates (for example, zeolites). These inorganic nanoparticles are chemically modified Preferably so that they carry electrostatic charges. The silicates are crushed ultrafine to nanoparticles and the silibinin component binds (is adsorbed) to the microporous surface of the nanoparticles. Organic nanoparticles include clusters or agglomerates of small proteins or oligopeptides or lipids. A protein carrier is, for example, protamine. Methods for the preparation of nanoparticles are known to those skilled in the art. For example, colloidal nanoparticles can be prepared as carriers for oral drug release by spraying the drug, that is, the silibinin component, together with suitable carrier materials under pressure, for example, at 60 ° C, through injectors that They are equipped with perforated filters (matrices) in the form of heavily cooled towers. Spontaneous cooling forms an amorphous phase consisting of nanoparticles. Such lipid nanoparticles, for example, can be prepared by this high pressure homogenization and subsequent spray cooling. Preferably the drug, that is, the silibinin component, is employed in the form of a solution in a suitable solvent or in the form of submicroparticles. The silybinin component can be sprayed and pressurized-homogenized, respectively, in admixture with a lipid carrier and a surfactant, for example, at 6 ° C. After the optional addition of fine fillers such as an external phase, as well as flow aids and other surfactants, the formulation thus obtained can be introduced into hard gelatin capsules. An example of these solid lipid nanoparticles comprises a core of the silibinin component, a suitable first emulsifier (for example, stearoyl-macrogolglyceride, such as Gelucire® 50/13) and, optionally, a suitable macromolecular non-ionic surfactant (for example, a poloxamer ). The formulation also preferably contains an external phase (coating) comprising a first surfactant (for example, Tween 20), aerosol and a second surfactant (for example, glyceryl palmosteaarate, such as Percirol®). Preferred embodiments Di to D6 of the solid solution are shown in the following table: % p D1 Ü2 Ü3 Ü4 Ü5 Ü6 Silibinin component 0.1-30 0.5-20 4.5 ± 3.0 4.5 ± 2.5 4.5 ± 2.0 4.5 ± 1.5 First emulsifier 10-99 20-95 75 ± 20 75 ± 15 75 ± 10 75 ± 7.5 Non-ionic macromolecular surfactant 0-50 0-40 15 ± 10 15 ± 7.5 15 ± 5 15 ± 2.5 First surfactant 0-10 0.1-7.5 1.5 ± 0.7 1.5 ± 0.5 1.5 ± 0.3 1.5 ± 0.2 aerosil 0-10 0.1-7.5 3.0 ± 2.0 3.0 ± 1.5 3.0 ± 1.0 3.0 ± 0.7 Second surfactant 0-10 0.1-7.5 1.5 ± 0.7 1.5 ± 0.5 1.5 ± 0.3 1.5 ± 0.2 The charged nanoparticles achieve a substantially faster appearance of the drug. The medicament contains the silibinin component Preferably in a dose of at least 10 mg, at least 15 mg, at least 20 mg, at least 25 mg, at least 50 mg, at least 75 mg, at least 100 mg, at least 125 mg, at least 150 mg, at least 175 mg or at least 200 mg; more Preferably at least 225 mg, at least 250 mg, at least 275 mg, at least 300 mg, at least 325 mg, at least 350 mg, at least 375 mg or at least 400 mg; even more preferably at least 425 mg, at least 450 mg, at least 475 mg, at least 500 mg, at least 525 mg, at least 550 mg, at least 575 mg or at least 600 mg; most preferably at least 625 mg, at least 650 mg, at least 675 mg, at least 700 mg, at least 725 mg, at least 750 mg, at least 775 mg or at least 800 mg; and in particular at least 825 mg, at least 850 mg, at least 875 mg, at least 900 mg, at least 925 mg, at least 950 mg, at least 975 mg, or at least 1,000 mg; in each case as an equivalent dose based on silibinin. The medicament contains the silybinin component Preferably in a dose of at least 1.0 mg / kg, more preferably at least 2.5 mg / kg, even more preferably at least 5.0 mg / kg, most preferably at least 7 , 5 mg / kg and in particular at least 10 mg / kg, at least 12.5 mg / kg, at least 15 mg / kg, at least 17.5 mg / kg, at least 20 mg / kg, at least 22.5 mg / kg, at least 25 mg / kg, at least 27.5 mg / kg or at least 30 mg / kg, based on weight body of the patient and in each case as an equivalent dose based on silibinin. Preferably, said dose is a daily dose. Therefore, when the medicine is adapted, for example, for twice daily administration, the respective daily dose is divided into two parts of the same amount. Similarly, when the medicine is adapted, for example, for administration three times a day, the respective daily dose is divided into three parts of the same amount. In a preferred embodiment, the daily dose of silibinin component is at least 5, more preferably at least 10, even more preferably at least 15 and most preferably at least 20 mg per kg of body weight, based on the equivalent weight of silibinin In a preferred embodiment, the daily dose of the silibinin component is 20 mg per kg body weight, based on the equivalent weight of silibinin. Therefore, when the medicament is adapted for once-daily administration, it preferably contains the total amount of the silibinin component, for example, 1,400 mg of silibinin for a patient having a body weight of 70 kg. When the medicament is adapted for twice daily administration, it preferably contains half the amount of the silibinin component, for example, 700 mg of silibinin for a patient having a body weight of 70 kg. When the medicament is adapted for administration three times a day, it preferably contains one third of the amount of the silibinin component, for example, 467 mg of silibinin for a patient having a body weight of 70 kg. When the medicament is adapted for administration four times a day, it preferably contains a quarter of the amount of the silibinin component, for example, 350 mg of 20 silibinin for a patient having a body weight of 70 kg. When the medicament is adapted for parenteral administration, preferably for infusion, a preferred treatment regimen comprises 4 equal infusions lasting 2 hours each. Preferably, after 4 hours the same infusion is repeated so that for every 24 hours a total of 4 infusions are administered. This regimen can be schematically abbreviated as "2-4-2-4-2-4-225 4", in which each figure indicates a number of hours and the underlined figures indicate the duration of an infusion while the underlined figures indicate a delay phase between two infusion intervals. Preferably, the treatment regimen is uniform, that is, for 24 hours all infusions are equally dosed for equal periods of time and the delay phases between consecutive infusions are also equal. 30 Following the above indications, the preferred parenteral administration regimens are summarized in the following table: Once a day 0.5-23.5; 1-23; 1.5-22.5; 2-22; 2.5-21.5; 3-21; 3.5-20.5; 4-20; 6-18; 12-12; 24; Twice daily 0.5-11.5-0.5-11.5: 1-11-1-11; 1.5-10.5-1.5-10.5: 2-10-2-10; 2.5-9.5-2.5-9.5: 3-9-3-9; 3.58.5-3.5-8.5, 4-8-4-8; 6-6-6-6; 8-4-8-4; Three times a day 0.5-7.5-0.5-7.5-0.5-7.5; 1-7-1-7-1-7; 1.5-6.5-1.5-6.5-1.5-6.5; 2-6-2-6-2-6; 2.5-5.5-2.5-5.52.5-5.5; 3-5-3-5-3-5; 3.5-4.5-3.5-4.5-3.5-4.5; 4-4-4-4-4-4; 6-2-6-2-6-2; Four times a day 0.5-5.5-0.5-5.5-0.5-5.5-0.5-5.5; 1-5-1-5-1-5-1-5; 1.5-4.5-1.5-4.5-1.5-4.5-1.5-4.5; 2-4-2-42-4-2-4; 2.5-3.5-2.5-3.5-2.5-3.5-2.5-3.5; 3-3-3-3-3-3-3-3; 3.5-2.5-3.5-2.5-3.5-2.5-3.5-2.5; and 4-2-4-2-4-2-4-2. In a preferred embodiment, the medicament is adapted for administration once, twice, three or four times a day so that the overall daily dose that is administered when the medicament is administered in the mode 35 prescribed, is in amounts of at least 300 mg, at least 325 mg, at least 350 mg, at least 375 mg or at minus 400 mg; more Preferably at least 425 mg, at least 450 mg, at least 475 mg, at least 500 mg, at least 525 mg, at least 550 mg, at least 575 mg or at least 600 mg; even more preferably at least 625 mg, at least 650 mg, at least 675 mg, at least 700 mg, at least 725 mg, at least 750 mg, at least 775 mg or at least 800 mg; even more preferably at least 825 mg, at least 850 mg, at 40 minus 875 mg, at least 900 mg, at least 925 mg, at least 950 mg, at least 975 mg, or at least 1,000 mg; most preferably at least 1,050 mg, at least 1,100 mg, at least 1,150 mg, at least 1,200 mg or at least 1,250 mg; and in particular at least 1,300 mg, at least 1,350 mg, at least 1,400 mg, at least 1,450 mg or at least 1,500 mg; in each case as an equivalent dose based on silibinin. Preferred pharmaceutical parameters AUC0-t, AUCt- ~, AUC0. »And AUC0.» (Corr.) (Preferably after 45 several infusions, for example, after 11 infusions; single dose: 12.5 mg / kg; dose daily: 4 infusions; total dose: 11 infusions) and are summarized as embodiments E a It is in the following table: 5 10 fifteen twenty 25 30 35 40 Four. Five E1 E2 E3 E4 E5 E6 E7 E8 mg h / ml mg h / ml mg h / ml mg h / ml mg h / ml mg h / ml mg h / ml mg h / ml AUC0-t 333 ± 200 333 ± 150 333 ± 125 333 ± 100 333 ± 80 333 ± 60 333 ± 40 333 ± 20 AUCt. ~ 322 ± 200 322 ± 150 322 ± 125 322 ± 100 322 ± 80 322 ± 60 322 ± 40 322 ± 20 AUCo- ~ 655 ± 200 655 ± 150 655 ± 125 655 ± 100 655 ± 80 655 ± 60 655 ± 40 655 ± 20 AUCo- ~ (corr.) 414 ± 200 414 ± 150 414 ± 125 414 ± 100 414 ± 80 414 ± 60 414 ± 40 414 ± 20 In a preferred embodiment of the invention, the medicament containing the silibinin component is adapted for an adjunctive therapy, preferably for immunomodulatory / antiviral combination therapies such as interferon / ribovarin. In a preferred embodiment, in addition to the silibinin component, the medicament contains an additional pharmaceutical product that is preferably suitable for the treatment of inflammatory liver diseases, particularly preferably for viral liver diseases, in particular for the treatment of hepatitis B or C . Preferably, the additional pharmaceutical product is selected from the group consisting of hepatic, lipotropic therapeutic agents [A05B]; nucleosides, nucleotides, exclusive reverse transcriptase inhibitors [J05AB]; interferons [L03AB] and monoclonal antibodies to HBV (hepatitis B virus). The references indicated in square brackets refer to the ATC index, preferably in the German version of 2007. Particularly preferably, the additional pharmaceutical product among the group consisting of arginine glutamate, citiolone, epomediol, ornihine oxoglurate, thidiacicarginine, myoinositol, methionine and N-acetyl-methionine, choline, ornithine aspartate, cyanidanol, thiopronin, betaine , cyanocobalamin, leucine, laevulose, acyclovir, idoxuridine, vidarabine, ribavirin, ganciclovir, famciclovir, valaciclovir, cidofovir, penciclovir, valganciclovir, brivudine, alpha interferon, beta interferon, gamma interferon, alpha-2 interferon, alpha-n1 interferon, alpha-n interferon interferon , interferon beta-1a, interferon beta-1b, interferon alfacon-1, peginterferon alfa-2b, peginterferon alfa-2a and interferon gamma 1b. In a preferred embodiment, the treatment of the patient with the silibinin component serves to support and / or prepare viral hepatitis, in particular hepatitis B or C, following this treatment with another pharmaceutical product that is preferably selected from the group consisting of arginine glutamate, silymarin, citiolone, epomediol, ornithine oxoglurate, thidiacicarginine, myoinositol, methionine and N-acetyl-methionine, choline, ornithine aspartate, cyanidanol, thiopronin, betaine, cyanocobalamin, leucinate, leucinate, leucinate idoxuridine, vidarabine, ribavirin, ganciclovir, famciclovir, valacyclovir, cidofovir, penciclovir, valganciclovir, brivudine, alpha interferon, beta interferon, gamma interferon, alpha2a interferon, alpha-2b interferon, alpha-n1 interferon, beta-1a interferon, beta-interferon 1b, interferon alfacon-1, peginterferon alfa-2b, peginterferon alfa-2a and interferon gamma 1b. Therefore, preferably following the treatment of viral hepatitis, in particular hepatitis C with the medicament containing the silibinin component, treatment of viral hepatitis, in particular hepatitis B or C, takes place with another medication. In a preferred embodiment, the medicament is formulated as a constituent of a sequential treatment, the medicament being initially administered for a first period Preferably parenterally and another medication being subsequently administered during a second period. Preferably, the first period comprises at least 2 days, more preferably at least 3 days, even more preferably at least 4 days, most preferably at least 5 days and, in particular, at least 6 days. Preferably, the second period comprises more days than the first period. Preferably, the second period comprises at least 2 days, more preferably at least 3 days, even more preferably at least 4 days, most preferably at least 5 days and in particular at least 6 days. In a particular embodiment, the second medicament contains a combination of ribavirin and pegylated interferon alfa and the second period comprises a time of 24 to 48 hours. Preferably, the other medicament contains one or more pharmaceutical products selected from the group consisting of arginine glutamate, silymarin, citiolone, epomediol, ornithine oxoglurate, thidiacicarginine, myoinositol, methionine and N-acetyl-methionine, choline, ornithine aspartate, cyanidanol, thiopronin, betaine, cyanocobalamin, leucine, laevulose, acyclovir, idoxuridine, vidarabine, ribavirin, ganciclovir, famciclovir, valacyclovir, cidofovir, penciclovir, valganciclovir, brivudine, alpha interferon, interferon alpha, interferon alpha interferon, alpha interferon gamma interferon -2b, interferon alfa-n1, interferon beta-1a, interferon beta-1b, interferon alfacon-1, peginterferon alfa-2b, peginterferon alfa-2a, interferon gamma 1b and monoclonal antibodies to HBV, particularly preferably an interferon and / or ribavirin and / or silymarin. If the other medicine contains a 5 10 fifteen twenty 25 30 35 40 Interferon, this is preferably pegylated interferon alpha (peginterferon alfa-2a or peginterferon alfa-2b). In a particularly preferred embodiment, the other medicament contains one or more pharmaceutical products selected from the group consisting of isosilibinin, silidianine, silicristine, taxifoline, isosylcycline, silimonin, silandrin, silihermine and neosilihermine, more preferably only a pharmaceutical product selected from the list. foregoing. Preferably, the other medicament contains a silibinin component as defined in relation to the previously written medicament which is administered during the first period and is essentially preferably free of at least one, preferably at least all of the aforementioned substances. In this regard, "essentially exempt" means that the residual content of the substance involved is preferably less than 2.0% by weight, more preferably less than 1.0% by weight, even more preferably less than 0.5% by weight. , most preferably less than 0.1% by weight and in particular less than 0.05% by weight, based on the total weight of the medicament. The other medicine may be formulated in principle for parenteral or oral administration. According to the invention, it is preferably formulated for another route of administration than that of the medicament that is administered during the first period. Particularly preferred, the other medicament is formulated for oral administration. In a particularly preferred embodiment according to the invention, the medicament that is administered during the first period is adapted for parenteral, preferably intravenous administration and the other medicament that is administered during the second period following the first period is adapted for oral administration. In a preferred embodiment, the treatment regime according to the invention comprises two phases that follow one another consecutively, namely, a first period and a second period. Preferably, during the first period the medication containing the silibinin component is preferably administered parenterally, but no other medication having a hepatic effect is administered simultaneously. During the second period, another medication is administered which preferably contains ribavirin and / or pegylated interferon alfa. In another embodiment, the medicament containing the silibinin component is also administered during the second period, preferably parenterally. In another preferred embodiment, the medicament containing the silibinin component is not administered during the second period, that is, only said other medicament is administered. Preferred embodiments F1 to F15 of the biphasic treatment regimen are summarized in the following table. No. of days F1 F2 F3 F4 F5 F 6 F7 F8 F9 F10 F11 F12 F13 F14 F15 First period > 1> 1> 2> 2> 2> 3> 3> 4> 3> 4> 4> 5> 5> 7> 7 Second period > 1> 2> 1> 2> 3> 2> 3> 3> 4> 4> 5> 4> 5> 7> 14 In another preferred embodiment, the treatment regime according to the invention comprises three phases that follow one another consecutively, namely, a first period, a second period and a third period. Preferably, during the first period the medication containing the silibinin component is administered, preferably parenterally, but no other medication having a hepatic effect is administered simultaneously. During the second period, another medicament containing Preferably ribavirin and / or pegylated alpha interferon is administered and the medicament containing the silibinin component is also administered during the second period, preferably parenterally. Preferably, during the third period said other medication is administered which preferably contains pegylated ribivirine and / or interferon alpha, but the medicament containing the silibinin component is not administered during the third period, that is, only said other medication is administered. Preferred embodiments G1 to G15 of the three-phase treatment regimen are summarized in the following table: No. of days G1 G2 G3 G4 G5 G6 G7 G8 G9 G10 G11 G12 G13 G14 G15 First period > 1> 1> 2> 1> 1> 2> 2> 2> 3> 4> 5> 6> 7> 14> 14 Second period > 1> 2> 1> 1> 2> 2> 1> 2> 3> 4> 5> 6> 7> 7> 14 Third period > 1> 1> 1> 2> 2> 1> 2> 2> 3> 4> 5> 6> 7> 7> 7 5 10 fifteen twenty 25 30 35 40 Four. Five they follow each other consecutively, namely a first period, a second period and a third period. Preferably, during the first period another medication is administered which preferably contains ribavirin and / or pegylated interferon alpha and the medication containing the silibinin component is not administered during the first period. During the second period, another medicament containing Preferably ribavirin and / or pegylated alpha interferon is administered and the medicament containing the silibinin component (co-administered) is also administered during the second period, preferably parenterally. Preferably, during the third period said other medication is administered which preferably contains pegylated ribivirine and / or interferon alpha, but the medication containing the silibinin component is not administered during the third period, that is, only said other medication is administered. In other words, according to this preferred embodiment, said other medicament which preferably contains ribavirin and / or pegylated interferon alpha is administered continuously, or during an intermediate period (= second period) the medicament containing the silibinin component is co-administered, preferably by parenteral route Preferred embodiments H1 to H15 of the three-phase treatment regimen are summarized in the following table: No. of days Hi H2 H3 H4 H5 H6 H7 H8 H9 H10 H11 H12 H13 H14 H15 First period > 1> 1> 2> 1> 1> 2> 2> 2> 3> 4> 5> 6> 7> 14> 14 Second period > 1> 2> 1> 1> 2> 2> 1> 2> 3> 4> 5> 6> 7> 7> 14 Third period > 1> 1> 1> 2> 2> 1> 2> 2> 3> 4> 5> 6> 7> 7> 7 Figure 10 shows various ways of co-administration of ribavirin and / or pegylated interferon alpha and the medicament containing the silibinin component (embodiments a1) to m2)). Each bar refers to a period of administration time. For example, according to embodiment fi), administration begins with ribavirin / peg interferon alfa and continues. During the intervening period, the silibinin component is co-administered. A further aspect of the invention relates to a medicament as described above, preferably adapted for parenteral administration, to treat viral hepatitis as described above. Still in a further aspect the invention relates to a test kit comprising at least one medicament according to the invention, containing a silibinin component and at least one other medicament. Both the medicament according to the invention, which contains a silibinin component, as the other medicament as described above, as in all preferred embodiments, are analogously applicable to the test kit according to the invention. In a preferred embodiment, the kit contains as many medications (individual dose units) as necessary in order to carry out a sequential therapy, the medication containing the silibinin component being initially administered during a first period and subsequently being administered the another medicine for a second period. Preferably, the first period comprises at least 2 days, more preferably at least 3 days, even more preferably at least 4 days, most preferably at least 5 days and, in particular, at least 6 days. Preferably the second period comprises at least 2 days, more preferably at least 3 days, even more preferably at least 4 days, most preferably at least 5 days and in particular at least 6 days. In a particularly preferred embodiment, the invention relates to the use of a silybinin component, preferably a silybinin ester, for the production of a medicament, which is formulated for parenteral administration, for the treatment of viral hepatitis C in non-people. sensitive with respect to ribavirin / interferon therapy, that is, in patients who do not respond to an immunomodulatory / antiviral combination therapy such as rebavirin / interferon therapy. A further aspect of the invention relates to a silybinin component, preferably a silybinin ester, preferably for parenteral administration, for the treatment of hepatitis C. Preferred embodiments of the aspect of the invention are apparent from the above description. of the preferred embodiments of the other aspects of the invention and, therefore, are not repeated. A further aspect of the invention relates to the treatment of hepatitis C, which comprises the administration, preferably parenteral administration, of a pharmaceutically effective amount of a silybinin component, preferably a silybinin ester, to a subject in need thereof. Preferred embodiments of the aspect of the invention are apparent from the above description of the preferred embodiments of the other aspects of the invention and, therefore, are not repeated. The following examples further illustrate the invention, but are not intended to limit its scope. Example 1: The silibinin component was administered parenterally in the form of C-2 ', 3-bi (hydrogensuccinate) of silibinin (Legalon Sil®, Madaus, Cologne) (hereinafter referred to as "silibinin"). Patients and methods: Patients Protocol 1 Protocol 2 N (male / female) 16 (14/2) 20 (17/3) Mean age (years ± SD) 49.9 ± 9.7 52.7 ± 12.8 Genotype (1/2/4) 15 / - / 1 17/1/2 Fibrosis phase: 0-2 3 10 3-4 13 7 Not available - 3 Preceding therapy PEG-interferon-alfa2a / RBV 14 18 PEG-interferon-alfa2b / RBV 2 4 Fall of registration at 12 weeks of the preceding therapy > 2 ** 3. 4 1-2 4 1 <1 5 12 Not available 2. 3 14 18 2 4 * some patients had more than one treatment cycle ** all were positive at week 24 5 Patients with insensitivity prior to the full dose of the combination therapy of peginteron / ribavirin were selected for these studies. The insensitivity was defined as the absence of a fall in the registry> 2 of viral load after 12 weeks of therapy and / or for not achieving a final response to treatment. Patients were required to have a liver biopsy done in both before inclusion in this study. Standard inclusion / exclusion criteria were applied for peginterferon / ribavirin therapy. Study Protocol: During the first phase of selection in the 35 days prior to the first dose of study drug, the eligibility of patients was established according to inclusion / exclusion criteria. All patients had at least one quantitative HCV-RNA trial in the 6 months prior to the selection phase. 15 Protocol 1: Patients first received daily 10 mg / kg of silibinin (Legalon Sil®, Madaus, Cologne) infused in 4 hours for 7 consecutive days. On day 1 blood was drawn to determine the reference oxidative stress parameters, every 30 minutes during the infusion and 2 hours after the end of the infusion. In 8 the treatment was changed to 140 mg of silymarin (Legalon®, Madaus, Cologne) TID by mouth in combination with 180 pg / week of Peg / Fna-2a (PEGASYS®; Roche, Basel) and 1-1.2 g / d of 20 ribavirin (COPEGUS®; Roche, Basel). Protocol 2: After obtaining the results of the first protocol, treatment with silibinin was prolonged for 2 weeks and different doses of silibinin were administered. The patients received first daily 5, 10, 15 or 20 mg / kg of silibinin infused in 4 hours for 14 consecutive days. On day 8 a treatment was started with 180 pg / week of Peg / Fna-2a and 1-1.2 g / d of ribavirin. After day 14, patients received 280 mg of silymarin (Legalon®, Madaus, Cologne) TID by mouth. During the 14-day infusion period blood was obtained daily for the determination of viral load. 5 In both protocols, in case of intolerance to PEG-IFN alpha2a or ribavirin, standard dose adjustment standards were used. Antiviral combination therapy was provided for a total of 24 weeks (with the option of stopping treatment in patients without a record drop> 2 at week 12); Those who were virologically sensitive at week 24 were offered to continue treatment for another 48 weeks. After the end of the infusion period, patients were tested after 2 and 4 weeks and 10 months monthly until the end of therapy at week 24. The protocol was approved by the ethical committee of the Medical University of Vienna. The details of the study were explained to the patients and they all signed an informed consent. Methods: The serum level of HCV-RNA was determined by the TaqMan PCR assay (Cobas 15 Ampliprep / Cobas TaqMan HCV assay; detection limit, 15 Ul / ml, Roche Diagnostics). The reactive reactive oxidative metabolites in blood were measured by the d-ROM assay (compounds derived from reactive oxygen metabolites; Diacron, Grosseto, Italy) and the amounts of antioxidants by the BAP assay (biological antioxidant potential; Diacron, Grosseto, Italy ) using the portable free radical combination system (FRAS 4, SEAC, Calenzano, Italy) beforehand, every 20 30 minutes during it (on day 1) and 2 hours after silibinin infusions. The d-ROM trial It measures reactive oxygen metabolites (mainly hydroperoxidases) released from plasma proteins by an acid buffer, which in the presence of iron generates alkoxyl and peroxyl radicals, according to the Fenton reaction. These radicals, in turn, are capable of oxidizing an alkyl-substituted aromatic amine (N, N-diethylparaphenylenediamine), thereby producing a pink derivative that is photometrically quantified at 505 nm. The results for reactive oxidative metabolites are expressed in the form of Caratelli units (Ucarr; normal: 250-300, 1 Ucarr = 0.08 mg of hydrogen peroxide / dl). The BAP test measures the intensity of decolorization of a solution of ferric chloride mixed with a thiocyanate derivative by the plasma sample photometrically added at 505 nm, which is proportional to the ability to reduce ferric ions by the amounts of antioxidants in plasma ( normal = 2,200 pM). The description of the tests by the manufacturer does not specify which substances are actually measured. Statistics: Optionally, the main variable outcome was the virological response defined as the percentage of patients who were negative for CRP at the end of treatment (week 24). Secondary efficacy variables were virological response rates at week 12, safety and tolerance of treatment with 35 PEG-IFN / ribavirin / silymarin, the reference quality of life at week 24, week 48, week 72 (SF - 36, severity scale of fatigue) and oxidative status after silibinin infusions. Due to the unexpected virological response force after 7 days of silibinin infusions, uptake was stopped and the study was redesigned based on virological response parameters using longer infusion periods and higher doses of silibinin. For the original study, the sample size was estimated 40 based on the Gehan two-phase design. According to previous studies, a response rate> 10% seems to guarantee further investigation of the treatment regimen. A total of 29 patients had to be recruited in the first phase (probability of error p = 5%). Results: Protocol 1: 45 16 selected non-sensitive persons were included (see table above for details). All patients received a full-dose treatment with pegylated interferon (12 peginterferon alfa 2a, 2 peginterferon alfa 2b) and ribavirin (1,000 - 1,200 mg / d) for at least 12 weeks. The parameters of oxidative stress measured were not altered during silibinin infusions (Figure 1). Serum HCV RNA fell in all patients of a SIL iv monotherapy (Figure 2) (reference: 6.59 ± 50 0.53, day 8: 5.26 ± 0.81 IU / ml record, [mean ± SD], p <0.001) with an average fall of the registry of 1.32 ± 0.55 in a week. In parallel, ALT decreased from 162 ± 133 to 118 ± 107 U / 1 (p = 0.004). In all patients, HCV RNA remained detectable at the start of PegIFN / RBv therapy. Three patients declined the PEGIFN / RBV combination therapy. In 11 of the remaining 13 patients, HCV RNA increased again after the end of silibinin infusions despite the onset of PegIFN / RBV. At week 55, all patients were still positive for HCV RNA, but 5 patients had a> 2 drop in the registry and continued treatment (Figure 3). None of them were negative for HCV RNA at week 24, one patient had a 5.5 drop in the registry and continued treatment of his own accord. 5 10 fifteen twenty 25 30 35 40 Four. Five fifty Protocol 2: Twenty selected non-sensitive people were included (see the table above for details). All patients received a full-dose treatment with pegylated interferon (18 peginterferon alfa 2a, 4 peginterferon alfa 2b; 2 patients received 2 treatment courses) and ribavirin (1,000 - 1,200 mg / d) for at least 12 weeks. Figure 4 shows the viral kinetic characteristics in these patients. The viral load dropped continuously. After 7 days of silibinin monotherapy the dose of 5 mg / kg was marginally effective (n = 3, registry drop 0.55 ± 0.5), while at 10 mg / kg (n = 19 [including patients of protocol 1], record drop 1.41 ± 0.59); 15 mg / kg (n = 5, registry drop 2.11 ± 1.15) and doses of 20 mg / day (n = 9, 3.02 ± 1.01) led to a highly significant decrease in viral load ( p <0.001). After 1 week of combined silibinin and peginterferon / ribavirin therapy the viral load decreased further (log drop: 5 mg / kg: 1.63 ± 0.78; 10 mg / kg: 4.16 ± 1.28; 15 mg / kg: 3.69 ± 1.29; 20 mg / kg: 4.8 ± 0.89; total p <0.0001 versus reference) (Figure 5). Two of the 5 patients in the 15 mg / kg group and 4 of the 9 patients in the 20 mg / kg group had HCV RNA <15 IU on day 15. The HCV RNA was <15 IU / ml in 8 and 7 patients at week 4 (week 5 of the study protocol) and week 12 (week 13 of the study protocol) after the start of PEGIFN / RBV, respectively. Antiviral combination therapy was continued for all patients (Figure 6). Security: Silibinin was generally well tolerated. Five patients complained of mild gastrointestinal symptoms (abdominal pain: 5, diarrhea: 2, nausea: 1), two of headaches and one of arthralgia. All these were assessed as mild by the patients and disappeared after the end of the infusions; No dosage changes were required. All patients in the 15 and 20 mg / kg groups appreciated a sensation of heat when the infusion began, which disappeared within 30 minutes without treatment. SAEs were not produced. Monotherapy showed no changes in hemoglobin, leukocytes, platelets and creatinine. Typical side effects of antiviral combination therapy were observed (including a patient suffering from increased dyspnea due to hemophilus influenzae-induced pneumonia, which required termination of peginterferon / ribavirin therapy after 8 weeks. This example demonstrates that parenteral administration of silibinin (C-2 ', 3-bis (hydrogensuccinate)) has considerable antiviral activity against the hepatitis C virus. These observations demonstrate the potential ability of this drug for hepatitis treatment. Chronic C, particularly in non-sensitive people. It was surprisingly found that silibinin iv (C-2 ', 3-bis (hydrogensuccinate)) is a potent antiviral agent in patients with chronic hepatitis C not sensitive to a standard antiviral combination therapy. Intravenous silibinin was well tolerated and no serious adverse effects were observed. The most commonly referred side effect was a transient sensation of heat. The antiviral effect was dose dependent but was not maintained after the end of the infusion period by the administration of silymarin. In comparison, similar amounts of silymarin given orally had no effect on HCV loading (A Gordon et al., J Gastroenterol Hepatol. 2006, 21, 275-80) reflecting differences in the bioavailability and metabolism of silibinin that resulted at much lower plasma levels. After oral dosing of silymarin, flavonolignans are rapidly glucuronidated and rapidly eliminated with short half-lives (Z Wen et al., Drug Metab Dispos. 2008, 36 (1), 65-72). Example 2: Patients were continuously treated with 180 pg of interferon alfa 2a and ribavirin based on weight. Despite this treatment, five patients were positive for HCV RNA after 24 hours of therapy: three male patients and two female patients; four patients with HCV genotype 1 and one patient with HCV genotype 3a; Three patients with cirrhosis. Four patients can be considered as devoid of immunity, while one patient can be considered as relapsed with respect to two previous therapies (24 and 48 weeks). In the course of continued treatment with 180 pg of peginterferon alfa 2a and ribavirin based on weight, all patients were treated at least once for 14 consecutive days with 20 mg / kg / d of silibinin iv. During this period, the combination therapy with peginterferon / ribavirin was continued. All 5 patients were negative for HCV RNA. Fig. 7 shows the result for an individual patient (male, 55 years old). As can be seen, peginterferon / ribavirin only causes a decrease in viral load from approximately one record of 5 10 fifteen twenty 25 30 35 7 Ul / ml to approximately a record of 4.5 Ul / ml after 24 weeks. Joint treatment with 20 mg / kg / day of bis (hydrogensuccinate) of silibinin i.v. for 14 days, however, it led to a huge decrease in viral load from a record of approximately 4.5 IU / ml to a value below the detection limit. After the first administration interval of parenteral silibinin bis (hydrogensuccinate), the viral load increased again to approximately 2 IU / ml which, however, could be permanently decreased below the detection limit by a second joint treatment with 20 mg / kg / day of silibinin bis (hydrogensuccinate) iv for 14 days Fig. 8 shows the result for another individual patient (female, 44 years old). As can be seen, peginterferon / ribavirin only causes a decrease in viral load from a record of approximately 7 IU / ml to a register of approximately 5 IU / ml after 30 weeks. Joint treatment with 20 mg / kg / day of bis (hydrogensuccinate) of silibinin i.v. for 14 days after 30 weeks, however, it led to a huge and permanent decrease in viral load from a record of approximately 4 IU / ml to a value below the detection limit. Fig. 9 shows the result for an individual patient (male, 52 years old). As can be seen, peginterferon / ribavirin causes an effective decrease in viral load from a record of approximately 5 IU / ml to a value close to the detection limit <15 IU / ml. The combined treatment with 20 mg / kg / day of bis (hydrogensuccinate) of silibinin for 14 days after 72 weeks caused an additional decrease in viral load well below the detection limit. These clinical trials demonstrate that parenteral treatment with a silibinin component for a comparatively short period of time supports and significantly improves conventional treatment using peginterferon / ribavirin. It seems that parenteral administration of the silibinin component (re) activates patients' susceptibility to conventional treatment by peginterferon / ribavirin (Figures 7 and 8) and / or improves the antiviral effect of conventional treatment by peginterferonon / ribavirin (Figure 9). Example 3: An in vivo study was conducted to characterize silibinin plasma concentration / time profiles in 8 patients suffering from chronic hepatitis C who received an infusion treatment i.v. 7 days with 20 mg silibinin / kg body weight (Legalon® SIL). For multiple doses of 20 mg / kg body weight, plasma concentration / time profiles and PK parameters of free and total silibinin were observed on day 1 (= single dose conditions) and compared with those on day 7 (= expected steady state conditions). Analytical Procedure: Study samples were analyzed using a validated HPLC-UV method. During the test period the analytical procedure was validated by two calibration curves per analytical experiment. The inspection of the chromatograms the data presented in the calibration curves and the quality control samples indicates that the result of the determinations of the total and free silibinin A and silibinin B concentrations for the study are reliable. The PK characteristics are summarized in the following table: Total silybinin Day 1 Day 7 Silibinin A Silibinin B Silibinin A Silibinin B AUC (0-m) [h ng / ml] 61,733 ± 27,489 13,745 ± 76040 - - AUC (0-tz) [h ng / ml] 50,019 ± 20,048 10,9038 ± 51,342 - - AUCss [h ng / ml] - - 84,299 ± 25,111 150,780 ± 47,780 Cmin [h ng / ml] - - 1,967 ± 831 3,311 ± 61,426 Cmax [ng / ml] 4,550 ± 928 9,539 ± 2,843 5,791 ± 977 11,083 ± 62,269 Cav [ng / ml] - - 3,512 ± 1,046 6,282 ± 1,991 t1 / 2 [ng / ml] 8.30 ± 2.26 8.29 ± 2.98 13.32 ± 3.66 12.02 ± 2.91 HVD [h] 9.28 ± 3.36 9.47 ± 3.29 15.20 ± 4.09 13.17 ± 3.62 MRT [h] 13.17 ± 3.74 13.44 ± 4.39 19.22 ± 5.28 17.34 ± 4.20 Total silybinin Day 1 Day 7 Silibinin A Silibinin B Silibinin A Silibinin B CL [ml / (h kg)] 0.435 ± 0.336 0.233 ± 0.237 0.269 ± 0.128 0.156 ± 0.091 Vz [ml / kg] 4.7 ± 1.3 2.4 ± 1.1 4.67 ± 0.73 2.43 ± 0.55 tmax [h] 4.14 ± 0.18 4.17 ± 0.18 - - % PTF [%] - - 121.89 ± 54.43 137.59 ± 52.51 Free silybinin Day 1 Day 7 Silibinin A Silibinin B Silibinin A Silibinin B AUC (0-D) [h ng / ml] 3,614 ± 1,648 753 ± 397 - - AUC (0-tz) [h ng / ml] 3,302 ± 1,551 559 ± 339 - - AUCss [h ng / ml] - - 4,095 ± 1,942 1,041 ± 627 Cmin [h ng / ml] - - 59 ± 40 3.3 ± 8.7 Cmax [ng / ml] 316 ± 108 90 ± 44 315 ± 119 120 ± 54 Cav [ng / ml] - - 171 ± 81 43 ± 26 t1 / 2 [ng / ml] 4.58 ± 1.35 5.16 ± 4.96 6.85 ± 1.29 4.35 ± 1.66 HVD [h] 10.12 ± 4.29 6.09 ± 2.14 11.87 ± 2.64 7.17 ± 1.63 MRT [h] 8.49 ± 2.64 8.81 ± 6.12 9.88 ± 1.86 6.2762.39 CL [ml / (h kg)] 7.0 ± 4.2 44.8 ± 42.5 5.9 ± 2.9 26.8 ± 16.7 Vz [ml / kg] 51.4 ± 12.7 285.8 ± 163.9 55.3 ± 18.5 140.4 ± 49.0 tmax [h] 3.73 ± 1.22 4.03 ± 0.04 - - % PTF [%] - - 164.00 ± 45.33 305.24 ± 80.46 Example 4: An in vitro study was conducted to assess the cytotoxic potential capacity of silymarin, silybinin, 5-disodium salt of silybinin bis (hydrogensuccinate) and succinic acid through the XTT assay using the line L929 mouse cell (see D.A. Scudiero et al., Cancer Res. 48, 4827-33; O.S. Weislow et al., J. Natl. Cancer Inst., 81, 577-86; N.W. Roehm et al., J. Immunol. Methods, 142). The following concentrations of the test articles were tested: 9.77, 19.53, 39.06, 78.13, 156.25, 312.5, 625, 1,250 pg / ml. Complete medium (RPMI 1640 containing 10% FCS (v / v)) was used as a negative control. The solvent control for the test article was RPMI 1640 medium containing 10% (v / v) of FCS and 1% DMSO. The solvent control for the positive control was also RPMI 1640 medium which it contained 10% (v / v) of FCS and 10.0% (v / v) of deionized water. SDS was used as a positive control The following concentrations were applied: 3,125, 6.25, 12.5, 25, 50, 100, 125, 250 pg / ml. The incubation time was 24 hours at 37 ± 1.5 ° C. 15 The negative control and solvent control showed no reduction in cell viability. The positive control (SDS) induced a reduction related to different doses in cell viability. Toxic effects were observed following incubation with silymarin from 39.06 pg / ml to the highest concentration tested (1,250 pg / ml). The calculated XTT50 value is 35.2 pg / ml. Toxic effects were observed following incubation with silibinin from 78.13 pg / ml to the highest concentration tested (1,250 pg / ml). The calculated XTT50 value is 67.5 pg / ml. 5 10 fifteen twenty 25 No relevant cytotoxic effects were observed following incubation with sodium salt of silibinin bis (hydrogensucinate) to the highest concentration tested (1250 | jg / ml). Due to the lack of cytotoxicity, the value of XXT50 could not be calculated. No relevant cytotoxic effects were observed following incubation with succinic acid until the highest concentration tested (1250 jg / ml). Due to the lack of cytotoxicity, a value of XXT50 could not be calculated. These experiments showed that under the given conditions, the potential cytotoxic capacity of silymarin is almost 100% greater than the potential cytotoxic capacity of silybinin. Therefore, it can be expected that silibinin can be administered in higher doses than silymarin without causing serious adverse side effects. Example 5: NS5B RNA-dependent RNA polymerase (RdRp) is an essential enzyme for viral replication (S.B. Hwang et al., Virology 1997, 227, 439-46). The following pure compounds were tested in a cell-free enzyme assay for the detection of the RdRp activity of HCV: silibinin A, silibinin B, isosilibinin A, isosilibinin B, silicristinin, silidianine and the C-disodium salt silibinin ester -2 ', 3- bis (hydrogensuccinate) of silibinin (active ingredient of Legalon® SIL). Stock solutions (100 mM) of the compounds were prepared in 100% DMSO. The concentration of DMSO in all reactions remained constant at 5%. The target enzyme for the study was HCV NS5BA21 polymerase genotype J4 (1b). Figure 11 shows the data generated for the six purified constituents of silymarin (ie, silibinin A, silibinin B, isosilibinin A, isosilibinin B, silicristin and silidianine). Figure 12 shows the respective data for the silybinin ester. The silybinin ester was revealed as the most effective. The IC50 value for the silybinin ester was determined from the dose response curve of two measurements. The IC50 value determined 47 ± 14 jM. The curves were adjusted to the data points and the IC50 values were interpolated from the resulting curves using the SigmaPlot 8.0 software.
权利要求:
Claims (6) [1] 1. A silibinin component to be used in the treatment of hepatitis C in a patient who does not respond to a ribavirin / interferon therapy, in which the silibinin component is provided as a medication, in which the medication is essentially free of minus one of the substances 5 selected from the group consisting of silidianine, silicristine and isosilibinin, in which essentially free means that the residual content of the substance is less than 2.0% by weight, based on the total weight of the drug. [2] 2. The silibinin component for use according to claim 1, wherein the patient will undergo or have undergone a liver transplant. The silibinin component to be used according to claim 1 or 2, which is administered orally. [4] 4. The silibinin component for use according to any of the preceding claims, which is provided as a medicament selected from the group consisting of tablets, capsules, sugar-coated tablets, granules and sachets. [5] 5. The silibinin component for use according to any of the preceding claims, which is provided as a medicament containing a cyclodixtrin and / or a phospholipid. [6] 6. The silibinin component to be used according to any of the preceding claims, for administration once a day, twice a day or three times a day. [7] 7. The silybinin component to be used according to any of the preceding claims, which is administered in a dose of at least 50 mg, based on silybinin. The silibinin component for use according to any of the preceding claims, which is for reduction of the virus load in hepatitis C patients.
类似技术:
公开号 | 公开日 | 专利标题 ES2662700T3|2018-04-09|Silibinin component for hepatitis treatment KR100651110B1|2006-12-21|Pharmaceutical compositions containing terbinafin and use thereof US20120014912A1|2012-01-19|Palatable pharmaceutical composition KR20210100136A|2021-08-13|Highly active drug combination to treat hepatitis C virus CN101668520A|2010-03-10|Methods for the treatment of liver diseases RU2482844C2|2013-05-27|Silibilin component for treating hepatitis US20180021360A1|2018-01-25|Combination Therapy Regimen For Treatment Of Selected HCV Genotypes US20170087174A1|2017-03-30|Combination therapy regimen for treatment of hcv BRPI0820640A2|2021-05-11|silybinin component for the treatment of hepatitis CN112843053A|2021-05-28|NS5A inhibitor composition WO2018017989A1|2018-01-25|Combination therapy regimen for treatment of selected hcv genotypes WO2013168179A2|2013-11-14|Controlled release pharmaceutical formulations of antiviral agents
同族专利:
公开号 | 公开日 HK1142270A1|2010-12-03| KR101435242B1|2014-08-29| AU2008323156B2|2012-05-31| CA2703834A1|2009-05-22| DK2219642T3|2011-12-05| US20110182858A1|2011-07-28| ES2662700T3|2018-04-09| EP2219642B1|2011-09-21| US20130236420A1|2013-09-12| CN102300570A|2011-12-28| ES2374117T3|2012-02-13| PL2392326T3|2018-07-31| EP2392326B9|2018-09-05| JP2011503133A|2011-01-27| PL2219642T3|2012-02-29| PT2219642E|2011-10-18| KR20100094981A|2010-08-27| MX2010005107A|2010-05-27| US9248115B2|2016-02-02| WO2009062737A1|2009-05-22| EP2392326A1|2011-12-07| AT525070T|2011-10-15| JP5349486B2|2013-11-20| CA2703834C|2014-05-20| EP2219642A1|2010-08-25| AU2008323156A1|2009-05-22| CN102300570B|2015-01-14| EP2392326B1|2018-01-24|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题 US4061765A|1973-01-19|1977-12-06|Dr. Madaus & Co.|Polyhydroxyphenylchromanone salts and therapeutic composition| DE3442639C2|1984-11-22|1989-08-10|Dr. Madaus & Co, 5000 Koeln, De| DE3537656C2|1984-11-22|1989-03-16|Dr. Madaus Gmbh & Co, 5000 Koeln, De| IT1215291B|1985-07-17|1990-01-31|Inverni Della Beffa Spa|FLAVANOLIGNANI COMPLEXES WITH PHOSPHOLIPIDS, THEIR PREPARATION AND RELATED PHARMACEUTICAL COMPOSITIONS.| IT1241673B|1989-10-09|1994-01-27|Istituto Biochimico Italiano|INCLUSION COMPLEXES OF SILIBININ WITH CYCLODESTRINE, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM.| DE4401902C2|1994-01-24|2000-02-03|Madaus Ag|Use of flavolignans as an adjuvant in tumor therapy| JP2002504514A|1998-02-27|2002-02-12|ニュートラマックスラボラトリーズ,インコーポレイテッド|L-Ergothioneine, Nogeshi and S-adenosylmetheonin for prevention, treatment and repair of liver injury| WO1999055326A1|1998-04-30|1999-11-04|Vit-Immune, L.C.|Method of treatment of glutathione deficient mammals| CN1328463A|1998-11-24|2001-12-26|霍利斯-伊登医药公司|Use of 17-ketosteroid compounds, as well as derivatives, metabolites and precursors for treatment of hapatitis C type virus and other togavirus infections| US6849254B1|1999-04-19|2005-02-01|Schering Corporation|HCV combination therapy| WO2002067853A2|2000-10-06|2002-09-06|Probiochem, Llc|A combination and method of treatment of hiv and viral diseases, vascular disease and cancer utilizing a cox-2 inhibitor and cystine| US6699900B2|2001-04-07|2004-03-02|Jan E. Zielinski|Hydrophilic and lipophilic silibinin pro-forms| CN100451501C|2003-08-26|2009-01-14|株式会社东芝|Refrigerator| US7078064B2|2003-12-03|2006-07-18|George Zabrecky|Compositions and methods useful for treating and preventing chronic liver disease, chronic HCV infection and non-alcoholic steatohepatitis| CN1762345A|2005-09-29|2006-04-26|丛晓东|Silybinin injection and its preparation method| CN100389767C|2005-09-30|2008-05-28|天津药物研究院|Injectable stable pharmaceutical composition containing active ingredient silibinin and its salt| CN100534992C|2005-12-26|2009-09-02|浙江海正药业股份有限公司|Silybin esters derivatives and preparation and use thereof| CN101244059B|2008-03-31|2010-11-17|广州佳科生物科技有限公司|Compound silibinin injection, preparation method and application thereof| CN101439018A|2008-11-11|2009-05-27|扬子江药业集团四川海蓉药业有限公司|Silybin compound emulsion for intravenous injection and preparation method thereof|WO2010130460A1|2009-05-14|2010-11-18|Euromed S.A.|Amorphous silibinin for the treatment of viral hepatitis| WO2011051742A1|2009-10-28|2011-05-05|Modutech S.A.|Preparation comprising amino acids and plants and its activity in the alcohol detoxification| US8492386B2|2011-10-21|2013-07-23|Abbvie Inc.|Methods for treating HCV| DE202012012956U1|2011-10-21|2014-10-16|Abbvie Inc.|A combination of at least two direct-acting antiviral agents for use in the treatment of HCV, comprising ribavirin but not interferon| US8466159B2|2011-10-21|2013-06-18|Abbvie Inc.|Methods for treating HCV| PT107924A|2011-10-21|2014-12-03|Abbvie Inc|TREATMENT OF AAD COMBINATIONFOR USE IN HCV TREATMENT| WO2015112171A1|2014-01-27|2015-07-30|Kaohsiung Medical University|A silibinin nanoparticle and a method of treating hepatitis c thereof| CN106170877B|2014-02-26|2018-08-31|联邦科学和工业研究组织|The method for forming the photosensitive layer of perovskite light-sensitive unit| EP3203989B1|2014-10-09|2019-08-21|Distretto Tecnologico Sicilia Micro e Nano Sistemi S.C.A.R.L.|Nanostructured formulations for the delivery of silibinin and other active ingredients for treating ocular diseases| CN109715250A|2015-10-02|2019-05-03|科罗拉多州立大学董事会法人团体|Part silibinin preparation and application thereof| CA3022119A1|2016-04-28|2017-11-02|Emory University|Alkyne containing nucleotide and nucleoside therapeutic compositions and uses related thereto|
法律状态:
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 US98816807P| true| 2007-11-15|2007-11-15| US988168P|2007-11-15| EP07022187|2007-11-15| EP07022187|2007-11-15| EP08005459|2008-03-25| EP08005459|2008-03-25| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|