US20180258151A1 - Recombinant super-compound interferon and uses thereof - Google Patents

Recombinant super-compound interferon and uses thereof Download PDF

Info

Publication number
US20180258151A1
US20180258151A1 US15/910,165 US201815910165A US2018258151A1 US 20180258151 A1 US20180258151 A1 US 20180258151A1 US 201815910165 A US201815910165 A US 201815910165A US 2018258151 A1 US2018258151 A1 US 2018258151A1
Authority
US
United States
Prior art keywords
rsifn
recombinant interferon
interferon
virus
recombinant
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/910,165
Inventor
Guangwen Wei
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Superlab Far East Ltd
Original Assignee
Superlab Far East Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from CNB011043679A external-priority patent/CN1245215C/en
Priority claimed from US10/927,975 external-priority patent/US20050079579A1/en
Priority claimed from US11/077,813 external-priority patent/US20060035327A1/en
Application filed by Superlab Far East Ltd filed Critical Superlab Far East Ltd
Priority to US15/910,165 priority Critical patent/US20180258151A1/en
Assigned to SUPERLAB FAR EAST LIMITED reassignment SUPERLAB FAR EAST LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WEI, GUANGWEN
Publication of US20180258151A1 publication Critical patent/US20180258151A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/02Preparation of peptides or proteins having a known sequence of two or more amino acids, e.g. glutathione
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
    • Y02A50/463

Definitions

  • This invention is related to a field of bioengineering. Specifically this invention relates to a recombinant super-compound interferon (rSIFN-co) or its equivalent with changed spatial configuration, high efficacy and low side effects. Therefore, high dose of rSIFN-co may be used. This invention also relates to a process to produce said super-compound interferon (rSIFN-co) or a pharmaceutical composition comprising said super-compound interferon (rSIFN-co) or its equivalent, and uses of said interferon or composition for anti-viral and anti-tumor therapy.
  • rSIFN-co super-compound interferon
  • IFN-con is a new interferon molecule constructed with the most popular conservative amino acid found in natural human IFN- ⁇ subtypes using genetic engineering methods.
  • U.S. Pat. Nos. 4,695,623 and 4,897,471 have described it. IFN-con had been proven to have broad-spectrum IFN activity and virus- and tumor-inhibition and natural killer cell activity.
  • U.S. Pat. No. 5,372,808 by Amgen, Inc. addresses treatment INFERGEN® (interferon alfacon-1).
  • Chinese Patent No. 97193506.8 by Amgen, Inc. addresses re-treatment of INFERGEN® (interferon alfacon-1) on hepatitis C.
  • Chinese Patent No. 98114663.5 by Shenzhen Jiusheng Bio-engineering Ltd. addresses recombinant human consensus interferon- ⁇ treatment for hepatitis B and hepatitis C.
  • FDA United States Food and Drug Administration
  • IFN- ⁇ is commonly used in clinics to treat hepatitis B. IFN- ⁇ binds superficial cell membrane receptors, thus inhibiting DNA and RNA (ribonucleic acid) duplication and inducing some enzymes to prevent duplication of the virus in hepatitis-infected cells. All IFNs can inhibit DNA duplication of viruses, but they cannot inhibit the e and s antigen expression.
  • SARS severe acute respiratory syndrome
  • This disclosure describes recombinant super-compound interferon (rSIFN-co), method to produce the same and uses thereof.
  • the super-compound interferon disclosed herein is capable of inhibiting, preventing and/or treating the hepatitis viruses, SARS virus, or virus-induced upper respiratory diseases, the Influenza virus, for example Avian Influenza virus and Ebola virus.
  • rSIFN-co is effective in preventing and/or treating viral diseases and tumors with less side effects as compared to other available interferons.
  • This invention provides a recombinant super-compound interferon (rSIFN-co) and its equivalent with changed spatial configuration, high efficacy and low side effects. Therefore, high dose of rSIFN-co may be used.
  • This invention also provides artificial gene encoding for the super-compound interferon or its equivalent.
  • This invention provides a vector comprising the gene which codes for the super-compound interferon or its equivalent.
  • This invention provides an expression system comprising the vector comprising the gene which codes for the super-compound interferon or its equivalent.
  • This invention also provides a host cell comprising the vector comprising the gene which codes for the recombinant super-compound interferon (rSIFN-co) or its equivalent.
  • Said host cell may be eukaryotic or prokaryotic, such as E. Coli.
  • This invention provides a method for producing a recombinant super-compound interferon (rSIFN-co) with changed spatial configuration and enhanced antiviral activity comprising steps of:
  • This invention provides a method for inhibiting, preventing or treating viral diseases, or for inhibiting or treating tumors in a subject comprising administering to the subject an effective amount of the super-compound interferon or its equivalent.
  • This invention provides the above-described method wherein super-compound interferon is administered orally, via vein injection, muscle injection, peritoneal injection, subcutaneous injection, nasal or mucosal administration, or by inhalation via a respirator.
  • This invention provides the method to prevent or treat viral diseases wherein the viral diseases is hepatitis A, hepatitis B, hepatitis C, other types of hepatitis, infections of viruses caused by Epstein-Barr virus, Human Immunodeficiency Virus (HIV), Ebola virus, Severe Acute Respiratory Syndrome Virus (SARS), Influenza virus, Cytomegalovirus, herpes simplex viruses, or other types of herpes viruses, papovaviruses, poxviruses, picornaviruses, adenoviruses, rhinoviruses, human T-cell leukemia viruses I, or human T-cell leukemia viruses II, or human T-cell leukemia virus III.
  • the viral diseases is hepatitis A, hepatitis B, hepatitis C, other types of hepatitis, infections of viruses caused by Epstein-Barr virus, Human Immunodeficiency Virus (HIV), Ebola virus, Severe A
  • This invention provides the method to prevent or treat viral diseases wherein the viral diseases are Human Immunodeficiency Virus (HIV) and Ebola virus.
  • HIV Human Immunodeficiency Virus
  • Ebola virus Ebola virus
  • This invention provides a method for anti-hepatitis activities. It can inhibit HBV-DNA replication, HBsAg and HBeAg production.
  • This invention provides a method to prevent or treat upper respiratory infection diseases.
  • This invention provides a method to prevent or treat tumors or cancers wherein the tumor is skin cancer, basal cell carcinoma and malignant melanoma, renal cell carcinoma, liver cancer, thyroid cancer, rhinopharyngeal cancer, solid carcinoma, prostate cancer, stomach/abdominal cancer, esophageal cancer, rectal cancer, pancreatic cancer, breast cancer, ovarian cancer, and superficial bladder cancer, hemangioma, epidermoid carcinoma, cervical cancer, non-small-cell lung cancer, small-cell lung cancer, glioma, leucocythemia, acute leucocythemia and chronic leucocythemia, chronica myelocytic leukemia, hairy cell leukemia, lymphadenoma, multiple myeloma, polycythemia vera, or Kaposi's sarcoma.
  • the tumor is skin cancer, basal cell carcinoma and malignant melanoma, renal cell carcinoma, liver cancer, thyroid cancer, rhinopharyngeal cancer, solid carcinoma, prostate
  • This invention provides a method for preventing or treating virus-induced diseases in a subject comprising administering to the subject an effective amount of recombinant super-compound interferon or a functional equivalent thereof.
  • the super-compound interferon may be administered orally, via vein injection, muscle injection, peritoneal injection, subcutaneous injection, nasal or mucosal administration, or by inhalation via a respirator.
  • This invention provides a method for inhibiting the causative agent of virus-induced diseases, comprising contacting the causative agent with an effective amount of super-compound interferon or its equivalent.
  • This invention also provides a method for inhibiting virus-induced diseases, comprising contacting an effective amount of the super-compound interferon with said virus or cells. This contact could be direct or indirect.
  • This invention provides a composition comprising an effective amount of the super-compound interferon capable of inhibiting, preventing or treating virus-induced diseases, and a suitable carrier.
  • This invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of the recombinant super-compound interferon capable of inhibiting, preventing or treating virus-induced diseases in a subject, and a pharmaceutically acceptable carrier.
  • This invention provides a method for preventing or treating tumors in a subject comprising administering to the subject an effective amount of recombinant super-compound interferon or a functional equivalent thereof.
  • This invention provides a method for inhibiting tumors, comprising contacting the causative agent with an effective amount of super-compound interferon or its equivalent.
  • This invention also provides a method for inhibiting tumors, comprising contacting an effective amount of the super-compound interferon with said virus or cells. This contact could be direct or indirect.
  • This invention provides a composition comprising an effective amount of the super-compound interferon capable of inhibiting, preventing or treating tumors, and a suitable carrier.
  • This invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of the recombinant super-compound interferon capable of inhibiting, preventing or treating tumors in a subject, and a pharmaceutically acceptable carrier.
  • FIG. 1 rSIFN-co cDNA sequence (SEQ ID NO: 2 and 3) designed according to E. Coli . codon usage and deduced rSIFN-co amino acid sequence (SEQ ID NO: 1)
  • FIGS. 2A-B Sequence of another super-compound interferon ( FIG. 2A —SEQ ID NO: 4-6; FIG. 2B —SEQ ID NO: 7-9)
  • FIG. 3 Diagram of pLac T7 cloning vector plasmid
  • FIG. 4 Diagram of pHY-4 expression vector plasmid
  • FIG. 5 Construction process of expression plasmid pHY-5
  • FIG. 6 -A Circular Dichroism spectrum of INFERGEN®
  • Samples contains 30 ⁇ g/ml IFN-cont, 5.9 mg/ml of NaCl and 3.8 mg/ml of Na 2 PO 4 , pH7.0.
  • INFERGEN® interferon alfacon-1
  • Amgen Inc. also known as consensus interferon
  • HCV chronic hepatitis C virus
  • FIG. 6 -B Circular Dichroism spectrum of INFERGEN® From Reference [Journal of Interferon and Cytokine Research. 16:489-499(1996)]
  • Circular dichroism spectra of concensus interferon subforms Concensus interferon was fractionated using an anion exchange column. Samples were dialyzed into 10 mM sodium phosphate, pH 7.4. Measurements were made on Jasco J-170 spectopolarimeter, in a cell thermostat at 15° C. (——), acylated form; (--) cis terminal form; ( . . . ), met terminal form.
  • FIG. 6 -C Circular Dichroism spectrum of rSIFN-co
  • Samples contains 0.5 mg/ml rSIFN-co, 5.9 mg/ml of NaCl and 3.8 mg/ml of Na 2 PO 4 , pH7.0.
  • FIG. 6 -D Circular Dichroism spectrum of rSIFN-co
  • Samples contains 30 ⁇ g/ml rSIFN-co, 5.9 mg/ml of NaCl and 3.8 mg/ml of Na 2 PO 4 , pH7.0.
  • FIG. 7 rSIFN-co Crystal I
  • FIG. 8 rSIFN-co Crystal II
  • FIG. 9 The X-ray Diffraction of rSIFN-co Crystal
  • FIG. 10 Comparison of Inhibition Effects of Different Interferons on HBV Gene Expression
  • FIG. 11A-C Recombinant Super-Compound Interferon Spray
  • Width 25 mm (bottom), 6 mm (top)
  • FIG. 11D Recombinant Super-Compound Interferon Spray
  • FIG. 12 Comparison of Anti-SARS Activity of Interferons:
  • Left top panel is negative control i.e. no virus added.
  • Right top panel is positive control i.e. virus is added, but no rSIFN-co added.
  • Left bottom panel is rSIFN-co with SARS Virus.
  • Right bottom panel is ⁇ IFN.
  • This figure is the record of body temperature changes of 5 patients in Group A.
  • This figure is the record of body temperature changes of the other 5 patients in Group A.
  • FIGS. 13B-1 Curves of Changes of Body Temperature in Group B (5 patients)
  • This figure is the record of body temperature changes of 5 patients in Group B.
  • FIGS. 13B-2 Curves of Changes of Body Temperature in Group B (6 patients)
  • This figure is the record of body temperature changes of the other 6 patients in Group B.
  • FIG. 15 Graph of Inhibition of Drug Resistant HIV by rSIFN-co using EXCEL and Luciferase as Y axis and concentration of rSIFN-co as X axis. A clear inverse dose-dependent response has been shown.
  • FIG. 16 rSIFN-co Inhibition of Influenza Virus: on the left, the control well is shown with Influenza virus added and without interferon, the cells had obvious CPE, such as rounding of cells, cell necroses, decrease in reflective light and sloughing off.
  • FIGS. 17A-H Clinical Report of a patient with mammary and ovarian cancers. These figures show an obvious anti-cancer effect of rSIFN-co on this patient.
  • This invention provides a recombinant super-compound interferon (rSIFN-co) or an equivalent thereof with changed spatial configuration.
  • rSIFN-co super-compound interferon
  • This invention reveals that proteins with the same primary sequence might have different biological activities. As illustrated in this application, proteins with identical amino acid sequences may have different activities. The efficacy of these proteins may sometimes be improved and, sometimes, proteins with changed spatial configuration would reveal new function.
  • equivalents are molecules which are similar in function to the compound interferon.
  • An equivalent could be a deletion, substitution, or replacement mutant of the original sequence.
  • Mimics could be a peptide, polypeptide or a small chemical entity.
  • the recombinant super-compound interferon (rSIFN-co) disclosed has higher efficacy than ⁇ , ⁇ , ⁇ , ⁇ or a combination thereof and as compared to the interferons disclosed in U.S. Pat. Nos. 4,695,623 and 4,897,471.
  • This recombinant super-compound interferon (rSIFN-co) is believed to have unique secondary or tertiary structure, wherein the 3-dimensional change is the result of changes in its production process. (See e.g. FIG. 6 .)
  • the recombinant super-compound interferon (rSIFN-co) possesses lower side effects when compared with other interferons. These lower side effects allow for higher dosages to be used on patients in need of interferon treatments. These lower side effects open the possibility of using rSIFN-co for prevention and/or treatment of other diseases. Accordingly, this invention provides the recombinant super-compound interferon (rSIFN-co) with less side effects when administered to a subject.
  • This invention provides recombinant super-compound interferon (rSIFN-co) with less side effects as compared to all currently available interferons.
  • This invention also provides artificial gene encoding for the super-compound interferon or its equivalent. It is within the ordinary skill to design an artificial gene. Many methods for generating nucleotide sequence and other molecular biology techniques have been described previously. See for example, Joseph Sambrook and David W. Russell, Molecular Cloning: A laboratory Manual, December 2000, published by Cold Spring Harbor Laboratory Press.
  • the recombinant super-compound interferon may also be produced with its gene as artificially synthesized cDNA with adjustment of its sequence from the wild-type according to codon preference of E. Coli . Extensive discussion of said codon usage (preference) may be found in U.S. Pat. No. 4,695,623. See e.g. column 6, line 41-column 7, line 35.
  • This invention provides an expression system comprising the vector comprising the gene which codes for the super-compound interferon or its equivalent.
  • the cells include, but are not limited to, prokaryotic or eukaryotic cells.
  • This invention also provides a host cell comprising the vector comprising the gene which codes for the recombinant super-compound interferon (rSIFN-co) or its equivalent.
  • This invention provides the method for producing recombinant super-compound interferon (rSIFN-co), further comprising recovery of the expressed interferon.
  • the above-described recombinant super-compound interferon may be produced by a high-efficiency expression system which uses a special promoter, enhancer or other regulatory element.
  • the promoter is inducible.
  • Said inducible promoter includes but is not limited to P BAD , heat shock promoters or heavy metal inducible promoters.
  • Heat shock promoters are activated by physical means, while other promoters are activated by chemical means, for example IPTG or Tetracyclin. IPTG is added to the cells to activate the downstream gene or removed to inactivate the gene. Tetracyclin is used to induce promoters or to regulate the strength of promoters.
  • the promoter is P BAD . Since early nineties, the properties of the mechanism of expression and repression of P BAD by AraC have been studied extensively, and their interactions have been dissected at the molecular level. See Schleif, R. S. 1992 DNA looping. Annu. Rev. Biochem. 61:199-223.
  • the AraC protein is both a positive and negative regulator, when present, it turns on the transcription from the P BAD promoter, when absent, the transcription occurs at a very low rate. See Guzman, L. M. et al. (1995) J. Bact. 177: 4121-4130.
  • the efficacy and mechanism of P BAD promoter is well known by other ordinary skilled artisans and is commercially-available.
  • the commercially-available Invitrogen expression kit includes pBAD vectors' designed to provide precise control of expression levels.
  • the araBAD promoter initiates gene expression. It's both positively and negatively regulated by the product of the araC gene, a transcriptional regulator that forms a complex with L-arabinose.
  • the AraC dimer contacts the O2 and I1 half sites of the araBAD operon, forming a 210 bp DNA loop.
  • two events are required: first, Arabinose binds to AraC. The protein releases the O2 site and binds the I2 site, which is adjacent to the I1 site. This releases the DNA loop and allows transcription to begin.
  • the cAMP activator protein (CAP)-cAMP complex binds to the DNA and stimulates binding of AraC to I1 and I2. Basal expression levels can be repressed by introducing glucose to the growth medium. Glucose acts by lowering cAMP levels, which in turn decreases the binding of CAP. As cAMP levels are lowered, transcriptional activation is decreased. Invitrogen's pBAD vectors are specifically designed for maximum expression and ease of use.
  • pBAD102/D-TOPO® pBAD102/D-TOPO®
  • pBAD202/D-TOPO® pBAD-TOPO®
  • pBAD-TOPO® pBAD/Thio-TOPO®
  • pBAD/His pBAD/Myc-His
  • pBAD-DEST49 pBAD/gIII
  • pBAD/Thio-E with the following features in all pBAD vectors:
  • the inducible promoters include but are not limited to heat shock promoters or heavy metal inducible promoters.
  • This invention provides a process for production of recombinant super-compound interferon (rSIFN-co) comprising introducing an artificial gene with selected codon preference into an appropriate host, culturing said introduced host in an appropriate condition for the expression of said compound interferon and harvesting the expressed compound interferon.
  • rSIFN-co super-compound interferon
  • the process may comprise extraction of super recombinant super-compound interferon (rSIFN-co) from fermentation broth, collection of inclusion bodies, denaturation and renaturation of the harvested protein.
  • rSIFN-co super recombinant super-compound interferon
  • the process may maintain the high efficacy even when the recombinant super-compound interferon (rSIFN-co) is used with an agent and in a particular concentration.
  • the process also comprises separation and purification of the recombinant super-compound interferon (rSIFN-co).
  • the process further comprises lyophilization of the purified recombinant super-compound interferon (rSIFN-co).
  • the process comprises production of liquid injection of recombinant super-compound interferon (rSIFN-co).
  • rSIFN-co recombinant super-compound interferon
  • the IFN DNA was redesigned according to the E. Coli . codon usage and then the rSIFN-co gene was artificially synthesized.
  • rSIFN-co cDNA was cloned into the high-expression vector of E. Coli . by DNA recombinant techniques, and a high expression of rSIFN-co was gained by using of induce/activate-mechanism of L-arabinose to activate the transcription of P BAD promoter.
  • arabinose induction/activation system Compared with usual thermo-induction, pH induction and IPTG induction systems of genetic engineering, arabinose induction/activation system has some advantages: (1) Common systems relieve promoter function by creating a “derepression” pattern. Promoters then induce downstream gene expression. Temperature and pH change and the addition of IPTG cannot activate promoters directly. In the system disclosed herein, L-arabinose not only deactivates and represses but also activates the transcription of P BAD promoter which induces a high expression of rSIFN-co. Therefore, the arabinose induction/activation system is a more effective expression system. (2) The relationship between Exogenous and L-arabinose dosage is linear. This means the concentration of arabinose can be changed to adjust the expression level of the exogenous gene.
  • This embodiment creates an effective and resistant rSIFN-co-expressing E. Coli . engineering strain with an L-arabinose induction/activation system.
  • the strain is cultivated and fermented under suitable conditions to harvest the bacterial bodies.
  • Inclusion bodies are then purified after destroying bacteria and washing repeatedly.
  • mass of high-purity, spatial-configuration-changed rSIFN-co protein for this invention and for clinical treatment was gained from denaturation and renaturation of inclusion bodies and a series of purification steps. Said purification would not affect the biological activity of the purified protein.
  • rSIFN-co recombinant super-compound interferon
  • This invention provides a method for treating or preventing viral diseases or tumors in a subject comprising administering to the subject an effective amount of the recombinant super-compound interferon (rSIFN-co) or its equivalent.
  • viral diseases include, but are not limited to, hepatitis A, hepatitis B, hepatitis C, other types of hepatitis, infections caused by Epstein-Barr virus, Human Immunodeficiency Virus (HIV), Ebola virus, Severe Acute Respiratory Syndrome Virus (SARS), Influenza virus, Cytomegalovirus, herpes simplex viruses, other herpes viruses, papovaviruses, poxviruses, picornaviruses, adenoviruses, rhinoviruses, human T-cell leukemia virus I, human T-cell leukemia virus II, or human T-cell leukemia virus III.
  • HIV Human Immunodeficiency Virus
  • SARS Severe Acute Respiratory Syndrome Virus
  • Influenza virus Cytomegalovirus
  • herpes simplex viruses other herpes viruses
  • papovaviruses poxviruses
  • picornaviruses picornaviruses
  • the effective amount is at nanogram level.
  • the virus is Human Immunodeficiency Virus and the effective amount is as low as 4 nanograms per milliliter.
  • the virus is Influenza and the effective amount is as low as 10 nanogram per milliliter.
  • the recombinant super-compound interferon inhibits the DNA duplication and secretion of HBsAg and HBeAg of Hepatitis B Virus.
  • SARS Severe Acute Respiratory Syndrome Virus
  • This invention provides a method for preventing or treating Severe Acute Respiratory Syndrome, or virus-induced upper respiratory diseases, of a subject comprising administering to the subject an effective amount of recombinant super-compound interferon (rSIFN-co) or a functional equivalent thereof.
  • the interferon is ⁇ , ⁇ , ⁇ , ⁇ or a combination thereof.
  • the recombinant super-compound interferon may be administered orally, via vein injection, muscle injection, peritoneal injection, subcutaneous injection, nasal or mucosal administration, or by inhalation via a spray or a respirator.
  • rSIFN-co is administered subcutaneously or intramuscularly at a dose of higher than or equal to 10 Million International Unit per square meter of surface area.
  • rSIFN-co is administered subcutaneously or intramuscularly at a dose of higher than or equal to 20 Million International Unit per square meter of surface area.
  • the interferon is delivered by a spray device. In a specific embodiment, the device is described in FIG. 11 . In one of the embodiments, the interferon is lyophilized.
  • This invention provides a method for inhibiting the causative agent of Severe Acute Respiratory Syndrome, or virus-induced upper respiratory diseases, comprising contacting the agent with an effective amount of recombinant super-compound interferon (rSIFN-co) or its equivalent.
  • rSIFN-co super-compound interferon
  • the causative agent of SARS is a virus. See eg. Rota et al (2003), Characterization of a Novel Coronavirus Associated with Severe Acute Respiratory Syndrome. Science 1085952 and Marra, et al. (2003), The Genome Sequence of the SARS-Associated Coronavirus. Science 1085853.
  • This invention also provides a method for inhibiting Severe Acute Respiratory Syndrome virus or Severe Acute Respiratory Syndrome virus-infected cells, or virus-induced upper respiratory diseases, or cells infected with viruses capable of inducing upper respiratory diseases, comprising contacting an effective amount of the recombinant super-compound interferon (rSIFN-co) with said virus or cell.
  • This contact could be direct or indirect.
  • This invention provides a composition comprising an effective amount of the recombinant super-compound interferon (rSIFN-co) capable of inhibiting Severe Acute Respiratory Syndrome virus or Severe Acute Respiratory Syndrome virus-infected cells, or virus-induced upper respiratory diseases, or cells infected with viruses capable of inducing upper respiratory diseases, and a suitable carrier.
  • rSIFN-co super-compound interferon
  • This invention provides a composition comprising an effective amount of the super-compound interferon capable of preventing or treating Severe Acute Respiratory Syndrome, or virus-induced upper respiratory diseases, of a subject and a suitable carrier.
  • This invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of the recombinant super-compound interferon (rSIFN-co) capable of inhibiting Severe Acute Respiratory Syndrome virus or Severe Acute Respiratory Syndrome virus-infected cells, or virus-induced upper respiratory diseases, and a pharmaceutically acceptable carrier.
  • rSIFN-co super-compound interferon
  • This invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of the recombinant super-compound interferon (rSIFN-co) capable of preventing or treating Severe Acute Respiratory Syndrome, or virus-induced upper respiratory diseases, in a subject and a pharmaceutically acceptable carrier.
  • rSIFN-co super-compound interferon
  • This invention provides a device to deliver the above-described pharmaceutical composition.
  • the subject is a human.
  • the super-compound interferon can be used in other animals or mammals.
  • This invention provides a method for preventing Severe Acute Respiratory Syndrome or virus-induced upper respiratory diseases, in humans comprising application of the super-compound interferon three times a day via a spray which contains twenty micrograms of interferon, equal to ten million units of activity in three milliliter.
  • VURI Viral Upper Respiratory Infection
  • Viral upper respiratory infection alternative names common cold, colds. This is a contagious viral infection of the upper respiratory tract characterized by inflammation of the mucous membranes, sneezing, and a sore throat. It is usually caused by over 200 different viruses, known as rhinoviruses. Colds are not caused by the same viruses responsible for Influenza. Colds are spread through droplets from the coughing or sneezing of others with a cold or by hand contact with objects contaminated by someone with a cold. The incidence of colds is highest among children, and the incidence decreases with age because immunity to the virus causing the cold occurs after the illness. Gradually, immunity to a wide variety of viruses that cause colds is developed in adults. Children may have 10 colds a year, and adults may have 3 colds a year.
  • URIs upper respiratory tract infections
  • coronaviruses 88% or the respiratory syncytial virus (7%) may also lead to infection.
  • Other viruses such as influenza (6%), parainfluenza, and adenoviruses, may produce respiratory symptoms, but these are often associated with pneumonia, fever, or chills.
  • the common cold has a variety of overt symptoms, including malaise, nasal stuffiness, rhinorrhea, nonproductive cough, mild sore throat, and, in some cases, a low-grade fever. Because of the similarity of symptoms, a cold may be mistaken for perennial allergic rhinitis, but allergies can usually be ruled out because of the differences in chronicity.
  • Aerosolized beta2 Reverse Raises heart rate and agonists eg, postinflammatory may cause tremor albuterol
  • bronchospasm Alcohol-based liquid Treat multiple Potential drowsiness and combination products symptoms coordination problems
  • Alphal agonists Decongestion May cause tachycardia, (oral) (eg, nervousness, transient pseudoephedrine, stimulation, dizziness, phenylpropanolamine) drowsiness, elevation of blood pressure
  • Anticholinergic Drying May cause nasal dryness compounds: and occasional epistaxis Ipratropium bromide (topical) Other Drying May cause orthostasis, anticholinergics dysfunction of heat (eg, regulation, dry mouth, methscopolamine, constipation atropine, hyoscyamine)
  • Antihistamines Drying Drowsiness, dry mouth, (oral) (eg, orthostatic hypertension chlorpheni
  • URI Nearly 70 ⁇ 80% URI are caused by viruses such as respiratory Syncytical virus, adenovirus, rhinovirous, cox-sackie virus, corona virus and its variant, influenza A virus and its variant, influenza B virus and its variant, parainfluenza virus and its variant, or enterovirus and its variant.
  • viruses such as respiratory Syncytical virus, adenovirus, rhinovirous, cox-sackie virus, corona virus and its variant, influenza A virus and its variant, influenza B virus and its variant, parainfluenza virus and its variant, or enterovirus and its variant.
  • a main cause of URI in adults is from rhinovirous.
  • respiratory syncytical virus and parainfluenza virus are two leading causes of URI.
  • rSIFN-co Recombinant super-compound interferon
  • recombinant super-compound interferon rSIFN-co
  • rSIFN-co recombinant super-compound interferon
  • this invention provides a method for inhibiting, preventing or treating virus replication or virus-infected cells by contacting said virus or infected cells with an effective amount of the recombinant super-compound interferon (rSIFN-co) or its equivalent.
  • This recombinant super-compound interferon (rSIFN-co) is useful in inhibiting, preventing or treating the following cancers or tumors:
  • this invention provides a method for inhibiting tumor or cancer cell growth by contacting the recombinant super-compound interferon (rSIFN-co) or its equivalent with said tumor or cancer cells.
  • This invention also provides the produced super-compound interferon by the above processes.
  • This invention provides a composition comprising recombinant super-compound interferon (rSIFN-co) or its equivalent and a suitable carrier.
  • This invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the recombinant super-compound interferon (rSIFN-co) or its equivalent and a pharmaceutically acceptable carrier.
  • This invention provides the above-described method wherein recombinant super-compound interferon (rSIFN-co) is administered via orally via vein injection, muscle injection, peritoneal injection, subcutaneous injection, nasal or mucosal administration, or by inhalation via a spray or a respirator.
  • rSIFN-co recombinant super-compound interferon
  • This invention provides the above-described method wherein recombinant super-compound interferon (rSIFN-co) is administered following the protocol of injections of 9 ⁇ g, 15 ⁇ g or 24 ⁇ g every two days, 3 times a week, for 24 weeks.
  • rSIFN-co recombinant super-compound interferon
  • rSIFN-co recombinant super-compound interferon
  • One objective of this invention is to offer a preparation of recombinant super-compound interferon (rSIFN-co) to directly inhibit the DNA duplication of hepatitis B viruses and the secretion of HBeAg and HBsAg of hepatitis B and decrease them to normal levels.
  • rSIFN-co super-compound interferon
  • the medicine carrier could be any acceptable medicine carrier, including carbohydrates, cellulosum, adhesive, collapse, emollient, filling, add-dissolving agent, amortization, preservative, thickening agent, matching, etc.
  • This invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising the above composition and a pharmaceutically acceptable carrier.
  • “pharmaceutically acceptable carriers” means any of the standard pharmaceutical carriers.
  • suitable carriers are well known in the art and may include, but are not limited to, any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution and various wetting agents.
  • Other carriers may include additives used in tablets, granules, capsules, etc.
  • Such carriers contain excipients such as starch, milk, sugar, certain types of clay, gelatin, stearic acid or salts thereof, magnesium or calcium stearate, talc, vegetable fats or oils, gum, glycols or other known excipients.
  • Such carriers may also include flavor and color additives or other ingredients. Compositions comprising such carriers are formulated by well-known conventional methods.
  • Pegylation is the process by which polyethylene glycol chains are attached to protein and peptide drugs to increase pharmacokinetics by shielding these proteins and peptide drugs from proteolytic enzymes. See Harris and Chess, Effect of pegylation on pharmaceuticals . Nat Rev Drug Discov. 2003 March; 2(3):214-21.
  • Pegylations is a well-established method for increasing the circulating half-life of protein and liposomal pharmaceuticals based on large hydrodynamic volume of polyethylene glycols. These polyethylene glycols shield the proteins and peptide drugs from renal clearance, enzymatic degradation and immune system recognition, thus their half-life and making them more acceptable to patients. See Molineux, Pegylation: engineering improved pharmaceuticals for enhanced therapy . Cancer Treat Rev. 2002 April; 28 Suppl A: 13-6. The author concludes that pegylation has beneficial effect on the quality of life of cancer patients.
  • Pegylation of the interferon increases the amount of time the interferon remains in the body by increasing the size of the interferon molecule by decreasing the rate of absorption, prolonging the half-life and the rate of interferon clearance.
  • the duration of biological activity is increased with pegylated interferon over nonpegylated interferon, thus providing an advantage over nonpegylated interferons with less frequent administration and comparable tolerability.
  • the author states that monotherapy with pegylated interferon produces a better response in some patients than monotherapy with the nonpegylated formulation. See Baker, Pegylated Interferons . Rev Gastroenterol Disord. 2001; 1(2):87-99.
  • Sustained release delivery matrices and liposomes maybe used with rSIFN-co to create sustained release and controlled release formulation. See Robinson and Talmadge, Sustained Release of Growth Factors . In Vivo 2002 November-December; 16(6): 535-40. The authors state that both pegylation and sustained release delivery matrices and liposomes improve the pharmacokinetic and pharmacodynamic properties of recombinant molecules, and thus by improving clinical efficacy these approaches increase patient compliance.
  • This invention provides recombinant super-compound interferon (rSIFN-co) comprising an agent or encapsulated by an agent, capable of affecting the half-life or delivery of said interferon.
  • this agent is polyethylene glycol (PEG).
  • This invention further provides a method for treating or preventing viral diseases or tumors in a subject comprising administering to the subject an effective amount of the recombinant super-compound interferon (rSIFN-co) or its equivalent comprising an agent or encapsulated by an agent, capable of affecting the half-life or delivery of said interferon.
  • this agent is polyethylene glycol (PEG).
  • IFN-con is a new interferon molecule constructed according to conservative amino acids in human IFN- ⁇ subtype using genetic engineering methods. It has been proven that IFN-con has broad-spectrum IFN activity, such as high antivirus and tumor inhibition activity, especially for effectively treating hepatitis C.
  • E. Coli . codon was used to redesign rSIFN-co cDNA and then artificially synthesize cDNA of rSIFN-co from published INFERGEN® (interferon alfacon-1) DNA sequences and deduced amino acid sequences ( FIG. 1 ).
  • rSIFN-co cDNA was cloned into E. Coli . high-expression vector, and L-arabinose, which can activate strong P BAD promoter in vectors, was used to induce high expression of rSIFN-co gene.
  • rSIFN-co cDNA was redesigned according to the codon usage of E. Coli . to achieve high expression in E. Coli .
  • Deduced amino acid sequence (SEQ ID NO: 1) from the redesigned cDNA sequence (SEQ ID NO: 2 and 3) of rSIFN-co is completely coincidental with primitive amino acid sequence of published INFERGEN® (interferon alfacon-1) ( FIG. 1 ).
  • Two semi-moleculars can be directly synthesized: rSIFN-co cDNA 5′-terminus 280 bp (fragment I) and 3′-terminus 268 bp (fragment II) by PCR. There are 41 bp overlapping among fragment II and fragment I.
  • Oligomer A (SEQ ID NO: 10) 5′ATGTGCGACCTGCCGCAGACCCACTCCCTGGGTAACCGTCGTGCTCTG ATCCTGCTGGCTCAGATGCGTCGTATCTCCCCGTTCTCCTGCCTGAAAGA CCGTCACGAC3′
  • Oligomer B (SEQ ID NO: 11) 5′CTGAAAGACCGTCACGACTTCGGTTTCCCGCAGGAAGAATTCGACGGT AACCAGTTCCAGAAAGCTCAGGCTATCTCCGTTCTGCACGAAATGATCCA GCAGACCTTC3′
  • Oligomer C (SEQ ID NO: 12) 5′GCTGCTGGTACAGTTCGGTGTAGAATTTTTCCAGCAGGGATTCGTCCC AAGCAGCGGAGGAGTCTTTGGTGGAGAACAGGTTGAAGGTCTGCTGGATC ATTTC3′
  • Oligomer D (SEQ ID NO: 13) 5′ATCCCTGCTGGAAAAATTCTACACCGAACTGTACCAGCAGCTGAACGA CC
  • PCR I for Fragment I oligodeoxynucleotide B as template, oligodeoxynucleotide A and C as primers, synthesized 280 bp Fragment I.
  • PCR I mixture (units: ⁇ l) sterilized distilled water 39 10 ⁇ Pfu buffer (Stratagen American Ltd.) 5 dNTP mixture (dNTP concentration 2.5 mmol/L) 2 Oligomer A primer (25 ⁇ mol/L) 1 Oligomer C primer (25 ⁇ mol/L) 1 Oligomer B template (1 ⁇ mol/L) 1 Pfu DNA polymerase (Stratagen American Ltd.) (25 U/ ⁇ l) 1 Total volume 50 ⁇ l
  • PCR II mixture (units: ⁇ l) sterilized distilled water 39 10 ⁇ Pfu buffer (Stratagen American Ltd.) 5 dNTP mixture (dNTP concentration 2.5 mmol/L) 2 Oligomer D primer (25 ⁇ mol/L) 1 Oligomer F primer (25 ⁇ mol/L) 1 Oligomer E template (1 ⁇ mol/L) 1 Pfu DNA polymerase (Stratagen American Ltd.) (25 U/ ⁇ l) 1 Total volume 50 ⁇ l
  • Fragment I and II were assembled together to get the complete cDNA molecular sequence of rSIFN-co using the overlapping and extending PCR method. Restriction enzyme Nde I and Pst I were introduced to clone rSIFN-co cDNA sequence into plasmid.
  • Oligomer G (SEQ ID NO: 16) 5′ATCGGCCATATGTGCGACCTGCCGCAGACCC3′
  • Oligomer H (SEQ ID NO: 17) 5′ACTGCCAGGCTGCAGTTATTCTTTACGACGCAGACGTTCC3′
  • PCR mixture (units: ⁇ l) sterilized distilled water 38 10 ⁇ Pfu buffer (Stratagen American Ltd.) 5 dNTP mixture (dNTP concentration 2.5 mmol/L) 2 primer G (25 ⁇ mol/L) 1 primer H (25 ⁇ mol/L) 1 *fragment I preduction (1 ⁇ mol/L) 1 *fragment II preduction (1 ⁇ mol/L) 1 Pfu DNA polymerase (Stratagen American Ltd.) (2.5 U/ ⁇ l) 1 Total volume 50 ⁇
  • pLac T7 plasmid as cloning vector.
  • pLac T7 plasmid is reconstructed with pBluescript II KS(+) plasmid produced by Stratagen ( FIG. 3 ).
  • N-Cys-Asp-Leu-Pro-Gln-Thr-His-Ser-Leu-Gly-Asn-Arg-Arg-Ala-Leu- SEQ ID NO: 18
  • the pHY-4 plasmid is double digested with Nde I-Xba I. Run 1% agarose gel electrophoresis and purify the 715 bp fragment. Ligate the rSIFN-co and pHY-4 fragments with T4 DNA ligase to construct the recombinant plasmid (See FIG. 4 ). Transform DH 5 ⁇ competent cells with the recombinant plasmid. Spread the transformed cells on LB plate with Amp, 37° C. culture overnight.
  • rSIFN-co gene is under the control of strong promoter P BAD .
  • This promoter is positively and negatively regulated by the product of the gene araC.
  • AraC is a transcriptional regulator that forms a complex with arabinose. In the absence of arabinose, the AraC dimer binds O 2 and I 1 , forming a 210 bp loop. This conformation leads to a complete inhibition of transcription. In the presence of arabinose, the dimer is released from O 2 and binds I 1 and I 2 leading to transcription.
  • rSIFN-co expression level in PVIII is more than 50% of the total E. Coli . protein.
  • rSIFN-CO is a new interferon molecule artificially built according to the conservative amino acid of human a interferons. It has been proven as an effective anti-hepatitis drug. In order to get enough pure rSIFN-co protein, a stable recombinant E. Coli . strain which highly expresses rSIFN-co protein was constructed.
  • the rSIFN-co cDNA was cloned into E. Coli . high-expression vector pHY-4 plasmid to construct the recombinant plasmid pHY-5.
  • E. Coli . LMG194 strain was further transformed with pHY-4 plasmid to get stable rSIFN-co high-expression transformant. This transformant was cultured for 30 generations.
  • the heredity of pHY-5 recombinant plasmid in E. Coli . LMG194 was normal and stable, and the expression of rSIFN-co was high and steady.
  • E. Coli . LMG194 which contains recombinant pHY-5 plasmid, is actually an ideal high-expression engineering strain.
  • Chelating SepharoseTM fast flow Add PB buffer of 0.2 mol/L (pH 6.6) and NaCl of 4 mol/L in the solution from HS to adjust solution pH to pH 6.0 and NaCl concentration to 1 mol/L.
  • LB Media 1 L Tryptone 10 g Yeast extracts 5 g NaCl 10 g RM Media: 1 L Casein 20 g MgCl 1 mmol/L (0.203 g) Na 2 HPO 4 4 g; KH 2 PO 4 3 g, NaCl 0.5 g NH 4 Cl 1 g
  • the buffer was changed to PBS (pH 7.0) along with the step of condensing by POROS HS/M.
  • PBS pH 7.0
  • POROS HS/M POROS HS/M
  • the quality of the product is controlled according to “Chinese Requirements for Biologics.”
  • Samples were supplied by Sichuan Huiyang Life-engineering Ltd., Sichuan province. Lot: 990101-03, 990101-05, 990102-03, 990102-05, 990103-03, 990103-05
  • Test samples at 2 ⁇ 8° C. The test samples were put into a 2 ⁇ 8° C. refrigerator, then the above items of these samples were respectively tested in the 1 st , 3 rd , 6 th , 9 th , 12 th , 18 th , 24 th , 30 th , 36 th month. The results were recorded.
  • Test samples at 25° C. The test samples were put into a thermostat at 25° C., then the above items of these samples were respectively tested in the 1 st , 3 rd , 6 th , 9 th , 12 th , 18 th , 24 th , 30 th month. The results were recorded.
  • Test samples at 37° C. The test samples were put into a thermostat at 37° C., then the above items of these samples were respectively tested in the 1 st , 3 rd , 6 th , 9 th , 12 th , 18 th , 24 th month. The results were recorded.
  • the lyophilized powder of recombinant super-compound interferon for injection should be better stored and transported at low temperatures. Without such conditions, the product can also be stored for short periods (i.e., 3 months) at room temperature.
  • the purity of the product (rSIFN-co) from this production procedure is shown to 95% under the test of SDS-PAGE where molecular weight is 14.5 Kda.
  • the reverse phase HPLC shows a single peak and the purity is up to 97%. Its specific activity is up to 1 ⁇ 10 9 IU/mg protein.
  • Preparation technique Weigh materials according to recipe. Dissolve with sterile and pyrogen-free water. Filter through 0.22 ⁇ m membrane to de-bacterialize, preserve at 6-10° C. Fill in vials after affirming they are sterile and pyrogen-free, 0.3 ml/vial or 0.5 ml/vial, and lyophilize in freeze dryer.
  • Preparation Weigh materials according to recipe. Add to desired level with sterile and pyrogen-free water. Filter through 0.22 ⁇ m membrane to de-bacterialize, preserve at 6-10° C. Fill in airtight vial after affirming it is sterile and non-pyrogen at 0.3 ml/vial or 0.5 ml/vial. Store at 2-10° C., and protect from light.
  • mice with large dose 150 ⁇ g/kg, equal to 1000 times of the normal dose per kilo used in treatment of adult patients
  • rSIFN-co at one time by intramuscular injection. Then observe and record their deaths and toxic reactions. Results show that: 24 hours after injection, no abnormal reaction had been recorded.
  • the organs of the animals which had been selected to be killed also had no signs of abnormal changes. Those remaining mice were all kept alive and were normal after two weeks.
  • the weights of mice in the experimental group and control group all increased, and the ratio of increase showed no obvious difference between the two groups (P>0.05) according to their weights on the fourteenth day. No abnormal changes were seen from the main organs of those mice after two weeks.
  • mice 40 healthy adult mice, weighing 18-22 g, half male and half female, qualified by Sichuan experiment animal control center.
  • rSIFN-co was administered i.m. in saline.
  • mice Separate the 40 mice into two groups randomly, one for experimental medicine, another for control. Inject medicines or saline at the same ratio (0.1 ml/10 g) through muscle to each mouse according to which group they belong. (150 ⁇ g/kg of rSIFN-co for experimental group; and saline for control group). After injection, observe and record acute toxicity shown in mice. Kill half of the mice (male and female each half) to check whether there were any abnormal pathologic changes in their main organs, such as heart, spleen, liver, lung, kidney, adrenal gland, stomach, duodenum, etc. after 24 hours. Those that remain are kept and observed until the fourteenth day. Weigh all mice, kill them, and then observe the appearance of the organs listed above to see if there are any abnormalities. Take pathological tissue and examine it, using the examination to assess the difference in weight increases in the two groups.
  • all mice lived well. They ate, drank, exercised, and excreted normally and showed normal hair conditions. None of them died.
  • the observation of the main organs of the randomly selected mice shows no abnormal changes 24 hours after injection. 14 days after injection, all remaining mice were killed. Autopsies also showed no changes. The weights of mice in the two groups all increased, but no obvious difference was shown when accessed with statistic method (p>0.05). See Table 6.1:
  • mice Under conditions of this experiment, there were no toxic reactions in all mice after injection of rSIFN-co with 150 ⁇ g/kg. The conclusion can be reached that the maximum tolerable dose of i.m. in mice is 150 ⁇ g/kg, which is equal to 1000 times the normal dose per kilo used in treatment of adult patients.
  • rSIFN-co Drug was white in color with produced no precipitation when water was added.
  • the pH value was 6.9 while the standard was between 6.5 to 7.5.
  • the water content of rSIFN-co was 2.3% while the standard was smaller than 3.0%.
  • Test for bacteria showed no bacterial grown.
  • rSIFN-co passed pyrogen test.
  • the toxicity test on mice showed no harm. Mice were alive and gained weight.
  • the specific activity test was 6.0 ⁇ 10 6 IU/vial while the standard was between 3.6 ⁇ 10 6 IU/vial to 6.8 ⁇ 10 6 IU/vial.
  • the identification test was positive.
  • Clinical Report 1 Evidence of effectiveness of rSIFN-co in healing cancer. See FIGS. 17A-H .
  • the ultra sound inspection showed an enlarged right ovary and abdominal fluid.
  • the patient was suspected of having ovarian cancer.
  • the patient was treated with rSFIN-co starting Jul. 14, 2004. She was injected with 15 ⁇ g of rSFIN-co on Jul. 14, 2004, Jul. 16, 2004, Jul. 18, 2004, Jul. 20, 2004 and Jul. 22, 2004 respectively. She began chemotherapy on Jul. 22, 2004. On Aug. 3, 2004 abdominal surgery was performed. It was expected that her abdominal water would be more than 2000 ml. However, only 200 ml were recorded. On Aug. 4, 2004 the examination results showed she had mammary gland cancer, ovarian cancer of right and left ovary and lymphoma. She was treated with rSIFN-co and chemotherapy at the same time. She did not have operation on mammary glands.
  • PET Imaging On Feb. 25, 2005 PET imaging report on Feb. 25, 2005 of this 43 years old patient diagnosis with left side ovary cancer and was treated with rSIFN-co since Jul. 14, 2004; PET imaging was done at PET Center of the Daping Hospital, Third Military Medical University of PRC.
  • the image of heart was clear.
  • Clinical Report 2 A kidney cancer patient was treated in the following manner. In a half-month period, the patient was given 3 injections of 9 ⁇ g of rSIFN-co and 3 injections of 15 ⁇ g of rSIFN-co. In the one and a half months following these injections, he received 24 ⁇ g injections of rSIFN-co every day. A kidney biopsy showed no metastasis after this course of treatment. The patient showed a full recovery. Every half year after recovery, the patient received 15 ⁇ g injections of rSIFN-co 15 times over a one-month period.
  • Solvent and Dispensing Method Add 1 ml saline into each vial, dissolve, and mix with MEM culture medium at different concentrations. Mix on the spot.
  • Control drugs IFN- ⁇ 2b (Intron A) as lyophilized powder, purchased from Schering Plough. 3 ⁇ 10 6 IU each, mix to 3 ⁇ 10 6 IU/ml with culture medium; INFERGEN® (liquid solution), purchased from Amgen, 9 ⁇ g, 0.3 ml each, equal to 9 ⁇ 10 6 IU, and mix to 9 ⁇ 10 6 IU/ml with culture medium preserve at 4° C.; 2.2.15 cell: 2.2.15 cell line of hepatoma (Hep G2) cloned and transfected by HBV DNA, constructed by Mount Sinai Medical Center.
  • MEM powder Gibco American Ltd. cattle fetal blood serum, HycloneLab American Ltd. G-418 (Geneticin); MEM dispensing, Gibco American Ltd.; L-Glutamyl, imported and packaged by JING KE Chemical Ltd.; HBsAg and HBeAg solid-phase radioimmunoassay box, Northward Reagent Institute of Chinese Isotope Ltd.; Biograncetina, Northern China Medicine; And Lipofectin, Gibco American Ltd.
  • 2.2.15 cell culture Added 0.25% pancreatic enzyme into culture box with full of 2.2.15 cell, digest at 37° C. for 3 minutes, and add culture medium to stop digest and disturb it to disperse the cells, reproduce with ratio of 1:3. They will reach full growth in 10 days.
  • Toxicity test Set groups of different concentrations and a control group in which cells are not acted on with medicine. Digest cells, and dispense to a 100,000 cell/ml solution. Inoculate to 96-well culture board, 200 ⁇ l each well, culture at 37° C. for 24 h with 5% CO 2 . Test when simple cell layer grows.
  • Dispense rSIFN-co to 1.8 ⁇ 10 7 IU/ml solution then prepare a series of solutions diluted at two-fold gradients. Add into 96-well culture board, 3 wells per concentration. Change the solution every 4 days. Test cytopathic effect by microscope after 8 days. Fully destroy as 4, 75% as 3, 50% as 2, 25% as 1, zero as 0. Calculate average cell lesion and inhibition rate of different concentrations. Calculate TC 50 and TC 0 according to the Reed Muench method.
  • TC 50 Antilog ⁇ ( B + 50 - B A - B ⁇ C )
  • A log>50% medicine concentration
  • B log ⁇ 50% medicine concentration
  • C log dilution power
  • HBeAg and HBsAg Separate into positive and negative HBeAg and HBsAg contrast groups, cell contrast group and medicine concentration groups. Inoculate 700,000 cells/ml of 2.2.15 cell into 6-well culture board, 3 ml each well, culture at 37° C. for 24 h with 5% CO 2 , then prepare 5 gradiently diluted solutions with 3-fold as the grade (Prepare 5 solutions, each with a different protein concentration.
  • the concentration of Solution 2 is 3 times lower than that of Solution 1, the concentration of Solution 3 is 3 times lower than that of Solution 2, etc.) 4.5 ⁇ 10 6 IU/ml, 1.5 ⁇ 10 6 IU/ml, 0.5 ⁇ 10 6 IU/ml, 0.17 ⁇ 10 6 IU/ml, and 0.056 ⁇ 10 6 IU/ml, 1 well per concentration, culture at 37° C. for 24 h with 5% CO 2 . Change solutions every 4 days using the same solution. Collect all culture medium on the 8 th day. Preserve at ⁇ 20° C. Repeat test 3 times to estimate HBsAg and HBeAg with solid-phase radioimmunoassay box (Northward Reagent Institute of Chinese Isotope Ltd.). Estimate cpm value of each well with a ⁇ -accounting machine.
  • Effects calculation Calculate cpm mean value of contrast groups and different-concentration groups and their standard deviation, P/N value such as inhibition rate, IC50 and SI.
  • Antigen ⁇ ⁇ inhibition ⁇ ⁇ IC 50 Antilog ⁇ ⁇ ( B + 50 + B A - B ⁇ C )
  • A log>50% medicine concentration
  • B log ⁇ 50% medicine concentration
  • C log dilution power
  • Southern blot (1) HBV-DNA extract in 2.2.15 cell: Culture cell 8 days. Exsuction culture medium (Separate cells from culture medium by means of draining the culture medium.). Add lysis buffer to break cells, then extract 2 times with a mixture of phenol, chloroform and isoamyl alcohol (1:1:1), 10,000 g centrifuge. Collect the supernatant adding anhydrous alcohol to deposit nucleic acid. Vacuum draw, re-dissolve into 20 ⁇ l TE buffer. (2) Electrophoresis: Add 6 ⁇ DNA loading buffer, electrophoresis on 1.5% agarose gel, IV/cm, at fixed pressure for 14-18 h.
  • Denaturation and hybridization respectively dip gel into HCl, denaturaion buffer and neutralization buffer.
  • Transmembrane Make an orderly transfer of DNA to Hybond-N membrane. Bake, hybridize and expose with dot blot hybridization. Scan and analyze relative density with gel-pro software. Calculate inhibition rate and IC 50 .
  • Results from Tables 4.1, 4.2 and 4.3 show: After maximum innocuous concentration exponent culturing for 8 days with 2.2.15 cell, the maxima is 9.0 ⁇ 0 ⁇ 10 6 IU/ml average inhibition rate of maximum innocuous concentration rSIFN-co to HBeAg is 46.0 ⁇ 5.25% (P ⁇ 0.001), IC 50 is 4.54 ⁇ 1.32 ⁇ 10 6 IU/ml, SI is 3.96; rate to HBsAg is 44.8 ⁇ 6.6%, IC 50 is 6.49 ⁇ 0.42 ⁇ 10 6 IU/ml, SI is 2.77. This shows that rSIFN-co can significantly inhibit the activity of HBeAg and HBsAg, but that the IFN of the contrast group and INFERGEN® cannot. It has also been proven in clinic that rSIFN-co can decrease HBeAg and HBsAg or return them to normal levels.
  • the recombinant super-compound interferon (rSIFN-co) is an invention for viral disease therapy, especially for hepatitis. Meanwhile, it can inhibit the activity of EB viruses, VSV, Herpes simplex viruses, cornaviruses, measles viruses, et al. Using Wish cells/VSV system as the assay for anti-virus activity, the results showed that: the other rIFN, was 0.9 ⁇ 10 8 IU/mg, Intron A was 2.0 ⁇ 10 8 IU/mg and rSIFN-co was 9 ⁇ 10 8 IU/mg. The anti-viral activity of rSIFN-co is much higher than those of the former two.
  • Standard of Patients Selection Standards 1-4 are effective for both treatment with rSIFN-co (9 ⁇ g) and IFN- ⁇ 1b (5MU, 50 ⁇ g), and Standard 1-5 are for rSIFN-co (15 ⁇ g) treatment.
  • Group C the cases were prior treatment of chronic active hepatitis B with other IFNs (3MU or 5MU) that failed or relapsed and then were treated with rSIFN-co (15 ⁇ g), subcutaneous injection, every one day, for 24 weeks. The total cases were 13. After 12 weeks treatment, 7 of 13 (53.85%) were effective. 3 of 13 (23.08%) HBeAg transferred to negative; 7 of 13 (53.85%) HBV-DNA transferred to negative; 11 of 13 (84.62%) heptal functions recovered to normal.
  • the side effects of IFN include fever, nausea, myalgia, anorexia, hair loss, leucopenia and thrombocytopenia, etc.
  • the maximum dose of IFN- ⁇ 1b is 5MIU per time; the routine dose is 3 MIU.
  • 90% patients have I-II degree (WHO standard) side effects. They had fever lower than 38° C., nausea, myalgia, anorexia, etc.
  • the maximum dose of rSIFN-co is 24 ⁇ g, subcutaneous injection, every one day for 3 months.
  • the routine dose is 9 ⁇ g.
  • I-II degree WHO standard
  • side effects including fever below 38° C., nausea, myalgia, anorexia, leucopenia and slight thrombocytopenia.
  • maximum dosage about 50% patients suffered from leucopenia and thrombocytopenia after using rSIFN-co one month, but those side effects disappeared after stopping treatment for one week. It is safe for continued use.
  • Hepatitis B virus (HBV) DNA contains consensus elements for transactivating proteins whose binding activity is regulated by interferons.
  • Treatment of HBV-infected hepatocytes with interferons leads to inhibition of HBV gene expression.
  • the aim of the present study was to characterize the effects of different interferons on HBV regulated transcription. Using transient transfection of human hepatoma cells with reporter plasmids containing the firefly luciferase gene under the control of HBV-Enhancer EnH I, Enh II and core promoter, Applicant studied the biological activities of three different interferons on transcription.
  • Interferons IFN-con1 (INFERGEN®), IFN-Hui-Yang (rSIFN-co) and IFN ⁇ -2b (Intron A).
  • Reporter plasmid The DNA fragments containing HBV-Enhancer EnH I, Enh II and core promoter were prepared using PCR and blunt-end cloned into the Smal I site of the promoter- and enhancer-less firefly luciferase reporter plasmid pGL3-Basic (Promega, Wis., USA). The resulting reporter plasmid was named as pGL3-HBV-Luc. 3.
  • HepG2 cells were cultured in DMEM medium supplemented with 10% FBS and 100 U/ml penicillin and 100 ug/ml streptomycin. The cells were kept in 30° C., 5% CO2 incubator. The cells were transfected with pGL3-HBV-Luc reporter plasmid using Boehringer's Lipofectin transfection kit. After 18 hours, the medium containing transfection reagents was removed and fresh medium was added with or without interferons. The cells were kept in culture for another 48 hours. 4. Luciferase Assay: Forty-eight hours after addition of interferon, the cells were harvested and cell lysis were prepared. The protein concentration of cell lysates were measured using Bio-Rad Protein Assay kit. The luciferase activity was measured using Promega's Luciferase Reporter Assay Systems according to the instructions of manufacturer.
  • the mechanism is interruption of the combination reaction between the IFN and the correspondent receptor, and inducement of the expression of 2′5′-A synthesizenzyme, protein kinase R in the target cell, therefore inhibiting expression of the viral protein.
  • IFN can induce expression of various anti-virus proteins to inhibit the reproduce of viral proteins, enhance the function of Natural Killer (NK) cell and other Immune regulative functions, and inhibit the invasion of viruses.
  • Acute Toxicity All mice are alive after the maximum dose (1000 times to human dose) subcutaneous injection, did not observe LD50.
  • Dosage and Administration Spray to both nasal cavity and throat, three times a day.
  • Adverse Reactions There was no report of adverse reactions from the rSIFN-co spray. It did not induce allergy. If the stimulation is occasional, adverse gastrointestinal reaction is small, and no other obvious adverse reaction was noted during treatment, it is safe to continue use. All reactions will resolve themselves. Warning: Patients allergic to IFN and productions of E. Coli . cannot use this product.
  • Precautions Before first use, spray twice to expel the air. If there is any cloudy precipitation material, if the product is expired, or there is material on the vial, do not use it.
  • Pediatric Use It is unclear.
  • Geriatric Use It is unclear.
  • CPE Cytopathic Effect
  • Assay of TCID 50 100 ⁇ l of Vero E 6 cells were plated in 96-well plates at 2 ⁇ 10 4 cells per well. After 24 hr incubation at 37° C., Vero E6 monolayer cells were treated with 9 levels of SARS-associated coronavirus dilution by 10-fold dilution, 4 wells per dilution. The cells were incubated at 37° C. and 5% CO 2 .
  • CPE cytopathic effect
  • CPE was examined daily by microscopy. CPE less than 25% was determined as +, 26-50% as ++, 51-75% as +++, 76-100% as ++++. CPE was recorded. Then TCID 50 was calculated by Reed-Muench method.
  • Vero E 6 cells were inoculated into 96-well plates at 2 ⁇ 10 4 cells (100 ul) per well. After 24-hr incubation at 37° C., cells grew up to monolayer. The medicine was diluted into 36, 18, 9, 4.5, 2.259 ⁇ g/ml (final concentration) and added into wells each for 4 wells. The normal cells as control group were set. CPE of medicine group was daily observed during 5-day period, and then the concentration of medicine exhibiting no toxicity was determined. CPE Assay of the Activity of the Medicine against SARS-Associated Coronavirus: 100 ⁇ l of Vero E 6 cells were plated in 96-well plates at 2 ⁇ 10 4 cells per well.
  • Toxicity of Virus TCID 50 of virus was 10 ⁇ 8 .
  • Cytotoxicity of Medicine the concentration of Recombinant compound interferon exhibiting no cytotoxicity was 18 ⁇ g/ml, the cells shape was similar with the control group, and no cytopathic effect was exhibited.
  • rSIFN-co New-type recombinant compound interferon
  • 618 ⁇ g/ml supplied by Huiyang Life Engineering Ltd., SiChuan province; Alfaron (recombinant interferon ⁇ -2b injection), supplied by Tianjin Hualida Biotechnology Co., Ltd. 30 ug/vial (300,0000 IU/vial), Lot Number:20030105.
  • Cells Vero E 6 , supplied by Molecular Biology Department of Institute of microbiology and epidemiology, Academy of Military Medical Science.
  • Virus SARS-associated coronavirus, BJ-01, supplied by Molecular Biology Department of Institute of microbiology and epidemiology, Academy of Military Medical Science.
  • TCID 50 was Measured with CPE Assay: Vero E 6 cells were inoculated in 96-well plates at 2 ⁇ 10 4 cells (100 ⁇ l) per well. After a 24-hr incubation at 37° C., Vero E6 monolayers were treated with 9 levels of SARS-associated coronavirus dilution by 10 times decreasing, each dilution per 4 wells. The cells were incubated at 37° C. and 5% carbon dioxide. CPE was examined daily by phase-contrast microscopy. CPE less than 25% was determined as +, 26-50% as ++, 51-75% as +++, 76-100% as ++++. CPE was recorded. Then TCID 50 was calculated by Reed-Muench method.
  • TC 50 of IFNs were Measured by MTT Assay: Vero E 6 cells were inoculated in 96-well plates at 2 ⁇ 10 4 cells per well (100 ⁇ l). After 24-hr incubation at 37° C., the supernatant liquid was removed when cells grew up to monolayer, then Vero E 6 was treated with different concentration of IFNs, each dilution per 4 wells. Normal group was set. After 5-day observation, the cells were mixed with MTT for 4 hours. After that, remove the liquid, and then thereafter DMSO were added into cells for 0.5 hour. The OD 570 nm was measured by microplate reader. Finally, TC 50 was calculated by Reed-Muench method.
  • the Activity of the INFs against SARS-Associated Coronavirus was Measured with MTT Assay: 100 ⁇ l of Vero E 6 cells were inoculated in 96-well plates at 2 ⁇ 10 4 cells per well. After 24-hr incubation 37° C., cells became monolayer. The medicine dilution at the concentration of exhibiting no cytotoxicity was 5 times decreasing and there were 5 levels of dilution. Then each dilution was added to 4 wells, 100 ul per well. After 24-hour incubation at 37° C. and 5% CO 2 , IFN solution was removed, then different concentrations of virus dilution (10000, 1000, 100 TCID 50 ) were added into dishes, 4 wells per dilution.
  • the cells were divided into the normal group, the medicine control group, and the different dilution of virus control group (10000, 1000, 100 TCID 50 ).
  • the cells were incubated at 37° C. and 5% CO 2 for 48-72 hr, until cytopathic effect was exhibited in the virus control group, CPE was recorded (CPE less than 25% was determined as +, 26-50% as ++, 51-75% as +++, 76-100% as ++++, normal cell as ⁇ ).
  • the growth ability of cells was measured with MTT assay, and then the antivirus effect of the INFs was evaluated. The experiment was repeated 3 times.
  • IC 50 of the medicine was calculated by Reed-Muench method.
  • TCID 50 of Virus TCID 50 of virus was 10 ⁇ 7 .
  • TC 50 of IFNs The concentration of new-type recombinant compound interferon (rSIFN-co) exhibiting no cytotoxicity was 100 ⁇ g/ml, and that of recombinant IFN ⁇ -2b was 12.5 ⁇ g/ml, the cells shape was identical with the normal group at that concentration.
  • TC50 of new-type recombinant compound interferon (rSIFN-co) was 139.18 ⁇ g/ml, that of recombinant IFN ⁇ -2b was 17.18 ⁇ g/ml.
  • IC 50 of new-type recombinant compound interferon on SARS-associated coronavirus at the concentration of 10000, 1000, and 100 was 0.92, 0.18, and 0.10 ⁇ g/ml in three experiments, TI of that was 151.28, 773.22, and 1391.80 respectively.
  • IC 50 of IFN ⁇ -2b was 4.75, 1.16, and 0.28 ⁇ g/ml, TI (treatment index) of that was 3.62, 14.78, 61.36 respectively.
  • the side effects includes: nausea, muscle soreness, loss of appetite, hair loss, hypoleucocytosis (hypoleukmia; hypoleukocytosis; hypoleukia), and decrease in blood platelet, etc.
  • Sample patients are divided into two groups. 10 patients in Group A were injected with 9 ⁇ g rSIFN-co. 11 patients in Group B were injected with 9 ⁇ g INFERGEN®. Both groups were monitored for 48 hours after injections. First monitoring was recorded 1 hour after injection. After that, records were taken every 2 hours.
  • Table 11.1 is the comparison of side effects between patients being injected with 9 ⁇ g of rSIFN-co and 9 ⁇ g of INFERGEN®.
  • Ebola virus is a notoriously deadly virus that causes fearsome symptoms, the most prominent being high fever and massive internal bleeding. Ebola virus kills as many as 90% of the people it infects. It is one of the viruses capable of causing hemorrhagic (bloody) fever. There is no specific treatment for the disease. Currently, patients receive supportive therapy. This consists of balancing the patient's fluids and electrolytes, maintaining their oxygen level and blood pressure, and treating them for any complicating infections. Death can occur within 10 days of the onset of symptoms.

Abstract

This invention provides a method for treating upper respiratory tract infection in a subject, comprising administering to the subject an effective amount of a recombinant interferon, wherein the recombinant interferon has the amino acid sequence of SEQ ID NO:1 and is encoded by the nucleotide sequence SEQ ID NO:2. The upper respiratory tract infection can be induced by various viruses such as rhinovirus, coronavirus, respiratory syncytial virus, influenza, parainfluenza, adenoviruses, coxsackie virus, corona virus and enterovirus.

Description

  • The application is a continuation application of U.S. Ser. No. 12/246,153, filed Oct. 6, 2008 which is a continuation application of U.S. Ser. No. 11/077,813, filed Mar. 10, 2005, which is a continuation-in-part of U.S. Ser. No. 10/927,975, filed Aug. 26, 2004, which is a continuation-in-part of U.S. Ser. No. 10/650,365, filed Aug. 28, 2003, which is a continuation-in-part of Int'l App'l No. PCT/CN02/00128, filed Feb. 28, 2002, which claims priority of Chinese App'l No. 01104367.9, filed Feb. 28, 2001. The contents of the above applications are hereby incorporated in their entireties by reference into this application.
  • Throughout this application, various publications are referenced. Disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this invention pertains.
  • FIELD OF THE INVENTION
  • This invention is related to a field of bioengineering. Specifically this invention relates to a recombinant super-compound interferon (rSIFN-co) or its equivalent with changed spatial configuration, high efficacy and low side effects. Therefore, high dose of rSIFN-co may be used. This invention also relates to a process to produce said super-compound interferon (rSIFN-co) or a pharmaceutical composition comprising said super-compound interferon (rSIFN-co) or its equivalent, and uses of said interferon or composition for anti-viral and anti-tumor therapy.
  • BACKGROUND OF THE INVENTION
  • IFN-con is a new interferon molecule constructed with the most popular conservative amino acid found in natural human IFN-α subtypes using genetic engineering methods. U.S. Pat. Nos. 4,695,623 and 4,897,471 have described it. IFN-con had been proven to have broad-spectrum IFN activity and virus- and tumor-inhibition and natural killer cell activity. U.S. Pat. No. 5,372,808 by Amgen, Inc. addresses treatment INFERGEN® (interferon alfacon-1). Chinese Patent No. 97193506.8 by Amgen, Inc. addresses re-treatment of INFERGEN® (interferon alfacon-1) on hepatitis C. Chinese Patent No. 98114663.5 by Shenzhen Jiusheng Bio-engineering Ltd. addresses recombinant human consensus interferon-α treatment for hepatitis B and hepatitis C.
  • The United States Food and Drug Administration (FDA) authorized Amgen to produce INFERGEN® (interferon alfacon-1) with E. Coli. for clinical hepatitis C treatment at the end of 1997.
  • Hepatitis B patients can be identified when detecting HBsAg and the HBeAg. IFN-α is commonly used in clinics to treat hepatitis B. IFN-α binds superficial cell membrane receptors, thus inhibiting DNA and RNA (ribonucleic acid) duplication and inducing some enzymes to prevent duplication of the virus in hepatitis-infected cells. All IFNs can inhibit DNA duplication of viruses, but they cannot inhibit the e and s antigen expression.
  • An outbreak of atypical pneumonia, referred to as severe acute respiratory syndrome (SARS) and first identified in Guangdong Province, China, has spread to several countries. Similar cases were detected in patients in Hong Kong, Vietnam, and Canada from February and March 2003. The World Health Organization (WHO) issued a global alert for the illness. In mid-March 2003, SARS was documented in health care workers and household members who had cared for patients with severe respiratory illness in the Far East. Many of these cases could be traced through multiple chains of transmission to one health care worker from Guangdong Province who visited Hong Kong, where he was hospitalized with pneumonia and died. By late April 2003, thousands of SARS cases and hundreds of SARS-related deaths from over 25 countries around the world were reported to WHO. Most of these cases occurred through exposure to SARS patients in household or health care settings. This invention provides a method to prevent and/or treat SARS.
  • This disclosure describes recombinant super-compound interferon (rSIFN-co), method to produce the same and uses thereof. Particularly, the super-compound interferon disclosed herein is capable of inhibiting, preventing and/or treating the hepatitis viruses, SARS virus, or virus-induced upper respiratory diseases, the Influenza virus, for example Avian Influenza virus and Ebola virus.
  • In addition, rSIFN-co is effective in preventing and/or treating viral diseases and tumors with less side effects as compared to other available interferons.
  • SUMMARY OF THE INVENTION
  • This invention provides a recombinant super-compound interferon (rSIFN-co) and its equivalent with changed spatial configuration, high efficacy and low side effects. Therefore, high dose of rSIFN-co may be used.
  • This invention also provides artificial gene encoding for the super-compound interferon or its equivalent.
  • This invention provides a vector comprising the gene which codes for the super-compound interferon or its equivalent.
  • This invention provides an expression system comprising the vector comprising the gene which codes for the super-compound interferon or its equivalent. This invention also provides a host cell comprising the vector comprising the gene which codes for the recombinant super-compound interferon (rSIFN-co) or its equivalent. Said host cell may be eukaryotic or prokaryotic, such as E. Coli.
  • This invention provides a method for producing a recombinant super-compound interferon (rSIFN-co) with changed spatial configuration and enhanced antiviral activity comprising steps of:
    • (a) Introducing nucleic acid molecule which codes for said interferon with preferred codons for expression to an appropriate host; and
    • (b) Placing the introduced host in conditions allowing expression of said interferon.
  • This invention provides the method for producing recombinant super-compound interferon (rSIFN-co), further comprising recovery of the expressed interferon.
  • This invention provides a method for inhibiting, preventing or treating viral diseases, or for inhibiting or treating tumors in a subject comprising administering to the subject an effective amount of the super-compound interferon or its equivalent.
  • This invention provides the above-described method wherein super-compound interferon is administered orally, via vein injection, muscle injection, peritoneal injection, subcutaneous injection, nasal or mucosal administration, or by inhalation via a respirator.
  • This invention provides the method to prevent or treat viral diseases wherein the viral diseases is hepatitis A, hepatitis B, hepatitis C, other types of hepatitis, infections of viruses caused by Epstein-Barr virus, Human Immunodeficiency Virus (HIV), Ebola virus, Severe Acute Respiratory Syndrome Virus (SARS), Influenza virus, Cytomegalovirus, herpes simplex viruses, or other types of herpes viruses, papovaviruses, poxviruses, picornaviruses, adenoviruses, rhinoviruses, human T-cell leukemia viruses I, or human T-cell leukemia viruses II, or human T-cell leukemia virus III.
  • This invention provides the method to prevent or treat viral diseases wherein the viral diseases are Human Immunodeficiency Virus (HIV) and Ebola virus.
  • This invention provides a method for anti-hepatitis activities. It can inhibit HBV-DNA replication, HBsAg and HBeAg production.
  • This invention provides a method to prevent or treat upper respiratory infection diseases.
  • This invention provides a method to prevent or treat tumors or cancers wherein the tumor is skin cancer, basal cell carcinoma and malignant melanoma, renal cell carcinoma, liver cancer, thyroid cancer, rhinopharyngeal cancer, solid carcinoma, prostate cancer, stomach/abdominal cancer, esophageal cancer, rectal cancer, pancreatic cancer, breast cancer, ovarian cancer, and superficial bladder cancer, hemangioma, epidermoid carcinoma, cervical cancer, non-small-cell lung cancer, small-cell lung cancer, glioma, leucocythemia, acute leucocythemia and chronic leucocythemia, chronica myelocytic leukemia, hairy cell leukemia, lymphadenoma, multiple myeloma, polycythemia vera, or Kaposi's sarcoma.
  • This invention provides a method for preventing or treating virus-induced diseases in a subject comprising administering to the subject an effective amount of recombinant super-compound interferon or a functional equivalent thereof.
  • The super-compound interferon (rSIFN-co) may be administered orally, via vein injection, muscle injection, peritoneal injection, subcutaneous injection, nasal or mucosal administration, or by inhalation via a respirator.
  • This invention provides a method for inhibiting the causative agent of virus-induced diseases, comprising contacting the causative agent with an effective amount of super-compound interferon or its equivalent.
  • This invention also provides a method for inhibiting virus-induced diseases, comprising contacting an effective amount of the super-compound interferon with said virus or cells. This contact could be direct or indirect.
  • This invention provides a composition comprising an effective amount of the super-compound interferon capable of inhibiting, preventing or treating virus-induced diseases, and a suitable carrier.
  • This invention provides a pharmaceutical composition comprising an effective amount of the recombinant super-compound interferon capable of inhibiting, preventing or treating virus-induced diseases in a subject, and a pharmaceutically acceptable carrier.
  • This invention provides a method for preventing or treating tumors in a subject comprising administering to the subject an effective amount of recombinant super-compound interferon or a functional equivalent thereof.
  • This invention provides a method for inhibiting tumors, comprising contacting the causative agent with an effective amount of super-compound interferon or its equivalent.
  • This invention also provides a method for inhibiting tumors, comprising contacting an effective amount of the super-compound interferon with said virus or cells. This contact could be direct or indirect.
  • This invention provides a composition comprising an effective amount of the super-compound interferon capable of inhibiting, preventing or treating tumors, and a suitable carrier.
  • This invention provides a pharmaceutical composition comprising an effective amount of the recombinant super-compound interferon capable of inhibiting, preventing or treating tumors in a subject, and a pharmaceutically acceptable carrier.
  • DETAILED DESCRIPTION OF THE FIGURES
  • FIG. 1. rSIFN-co cDNA sequence (SEQ ID NO: 2 and 3) designed according to E. Coli. codon usage and deduced rSIFN-co amino acid sequence (SEQ ID NO: 1)
  • FIGS. 2A-B. Sequence of another super-compound interferon (FIG. 2A—SEQ ID NO: 4-6; FIG. 2B—SEQ ID NO: 7-9)
  • FIG. 3. Diagram of pLac T7 cloning vector plasmid
  • FIG. 4. Diagram of pHY-4 expression vector plasmid
  • FIG. 5. Construction process of expression plasmid pHY-5
  • FIG. 6-A. Circular Dichroism spectrum of INFERGEN®
  • (Tested by Analysis and Measurement Center of Sichuan University)
  • Spectrum range: 250 nm-190 nm
  • Sensitivity: 2 m°/cm
  • Light path: 0.20 cm
  • Equipment: Circular Dichroism J-500C
  • Samples: contains 30 μg/ml IFN-cont, 5.9 mg/ml of NaCl and 3.8 mg/ml of Na2PO4, pH7.0.
  • INFERGEN® (interferon alfacon-1), made by Amgen Inc., also known as consensus interferon, is marketed for the treatment of adults with chronic hepatitis C virus (HCV) infections. It is currently the only FDA-approved, bio-optimized interferon developed through rational drug design and the only interferon with data on the label specifically for non-responding or refractory patients. InterMune's sales force re-launched INFERGEN® in January 2002 with an active campaign to educate U.S. hepatologists about the safe and appropriate use of INFERGEN®, which represents new hope for the more than 50 percent of HCV patients who fail other currently available therapies.
  • FIG. 6-B. Circular Dichroism spectrum of INFERGEN® From Reference [Journal of Interferon and Cytokine Research. 16:489-499(1996)]
  • Circular dichroism spectra of concensus interferon subforms. Concensus interferon was fractionated using an anion exchange column. Samples were dialyzed into 10 mM sodium phosphate, pH 7.4. Measurements were made on Jasco J-170 spectopolarimeter, in a cell thermostat at 15° C. (——), acylated form; (--) cis terminal form; ( . . . ), met terminal form. A. Far UV Spectrum. B. Near UV Spectrum.
  • FIG. 6-C. Circular Dichroism spectrum of rSIFN-co
  • Spectrum range: 320 nm-250 nm
  • Sensitivity: 2 m°/cm
  • Light path: 2 cm
  • Equipment: Circular Dichroism J-500C
  • Samples: contains 0.5 mg/ml rSIFN-co, 5.9 mg/ml of NaCl and 3.8 mg/ml of Na2PO4, pH7.0.
  • FIG. 6-D. Circular Dichroism spectrum of rSIFN-co
  • Spectrum range: 250 nm-190 nm
  • Sensitivity: 2 m°/cm
  • Light path: 0.20 cm
  • Equipment: Circular Dichroism J-500C
  • Samples: contains 30 μg/ml rSIFN-co, 5.9 mg/ml of NaCl and 3.8 mg/ml of Na2PO4, pH7.0.
  • Clearly, as evidenced by the above spectra, the secondary or even tertiary structure of rSIFN-co is different from INFERGEN®.
  • FIG. 7. rSIFN-co Crystal I
  • FIG. 8. rSIFN-co Crystal II
  • FIG. 9. The X-ray Diffraction of rSIFN-co Crystal
  • FIG. 10. Comparison of Inhibition Effects of Different Interferons on HBV Gene Expression
  • FIG. 11A-C. Recombinant Super-Compound Interferon Spray
  • Height: 90 mm
  • Width: 25 mm (bottom), 6 mm (top)
  • Weight: 9 g
  • Volume delivery: 0.1 ml
  • FIG. 11D. Recombinant Super-Compound Interferon Spray
  • When using the spray for the first time, take off the cap and discharge in the air several times until some liquid squirts out. Do not need to test spray for subsequent uses. To use, follow the illustrations shown in the figure, i.e.: (1) Pre-spray and (2) Press down on the nozzle to release the medication.
  • FIG. 12. Comparison of Anti-SARS Activity of Interferons:
  • Left top panel is negative control i.e. no virus added. Right top panel is positive control i.e. virus is added, but no rSIFN-co added. Left bottom panel is rSIFN-co with SARS Virus. Right bottom panel is αIFN.
  • FIGS. 13A-1. Curves of Changes of Body Temperature in Group A (5 patients)
  • This figure is the record of body temperature changes of 5 patients in Group A.
  • FIGS. 13A-2. Curves of Changes of Body Temperature in Group A (5 patients)
  • This figure is the record of body temperature changes of the other 5 patients in Group A.
  • FIGS. 13B-1. Curves of Changes of Body Temperature in Group B (5 patients)
  • This figure is the record of body temperature changes of 5 patients in Group B.
  • FIGS. 13B-2. Curves of Changes of Body Temperature in Group B (6 patients)
  • This figure is the record of body temperature changes of the other 6 patients in Group B.
  • FIG. 14. Graph of Inhibition of Wild-Type HIV by rSIFN-co using EXCEL and Luciferase as Y axis and concentration of rSIFN-co as X axis. A clear inverse dose-dependent response has been shown.
  • FIG. 15. Graph of Inhibition of Drug Resistant HIV by rSIFN-co using EXCEL and Luciferase as Y axis and concentration of rSIFN-co as X axis. A clear inverse dose-dependent response has been shown.
  • FIG. 16. rSIFN-co Inhibition of Influenza Virus: on the left, the control well is shown with Influenza virus added and without interferon, the cells had obvious CPE, such as rounding of cells, cell necroses, decrease in reflective light and sloughing off.
  • On the right, the experimental wells is shown containing Influenza virus and rSIFN-co at concentration 10 nanogram per milliliter (ng/ml) had morphology comparable to normal cells.
  • FIGS. 17A-H. Clinical Report of a patient with mammary and ovarian cancers. These figures show an obvious anti-cancer effect of rSIFN-co on this patient.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Recombinant Super-Compound Interferon (rSIFN-co)
  • This invention provides a recombinant super-compound interferon (rSIFN-co) or an equivalent thereof with changed spatial configuration. This invention reveals that proteins with the same primary sequence might have different biological activities. As illustrated in this application, proteins with identical amino acid sequences may have different activities. The efficacy of these proteins may sometimes be improved and, sometimes, proteins with changed spatial configuration would reveal new function.
  • As defined herein, equivalents are molecules which are similar in function to the compound interferon. An equivalent could be a deletion, substitution, or replacement mutant of the original sequence. Alternatively, it is also the intention of this invention to cover mimics of the recombinant super-compound interferon (rSIFN-co). Mimics could be a peptide, polypeptide or a small chemical entity.
  • The recombinant super-compound interferon (rSIFN-co) described herein includes but is not limited to interferon α, β, γ or ω. In an embodiment, it is IFN-1α, IFN-2β or other mutants.
  • In another embodiment, the recombinant super-compound interferon (rSIFN-co) disclosed has higher efficacy than α, β, γ, ω or a combination thereof and as compared to the interferons disclosed in U.S. Pat. Nos. 4,695,623 and 4,897,471. This recombinant super-compound interferon (rSIFN-co) is believed to have unique secondary or tertiary structure, wherein the 3-dimensional change is the result of changes in its production process. (See e.g. FIG. 6.)
  • The recombinant super-compound interferon (rSIFN-co) described herein has spatial structure change(s) resulting from the changes of its production process.
  • Lower Side Effects
  • The recombinant super-compound interferon (rSIFN-co) possesses lower side effects when compared with other interferons. These lower side effects allow for higher dosages to be used on patients in need of interferon treatments. These lower side effects open the possibility of using rSIFN-co for prevention and/or treatment of other diseases. Accordingly, this invention provides the recombinant super-compound interferon (rSIFN-co) with less side effects when administered to a subject.
  • This invention provides recombinant super-compound interferon (rSIFN-co) with less side effects as compared to all currently available interferons.
  • This invention further provides a method for treating or preventing viral diseases or tumors in a subject comprising administering to the subject an effective amount of the rSIFN-co with less side effects as compared to all currently available interferons. Therefore, high dose of rSIFN-co may be used. In an embodiment, the effective amount of recombinant super-compound interferon is in nanogram level.
  • Process to Produce rSIFN-co
  • Artificial Gene
  • This invention also provides artificial gene encoding for the super-compound interferon or its equivalent. It is within the ordinary skill to design an artificial gene. Many methods for generating nucleotide sequence and other molecular biology techniques have been described previously. See for example, Joseph Sambrook and David W. Russell, Molecular Cloning: A laboratory Manual, December 2000, published by Cold Spring Harbor Laboratory Press.
  • The recombinant super-compound interferon (rSIFN-co) may also be produced with its gene as artificially synthesized cDNA with adjustment of its sequence from the wild-type according to codon preference of E. Coli. Extensive discussion of said codon usage (preference) may be found in U.S. Pat. No. 4,695,623. See e.g. column 6, line 41-column 7, line 35.
  • Vector
  • This invention provides a vector comprising the gene which codes for the super-compound interferon or its equivalent.
  • This invention provides an expression system comprising the vector comprising the gene which codes for the super-compound interferon or its equivalent. The cells include, but are not limited to, prokaryotic or eukaryotic cells.
  • This invention also provides a host cell comprising the vector comprising the gene which codes for the recombinant super-compound interferon (rSIFN-co) or its equivalent.
  • This invention provides a method for producing a recombinant super-compound interferon (rSIFN-co) with changed spatial configuration and enhanced antiviral activity comprising steps of:
    • (a) Introducing nucleic acid molecule which codes for said interferon with preferred codons for expression to an appropriate host; and
    • (c) Placing the introduced host in conditions allowing expression of said interferon.
  • This invention provides the method for producing recombinant super-compound interferon (rSIFN-co), further comprising recovery of the expressed interferon.
  • Expression System
  • The above-described recombinant super-compound interferon (rSIFN-co) may be produced by a high-efficiency expression system which uses a special promoter, enhancer or other regulatory element. In an embodiment the promoter is inducible. Said inducible promoter includes but is not limited to PBAD, heat shock promoters or heavy metal inducible promoters. Heat shock promoters are activated by physical means, while other promoters are activated by chemical means, for example IPTG or Tetracyclin. IPTG is added to the cells to activate the downstream gene or removed to inactivate the gene. Tetracyclin is used to induce promoters or to regulate the strength of promoters.
  • In an embodiment the promoter is PBAD. Since early nineties, the properties of the mechanism of expression and repression of PBAD by AraC have been studied extensively, and their interactions have been dissected at the molecular level. See Schleif, R. S. 1992 DNA looping. Annu. Rev. Biochem. 61:199-223. The AraC protein is both a positive and negative regulator, when present, it turns on the transcription from the PBAD promoter, when absent, the transcription occurs at a very low rate. See Guzman, L. M. et al. (1995) J. Bact. 177: 4121-4130. The efficacy and mechanism of PBAD promoter is well known by other ordinary skilled artisans and is commercially-available.
  • The commercially-available Invitrogen expression kit includes pBAD vectors' designed to provide precise control of expression levels. The araBAD promoter initiates gene expression. It's both positively and negatively regulated by the product of the araC gene, a transcriptional regulator that forms a complex with L-arabinose. In the absence of arabinose, the AraC dimer contacts the O2 and I1 half sites of the araBAD operon, forming a 210 bp DNA loop. For maximum transcriptional activation, two events are required: first, Arabinose binds to AraC. The protein releases the O2 site and binds the I2 site, which is adjacent to the I1 site. This releases the DNA loop and allows transcription to begin. Second, the cAMP activator protein (CAP)-cAMP complex binds to the DNA and stimulates binding of AraC to I1 and I2. Basal expression levels can be repressed by introducing glucose to the growth medium. Glucose acts by lowering cAMP levels, which in turn decreases the binding of CAP. As cAMP levels are lowered, transcriptional activation is decreased. Invitrogen's pBAD vectors are specifically designed for maximum expression and ease of use.
  • Nine pBAD vectors are currently available: pBAD102/D-TOPO®, pBAD202/D-TOPO®, pBAD-TOPO®, pBAD/Thio-TOPO®, pBAD/His, pBAD/Myc-His, pBAD-DEST49, pBAD/gIII and pBAD/Thio-E. with the following features in all pBAD vectors:
      • 1. araBAD promoter for dose-dependent regulation
      • 2. araC gene for tight control of the araBAD promoter
      • 3. Optimized ribosome binding site for increased translation efficiency
      • 4. rrnB transcription termination region for efficient transcript
  • The inducible promoters include but are not limited to heat shock promoters or heavy metal inducible promoters.
  • This invention provides a process for production of recombinant super-compound interferon (rSIFN-co) comprising introducing an artificial gene with selected codon preference into an appropriate host, culturing said introduced host in an appropriate condition for the expression of said compound interferon and harvesting the expressed compound interferon.
  • The process may comprise extraction of super recombinant super-compound interferon (rSIFN-co) from fermentation broth, collection of inclusion bodies, denaturation and renaturation of the harvested protein.
  • The process may maintain the high efficacy even when the recombinant super-compound interferon (rSIFN-co) is used with an agent and in a particular concentration. The process also comprises separation and purification of the recombinant super-compound interferon (rSIFN-co). The process further comprises lyophilization of the purified recombinant super-compound interferon (rSIFN-co). The process comprises production of liquid injection of recombinant super-compound interferon (rSIFN-co).
  • In one embodiment, recombinant super-compound interferon (rSIFN-co) was produced with recombinant techniques. On the condition of fixed amino acid sequence, the IFN DNA was redesigned according to the E. Coli. codon usage and then the rSIFN-co gene was artificially synthesized. rSIFN-co cDNA was cloned into the high-expression vector of E. Coli. by DNA recombinant techniques, and a high expression of rSIFN-co was gained by using of induce/activate-mechanism of L-arabinose to activate the transcription of PBAD promoter.
  • Compared with usual thermo-induction, pH induction and IPTG induction systems of genetic engineering, arabinose induction/activation system has some advantages: (1) Common systems relieve promoter function by creating a “derepression” pattern. Promoters then induce downstream gene expression. Temperature and pH change and the addition of IPTG cannot activate promoters directly. In the system disclosed herein, L-arabinose not only deactivates and represses but also activates the transcription of PBAD promoter which induces a high expression of rSIFN-co. Therefore, the arabinose induction/activation system is a more effective expression system. (2) The relationship between Exogenous and L-arabinose dosage is linear. This means the concentration of arabinose can be changed to adjust the expression level of the exogenous gene. Therefore, it is easier to control the exogenous gene expression level in E. Coli. by arabinose than by changing temperature and pH value. This characteristic is significant for the formation of inclusion bodies. (3) L-arabinose is resourceful, cheap and safe, which, on the contrary, are the disadvantages of other inducers such as IPTG.
  • This embodiment creates an effective and resistant rSIFN-co-expressing E. Coli. engineering strain with an L-arabinose induction/activation system. The strain is cultivated and fermented under suitable conditions to harvest the bacterial bodies. Inclusion bodies are then purified after destroying bacteria and washing repeatedly. The end result, mass of high-purity, spatial-configuration-changed rSIFN-co protein for this invention and for clinical treatment, was gained from denaturation and renaturation of inclusion bodies and a series of purification steps. Said purification would not affect the biological activity of the purified protein.
  • The above-described recombinant super-compound interferon (rSIFN-co) possesses anti-viral or anti-tumor activity, and; therefore, is useful in inhibiting, preventing and treating viral diseases, inhibiting or treating tumors, or cancers.
  • Viral Diseases
  • This invention provides a method for treating or preventing viral diseases or tumors in a subject comprising administering to the subject an effective amount of the recombinant super-compound interferon (rSIFN-co) or its equivalent.
  • As used herein, viral diseases include, but are not limited to, hepatitis A, hepatitis B, hepatitis C, other types of hepatitis, infections caused by Epstein-Barr virus, Human Immunodeficiency Virus (HIV), Ebola virus, Severe Acute Respiratory Syndrome Virus (SARS), Influenza virus, Cytomegalovirus, herpes simplex viruses, other herpes viruses, papovaviruses, poxviruses, picornaviruses, adenoviruses, rhinoviruses, human T-cell leukemia virus I, human T-cell leukemia virus II, or human T-cell leukemia virus III.
  • In an embodiment, the effective amount is at nanogram level. In another embodiment, the virus is Human Immunodeficiency Virus and the effective amount is as low as 4 nanograms per milliliter. In another embodiment, the virus is Influenza and the effective amount is as low as 10 nanogram per milliliter.
  • Inhibition of DNA Replication and Secretion of of HBsAg and HBeAg of Hepatitis B Virus.
  • The recombinant super-compound interferon (rSIFN-co) inhibits the DNA duplication and secretion of HBsAg and HBeAg of Hepatitis B Virus.
  • Severe Acute Respiratory Syndrome Virus (SARS)
  • This invention provides a method for preventing or treating Severe Acute Respiratory Syndrome, or virus-induced upper respiratory diseases, of a subject comprising administering to the subject an effective amount of recombinant super-compound interferon (rSIFN-co) or a functional equivalent thereof. In an embodiment of the above method, the interferon is α, β, γ, ω or a combination thereof.
  • The recombinant super-compound interferon (rSIFN-co) may be administered orally, via vein injection, muscle injection, peritoneal injection, subcutaneous injection, nasal or mucosal administration, or by inhalation via a spray or a respirator. In an embodiment rSIFN-co is administered subcutaneously or intramuscularly at a dose of higher than or equal to 10 Million International Unit per square meter of surface area. In another embodiment rSIFN-co is administered subcutaneously or intramuscularly at a dose of higher than or equal to 20 Million International Unit per square meter of surface area. In an embodiment, the interferon is delivered by a spray device. In a specific embodiment, the device is described in FIG. 11. In one of the embodiments, the interferon is lyophilized.
  • This invention provides a method for inhibiting the causative agent of Severe Acute Respiratory Syndrome, or virus-induced upper respiratory diseases, comprising contacting the agent with an effective amount of recombinant super-compound interferon (rSIFN-co) or its equivalent.
  • It is determined that the causative agent of SARS is a virus. See eg. Rota et al (2003), Characterization of a Novel Coronavirus Associated with Severe Acute Respiratory Syndrome. Science 1085952 and Marra, et al. (2003), The Genome Sequence of the SARS-Associated Coronavirus. Science 1085853.
  • This invention also provides a method for inhibiting Severe Acute Respiratory Syndrome virus or Severe Acute Respiratory Syndrome virus-infected cells, or virus-induced upper respiratory diseases, or cells infected with viruses capable of inducing upper respiratory diseases, comprising contacting an effective amount of the recombinant super-compound interferon (rSIFN-co) with said virus or cell. This contact could be direct or indirect.
  • This invention provides a composition comprising an effective amount of the recombinant super-compound interferon (rSIFN-co) capable of inhibiting Severe Acute Respiratory Syndrome virus or Severe Acute Respiratory Syndrome virus-infected cells, or virus-induced upper respiratory diseases, or cells infected with viruses capable of inducing upper respiratory diseases, and a suitable carrier.
  • This invention provides a composition comprising an effective amount of the super-compound interferon capable of preventing or treating Severe Acute Respiratory Syndrome, or virus-induced upper respiratory diseases, of a subject and a suitable carrier.
  • This invention provides a pharmaceutical composition comprising an effective amount of the recombinant super-compound interferon (rSIFN-co) capable of inhibiting Severe Acute Respiratory Syndrome virus or Severe Acute Respiratory Syndrome virus-infected cells, or virus-induced upper respiratory diseases, and a pharmaceutically acceptable carrier.
  • This invention provides a pharmaceutical composition comprising an effective amount of the recombinant super-compound interferon (rSIFN-co) capable of preventing or treating Severe Acute Respiratory Syndrome, or virus-induced upper respiratory diseases, in a subject and a pharmaceutically acceptable carrier.
  • This invention provides a device to deliver the above-described pharmaceutical composition.
  • In a preferred embodiment, the subject is a human. As it can easily be appreciated, the super-compound interferon can be used in other animals or mammals.
  • This invention provides a method for preventing Severe Acute Respiratory Syndrome or virus-induced upper respiratory diseases, in humans comprising application of the super-compound interferon three times a day via a spray which contains twenty micrograms of interferon, equal to ten million units of activity in three milliliter.
  • Viral Upper Respiratory Infection (VURI)
  • Viral upper respiratory infection, alternative names common cold, colds. This is a contagious viral infection of the upper respiratory tract characterized by inflammation of the mucous membranes, sneezing, and a sore throat. It is usually caused by over 200 different viruses, known as rhinoviruses. Colds are not caused by the same viruses responsible for Influenza. Colds are spread through droplets from the coughing or sneezing of others with a cold or by hand contact with objects contaminated by someone with a cold. The incidence of colds is highest among children, and the incidence decreases with age because immunity to the virus causing the cold occurs after the illness. Gradually, immunity to a wide variety of viruses that cause colds is developed in adults. Children may have 10 colds a year, and adults may have 3 colds a year.
  • The U.S. Centers for Disease Control and Prevention have estimated that the average annual incidence of upper respiratory tract infections (URIs) in the United States is 429 million episodes, resulting in more than $2.5 billion in direct and indirect healthcare costs. The common cold is most often caused by one of several hundred rhinoviruses (52%), but coronaviruses (8%) or the respiratory syncytial virus (7%) may also lead to infection. Other viruses, such as influenza (6%), parainfluenza, and adenoviruses, may produce respiratory symptoms, but these are often associated with pneumonia, fever, or chills.
  • Colds occur in a seasonal pattern that usually begins in mid-September and concludes in late April to early May. The common cold is quite contagious and can be transmitted by either person-to-person contact or airborne droplets. Upper respiratory symptoms usually begin 1 to 2 days after exposure and generally last 1 to 2 weeks, even though viral shedding and contagion can continue for 2 to 3 more weeks. Symptoms may persist with the occurrence of complications such as sinusitis or lower respiratory involvement such as bronchitis or pneumonia.
  • The common cold has a variety of overt symptoms, including malaise, nasal stuffiness, rhinorrhea, nonproductive cough, mild sore throat, and, in some cases, a low-grade fever. Because of the similarity of symptoms, a cold may be mistaken for perennial allergic rhinitis, but allergies can usually be ruled out because of the differences in chronicity.
  • If a patient presents with a viral URI, the spectrum of remedies is extensive. Since most of these infections are self-limiting, clinicians usually recommend rest and fluids, but other treatments include environmental and nutritional therapies, over-the-counter and prescription decongestant and antihistamine products, new antihistamine and anticholinergic nasal formulations, and antibiotics. Table 1 lists commonly used cough and cold medications and their side effects.
  • TABLE 1
    A Profile of Common Cough and Cold
    Medications and Their Side Effects
    Side Effects and
    Medication Purpose Special Considerations
    Aerosolized beta2 Reverse Raises heart rate and
    agonists (eg, postinflammatory may cause tremor
    albuterol) bronchospasm
    Alcohol-based liquid Treat multiple Potential drowsiness and
    combination products symptoms coordination problems
    Alphal agonists Decongestion May cause tachycardia,
    (oral) (eg, nervousness, transient
    pseudoephedrine, stimulation, dizziness,
    phenylpropanolamine) drowsiness, elevation of
    blood pressure
    Anticholinergic Drying May cause nasal dryness
    compounds: and occasional epistaxis
    Ipratropium
    bromide
    (topical)
    Other Drying May cause orthostasis,
    anticholinergics dysfunction of heat
    (eg, regulation, dry mouth,
    methscopolamine, constipation
    atropine,
    hyoscyamine)
    Antihistamines Drying Drowsiness, dry mouth,
    (oral) (eg, orthostatic hypertension
    chlorpheniramine,
    diphenhydramine)
    Benzonatate capsules Cough suppression, Chewing can numb the
    local anesthesia mouth; can cause
    sedation, dizziness
    Codeine, hydrocodone Cough suppression Drowsiness, constipation,
    nausea
    Dextromethorphan Cough suppression Drowsiness possible, but
    side effects uncommon
    Guaifenesin Promote No side effects; must be
    expectoration taken with lots of water
    (mucolysis) to improve efficacy
    Topical Decongestion Local burning; prolonged
    decongestants (eg, use may cause dependence
    oxymetazoline,
    phenylephrine)
    Zinc and vitamin C Possible reduction Possible taste disturbance,
    lozenges in symptom increase of oxalate stones
    severity and if susceptible
    duration
  • Prevention and Treatment of Upper Respiratory Tract Infections (URI)
  • Nearly 70˜80% URI are caused by viruses such as respiratory Syncytical virus, adenovirus, rhinovirous, cox-sackie virus, corona virus and its variant, influenza A virus and its variant, influenza B virus and its variant, parainfluenza virus and its variant, or enterovirus and its variant. A main cause of URI in adults is from rhinovirous. For children, respiratory syncytical virus and parainfluenza virus are two leading causes of URI.
  • Recombinant super-compound interferon (rSIFN-co) plays an important role in the fight against virus that causes URI. Super-compound interferon gains its anti-virus effects mainly via two mechanisms:
      • 1. Attach to surface of sensitive cells and induce them to produce anti-virus protein, then block the duplication and reproduction of viruses in vivo.
      • 2. recombinant super-compound interferon (rSIFN-co) can adjust immune response, including T-cell immune response, activity of NK cell, the phagocytosis function of monokaryon, and even formation of some antibodies in vivo.
  • In treatment for URI, recombinant super-compound interferon (rSIFN-co) can be directly applied to the affected area via a spray or a respiration. This method of treatment allows the interferon to reach the target cells first hand. Consequently, marketing the supply as a spray, rather than via oral or injection, would be safer and more effective for administrating the interferon.
  • Prevention and Treatment of SARS
  • With the consent of the Sichuan (a province in China) working group on SARS prevention and control, the distribution of recombinant super-compound interferon (rSIFN-co) began in May of 2003. Super-compound interferon spray was allocated to doctors and nurses in hospitals, populated areas with a high risk for SARS, and to the National research group on prevention and control of SARS. Among the 3,000 users as of Dec. 19, 2003, there were no reports of any side effects connected to the use of the spray. Furthermore, none of the doctors and nurses, the people of Sichuan Province, or other organizations that have used the Super-compound interferon spray has been infected by SARS.
  • Therefore, this invention provides a method for inhibiting, preventing or treating virus replication or virus-infected cells by contacting said virus or infected cells with an effective amount of the recombinant super-compound interferon (rSIFN-co) or its equivalent.
  • Prevention and Treatment of Tumors
  • This recombinant super-compound interferon (rSIFN-co) is useful in inhibiting, preventing or treating the following cancers or tumors:
  • Cancer Skin Cancer Basal Cell Carcinoma
    Malignant Melanoma
    Renal cell carcinoma
    Liver Cancer
    Thyroid Cancer
    Rhinopharyngeal Cancer
    Solid Carcinoma Prostate Cancer
    Stomach/Abdominal Cancer
    Esophageal Cancer
    Rectal Cancer
    Pancreatic Cancer
    Breast Cancer
    Ovarian Cancer &
    Superficial Bladder
    Cancer
    Hemangioma
    Epidermoid Carcinoma Cervical Cancer
    Non-small Cell Lung Cancer
    Small Cell Lung Cancer
    Glioma
    Malignant Leucocythemia Acute Leucocythemia
    Hemal Chronic Leucocythemia
    Disease Chronic Myelocytic
    Leukemia
    Hairy Cell Leukemia
    Lymphadenoma
    Multiple Myeloma
    Polycythemia Vera
    Others Kaposi's Sarcoma
  • Accordingly, this invention provides a method for inhibiting tumor or cancer cell growth by contacting the recombinant super-compound interferon (rSIFN-co) or its equivalent with said tumor or cancer cells.
  • Formulation and Route of Administration
  • This invention also provides the produced super-compound interferon by the above processes.
  • This invention provides a composition comprising recombinant super-compound interferon (rSIFN-co) or its equivalent and a suitable carrier.
  • This invention provides a pharmaceutical composition comprising the recombinant super-compound interferon (rSIFN-co) or its equivalent and a pharmaceutically acceptable carrier.
  • This invention provides the above-described method wherein recombinant super-compound interferon (rSIFN-co) is administered via orally via vein injection, muscle injection, peritoneal injection, subcutaneous injection, nasal or mucosal administration, or by inhalation via a spray or a respirator.
  • This invention provides the above-described method wherein recombinant super-compound interferon (rSIFN-co) is administered following the protocol of injections of 9 μg, 15 μg or 24 μg every two days, 3 times a week, for 24 weeks.
  • It was surprising to find that recombinant super-compound interferon (rSIFN-co), the spatial structure of which has been changed, is not only a preparation to inhibit the DNA duplication of hepatitis B, but to inhibit the secretion of HBsAg and HBeAg on 2.2.15 cells.
  • One objective of this invention is to offer a preparation of recombinant super-compound interferon (rSIFN-co) to directly inhibit the DNA duplication of hepatitis B viruses and the secretion of HBeAg and HBsAg of hepatitis B and decrease them to normal levels.
  • Formulation
  • The following are some rSIFN-co preparations: tablets, capsules, liquids for oral consumption, pastes, injections, sprays, suppositories, and solutions. Injections are recommended. It is common to subcutaneously inject or vein-inject the medicine. The medicine carrier could be any acceptable medicine carrier, including carbohydrates, cellulosum, adhesive, collapse, emollient, filling, add-dissolving agent, amortization, preservative, thickening agent, matching, etc.
  • This invention also provides a pharmaceutical composition comprising the above composition and a pharmaceutically acceptable carrier.
  • For the purposes of this invention, “pharmaceutically acceptable carriers” means any of the standard pharmaceutical carriers. Examples of suitable carriers are well known in the art and may include, but are not limited to, any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution and various wetting agents. Other carriers may include additives used in tablets, granules, capsules, etc. Typically such carriers contain excipients such as starch, milk, sugar, certain types of clay, gelatin, stearic acid or salts thereof, magnesium or calcium stearate, talc, vegetable fats or oils, gum, glycols or other known excipients. Such carriers may also include flavor and color additives or other ingredients. Compositions comprising such carriers are formulated by well-known conventional methods.
  • Increase of the Half-Life of rSIFN-co
  • Pegylation
  • Pegylation is the process by which polyethylene glycol chains are attached to protein and peptide drugs to increase pharmacokinetics by shielding these proteins and peptide drugs from proteolytic enzymes. See Harris and Chess, Effect of pegylation on pharmaceuticals. Nat Rev Drug Discov. 2003 March; 2(3):214-21.
  • Pegylations is a well-established method for increasing the circulating half-life of protein and liposomal pharmaceuticals based on large hydrodynamic volume of polyethylene glycols. These polyethylene glycols shield the proteins and peptide drugs from renal clearance, enzymatic degradation and immune system recognition, thus their half-life and making them more acceptable to patients. See Molineux, Pegylation: engineering improved pharmaceuticals for enhanced therapy. Cancer Treat Rev. 2002 April; 28 Suppl A: 13-6. The author concludes that pegylation has beneficial effect on the quality of life of cancer patients.
  • Pegylation of the interferon increases the amount of time the interferon remains in the body by increasing the size of the interferon molecule by decreasing the rate of absorption, prolonging the half-life and the rate of interferon clearance. Thus, the duration of biological activity is increased with pegylated interferon over nonpegylated interferon, thus providing an advantage over nonpegylated interferons with less frequent administration and comparable tolerability. The author states that monotherapy with pegylated interferon produces a better response in some patients than monotherapy with the nonpegylated formulation. See Baker, Pegylated Interferons. Rev Gastroenterol Disord. 2001; 1(2):87-99.
  • Sustained Release or Controlled Release
  • Sustained release delivery matrices and liposomes maybe used with rSIFN-co to create sustained release and controlled release formulation. See Robinson and Talmadge, Sustained Release of Growth Factors. In Vivo 2002 November-December; 16(6): 535-40. The authors state that both pegylation and sustained release delivery matrices and liposomes improve the pharmacokinetic and pharmacodynamic properties of recombinant molecules, and thus by improving clinical efficacy these approaches increase patient compliance.
  • This invention provides recombinant super-compound interferon (rSIFN-co) comprising an agent or encapsulated by an agent, capable of affecting the half-life or delivery of said interferon. In an embodiment this agent is polyethylene glycol (PEG).
  • This invention further provides a method for treating or preventing viral diseases or tumors in a subject comprising administering to the subject an effective amount of the recombinant super-compound interferon (rSIFN-co) or its equivalent comprising an agent or encapsulated by an agent, capable of affecting the half-life or delivery of said interferon. In an embodiment this agent is polyethylene glycol (PEG).
  • This invention will be better understood from the examples which follow. However, one skilled in the art will readily appreciate that the specific methods and results discussed are merely illustrative of the invention as described more fully in the claims which follow thereafter.
  • EXPERIMENTAL DETAILS
  • IFN-con is a new interferon molecule constructed according to conservative amino acids in human IFN-α subtype using genetic engineering methods. It has been proven that IFN-con has broad-spectrum IFN activity, such as high antivirus and tumor inhibition activity, especially for effectively treating hepatitis C.
  • E. Coli. codon was used to redesign rSIFN-co cDNA and then artificially synthesize cDNA of rSIFN-co from published INFERGEN® (interferon alfacon-1) DNA sequences and deduced amino acid sequences (FIG. 1).
  • In order to get pure rSIFN-co protein, rSIFN-co cDNA was cloned into E. Coli. high-expression vector, and L-arabinose, which can activate strong PBAD promoter in vectors, was used to induce high expression of rSIFN-co gene.
  • Example 1
  • Synthesis of E. Coli. cDNA Sequence
    Redesign of rSIFN-co cDNA Sequence
  • rSIFN-co cDNA was redesigned according to the codon usage of E. Coli. to achieve high expression in E. Coli. Deduced amino acid sequence (SEQ ID NO: 1) from the redesigned cDNA sequence (SEQ ID NO: 2 and 3) of rSIFN-co is completely coincidental with primitive amino acid sequence of published INFERGEN® (interferon alfacon-1) (FIG. 1).
  • rSIFN-co cDNA Sequence Synthesis
    rSIFN-co cDNA 5′-Terminus and 3′-Terminus Semi-Molecular Synthesis
  • Two semi-moleculars can be directly synthesized: rSIFN-co cDNA 5′-terminus 280 bp (fragment I) and 3′-terminus 268 bp (fragment II) by PCR. There are 41 bp overlapping among fragment II and fragment I.
  • (1) Chemical synthesis oligodeoxynucleotide fragment:
  • Oligomer A:
    (SEQ ID NO: 10)
    5′ATGTGCGACCTGCCGCAGACCCACTCCCTGGGTAACCGTCGTGCTCTG
    ATCCTGCTGGCTCAGATGCGTCGTATCTCCCCGTTCTCCTGCCTGAAAGA
    CCGTCACGAC3′
    Oligomer B:
    (SEQ ID NO: 11)
    5′CTGAAAGACCGTCACGACTTCGGTTTCCCGCAGGAAGAATTCGACGGT
    AACCAGTTCCAGAAAGCTCAGGCTATCTCCGTTCTGCACGAAATGATCCA
    GCAGACCTTC3′
    Oligomer C:
    (SEQ ID NO: 12)
    5′GCTGCTGGTACAGTTCGGTGTAGAATTTTTCCAGCAGGGATTCGTCCC
    AAGCAGCGGAGGAGTCTTTGGTGGAGAACAGGTTGAAGGTCTGCTGGATC
    ATTTC3′
    Oligomer D:
    (SEQ ID NO: 13)
    5′ATCCCTGCTGGAAAAATTCTACACCGAACTGTACCAGCAGCTGAACGA
    CCTGGAAGCTTGCGTTATCCAGGAAGTTGGTGTTGAAGAAACCCCGCTGA
    TGAAC3′
    Oligomer E:
    (SEQ ID NO: 14)
    5′GAAGAAACCCCGCTGATGAACGTTGACTCCATCCTGGCTGTTAAAAAA
    TACTTCCAGCGTATCACCCTGTACCTGACCGAAAAAAAATACTCCCCGTG
    CGCTTGGG3′
    Oligomer F:
    (SEQ ID NO: 15)
    5′TTATTCTTTACGACGCAGACGTTCCTGCAGGTTGGTGGACAGGGAGAA
    GGAACGCATGATTTCAGCACGAACAACTTCCCAAGCGCACGGGGAGTATT
    TTTTTTCGGTCAGG3′
  • PCR I for Fragment I: oligodeoxynucleotide B as template, oligodeoxynucleotide A and C as primers, synthesized 280 bp Fragment I.
  • PCR I mixture (units: μl)
    sterilized distilled water 39
    10 × Pfu buffer (Stratagen American Ltd.) 5
    dNTP mixture (dNTP concentration 2.5 mmol/L) 2
    Oligomer A primer (25 μmol/L) 1
    Oligomer C primer (25 μmol/L) 1
    Oligomer B template (1 μmol/L) 1
    Pfu DNA polymerase (Stratagen American Ltd.) (25 U/μl) 1
    Total volume 50 μl
      • PCR cycle:
      • 95° C.2 m→(95° C.45 s→65° C.1 m→72° C.1 m)×25 cycle→72° C.10 m→4° C.
      • PCR II for Fragment II: oligodeoxynucleotide E as template, oligodeoxynucleotide D and F as primers, synthesized 268 bp Fragment II.
  • PCR II mixture (units: μl)
    sterilized distilled water 39
    10 × Pfu buffer (Stratagen American Ltd.) 5
    dNTP mixture (dNTP concentration 2.5 mmol/L) 2
    Oligomer D primer (25 μmol/L) 1
    Oligomer F primer (25 μmol/L) 1
    Oligomer E template (1 μmol/L) 1
    Pfu DNA polymerase (Stratagen American Ltd.) (25 U/μl) 1
    Total volume 50 μl
      • PCR cycle: the same as PCR I
        Assembling of rSIFN-co cDNA
  • Fragment I and II were assembled together to get the complete cDNA molecular sequence of rSIFN-co using the overlapping and extending PCR method. Restriction enzyme Nde I and Pst I were introduced to clone rSIFN-co cDNA sequence into plasmid.
      • (1) Chemical synthesis primers
  • Oligomer G:
    (SEQ ID NO: 16)
    5′ATCGGCCATATGTGCGACCTGCCGCAGACCC3′
    Oligomer H:
    (SEQ ID NO: 17)
    5′ACTGCCAGGCTGCAGTTATTCTTTACGACGCAGACGTTCC3′
      • (2) Overlapping and extending PCR
  • PCR mixture (units: μl)
    sterilized distilled water 38
    10 × Pfu buffer (Stratagen American Ltd.) 5
    dNTP mixture (dNTP concentration 2.5 mmol/L) 2
    primer G (25 μmol/L) 1
    primer H (25 μmol/L) 1
    *fragment I preduction (1 μmol/L) 1
    *fragment II preduction (1 μmol/L) 1
    Pfu DNA polymerase (Stratagen American Ltd.) (2.5 U/μl) 1
    Total volume 50μ
      • PCR cycle: the same as PCR I
        rSIFN-co Gene Clone and Sequence Analysis
  • pLac T7 plasmid as cloning vector. pLac T7 plasmid is reconstructed with pBluescript II KS(+) plasmid produced by Stratagen (FIG. 3).
  • Purified PCR production of rSIFN-co cDNA with StrataPrep PCR purification kit. Digest cDNA and pLac T7 plasmid with NdeI and PstI. Run 1% agarose gel electrophoresis and separate these double-digested DNA fragments. Recover 507 bp long rSIFN-co DNA fragment and 2.9 kb plasmid DNA fragment. Ligate these fragments by T4 DNA ligase to form a recombinant plasmid. Transform DHcompetent cells (Gibco) with the recombinant plasmid, culture at 37° C. overnight. Identify the positive recombinant colony, named pHY-1.
  • Run DNA sequencing with SequiTherm™ Cycle Sequencing Kit produced by American Epicentre Technologies Ltd using L1-COR Model 4000L. Primers are T7 and T3 common sequence primer, the DNA sequencing result matches theoretic design.
  • Purify the rSIFN-co, sequence the N-terminus amino acids, the N-terminus amino acid sequence matches experimental design which is as follows: N-Cys-Asp-Leu-Pro-Gln-Thr-His-Ser-Leu-Gly-Asn-Arg-Arg-Ala-Leu- (SEQ ID NO: 18)
  • Construction, Transformation, Identification, and Hereditary Stability of Expression Vector Construction and Transformation of Expression Vector
  • Digested E. Coli. expression vector pHY-4 (see FIG. 3) with Nde I to linearize and subsequently digest with Xba I. Run 1% agarose gel electrophoresis, and purify the 4.8 kb pHY-4 Nde I-Xba I digest fragment with QIAEX II kit produced by QIAGEN Germany Ltd.
  • At the same time, the pHY-4 plasmid is double digested with Nde I-Xba I. Run 1% agarose gel electrophoresis and purify the 715 bp fragment. Ligate the rSIFN-co and pHY-4 fragments with T4 DNA ligase to construct the recombinant plasmid (See FIG. 4). Transform DHcompetent cells with the recombinant plasmid. Spread the transformed cells on LB plate with Amp, 37° C. culture overnight.
  • Positive Cloning Strain Screening
  • Randomly choose E. Coli. colonies from above LB-plate, screening the positive strains containing recombinant vector by endonuclease digesting and PCR analysis. Name one of the positive recombinant plasmid pHY-5, and name the strain containing pHY-5 plasmid PVIII. Amplify and store the positive strain with glycerol in −80° C.
  • High Expression of rSIFN-co Gene in E. Coli.
  • In pHY-5 plasmid, rSIFN-co gene is under the control of strong promoter PBAD. This promoter is positively and negatively regulated by the product of the gene araC. AraC is a transcriptional regulator that forms a complex with arabinose. In the absence of arabinose, the AraC dimer binds O2 and I1, forming a 210 bp loop. This conformation leads to a complete inhibition of transcription. In the presence of arabinose, the dimer is released from O2 and binds I1 and I2 leading to transcription. Arabinose binding deactivates, represses, and even activates the transcription of PBAD promoter, which stimulates PBAD, inducing high expression of rSIFN-co. rSIFN-co expression level in PVIII is more than 50% of the total E. Coli. protein.
  • SUMMARY
  • rSIFN-CO is a new interferon molecule artificially built according to the conservative amino acid of human a interferons. It has been proven as an effective anti-hepatitis drug. In order to get enough pure rSIFN-co protein, a stable recombinant E. Coli. strain which highly expresses rSIFN-co protein was constructed.
  • First, according to published INFERGEN® (interferon alfacon-1) amino acid sequence, E. Coli. codon was used to synthesize the whole cDNA of rSIFN-co. This DNA fragment was sequenced, proving that the 501 bp codon sequence and TAA termination codon sequence are valid and identical to theocratic design. Subsequent analysis revealed that the N-terminus amino acid sequence and amino acid composed of rSIFN-co produced by the recombinant strain were both identical to the prediction.
  • The rSIFN-co cDNA was cloned into E. Coli. high-expression vector pHY-4 plasmid to construct the recombinant plasmid pHY-5. E. Coli. LMG194 strain was further transformed with pHY-4 plasmid to get stable rSIFN-co high-expression transformant. This transformant was cultured for 30 generations. The heredity of pHY-5 recombinant plasmid in E. Coli. LMG194 was normal and stable, and the expression of rSIFN-co was high and steady.
  • E. Coli. LMG194, which contains recombinant pHY-5 plasmid, is actually an ideal high-expression engineering strain.
  • REFERENCES
    • 1. Blatt L M, Davis J M, Klein S B. et al. The biologic activity and molecular characterization of a novel synthetic interferon-alpha species, consensus interferon. Journal of Interferon and Cytokine Research, 1996; 16(7):489-499.
    • 2. Alton, K. et al: Production characterization and biological effects of recombinant DNA derived human IFN-α and IFN-γ analogs. In: De Maeger E, Schellekens H. eds. The Biology of Interferon System. 2nd ed. Amsterdam: Elsevier Science Publishers, 1983: 119-128
    • 3. Pfeffer L M. Biologic activity of natural and synthetic type 1 interferons. Seminars in Oncology, 1997; 24 (3 suppl 9):S9-63-59-69.
    • 4. Ozes O N, Reiter Z, Klein S, et al. A comparison of interferon-con1 with natural recombinant interferons-α antiviral, antiproliferative, and natural killer-inducing activities. J. Interferon Res., 1992; 12:55-59.
    • 5. Heathcote E J L, Keeffe E B, Lee S S, et al. Re-treatment of chronic hepatitis C with consensus interferon. Hepatology, 1998; 27(4):1136-1143.
    • 6. Klein M L, Bartley T D, Lai P H, et al. Structural characterization of recombinant consensus interferon-alpha. Journal of Chromatography, 1988; 454:205-215.
    • 7. The Wisconsin Package, by Genetics Computer Group, Inc. Copyright 1992, Medison, Wis., USA
    • 8. Nishimura, A et al: A rapid and highly efficient method for preparation of competent E. coli cells. Nuclei. Acids Res. 1990, 18:6169
    • 9. All molecular cloning techniques used are from: Sambrook, J., E. F. Fritsch and T. Maniatis. Molecular Cloning:A laboratory manual, 2nd ed. CSH Laboratory Press, Cold Spring Harbour, N.Y. 1989.
    • 10. Guzman, L. M et al: Tight regulation, modulation, and high-level express-ion by vectors containing the arabinose PBAD promoter. J. Bacteriol. 1995, 177: 4121˜4130.
  • rSIFN-co cDNA SEQUENCE DESIGNED ACCORDING TO E. COLI. CODON
    USAGE AND DEDUCED rSIFN-co AMINO ACID SEQUENCE
    5′   11     21    31    41  51
    +1 M C D L P Q T H S L G N R R A L I L L A
    1 ATGTGCGACC TGCCGCAGAC CCACTCCCTG GGTAACCGTC GTGCTCTGAT CCTGCTGGCT
    TACACGCTGG ACGGCGTCTG GGTGAGGGAC CCATTGGCAG CACGAGACTA GGACGACCGA
    5′   71    81    91   101   111
    +1 Q M R R I S P F S C L K D R H D F G F P
    61 CAGATGCGTC GTATCTCCCC GTTCTCCTGC CTGAAAGACC GTCACGACTT CGGTTTCCCG
    GTCTACGCAG CATAGAGGGG CAAGAGGACG GACTTTCTGG CAGTGCTGAA GCCAAAGGGC
    5′  131   141   151   161   171
    +1 Q E E F D G N Q F Q K A Q A I S V L H E
    121 CAGGAAGAAT TCGACGGTAA CCAGTTCCAG AAAGCTCAGG CTATCTCCGT TCTGCACGAA
    GTCCTTCTTA AGCTGCCATT GGTCAAGGTC TTTCGAGTCC GATAGAGGCA AGACGTGCTT
    5′  191   201    211   221   231
    +1 M I Q Q T F N L F S T K D S S A A W D E
    181 ATGATCCAGC AGACCTTCAA CCTGTTCTCC ACCAAAGACT CCTCCGCTGC TTGGGACGAA
    TACTAGGTCG TCTGGAAGTT GGACAAGAGG TGGTTTCTGA GGAGGCGACG AACCCTGCTT
    5′   251   261    271  281   291
    +1 S L L E K F Y T E L Y Q Q L N D L E A C
    241 TCCCTGCTGG AAAAATTCTA CACCGAACTG TACCAGCAGC TGAACGACCT GGAAGCTTGC
    AGGGACGACC TTTTTAAGAT GTGGCTTGAC ATGGTCGTCG ACTTGCTGGA CCTTCGAACG
    5′              311         321       331         341   351
    +1  V I Q        E V G V     E E T P    L M N       V D S I L A
    301 GTTATCCAGG AAGTTGGTGT TGAAGAAACC CCGCTGATGA ACGTTGACTC CATCCTGGCT
    CAATAGGTCC TTCAACCACA ACTTCTTTGG GGCGACTACT TGCAACTGAG GTAGGACCGA
    5′      371   381    391    401   411
    +1 V K K Y F Q R I T L Y L T E K K Y S P C
    361 GTTAAAAAAT ACTTCCAGCG TATCACCCTG TACCTGACCG AAAAAAAATA CTCCCCGTGC
    CAATTTTTTA TGAAGGTCGC ATAGTGGGAC ATGGACTGGC TTTTTTTTAT GAGGGGCACG
    5′      431     441  451    461   471
    +1 A W E V V R A E I M R S F S L S T N L Q
    421 GCTTGGGAAG TTGTTCGTGC TGAAATCATG CGTTCCTTCT CCCTGTCCAC CAACCTGCAG
    CGAACCCTTC AACAAGCACG ACTTTAGTAC GCAAGGAAGA GGGACAGGTG GTTGGACGTC
    5′       491  501
    +1   E R L R R K E # (SEQ ID NO: 1)
    481 GAACGTCTGC GTCGTAAAGA ATAA (SEQ ID NO: 2)
    CTTGCAGACG CAGCATTTCT TATT (SEQ ID NO: 3)
  • Example 2
  • Separation and Purification of rSIFN-co
  • 1. Fermentation
  • Inoculate the recombinant strain in LB media, shaking (200 rpm) under 37° C. overnight (approximate 18 h), then add 30% glycerol to the fermentation broth to get final concentration of 15%, allotted to 1 ml tube and kept in −20° C. as seed for production.
  • Add 1% of the seed to LB media, shaking (200 rpm) under 37° C. overnight to enlarge the scale of the seed, then add to RM media with a ratio of 10%, culturing under 37° C. Add arabinose (20% solution) to 0.02% as an inductor when the OD600 reaches about 2.0. 4 hours after that, stop the culture process, collect the bacteria by centrifuge, resuspend the pellet with buffer A, and keep in −20° C. overnight. Thaw and break the bacteria by homogenizer, then centrifuge. Wash the pellet with buffer B, buffer C, and distilled water to get a relatively pure inclusion bodies.
  • 2. Denaturation and Renaturation
  • Dissolve the inclusion body in Guanidine-HCl (or urea) of 6 mol/L. The solution will be a little cloudy. Centrifuge it at a speed of 10000 rpm. Determine the protein concentration of the supernatant. This supernatant is called “denaturation solution.” Add the denaturation solution to renaturation buffer, and keep the final protein concentration under 0.3 mg/ml. It is better to add the totally denatured solution in three steps instead of one step. Keep the solution overnight under 4° C. Afterwards, dialyze 10 mol/L, 5 mol/L PB buffer and distilled water, then adjust its pH by 2 mol/L HAc—NaAc. Let it stand, then filtrate.
  • 3. Purification
      • POROS HS/M anion exchange chromatography:
  • Figure US20180258151A1-20180913-C00001
  • Chelating Sepharose™ fast flow: Add PB buffer of 0.2 mol/L (pH 6.6) and NaCl of 4 mol/L in the solution from HS to adjust solution pH to pH 6.0 and NaCl concentration to 1 mol/L.
  • Figure US20180258151A1-20180913-C00002
  • Condense the eluted solution by POROS HS/M. Sometimes a purification by sephacryl S-100 step can be added to meet stricter purity requirements.
  • Note:
    • Buffer A: 100 mmol/L Tris-HCl, pH 7.5-10 mmol/L EDTA-100 mmol/L NaCl
    • Buffer B: 50 mmol/L Tris-HCl, pH 7.5-1 mol/L Urea-10 mmol/L EDTA-0.5% Triton X-100
    • Buffer C: 50 mmol/L Tris-HCl, pH 7.5-2 mol/L Urea-10 mmol/L EDTA-0.5% Triton X-100
    • Buffer D: 1 mol/L NaCl - - - 50 mmol/L Na2HPO4 (pH 5.5)
    • Buffer E: 1 mol/L NaCl - - - 50 mmol/L Na2HPO4 (pH 5.0)
    • Buffer F: 1 mol/L NaCl - - - 50 mmol/L Na2HPO4 (pH 4.0)
    • Buffer G: 1 mol/L NaCl - - - 50 mmol/L Na2HPO4 (pH 3.6)
    • Renaturation buffer: 0.5 mol/L Arginine-150 mmol/L Tris-HCl, pH 7.5-0.2 mmol/L EDTA
  • LB Media: 1 L
    Tryptone 10 g
    Yeast extracts 5 g
    NaCl 10 g
    RM Media: 1 L
    Casein 20 g
    MgCl 1 mmol/L (0.203 g)
    Na2HPO4 4 g;
    KH2PO4 3 g,
    NaCl 0.5 g
    NH4Cl 1 g
  • After purification, the buffer was changed to PBS (pH 7.0) along with the step of condensing by POROS HS/M. This is called the “Protein Stock Solution.” It can be directly used in the preparation of injections or sprays, or stored at 2-8° C.
  • Formula for injection:
  • Solution Lyophilized powder
    Solution of rSIFN-co 34.5 μg/ml 34.5 μg/ml
    PB (pH 7.0) 25 mmol/L 10 mmol/L
    Glycine 0.4 mol/L
    NaCl 0.1 mol/L
  • For spray:
  • EDTA 0.01%
    Tween
    80 0.05%
    Trisodium citrate
    10 mmol/L
    Glycerol 1.26%
    Sodium Chloride 0.03%
    Phenylmethanol 0.5%
    HSA 0.1%
    rSIFN-co 10 μg/ml
  • Quality Control Process
  • During purification, tests for protein content, protein purity, specific activity and pyrogen are conducted after each step. When the stock solution is obtained, all the tests listed in the table are done one after the other.
  • The quality of the product is controlled according to “Chinese Requirements for Biologics.”
  • 1. Original protein solution
  • Item of Test Method
    Protein Stock Solution:
    Test for Protein Content Lowry
    Test for Protein Purity Non-reductive SDS-PAGE
    (sodium dodecyl sulfate
    polyacrylamide gel
    electrophoresis)
    HPLC Analysis
    Test for Molecular Weights Reductive SDS-PAGE
    Test for Specific Activity According to Method in
    “Specific Activity Test of
    Interferon
    Test for Leftover Exogenetic Using DNA Labeling and
    DNA Detection Kit
    Test for Activity of According to Method in
    Leftover Antibiotics “Chemical and Other Test
    Methods for Biologics”
    Test for Bacterial Endotoxin According to Method in
    “Requirements for Bacterial
    Endotoxin Test of Biologics”
    Test for Isoelectronic Point Isoelectric Focusing
    Electrophoresis
    Test for Identify UV spectrum (range of
    Characteristics of the wavelength: 190-380 nm)
    Protein
    Peptide Mapping (hydrolyzed
    by pancreatic enzyme,
    analyzed by C-18 column)
    N-terminal Sequence Test
    C-terminal Sequence Test
    Circular Dichroism
    Amino Acid Analysis
    Semi-finished Product
    Test for Bacterial Endotoxin According to Method in
    “Requirements for Bacterial
    Endotoxin Test of Biologics”
    Product
    Appearance Check
    Chemical According to Method in
    “Chemical and Other Test
    Methods for Biologics”
    Test for Specific Activity According to Method in
    “Specific Activity Test of
    Interferon
    Sterility Test According to Method in “c”
    Abnormal Toxicity Test Test on Mouse
    Pyrogen Test According to Method in
    “Requirements for Pyrogen
    Test of Biologics”
    Test for Stability of
    Product
    Note:
    “Chemical and Other Test Methods for Biologics”, “Requirements for Pyrogen Test of Biologics” and “Requirements for Bacterial Endotoxin Test of Biologics” all can be found in the “Chinese Requirements for Biologics.” “Chinese Requirements for Biologics,” PAN Zhengan, ZHANG Xinhui, DUAN Zhibing, et al. Chinese Biologics Standardization committee. Published by Chemical Industry Publishing Company, 2000.
  • Example 3 Stability of Lyophilized Powder of Recombinant Super-Compound Interferon Injection
  • The stability experiments were carried out with samples of lyophilized powder of recombinant super-compound interferon (rSIFN-co) injection in two specifications and three batches. The experiments started in April 2000.
  • 1. Sample Source
  • Samples were supplied by Sichuan Huiyang Life-engineering Ltd., Sichuan Province. Lot: 990101-03, 990101-05, 990102-03, 990102-05, 990103-03, 990103-05
  • 2. Sample Specifications
  • Every sample in this experiment should conform with the requirements in the table below.
  • TABLE 1
    Standard of Samples in Experiment
    Items Standards
    1. Appearance white loose powder
    2. Dissolving dissolve rapidly in injection water(within
    time 2 min) at room temperature
    3. Clarity colorless liquid or with little milk-like
    glisten; should not be cloudy, impurity or
    with indiscernible deposit
    4. pH value 6.5~7.5
    5. Potency (IU/dose) 80%~150% of indicated quantity (9 μg: 4.5 ×
    106 IU, 15 μg: 7.5 × 106 IU)
    6. Moisture no more than 3.0% (W/W)
  • 3. Experimental Content
  • Test samples at 2˜8° C.: The test samples were put into a 2˜8° C. refrigerator, then the above items of these samples were respectively tested in the 1st, 3rd, 6th, 9th, 12th, 18th, 24th, 30th, 36th month. The results were recorded.
  • Test samples at 25° C.: The test samples were put into a thermostat at 25° C., then the above items of these samples were respectively tested in the 1st, 3rd, 6th, 9th, 12th, 18th, 24th, 30th month. The results were recorded.
  • Test samples at 37° C.: The test samples were put into a thermostat at 37° C., then the above items of these samples were respectively tested in the 1st, 3rd, 6th, 9th, 12th, 18th, 24th month. The results were recorded.
  • 4. Results and Conclusion
      • 1) At 37° C., according to data collected at designated points during testing and compared with data before testing, the potency began descending from the 6th month and the changes in the three batches were similar. The appearance of other items had no changes.
  • 2) At 25° C., according to data collected at designated points during testing and compared with data before the testing, the potency only had a little change, and the changes in the three batches were similar. The appearance of other items had no changes.
  • 3) At 2-8° C., according to data collected at designated points during testing and compared with data before testing, the potency of the three batches all were stable. The appearance of other items also had no changes.
  • In conclusion, it is suggested that the lyophilized powder of recombinant super-compound interferon for injection should be better stored and transported at low temperatures. Without such conditions, the product can also be stored for short periods (i.e., 3 months) at room temperature.
  • Example 3.5
  • Production Flow Chart of rSIFN-co
  • 1. Production
      • 1.1 Fermentation
        • Use mixture of LB+M9 as culturing medium. The amount of innoculum will be 1.5%. Agitate to OD600=0.4 (about 3.5 hours) under 32° C., then raise temperature to 42° C. Continue the agitation for another 6 hours, the expression of rSIFN-co will reach the maximum level. The examination under scanning of the gel resulting from SDS-PAGE shows that the level of expression is up to 57%, which is the highest standard in China.
      • 1.2 Purification
  • Figure US20180258151A1-20180913-C00003
  • The purity of the product (rSIFN-co) from this production procedure is shown to 95% under the test of SDS-PAGE where molecular weight is 14.5 Kda. The reverse phase HPLC shows a single peak and the purity is up to 97%. Its specific activity is up to 1×109 IU/mg protein.
      • 1.3 Packaging and Inspection
        • After HPLC purification, 2% human serum albumin, 1% sucrose and 1% glucose are added to the rSIFN-co. It is then separated and lyophilized into injection sample. When tested under the Wish-VVS inspection system, the result was 4.5×108 IU. When tested with aseptic inspection and pyrogen inspection under the standard requirement of China, the results were negative. This result complies with the requirements for IV injection.
    2. Quality Control
      • 2.1 Biological characteristics
        • (1) When using LB+M9 to cultivate bacteria, the characteristics should match with the typical characteristics of E-coli bacteria. No other bacteria were detected.
        • (2) When smeared for Gram staining and inspected under a microscope, it is bacteria-negative.
        • (3) Reaction to antibiotics is the same as those original bacteria.
        • (4) Electron microscope inspection shows typical characteristics of E-coli bacteria. No mycoplasma, virus spore or other micro pollutes was detected.
        • (5) Biochemical reaction test shows characteristics of E-coli bacteria.
      • 2.2 Quality control of interferon expression
        • (1) Interferon expression (cultivated in an agitating platform) matches the amount of expression in original input bacteria.
        • (2) When tested with anti-interferon serum, a reaction is shown.
        • (3) Plasmid inspection: Restriction digest matched with the original plasmid.
      • 2.3 Bacteria strain product
        • Bacteria strain product denotes the specimen from the original bacteria strain that was produced from the procedures shown in 1.2.
        • The bacteria strain product should be inspected as follows to make sure there is no derivation: Use LB to plate 2-3 pieces and cultivate. Separate and take 5-10 bacteria groups for the test of interferon expression. Repeat the test at least two (2) times. Only use the one which shows the highest % to be the bacteria strain product.
      • 2.4 Inoculum
        • The inoculum denotes the chosen bacteria strain product after fermentation. The amount, cultivation time and most appropriate OD value of inoculum can be decided according to bacteria strain. An anti-polluted bacteria procedure should apply for whatever inoculum would be produced.
      • 2.5 Growing of bacteria strain
        • Growing of bacteria strain would be done in a Bacteria Free room environment where no more than one bacterium is growing in the same room. Same culturing medium will be used for both bacteria strain and inoculum. The one used in rSIFN-co is LB.
      • 2.6 Fermentation
        • (1) Fermentation only takes place in a clean fermentation room with a single bacteria fermentation environment.
        • (2) Cleaning of fermentation container and tube is done twice, before and after the insertion of culturing medium. Then, the container should be frozen to reach the appropriate temperature for inoculum.
        • (3) Avoid using antibiotic which might affect cell growth in the culturing medium.
        • (4) Fermentation parameters like temperature, pH value, dissolved oxygen and time required could be varied according to different types of bacterial strains.
      • 2.7 Bacteria collection
        • (1) Centrifuge the bacteria solution to collect bacteria or use another method. All apparatus should be cleaned before and after the operation. The waste solution should be drained after the cleaning procedure.
        • (2) The bacteria should be kept under 4-8° C. if they are going to be split within 24 hours. Otherwise, they should be kept under −30° C. Those are kept under such conditions can be used within 6 months.
      • 2.8 Bacteria cell lysis
        • (1) Use appropriate buffer solution to balance the bacteria strain. Cell lysis can be done by physical, chemical or biological methods. Use centrifuge to precipitate the bacteria and apply cleaning solutions.
        • (2) If the chemical method is used to split cells, no solutions harmful to human beings should be used.
      • 2.9 Purification
        • (1) Purification will get rid of most of the non-interferon contents. In the process of purification, no toxic materials should be found if extra elements are added.
        • (2) If using antibody affinity chromatography for purification, there should be an indication of the source and degree of purity. Also, inspection of small quality IgG should be performed.
        • (3) During the process of purification, clearance of pyrogen is critical. All apparatus should be checked to eliminate this interference.
        • (4) The highly concentrated interferon is known as “intermediate product”. After inspection and tests, add albumin to raise the concentration to 2% which is now known as “albumin intermediate product”. After examination and tests, it should be kept at −30° C. and never thawed before use. This product should be used within 6 months.
        • (5) The albumin that is used in this process should also fulfill tests and requirements such as: negativity under RBSAG inspection and an indication of the ratio among monomer, dimer and polymer.
      • 2.10 Production into tube product
        • (1) Filtration: Use 0.22μ membrane to filter the bacteria. The product should be handled with aseptic techniques. Samples should be taken to test the value of the interferon.
        • (2) Dilution: Dilute the albumin intermediate product with 2% diluent. No preservative should be added. The product can be lyophilized after the aseptic inspection and pyrogen inspection.
      • 2.11 Lyophilization
        • The lyophilization should not affect the activity of interferon, and the water content of said lyophilite will be maintained.
      • 2.12 Inspection
        • There are two types of rSIFN-co made. One is for injection and the other for topical use. The specifications for the two are different. There are intermediate products and final products for each type. In the injection type, intermediate products include purified interferon, albumin intermediate product, and bacteria free albumin intermediate product. Final product from the injection type will denote only lyophilized product. The intermediate product in the topical type denotes only purified interferon. The final product from the topical type denotes only separated packed liquid formed lyophilized products.
      • 2.13 Packaging
        • There is different packaging for the injection type and the topical type.
      • 2.14 Storage
        • The product should be kept at 4° C. The purification solution should not be stored in a frozen state.
      • 2.15 Expiration
        • The expiration period is two (2) years after the lyophilization procedure for lyophilized products. The expiration period is 6 months after individual packing for liquidated products.
    Example 4
  • Preparation of rSIFN-co
  • Preparation of Lyophilized Injection
      • Lyophilized powder
  • Stock Solution of 34.5 μg/ml
    rSIFN-co
    PB (pH 7.0)   10 mmol/L
    Glycine  0.4 mol/L
  • Preparation technique: Weigh materials according to recipe. Dissolve with sterile and pyrogen-free water. Filter through 0.22 μm membrane to de-bacterialize, preserve at 6-10° C. Fill in vials after affirming they are sterile and pyrogen-free, 0.3 ml/vial or 0.5 ml/vial, and lyophilize in freeze dryer.
  • Preparation of Liquid Injection
      • Solution
  • Stock Solution of 34.5 μg/ml
    rSIFN-co
    PB (pH 7.0)   25 mmol/L
    NaCl  0.1 mol/L
  • Preparation: Weigh materials according to recipe. Add to desired level with sterile and pyrogen-free water. Filter through 0.22 μm membrane to de-bacterialize, preserve at 6-10° C. Fill in airtight vial after affirming it is sterile and non-pyrogen at 0.3 ml/vial or 0.5 ml/vial. Store at 2-10° C., and protect from light.
  • Example 4.5
  • Acute Toxicity of rSIFN-co
  • Treat mice with large dose (150 μg/kg, equal to 1000 times of the normal dose per kilo used in treatment of adult patients) of rSIFN-co at one time by intramuscular injection. Then observe and record their deaths and toxic reactions. Results show that: 24 hours after injection, no abnormal reaction had been recorded. The organs of the animals which had been selected to be killed also had no signs of abnormal changes. Those remaining mice were all kept alive and were normal after two weeks. The weights of mice in the experimental group and control group all increased, and the ratio of increase showed no obvious difference between the two groups (P>0.05) according to their weights on the fourteenth day. No abnormal changes were seen from the main organs of those mice after two weeks.
  • 1. Experimental Material 1.1 Animals
  • 40 healthy adult mice, weighing 18-22 g, half male and half female, qualified by Sichuan experiment animal control center.
  • 1.2 Medicines
  • rSIFN-co (Provided by Sichuan Huiyang Life-engineering Ltd.) sterilized solution, 0.15 mg/ml, Lot: 981201
  • rSIFN-co was administered i.m. in saline.
  • 2. Method
  • Separate the 40 mice into two groups randomly, one for experimental medicine, another for control. Inject medicines or saline at the same ratio (0.1 ml/10 g) through muscle to each mouse according to which group they belong. (150 μg/kg of rSIFN-co for experimental group; and saline for control group). After injection, observe and record acute toxicity shown in mice. Kill half of the mice (male and female each half) to check whether there were any abnormal pathologic changes in their main organs, such as heart, spleen, liver, lung, kidney, adrenal gland, stomach, duodenum, etc. after 24 hours. Those that remain are kept and observed until the fourteenth day. Weigh all mice, kill them, and then observe the appearance of the organs listed above to see if there are any abnormalities. Take pathological tissue and examine it, using the examination to assess the difference in weight increases in the two groups.
  • 3. Results
  • Results show that there was no acute toxicity seen after all mice were treated with i.m. rSIFN-co with 150 μg/kg at a time, equal to 1000 times the normal dose per kilo used in treatment of adult patients. In the 14 days after injection, all mice lived well. They ate, drank, exercised, and excreted normally and showed normal hair conditions. None of them died. The observation of the main organs of the randomly selected mice shows no abnormal changes 24 hours after injection. 14 days after injection, all remaining mice were killed. Autopsies also showed no changes. The weights of mice in the two groups all increased, but no obvious difference was shown when accessed with statistic method (p>0.05). See Table 6.1:
  • TABLE 6.1
    Influence to weights of mice after injection of rSIFN-co
    Weights Weights Increased
    before after value of
    injection injection weights
    Group Dose Animal (g) (g) (g)
    Control 0 20 19.8 ± 1.7 30.8 ± 2.8 11.0 ± 2.9
    rSIFN-co 150 20 19.4 ± 1.7 32.1 ± 3.3 12.7 ± 4.3
  • 4. Conclusion
  • Under conditions of this experiment, there were no toxic reactions in all mice after injection of rSIFN-co with 150 μg/kg. The conclusion can be reached that the maximum tolerable dose of i.m. in mice is 150 μg/kg, which is equal to 1000 times the normal dose per kilo used in treatment of adult patients.
  • 2002 rSIFN-co Drug Inspection Report: Nov. 14, 2002 rSIFN-co Drug Inspection Report by China Drugs & Biological Products Inspection Laboratory.
  • On Nov. 14, 2000, 80 vials of rSIFN-co each containing 9 μg (micrograms) provided by Sichuan Biotechnology Research Center were tested. rSIFN-co Drug was white in color with produced no precipitation when water was added. The pH value was 6.9 while the standard was between 6.5 to 7.5. The water content of rSIFN-co was 2.3% while the standard was smaller than 3.0%. Test for bacteria showed no bacterial grown. rSIFN-co passed pyrogen test. The toxicity test on mice showed no harm. Mice were alive and gained weight. The specific activity test was 6.0×106 IU/vial while the standard was between 3.6×106 IU/vial to 6.8×106 IU/vial. The identification test was positive.
  • Example 5
  • Crystal Growth of rSIFN-co and Test of Crystallography Parameter
  • Crystal of rSIFN-co. Two types of crystal were found after systematically trial and experiment. (See FIGS. 7-9)
  • 1. Crystal Growth
      • Dissolve rSIFN-co protein with pure water (H2O) to 3 mg/ml in density. Search of crystallization by using Hampton Research Crystal Screen I and II which was made by Hampton Company. By using Drop Suspension Diffusion Method, liquid 500 μl, drop 1 μl protein+1 μl liquid, in 293K temperature. First 2 different types of small crystals were found as listed in Table 12.1.
  • TABLE 12.1
    Screen of rSIFN-co Crystallin
    Condition
    I II
    Diluent 0.1M Tris-HCl 0.1M HEPES
    PH = 8.75 PH = 7.13
    Precipitant 17.5% (w/v) PEG550 MME 10% (w/v)PEG6K
    Additives 0.1M NaCl 3% (w/v)MPD
    Temperature 293K 293K
    Crystal Size (mm) 0.2 × 0.2 × 0.1 0.6 × 0.02 × 0.02
    Crystallogram FIG. 7 FIG. 8
  • 2. Data Collection and Processing
      • Crystal I was used to collect X-Ray diffraction data and preliminary analysis of crystallography. Parameters were also tested. The diffraction data was collected under room temperature. Crystal I (Condition I) was inserted into a thin siliconized wall tube. Using BrukerAXS Smart CCD detector, the light source is CuKα (λ=1.5418 Å) generated by Nonius FR591 X-ray generator. Light power 2000 KW (40 kv×50 mA), wave length 1.00 Å, under explosion 60 second, Δφ=2°, the distance between crystal and detector was 50 mm. Data was processed for using Proteum Procedure Package by Bruker Company. See FIG. 9 for crystal diffraction pattern (partially). See Table 12.2 for the result of the process.
  • TABLE 12.2
    Results of Crystallography Parameters
    Parameters
    a (Å) 82.67
    b (Å) 108.04
    c (Å) 135.01
    α (Å) 90.00
    β (Å) 90.00
    γ (Å) 98.35
      • Space Group P2 or P21
      • Sharpness of separation 5 Å
      • Asymmetric molecule #10
      • Dissolution 57.6%
  • Besides, there was no crystal growth of rSIFN-co based on previous publications. The closest result to the rSIFN-co was huIFN-α2b but the screen was very complicated. After seeding 3 times, crystal grew to 0.5×0.5×0.3 mm, sharpness of separation was 2.9 Å, space group was P21. The crystals were also big, asymmetric molecule number was 6, and dissolution was about 60%.
  • Clinical Report 1: Evidence of effectiveness of rSIFN-co in healing cancer. See FIGS. 17A-H.
  • The ultra sound inspection showed an enlarged right ovary and abdominal fluid. The patient was suspected of having ovarian cancer.
  • Western China No. 2 Hospital reported a patient with ovarian cancer and breast gland cancer diagnosed on Jul. 14, 2004. Her serum contained CA-125>600 U/ml and CA-153>250 U/ml. Also 2000 ml abdominal water was found. On Jul. 16, 2004, malignant cancer cells and low differential gland cancer cells (likely a low graded differential Adenocarcinoma) were found from the abdominal water and cancer cells and death materials were found from the mammary gland check up. On Aug. 4, 2004, it was concluded diagnosis as ovary cancer.
  • The patient was treated with rSFIN-co starting Jul. 14, 2004. She was injected with 15 μg of rSFIN-co on Jul. 14, 2004, Jul. 16, 2004, Jul. 18, 2004, Jul. 20, 2004 and Jul. 22, 2004 respectively. She began chemotherapy on Jul. 22, 2004. On Aug. 3, 2004 abdominal surgery was performed. It was expected that her abdominal water would be more than 2000 ml. However, only 200 ml were recorded. On Aug. 4, 2004 the examination results showed she had mammary gland cancer, ovarian cancer of right and left ovary and lymphoma. She was treated with rSIFN-co and chemotherapy at the same time. She did not have operation on mammary glands.
  • On Dec. 27, 2004 the examination report showed her CA-125 dropped to 5 U/ml and CA-153 dropped to 13 U/ml. On Feb. 25, 2005, her PET examination report from Daping Hospital, Third Military Medical University of PRC showed there was no obvious abnormal difference on metabolic reactions on her body and brain. The symptoms of her mammary gland cancer disappeared. No traces of cancer were found.
  • PET Imaging: On Feb. 25, 2005 PET imaging report on Feb. 25, 2005 of this 43 years old patient diagnosis with left side ovary cancer and was treated with rSIFN-co since Jul. 14, 2004; PET imaging was done at PET Center of the Daping Hospital, Third Military Medical University of PRC.
  • Fasting patient was intravenously injected with 18F-FDG14.8 mCi. Brain images were taken 50 minutes after injection. The images were clear, no obvious abnormal increase or decrease of radiation were observed on cerebral epidermis, both sides of cerebellum, both sides of hypothalamus and basal.
      • Whole body imaging was done 60 minutes after injection. The images were clear. No obvious abnormal increase or decrease of radiation on neck, lungs, mediastinum, liver, both sides of adrenals, abdominal lymph gland, pelvic cavity, bones.
  • The image of heart was clear.
  • Result: The FDG-PET images of the whole body and brain did not show abnormal FDG metabolic increase or decrease after five-and-half (5.5) months of rSIFN-co treatment of ovarian ovary cancer.
  • Conclusion: Comparison of CA-153 and CA-125 levels before and after rSFIN-co treatment evidenced that rSFIN-co is effective against breast and ovarian cancer.
  • Clinical Report 2: A kidney cancer patient was treated in the following manner. In a half-month period, the patient was given 3 injections of 9 μg of rSIFN-co and 3 injections of 15 μg of rSIFN-co. In the one and a half months following these injections, he received 24 μg injections of rSIFN-co every day. A kidney biopsy showed no metastasis after this course of treatment. The patient showed a full recovery. Every half year after recovery, the patient received 15 μg injections of rSIFN-co 15 times over a one-month period.
  • Example 6
  • rSIFN-co Inhibits HBV-DNA Duplication and Secretion of HBsAg and HBeAg.
  • Materials
  • Solvent and Dispensing Method: Add 1 ml saline into each vial, dissolve, and mix with MEM culture medium at different concentrations. Mix on the spot.
  • Control drugs: IFN-α2b (Intron A) as lyophilized powder, purchased from Schering Plough. 3×106IU each, mix to 3×106IU/ml with culture medium; INFERGEN® (liquid solution), purchased from Amgen, 9 μg, 0.3 ml each, equal to 9×106IU, and mix to 9×106IU/ml with culture medium preserve at 4° C.; 2.2.15 cell: 2.2.15 cell line of hepatoma (Hep G2) cloned and transfected by HBV DNA, constructed by Mount Sinai Medical Center.
  • Reagent: MEM powder, Gibco American Ltd. cattle fetal blood serum, HycloneLab American Ltd. G-418 (Geneticin); MEM dispensing, Gibco American Ltd.; L-Glutamyl, imported and packaged by JING KE Chemical Ltd.; HBsAg and HBeAg solid-phase radioimmunoassay box, Northward Reagent Institute of Chinese Isotope Ltd.; Biograncetina, Northern China Medicine; And Lipofectin, Gibco American Ltd.
  • Experimental goods and equipment: culture bottle, Denmark Tunclon™; 24-well and 96-well culture board, Corning American Ltd.; Carbon Dioxide hatching box, Shel-Lab American Ltd.; MEM culture medium 100 ml: 10% cattle fetal blood serum, 3% Glutamyl 1%, G418 380 μg/ml, biograncetina 50 U/ml.
  • Method:
  • 2.2.15 cell culture: Added 0.25% pancreatic enzyme into culture box with full of 2.2.15 cell, digest at 37° C. for 3 minutes, and add culture medium to stop digest and disturb it to disperse the cells, reproduce with ratio of 1:3. They will reach full growth in 10 days.
  • Toxicity test: Set groups of different concentrations and a control group in which cells are not acted on with medicine. Digest cells, and dispense to a 100,000 cell/ml solution. Inoculate to 96-well culture board, 200 μl each well, culture at 37° C. for 24 h with 5% CO2. Test when simple cell layer grows.
  • Dispense rSIFN-co to 1.8×107IU/ml solution, then prepare a series of solutions diluted at two-fold gradients. Add into 96-well culture board, 3 wells per concentration. Change the solution every 4 days. Test cytopathic effect by microscope after 8 days. Fully destroy as 4, 75% as 3, 50% as 2, 25% as 1, zero as 0. Calculate average cell lesion and inhibition rate of different concentrations. Calculate TC50 and TC0 according to the Reed Muench method.
  • TC 50 = Antilog ( B + 50 - B A - B × C )
  • A=log>50% medicine concentration, B=log<50% medicine concentration, C=log dilution power
  • Inhibition test for HBeAg and HBsAg: Separate into positive and negative HBeAg and HBsAg contrast groups, cell contrast group and medicine concentration groups. Inoculate 700,000 cells/ml of 2.2.15 cell into 6-well culture board, 3 ml each well, culture at 37° C. for 24 h with 5% CO2, then prepare 5 gradiently diluted solutions with 3-fold as the grade (Prepare 5 solutions, each with a different protein concentration. The concentration of Solution 2 is 3 times lower than that of Solution 1, the concentration of Solution 3 is 3 times lower than that of Solution 2, etc.) 4.5×106IU/ml, 1.5×106IU/ml, 0.5×106IU/ml, 0.17×106 IU/ml, and 0.056×106 IU/ml, 1 well per concentration, culture at 37° C. for 24 h with 5% CO2. Change solutions every 4 days using the same solution. Collect all culture medium on the 8th day. Preserve at −20° C. Repeat test 3 times to estimate HBsAg and HBeAg with solid-phase radioimmunoassay box (Northward Reagent Institute of Chinese Isotope Ltd.). Estimate cpm value of each well with a γ-accounting machine.
  • Effects calculation: Calculate cpm mean value of contrast groups and different-concentration groups and their standard deviation, P/N value such as inhibition rate, IC50 and SI.
  • 1 ) Antigen inhibition rate ( % ) = A - B A × 100
      • A=cpm of control group; B=cpm of test group;
      • 2) Counting the half-efficiency concentration of the medicine
  • Antigen inhibition IC 50 = Antilog ( B + 50 + B A - B × C )
  • A=log>50% medicine concentration, B=log<50% medicine concentration, C=log dilution power
      • 3) SI of interspace-conformation changed rSIFN-co effect on HBsAg and HBeAg in 2.2.15 cell culture
  • SI = TC 50 TC 50
      • 4) Estimate the differences in cpm of each dilution degree from the control group using student t test
  • Southern blot: (1) HBV-DNA extract in 2.2.15 cell: Culture cell 8 days. Exsuction culture medium (Separate cells from culture medium by means of draining the culture medium.). Add lysis buffer to break cells, then extract 2 times with a mixture of phenol, chloroform and isoamyl alcohol (1:1:1), 10,000 g centrifuge. Collect the supernatant adding anhydrous alcohol to deposit nucleic acid. Vacuum draw, re-dissolve into 20 μl TE buffer. (2) Electrophoresis: Add 6×DNA loading buffer, electrophoresis on 1.5% agarose gel, IV/cm, at fixed pressure for 14-18 h. (3) Denaturation and hybridization: respectively dip gel into HCl, denaturaion buffer and neutralization buffer. (4) Transmembrane: Make an orderly transfer of DNA to Hybond-N membrane. Bake, hybridize and expose with dot blot hybridization. Scan and analyze relative density with gel-pro software. Calculate inhibition rate and IC50.
  • Results
  • Results from Tables 4.1, 4.2 and 4.3 show: After maximum innocuous concentration exponent culturing for 8 days with 2.2.15 cell, the maxima is 9.0±0×106 IU/ml average inhibition rate of maximum innocuous concentration rSIFN-co to HBeAg is 46.0±5.25% (P<0.001), IC50 is 4.54±1.32×106IU/ml, SI is 3.96; rate to HBsAg is 44.8±6.6%, IC50 is 6.49±0.42×106IU/ml, SI is 2.77. This shows that rSIFN-co can significantly inhibit the activity of HBeAg and HBsAg, but that the IFN of the contrast group and INFERGEN® cannot. It has also been proven in clinic that rSIFN-co can decrease HBeAg and HBsAg or return them to normal levels.
  • TABLE 4.1
    Results of inhibition rate of rSIFN-co to HBsAg and HBeAg
    Inhibition rate Average Accumulated
    Concentration First Second Third First Second Third inhibition inhibition
    (×104 IU/ml) well well well well well well rate Accumulation 1-Accumulation rate
    First batch: (rSIFN-co)
    Inhibition effect to HBeAg
    900 9026 8976 10476 0.436227 0.43935 0.345659 0.407079 0.945909 0.592921 0.614693546
    300 9616 12082 10098 0.3993754 0.245347 0.369269 0.337997 0.5388299 1.254924 0.300392321
    100 9822 16002 12800 0.386508 0.0005 0.2005 0.195836 0.200833 2.059088 0.08867188
    33.33333 15770 19306 16824 0.014991 0 0 0.004997 0.0049969 3.054091 0.001633453
    11.11111 19172 22270 18934 0 0 0 0 0 4.054091 0
    Control Cell 16010 Blank 0 Dilution 3  IC50 602.74446016
    Inhibition effect to HBsAg
    900 7706 7240 7114 0.342155 0.381936 0.392693 0.372261 0.922258 0.627739 0.595006426
    300 8856 7778 9476 0.2439816 0.336008 0.191053 0.257014 0.5499972 1.370724 0.286349225
    100 10818 10720 10330 0.07649 0.084856 0.118149 0.093165 0.292983 2.27756 0.113977019
    33.33333 10744 11114 10570 0.082807 0.051221 0.097661 0.07723 0.1998179 3.20033 0.058767408
    11.11111 10672 9352 10810 0.088953 0.201639 0.077173 0.122588 0.122588 4.077742 0.02918541
    Control Cell 11714 Blank 0 Dilution 3 IC50 641.7736749
    Second batch: (rSIFN-co)
    Inhibition effect to HBeAg
    900 7818 8516 9350 0.554378 0.514592 0.467054 0.512008 1.371181 0.487992 0.737521972
    300 10344 10628 9160 0.4103967 0.394209 0.477884 0.427497 0.8591731 1.060496 0.447563245
    100 12296 14228 13262 0.299134 0.18901 0.244072 0.244072 0.4316522 1.816423 0.19201839
    33.33333 15364 17414 16188 0.124259 0.00741 0.77291 0.069653 0.1876045 2.74677 0.063933386
    11.11111 17386 13632 15406 0.009006 0.222982 0.121865 0.117951 0.117951 3.628819 0.03148073
    Control Cell 16962 Blank 0 Dilution 3 IC50 365.9357846
    Inhibition effect to HBsAg
    900 5784 6198 5792 0.498265 0.462353 0.497571 0.486063 0.893477 0.513937 0.634835847
    300 7150 8534 8318 0.379771 0.259715 0.278452 0.30598 0.4074138 1.207957 0.252210647
    100 9830 11212 10210 0.147294 0.027412 0.11433 0.096345 0.101434 2.111612 0.04583464
    33.33333 13942 12368 13478 0 0 0 0 0.0050891 3.111612 0.001632835
    11.11111 12418 11634 11352 0 0 0.015267 0.005089 0.005089 4.106523 0.001237728
    Control Cell Blank 0 Dilution 3 IC50 611.0919568
    Third batch: (rSIFN-co)
    Inhibition effect to HBeAg
    900 9702 9614 8110 0.428016 0.433204 0.521872 0.461031 1.316983 0.538969 0.709599543
    300 8914 10032 8870 0.4744723 0.40856 0.477066 0.453366 0.8559525 1.085603 0.440859127
    100 16312 12688 13934 0.038321 0.251975 0.178517 0.156271 0.402586 1.929332 0.172641621
    33.33333 15080 12814 13288 0.110954 0.244547 0.216602 0.190701 0.2463153 2.738631 0.082519158
    11.11111 21928 15366 15728 0 0.094093 0.072751 0.0055615 0.055615 3.683017 0.014875633
    Control Cell 17544 Blank 0 Dilution 3 IC50 382.0496935
    Inhibition effect to HBsAg
    900 5616 6228 5346 0.496864 0.442035 0.521054 0.486651 0.763125 0.513349 0.597838293
    300 8542 8590 7096 0.234725 0.230425 0.364272 0.276474 0.2764738 1.236875 0.182690031
    100 11420 11360 11394 0 0 0 0 0 2.236875 0
    33.33333 12656 11582 13110 0 0 0 0 0 0
    11.11111 13142 12336 13342 0 0 0 0 0 4.236875 0
    Control Cell 11528 Blank 0 Dilution 3 IC50 694.7027149
    HBeAg: Average IC50: 450.2434 SD: 132.315479
    HBsAg: Average IC50: 649.1894 SD: 42.29580
  • TABLE 4.2
    Results of inhibition rate of Intron A(IFN-α2b) to HBsAg and HBeAg
    Inhibition rate Average Accumulated
    Concentration First Second Third First Second Third inhibition inhibition
    (×104 IU/ml) well well well well well well rate Accumulation 1-Accumulation rate
    Inhibition effect to HBeAg
    300 14918 11724 9950 0 0.029711 0.176529 0.068747 0.068747 0.931253 0.068746724
    100 14868 16890 15182 0 0 0 0 0 1.931253 0
    33.33333 16760 21716 16400 0 0 0 0 0 2.931253 0
    11.11111 20854 15042 16168 0 0 0 0 0 3.931253 0
    3.703704 12083 12083 12083 0 0 0 0 0 4.931253 0
    Control Cell 17544 Blank 0 Dilution 3 IC50 FALSE
    Inhibition effect to HBsAg
    300 9226 8196 9658 0.152489 0.247106 0.521054 0.1708 0.189295 0.8292 0.185857736
    100 10946 10340 10828 0 0.050156 0.364272 0.018495 0.0184947 1.810705 0.010110817
    33.33333 12250 12980 13934 0 0 0 0 0 2.810705 0
    11.11111 12634 12342 12000 0 0 0 0 0 3.810705 0
    3.703704 10886 10886 10886 0 0 0 0 0 4.810705 0
    Control Cell 10886 Blank 0 Dilution 3 IC50 FALSE
  • TABLE 4.3
    Results of inhibition rate of INFERGEN ® to HBsAg and HBeAg
    Inhibition rate Average Accumulated
    Concentration First Second Third First Second Third inhibition inhibition
    (×104 IU/ml) well well well well well well rate Accumulation 1-Accumulation rate
    First batch: (INFERGEN ®)
    Inhibition effect to HBeAg
    900 14172 12156 17306 0.091655 0.220869 0 0.104175 0.306157 0.895825 0.254710274
    300 13390 12288 16252 0.1417767 0.212409 0 0.118062 0.2019827 1.777764 0.102024519
    100 14364 18834 14194 0.079349 0 0.090245 0.056531 0.083921 2.721232 0.029916678
    33.33333 15722 16034 16340 0 0 0 0 0.0273897 3.721232 0.007306592
    11.11111 17504 17652 14320 0 0 0.082169 0.02739 0.02739 4.693843 0.005801377
    Control Cell 15602 Blank 0 Dilution 3 IC50 FALSE
    Inhibition effect to HBsAg
    900 12080 11692 12234 0 0.01275 0 0.00425 0.025163 0.99575 0.024647111
    300 12840 11484 12350 0 0.030313 0 0.010104 0.0209125 1.985646 0.010422073
    100 12894 14696 15086 0 0 0 0 0.010808 2.985646 0.003606955
    33.33333 15032 12928 13020 0 0 0 0 0.0108081 3.985646 0.002704416
    11.11111 11794 11984 11508 0.004137 0 0.028287 0.010808 0.010808 4.974837 0.002167838
    Control Cell 11843 Blank 0 Dilution 3 IC50 FALSE
    Second batch: (INFERGEN ®)
    Inhibition effect to HBeAg
    900 6278 6376 6408 0.200051 0.187564 0.183486 0.190367 0.274635 0.809633 0.253290505
    300 7692 9092 6394 0.0198777 0 0.18527 0.068383 0.0842678 1.74125 0.046161005
    100 8960 7474 8190 0 0.047655 0 0.015885 0.015885 2.725365 0.005794856
    33.33333 8530 8144 9682 0 0 0 0 0 3.725365 0
    11.11111 7848 7848 7848 0 0 0 0 0 4.725365 0
    Control Cell 7848  Blank 0 Dilution 3 IC50 FALSE
    Inhibition effect to HBsAg
    900 12364 12268 12274 0.036171 0.043655 0.043187 0.041004 0.140162 0.958996 0.12751773
    300 11590 12708 13716 0.0965076 0.009355 0 0.035287 0.0991581 1.923709 0.0490186
    100 12448 13468 13982 0.029623 0 0 0.009874 0.063871 2.913834 0.02144964
    33.33333 12616 11346 12444 0.016526 0.115529 0.029935 0.053996 0.0539965 3.859838 0.013796309
    11.11111 12828 12828 12828 0 0 0 0 0 4.859838 0
    Control Cell 12828 Blank 0 Dilution 3 IC50 FALSE
    Third batch: (INFERGEN ®)
    Inhibition effect to HBeAg
    900 7240 6642 6158 0.064599 0.14186 0.204393 0.136951 0.217399 0.863049 0.201211735
    300 11072 8786 6902 0 0 0.108269 0.03609 0.0804479 1.82696 0.042176564
    100 7016 9726 7552 0.09354 0 0.024289 0.039276 0.044358 2.787683 0.015663017
    33.33333 7622 8866 8676 0.015245 0 0 0.005082 0.0050818 3.782601 0.001341671
    11.11111 7740 7740 7740 0 0 0 0 0 4.782601 0
    Control Cell 7740  Blank 0 Dilution 3 IC50 FALSE
    Inhibition effect to HBsAg
    900 11048 11856 11902 0.04775 0 0 0.015917 0.015917 0.984083 0.015916796
    300 13454 12896 11798 0 0 0 0 0 1.984083 0
    100 12846 13160 12546 0 0 0 0 0 2.984083 0
    33.33333 12680 12458 12360 0 0 0 0 0 3.984083 0
    11.11111 11602 11602 11602 0 0 0 0 0 4.984083 0
    Control Cell 11602 Blank 0 Dilution 3 IC50 FALSE
    HBeAg: Average IC50: 0 SD: 0
    HBsAg: Average IC50: 0 SD: 0
  • Example 7
  • The Clinic Effects of Recombinant Super-Compound Interferon (rSIFN-co)
  • The recombinant super-compound interferon (rSIFN-co) is an invention for viral disease therapy, especially for hepatitis. Meanwhile, it can inhibit the activity of EB viruses, VSV, Herpes simplex viruses, cornaviruses, measles viruses, et al. Using Wish cells/VSV system as the assay for anti-virus activity, the results showed that: the other rIFN, was 0.9×108 IU/mg, Intron A was 2.0×108 IU/mg and rSIFN-co was 9×108 IU/mg. The anti-viral activity of rSIFN-co is much higher than those of the former two.
  • Under the permission of the State Food and Drug Administration (SFDA), People's Republic of China, the clinical trials have taken place in West China Hospital, Sichuan University, the Second Hospital of Chongqing Medical University, the First Hospital of School of Medical, Zhejiang University since the February 2003. The clinical treatment which focuses on hepatitis B is conducted under the guidance of the mutilcenter, double-blind random test. IFN-α1b was used as control, and the primary results showed the following:
  • The Effect of rSIFN-co Compared with IFN-α1b in the Treatment of Chronic Active Hepatitis B
    1. Standard of Patients Selection: Standards 1-4 are effective for both treatment with rSIFN-co (9 μg) and IFN-α1b (5MU, 50 μg), and Standard 1-5 are for rSIFN-co (15 μg) treatment.
  • 1). Age: 18-65
  • 2). HBsAg-test positive over last six months, HBeAg-test positive, PCR assay, HBV-DNA copies ≥105/ml
    3). ALT≥two times the normal value
    4). Never received IFN treatment; or received the Lamividine treatment but failed or relapsed
    5) Once received other IFNs (3MU or 5MU) treatment six months ago following the standard of SFDA, but failed or relapsed
  • 2. Evaluation of the Effects:
  • In reference to the recommendations from the Tenth China National Committee of Virus Hepatitis and Hepatopathy, the effects were divided into three degrees according to the ALT level, HBV-DNA and HBeAg tests.
    • Response: ALT normal level, HBV-DNA negative, HBeAg negative
    • Partial response: ALT normal level, HBV-DNA or HBeAg negative
    • Non response: ALT, HBV-DNA and HBeAg unchanged
    • The response and partial response groups were considered effective cases.
    3. Results of Clinic Trial:
  • HBsAg HBeAg HBV-DNA Heptal
    Effective Transfer to Transfer to Transfer to function
    Period group Medicine cases Rate negative rate negative rate negative rate Recover rate
    8-12 A rSIFN-co (9 μg) 32 46.88 9.38 28.12 37.50 84.38
    week (15) (3) (9) (12) (27)
    B IFN-α1b 32 21.88 0.00  9.38 15.62 56.25
    (5 MU, 50 μg)  (7) (0) (3)  (5) (18)
    16-24 A rSIFN-co (9 μg) 64 54.69 7.81 25.00 34.38 90.62
    week (35) (5) (16)  (22) (58)
    B IFN-α1b 64 25.00 0.00  9.38 18.75 78.13
    (5 MU, 50 μg) (16) (0) (6) (12) (50)
    Group A: treatment with rSIFN-co(9 μg)
    Group B: treatment with IFN-α1b (5 MU, 50 μg)
  • In Group C, the cases were prior treatment of chronic active hepatitis B with other IFNs (3MU or 5MU) that failed or relapsed and then were treated with rSIFN-co (15 μg), subcutaneous injection, every one day, for 24 weeks. The total cases were 13. After 12 weeks treatment, 7 of 13 (53.85%) were effective. 3 of 13 (23.08%) HBeAg transferred to negative; 7 of 13 (53.85%) HBV-DNA transferred to negative; 11 of 13 (84.62%) heptal functions recovered to normal.
  • 4. The Side Effects of rSIFN-co Compared with IFN-α1b in the Treatment
  • The side effects of IFN include fever, nausea, myalgia, anorexia, hair loss, leucopenia and thrombocytopenia, etc. The maximum dose of IFN-α1b is 5MIU per time; the routine dose is 3 MIU. When taken the routine dose, 90% patients have I-II degree (WHO standard) side effects. They had fever lower than 38° C., nausea, myalgia, anorexia, etc. When taken at maximum dose, the rate of side effects did not rise obviously, but were more serious. The maximum dose of rSIFN-co is 24 μg, subcutaneous injection, every one day for 3 months. The routine dose is 9 μg. When routine doses were used, less than 50% of patients had I-II degree (WHO standard) side effects, including fever below 38° C., nausea, myalgia, anorexia, leucopenia and slight thrombocytopenia. With maximum dosage, about 50% patients suffered from leucopenia and thrombocytopenia after using rSIFN-co one month, but those side effects disappeared after stopping treatment for one week. It is safe for continued use.
  • The Observations of rSIFN-Co Treat Hepatitis C
  • 1. Standard of Patients Selection
      • 1) age: 18-65
      • 2) HCV antibody positive
      • 3) ALT≥1.5 times of the normal value, last more than 6 months
  • 2. Evaluation of the Effects:
      • Referring to the standard of INFERGEN® for treatment of hepatitis C and according to the ALT level and HCV-RNA test, divided the effects into three degree:
      • Response: ALT normal level, HCV-RNA negative
      • Partial response: ALT normal level, HCV-RNA unchanged
      • Non response: ALT and HCV-RNA unchanged
  • 3. Effects in Clinic
      • The clinical trial was done at the same time with hepatitis B treatment. 46 cases received the treatment, 9 μg each time, subcutaneous injection, every day for 24 weeks. After treatment, 26 of 46 (56.52%) have obvious effects, 12 of 46 (26.09%) HCV-RNA transferred to negative, 26 of 46 (56.52%) heptal functions recovered to normal.
    Example 8 Comparison of Inhibitory Effects of Different Interferons on HBV Gene Expression
  • Hepatitis B virus (HBV) DNA contains consensus elements for transactivating proteins whose binding activity is regulated by interferons. Treatment of HBV-infected hepatocytes with interferons leads to inhibition of HBV gene expression. The aim of the present study was to characterize the effects of different interferons on HBV regulated transcription. Using transient transfection of human hepatoma cells with reporter plasmids containing the firefly luciferase gene under the control of HBV-Enhancer EnH I, Enh II and core promoter, Applicant studied the biological activities of three different interferons on transcription.
  • Materials and Methods
  • 1. Interferons: IFN-con1 (INFERGEN®), IFN-Hui-Yang (rSIFN-co) and IFNα-2b (Intron A).
    2. Reporter plasmid: The DNA fragments containing HBV-Enhancer EnH I, Enh II and core promoter were prepared using PCR and blunt-end cloned into the Smal I site of the promoter- and enhancer-less firefly luciferase reporter plasmid pGL3-Basic (Promega, Wis., USA). The resulting reporter plasmid was named as pGL3-HBV-Luc.
    3. Cell Culture and DNA transfection: HepG2 cells were cultured in DMEM medium supplemented with 10% FBS and 100 U/ml penicillin and 100 ug/ml streptomycin. The cells were kept in 30° C., 5% CO2 incubator. The cells were transfected with pGL3-HBV-Luc reporter plasmid using Boehringer's Lipofectin transfection kit. After 18 hours, the medium containing transfection reagents was removed and fresh medium was added with or without interferons. The cells were kept in culture for another 48 hours.
    4. Luciferase Assay: Forty-eight hours after addition of interferon, the cells were harvested and cell lysis were prepared. The protein concentration of cell lysates were measured using Bio-Rad Protein Assay kit. The luciferase activity was measured using Promega's Luciferase Reporter Assay Systems according to the instructions of manufacturer.
  • Results Expression of Luciferase Activity in Different Interferon-Treated Cell Lysates
  • No treatment IFN-con1 rSIFN-co IFNα-2b
    100 65 32 73
  • This result shows that rSIFN-co inhibits most effectively on the expression of HBV gene expression of HB core Antigen. This data shows inhibitory effect of rSIFN-co is twice better than INFERGEN® and Intron A. See FIG. 10.
  • Example 9 Recombinant Super-Compound Interferon Spray Major Component: Recombinant Super Compound Interferon
  • Characteristic: Liquid, no insoluble material
    Pharmacology: Recombinant Super-Compound Interferon has a wide spectrum of anti-virus activity. Its effects are 5-20 times higher than those interferons (IFNs) which are available on the market. It can inhibit coronavirus growth in cell culture. In vitro test shows that rSIFN-co has an obvious anti-SARS virus activity. rSIFN-co effect to 10,000 and 1000 TCID50. The Inhibitory Indexes are 0.92 μg/ml and 0.18 μg/ml respectively. The Treatment Indexes (TI) are 151.28, 773.22 respectively. The mechanism is interruption of the combination reaction between the IFN and the correspondent receptor, and inducement of the expression of 2′5′-A synthesizenzyme, protein kinase R in the target cell, therefore inhibiting expression of the viral protein. IFN can induce expression of various anti-virus proteins to inhibit the reproduce of viral proteins, enhance the function of Natural Killer (NK) cell and other Immune regulative functions, and inhibit the invasion of viruses.
    Acute Toxicity: All mice are alive after the maximum dose (1000 times to human dose) subcutaneous injection, did not observe LD50.
  • Indication: Prevention of Severe Acute Respiratory Syndrome
  • Dosage and Administration: Spray to both nasal cavity and throat, three times a day.
    Adverse Reactions: There was no report of adverse reactions from the rSIFN-co spray. It did not induce allergy. If the stimulation is occasional, adverse gastrointestinal reaction is small, and no other obvious adverse reaction was noted during treatment, it is safe to continue use. All reactions will resolve themselves.
    Warning: Patients allergic to IFN and productions of E. Coli. cannot use this product.
    Precautions: Before first use, spray twice to expel the air. If there is any cloudy precipitation material, if the product is expired, or there is material on the vial, do not use it.
    Pediatric Use: It is unclear.
    Geriatric Use: It is unclear.
    Nursing Mothers and Pregnant Women: Use with care or under physician's supervision.
    Drug Interactions: It is unclear.
    Overdose: One-time dose of over 27 million of International Units have not produced any adverse effects.
    Supplied: 1 spray/pack, 20 ug (1×107IU)/3 ml. See FIGS. 11A-11D.
    Storage: Store at 4-8° C. Do not freeze, protect from light.
    Effective Period: Approximately one year
    Manufacture: Manufactured by Sichuan Huiyang life-engineering Ltd.
  • Address: 8 Yusa Road, Room 902, Building A Chengdu, 610017 Sichuan, P.R. China Example 9-A In Vitro Effect of a New-Style Recombinant Compound Interferon on SARS-Associated Coronavirus Sample Supplied by: Huiyang Life Engineering Lt Company, SiChuan Province
  • Experimenter: Molecular Biology Department, microorganism and epidemiology Institute, Academy of Military Medical Science
    Original Data: Preserved in archive of Molecular Biology Department, microorganism and epidemiology Institute, Academy of Military Medical Science
  • 1. Materials
  • Medicine: New-type recombinant compound interferon, 9 μg each, supplied by Huiyang Life Engineering Lt Company, SiChuan Province, Lot number: 20020501.
    Cells: Vero E6, supplied by Molecular Biology Department of Microorganism and Epidemiology Institute, Academy of Military Medical Science.
    Virus: SARS-associated coronavirus, BJ-01, supplied by Molecular Biology Department of Microorganism and Epidemiology Institute, Academy of Military Medical Science.
    Cell Medium: DMEM supplemented with 10% FBS.
    2. Condition Virus was measured in grade 3rd laboratory of biosafety
  • 3. Method
  • CPE (Cytopathic Effect) Assay of TCID50: 100 μl of Vero E6 cells were plated in 96-well plates at 2×104 cells per well. After 24 hr incubation at 37° C., Vero E6 monolayer cells were treated with 9 levels of SARS-associated coronavirus dilution by 10-fold dilution, 4 wells per dilution. The cells were incubated at 37° C. and 5% CO2. CPE (cytopathic effect) was examined daily by microscopy. CPE less than 25% was determined as +, 26-50% as ++, 51-75% as +++, 76-100% as ++++. CPE was recorded. Then TCID50 was calculated by Reed-Muench method.
    Cytotoxicity of Medicine: Vero E6 cells were inoculated into 96-well plates at 2×104 cells (100 ul) per well. After 24-hr incubation at 37° C., cells grew up to monolayer. The medicine was diluted into 36, 18, 9, 4.5, 2.259 μg/ml (final concentration) and added into wells each for 4 wells. The normal cells as control group were set. CPE of medicine group was daily observed during 5-day period, and then the concentration of medicine exhibiting no toxicity was determined.
    CPE Assay of the Activity of the Medicine Against SARS-Associated Coronavirus: 100 μl of Vero E6 cells were plated in 96-well plates at 2×104 cells per well. After 24 hr incubation at 37° C., cells grew up to monolayer. The medicine at the maximal concentration exhibiting no cytotoxicity was diluted into 5 levels by 2-fold dilution and added into wells (100 μl per well). By incubation with 5% CO2 at 37° C. for 24-hour, different concentration of virus (10−3, 10−4, 10−5) were added. After treatment with virus for 48-72 hours, CPE was examined (CPE less than 25% was determined as +, 26-50% as ++, 51-75% as +++, 76-100% as ++++, normal cell as −). The cells were divided into the normal group, the medicine control group, and the different dilution of virus control group, 4 wells per group. CPE was examined daily. Till cytopathic effect was obviously exhibited in the virus control group, the anti-virus activity of interferon was evaluated. The experiment was repeated. IC50 of the medicine was calculated by Reed-Muench method.
  • 4. Results
  • Toxicity of Virus: TCID50 of virus was 10−8.
    Cytotoxicity of Medicine: the concentration of Recombinant compound interferon exhibiting no cytotoxicity was 18 μg/ml, the cells shape was similar with the control group, and no cytopathic effect was exhibited.
  • The Anti-Virus Effect of the Medicine: Shown in Table 9-A.1 and Table 9-A.2
  • TABLE 9-A.1
    the anti-virus effect of new-type recombinant
    compound interferon (first experiment)
    Concentration of CPE at different
    IFN concentration of virus
    (μg/ml) 10−3 10−4 10−5
    18
     9
     4.5 ++
     2.25 +++ ++
     1.125 ++++ ++++ ++
    Virus control ++++ ++++ +++
    group
    Normal group
    Medicine control
    group
  • TABLE 9-A.2
    the anti-virus effect of new-type recombinant
    compound interferon (second experiment)
    Concentration of CPE at different
    IFN concentration of virus
    (μg/ml) 10−3 10−4 10−5
    18
     9
     4.5 +
     2.25 +++ ++
     1.125 ++++ ++++ ++
    Virus control ++++ ++++ ++++
    group
    Normal group
    Medicine control
    group
  • 5. Conclusion
  • The concentration of the new-type recombinant compound interferon exhibiting no cytotoxicity at 18 μg/ml. Its IC50 were 1.27, 2.25, and 4.04 μg/ml respectively according to the concentration of 10−5(1000TCID50), 10−4(1000TCID50), 10−3(100000TCID50) of SARS-associated coronavirus (Table 9-A.3).
  • TABLE 9-A.3
    IC50 of IFN at different concentrations of virus
    Dilution of virus IC50 of IFN(ug/ml)
    10−3 4.04
    10−4 2.25
    10−5 1.27
  • Principal: Jin-yan Wang Laboratory Assistant: Yan-hong Zhao, Xiao-guang Ji, Xiao-yu Li.
  • Original Data: Preserved in archives of Molecular Biology Department, microorganism and epidemiology Institute, Academy of Military Medical Science
  • Date: From May 12th to 30th, 2003 Example 9-B In Vitro Effect of a New-Type Recombinant Compound Interferon and Recombinant Interferon α-2b Injection on SARS-Associated Coronavirus
  • Sample (rSIFN-co) supplied by: Huiyang Life Engineering Ltd., Sichuan province
  • Experimenter: Molecular Biology Department, Institute of microbiology and epidemiology, Academy of Military Medical Science
  • Original data: Preserved in monument room of Molecular Biology Department, Institute of microbiology and epidemiology, Academy of Military Medical Science
  • 1. Materials
  • Medicine: New-type recombinant compound interferon (rSIFN-co), 618 μg/ml, supplied by Huiyang Life Engineering Ltd., SiChuan Province; Alfaron (recombinant interferon α-2b injection), supplied by Tianjin Hualida Biotechnology Co., Ltd. 30 ug/vial (300,0000 IU/vial), Lot Number:20030105.
    Cells: Vero E6, supplied by Molecular Biology Department of Institute of microbiology and epidemiology, Academy of Military Medical Science.
    Virus: SARS-associated coronavirus, BJ-01, supplied by Molecular Biology Department of Institute of microbiology and epidemiology, Academy of Military Medical Science.
    Condition: Viruses were measured in grade 3rd laboratory of biosafety
  • 2. Method
  • TCID50 was Measured with CPE Assay: Vero E6 cells were inoculated in 96-well plates at 2×104 cells (100 μl) per well. After a 24-hr incubation at 37° C., Vero E6 monolayers were treated with 9 levels of SARS-associated coronavirus dilution by 10 times decreasing, each dilution per 4 wells. The cells were incubated at 37° C. and 5% carbon dioxide. CPE was examined daily by phase-contrast microscopy. CPE less than 25% was determined as +, 26-50% as ++, 51-75% as +++, 76-100% as ++++. CPE was recorded. Then TCID50 was calculated by Reed-Muench method.
    TC50 of IFNs were Measured by MTT Assay: Vero E6 cells were inoculated in 96-well plates at 2×104 cells per well (100 μl). After 24-hr incubation at 37° C., the supernatant liquid was removed when cells grew up to monolayer, then Vero E6 was treated with different concentration of IFNs, each dilution per 4 wells. Normal group was set. After 5-day observation, the cells were mixed with MTT for 4 hours. After that, remove the liquid, and then thereafter DMSO were added into cells for 0.5 hour. The OD570 nm was measured by microplate reader. Finally, TC50 was calculated by Reed-Muench method.
    The Activity of the INFs Against SARS-Associated Coronavirus was Measured with MTT Assay: 100 μl of Vero E6 cells were inoculated in 96-well plates at 2×104 cells per well. After 24-hr incubation 37° C., cells became monolayer. The medicine dilution at the concentration of exhibiting no cytotoxicity was 5 times decreasing and there were 5 levels of dilution. Then each dilution was added to 4 wells, 100 ul per well. After 24-hour incubation at 37° C. and 5% CO2, IFN solution was removed, then different concentrations of virus dilution (10000, 1000, 100 TCID50) were added into dishes, 4 wells per dilution. The cells were divided into the normal group, the medicine control group, and the different dilution of virus control group (10000, 1000, 100 TCID50). The cells were incubated at 37° C. and 5% CO2 for 48-72 hr, until cytopathic effect was exhibited in the virus control group, CPE was recorded (CPE less than 25% was determined as +, 26-50% as ++, 51-75% as +++, 76-100% as ++++, normal cell as −). The growth ability of cells was measured with MTT assay, and then the antivirus effect of the INFs was evaluated. The experiment was repeated 3 times. IC50 of the medicine was calculated by Reed-Muench method.
  • 3. Results
  • TCID50 of Virus: TCID50 of virus was 10−7.
    TC50 of IFNs: The concentration of new-type recombinant compound interferon (rSIFN-co) exhibiting no cytotoxicity was 100 μg/ml, and that of recombinant IFNα-2b was 12.5 μg/ml, the cells shape was identical with the normal group at that concentration. TC50 of new-type recombinant compound interferon (rSIFN-co) was 139.18 μg/ml, that of recombinant IFNα-2b was 17.18 μg/ml.
  • TABLE 9-B.1
    TC50 of IFNs
    TC50(μg/ml) Mean value -
    1st 2nd 3rd (X ± SD,
    IFN experiment experiment experiment n = 3)
    new-type 141.42 125.96 150.08 139.18 ± 12.22
    recombinant
    compound
    interferon
    IFNα-2b 17.68 15.75 18.10 17.18 ± 1.25

    The Anti-Virus Effect of the Medicine: The anti-virus effects of two IFNs were observed in vitro. The results of the experiments are shown on the Table 9-B.2, and the results of TI are shown on the Table 9-B.3.
  • TABLE 9-B.2
    The anti-virus activity of IFNs
    Concentration IC50(μg/ml)
    of 1st 2nd 3rd Mean value -
    IFNs virus (TCID50) experiment experiment experiment (X ± SD, n = 3)
    new-type 10000 0.79 1.04 0.93 0.92 ± 0.12
    recombinant
    compound interferon
    IFNα-2b 5.04 4.56 4.65 4.75 ± 0.25
    new-type 1000 0.19 0.18 0.18 0.18 ± 0.01
    recombinant
    compound interferon
    IFNα-2b 1.18 1.19 1.12 1.16 ± 0.04
    new-type 100 0.08 0.10 0.11 0.10 ± 0.02
    recombinant
    compound interferon
    IFNα-2b 0.33 0.21 0.30 0.28 ± 0.06
  • TABLE 9-B.3
    The anti-virus activity of IFNs
    Concentration
    of TC50 IC50 TI
    IFNs virus (TCID50) (μg/ml) (μg/ml) (TC50/IC50)
    new-type 10000 139.18 0.92 151.28
    recombinant
    compound interferon
    IFNα-2b 17.18 4.75 3.62
    new-type 1000 139.18 0.18 773.22
    recombinant
    compound interferon
    IFNα-2b 17.18 1.16 14.78
    new-type 100 139.18 0.10 1391.80
    recombinant
    compound interferon
    IFNα-2b 17.18 0.28 61.36
  • 4. Conclusion
  • The protection effect of new-type recombinant compound interferon (rSIFN-co) and IFNα-2b on Vero E6 was observed in vitro, and the anti-virus ability of IFNs was manifested. IC50 of new-type recombinant compound interferon on SARS-associated coronavirus at the concentration of 10000, 1000, and 100 was 0.92, 0.18, and 0.10 μg/ml in three experiments, TI of that was 151.28, 773.22, and 1391.80 respectively. IC50 of IFNα-2b was 4.75, 1.16, and 0.28 μg/ml, TI (treatment index) of that was 3.62, 14.78, 61.36 respectively.
  • Most importantly, the two tests (See the above Examples 9A & 9B) of in vitro anti-SARS virus effect of rSIFN-co all testified that even the effective dose of rSIFN-co to inhibit SARS virus is ⅕ of that of Interferon α-2b which was used clinically in China at present, the Treatment Index (TI) of rSIFN-co is nearly 50 times of that of Interferon a-2b. (SEE: In vitro effect of a new-type recombinant compound interferon and recombinant interferon-α-2b injection on SARS-associated coronavirus. By The Institute of Microbiology & Epidemiology, Academy of Military Medical Science) Also, see FIG. 12.
  • Thirty thousand sprays of rSIFN-co had been used among front-line nurses and doctors, and people at high risk in Sichuan province. The result shows that none of the nurses and doctors infected SARS in Sichuan Province.
  • Principal: Jin-yan Wang Laboratory Assistant: Yan-hong Zhao, Xiao-guang Ji, Min Zhang, Jing-hua, Zhao. Date: From Jul. 1 to 30, 2003 Example 10
  • Side Effects and Changes in Body Temperature when Using rSIFN-co
  • There are usually more side effects to using interferon. The side effects includes: nausea, muscle soreness, loss of appetite, hair loss, hypoleucocytosis (hypoleukmia; hypoleukocytosis; hypoleukia), and decrease in blood platelet, etc.
  • Method
  • Sample patients are divided into two groups. 10 patients in Group A were injected with 9 μg rSIFN-co. 11 patients in Group B were injected with 9 μg INFERGEN®. Both groups were monitored for 48 hours after injections. First monitoring was recorded 1 hour after injection. After that, records were taken every 2 hours.
  • Table 11.1 is the comparison of side effects between patients being injected with 9 μg of rSIFN-co and 9 μg of INFERGEN®.
  • TABLE 11.1
    Side Effects
    rSIFN-co INFERGEN ®
    9 μg (Group A) 9 μg (Group B)
    Person: n = 10 Person: n = 11
    Body Systems Reactions Headcount Headcount
    In General Feeble 3 3
    Fever 3 6
    Sole heat 1
    frigolabile 3 4
    Leg 3
    strengthless
    Mild lumbago 2 1
    Body soreness 4 5
    Central Nervous Headache 3 6
    System/ Dizziness 2 11
    Peripheral Drowsiness 3
    Nervous System
    Gastroenterostomy Apoclesis
    1
    Celiodynia 1
    Diarrhea 1
    Musculoskeletal Myalgia 1 2
    system
    Arthralgia
    2
    Respiratory Stuffy nose 1
    system
    Paropsia Swollen Eyes 1
  • Results
  • For those patients who were injected with rSIFN-co, the side effects were minor. They had some common symptoms similar to flu, such as: headache, feebleness, frigolability, muscle soreness, hidrosis, arthralgia (arthrodynia; arthronalgia). The side effects of those patients whom were injected with INFERGEN® were worse than those injected with rSIFN-co.
  • From FIGS. 13A-1, 13A-2, 13B-1, and 13B-2, it was obvious that the body temperatures of sample patients in Group B were higher than the patients in Group A. It also reflected that the endurance of rSIFN-co was much better than INFERGEN®.
  • Example 11
  • Effects of Recombinant Super-Compound Interferon (rSIFN-Co) on Ebola Virus.
  • Background: Ebola virus is a notoriously deadly virus that causes fearsome symptoms, the most prominent being high fever and massive internal bleeding. Ebola virus kills as many as 90% of the people it infects. It is one of the viruses capable of causing hemorrhagic (bloody) fever. There is no specific treatment for the disease. Currently, patients receive supportive therapy. This consists of balancing the patient's fluids and electrolytes, maintaining their oxygen level and blood pressure, and treating them for any complicating infections. Death can occur within 10 days of the onset of symptoms.
  • 1. Materials
      • 1.1 Drugs: rSIFN-co, provided by Sichuan Biotechnology Research Center.
      • 1.2 Virus: Ebola, supplied by The Academy of Military Medical Science, Institute of Microbiology Epidemiology.
      • 1.3 Safety level of experiment: Viral experiments were carried under Biological Laboratory Safety System level 3.
      • 1.4 Animals: 60 BALB/c mice
  • 2 Method
      • 2.1 60 mice were randomly separated into 6 groups, each group consisting of 10 mice. Group 1 was treated with 1 μg/of rSIFN-co on the day of inoculation with Ebola virus. Group 2 was treated with 1 μg/of rSIFN-co day one (1) after inoculation with Ebola virus. Group 3 was treated with 1 μg/of rSIFN-co on the day two (2) after inoculation with Ebola virus. Group 4 was treated with 1 μg/of rSIFN-co on day three (3) after inoculation with Ebola virus. Group 5 was treated with 1 μg/of rSIFN-co on day four (4) after inoculation with Ebola virus. Group 6 was not treated with rSIFN-co and this is designated as the control group.
      • 2.2 Administration of the medication: 1 μg/of rSIFN-co was administered once a day for six (6) consecutive days.
  • 3 Results
      • All ten (10) mice in group 6 (control group) died. All mice in groups one (1), two (2) and three (3) survived with no observable toxic effect. In groups four (4) and five (5), showed some effects.
  • 4 Conclusion
      • Clearly these result show effectiveness of rSIFN-co against Ebola virus.
    Example 12
  • Anti-HIV Effects of Recombinant Super-Compound Interferon (rSIFN-co).
  • 1. Materials
      • 1.1 Wild-Type HIV
      • 1.2 Drug Resistant HIV
      • 1.3 293-CD4-CCR5 cells
      • 1.4 DMEM, Gibco
      • 1.5 Fetal Bovine Seru, Gibco
      • 1.6 rSIFN-co provided by Sichuan Biotechnology Research Center
      • 1.7 96-well plate, NUNC
      • 1.8 CO2 incubator
      • 1.9 Laminar Flow Hood
      • 1.10 Fluorometer
      • 1.11 UV Absorbance Meter
      • 1.12 Others
  • 2. Method
      • 2.1 293-CD4-CCR5 cells in exponential (log) phase were obtained, digested with 0.25% pancreatin, stained with Trypan blue stain to determine cell number and diluted with DMEM to concentration of 2.0×105 cells per milliliter (cell/ml).
      • 2.2 Each well of 96-well plate was filled with 100 μl (microliters) of 293-CD4-CCR5-DMEM suspension solution. The plate was placed into 5% carbon dioxide incubator at 37 degrees Celsius and observed the next day seventy percent (70%) of basal area of the well were recovered.
      • 2.3 After supernatant was removed, 100 μl (microliters) of different concentrations of rSIFN-co were added to each well. Two controls were used: Phosphate Buffered Saline (PBS) and Growth Media.
      • 2.4 The plate was placed into carbon dioxide incubator at 37 degrees Celsius for approximately 18 to 20 hours.
      • 2.5 Experimental wells: Different concentrations of the Wild-Type HIV and Drug Resistant HIV viruses were placed into each well at 100 μl (microliters) per well.
        • Control wells: No virus was added, only 100 μl (microliters) of DMEM per well.
      • 2.6 The plate was placed into carbon dioxide incubator at 37 degrees Celsius for approximately 24 hours.
      • 2.7 Routine Luciferase assay was performed and protein concentrations of the supernatants were measured. Luciferase was measured in RLU/mg units.
  • 3 Results
      • rSIFN-co can inhibit HIV at level of ≥4 nanograms per milliliter (ng/ml). See Table 4 and FIGS. 14-15. When using Luciferase as Y axis and concentration of rSIFN-co as X axis, using EXCEL, it is clear that at level of rSIFN-co ≥4 nanograms per milliliter (ng/ml), the level of Luciferase activities are obviously lower than in PBS and Medium controls. A clear inverse dose-dependent response has been shown.
  • TABLE 4
    Comparison of Inhibition of Wild-Type HIV and Drug
    Resistant HIV by rSIFN-co
    Concentration Luciferase Assay
    of rSIFN-co Wild-Type HIV Drug Resistant HIV
    Medium 13500 + 2000 18000 + 2000
    1 μg/ml 3000 + 200 2800 + 800
    500 ng/ml 3000 + 600 2800 + 900
    250 ng/ml 3400 + 400 4000 + 600
    125 ng/ml 4300 + 200 4100 + 600
    62.5 ng/ml 4300 + 400  4100 + 1000
    31 ng/ml 5000 + 800 5100 + 800
    15 ng/ml 7200 + 400  6000 + 1500
    7.5 ng/ml 7000 + 800  7700 + 1300
    4 ng/ml  9000 + 2000  8900 + 2000
    PBS 13000 + 3000 15100 + 2300
    Medium 16000 + 3600 19000 + 2500
  • 4 Conclusion: rSIFN-Co is Effective Against Both: Wild-Type HIV and Drug Resistant HIV.
  • Example 13
  • Anti-Influenza Effects of Recombinant Super-Compound Interferon (rSIFN-co).
  • 1. Materials
      • 1.1. 10-day old chick embryonic membrane cells
      • 1.2. SIFN-co provided by Sichuan Biotechnology Research Center
      • 1.3. Influenza virus provided by Molecular Biology Department of Institute of microbiology and epidemiology, Academy of Military Medical Science.
      • 1.4. DMEM, Gibco
      • 1.5. Newborn Calf Serum
      • 1.6. 96-well plate, NUNC
      • 1.7. CO2 incubator
      • 1.8. Laminar Flow Hood
      • 1.9. Inverted Microscope
      • 1.10. Others
  • 2. Method
      • 2.1 10-day old chick embryonic membrane cell in exponential (log) phase were obtained, digested with 0.25% pancreatin, stained with Trypan blue stain to determine cell number and diluted with DMEM to concentration of 2.0×105 cells per milliliter (cell/ml).
      • 2.2 Each well of 96-well plate was filled with 100 μl (microliters) of 293-CD4-CCR5-DMEM suspension solution. The plate was placed into carbon dioxide incubator at 37 degrees Celsius. The next day cells grew to a monolayer.
      • 2.3 After supernatant was removed, 100 μl (microliters) of different concentrations of rSIFN-co were added to each well. Two control wells: No rSIFN-co was added
      • 2.4 The plate was placed into carbon dioxide incubator at 37 degrees Celsius for approximately 18 to 20 hours.
      • 2.5 Experimental wells: Different concentrations of the Influenza virus were placed into each well at 100 microliters (μl) per well.
        • Control wells: No Influenza virus was added, only 100 μl (microliters) of DMEM per well.
      • 2.6 The plate was placed into carbon dioxide incubator at 37 degrees Celsius for approximately 24 hours.
      • 2.7 Cells were observed under inverted microscope.
  • 3. Results
      • 3.1 Under inverted microscope, the cells in the control well with Influenza virus added and without interferon had obvious CPE, such as rounding of cells, cell necroses, decrease in reflective light and sloughing off.
      • 3.2 Cells from the experimental wells containing rSIFN-co at concentration ≥10 nanogram per milliliter (ng/ml) had no CPE and morphology comparable to normal cells. See FIG. 16.
      • 3.3 Control Wells without Influenza virus added and without interferon did not have any CPE.
  • 4 Conclusion
      • At concentration ≥10 nanogram per milliliter (ng/ml) rSIFN-co is effective against Influenza virus.

Claims (19)

1-77. (canceled)
78. A method for treating an upper respiratory tract infection in a subject, comprising administering to the subject an effective amount of a recombinant interferon, wherein the recombinant interferon has the amino acid sequence of SEQ ID NO:1 and is encoded by the nucleotide sequence SEQ ID NO:2.
79. The method of claim 78, wherein the recombinant interferon is produced by a method comprising the step of expressing SEQ ID NO:2 in E. coli host cells.
80. The method of claim 78, wherein the upper respiratory tract infection is induced by a virus selected from the group consisting of rhinovirus, coronavirus, respiratory syncytial virus, influenza, parainfluenza, adenoviruses, cox-sackie virus, corona virus and enterovirus.
81. The method of claim 78, wherein the effective amount of the recombinant interferon comprises a dose in a range from nanogram to microgram.
82. The method of claim 78, wherein the recombinant interferon is administered orally, via vein injection, muscle injection, peritoneal injection, subcutaneous injection, nasal or mucosal administration, or by inhalation.
83. The method of claim 78, wherein the recombinant interferon is administered to the subject by spraying the recombinant interferon directly to the subject's nasal cavity or throat.
84. The method of claim 78, wherein the recombinant interferon is administered via a spray device containing twenty micrograms of the recombinant interferon in three millilitres three times a day.
85. The method of claim 78, wherein the recombinant interferon is administered following a protocol of 9 μg, 15 μg or 24 μg injection per day, 3 times a week, for a total of 24 weeks.
86. The method of claim 78, wherein the subject has one or more symptoms selected from the group consisting of malaise, nasal stuffiness, rhinorrhea, nonproductive cough, mild sore throat and low-grade fever.
87. The method of claim 78, wherein the recombinant interferon modulates one or more immune responses in the subject, said immune responses are selected from the group consisting of T-cell immune response, activity of natural killer cell, phagocytosis function of monokaryon and formation of antibodies.
88. A spray device for administering a recombinant interferon to a subject for treating an upper respiratory tract infection, comprising an effective amount of the recombinant interferon, wherein the recombinant interferon has the amino acid sequence of SEQ ID NO:1 and is encoded by the nucleotide sequence SEQ ID NO:2.
89. The spray device of claim 88, wherein the recombinant interferon is produced by a method comprising the step of expressing SEQ ID NO:2 in E. coli host cells.
90. The spray device of claim 88, wherein the recombinant interferon is formulated at a concentration of 10 μg/ml.
91. The spray device of claim 88, wherein the effective amount of the recombinant interferon comprises a dose in a range from nanogram to microgram.
92. The spray device of claim 88, containing twenty micrograms of the recombinant interferon.
93. The spray device of claim 92, wherein the recombinant interferon is formulated in a volume of three milliliters.
94. The spray device of claim 88, wherein the recombinant interferon is formulated in an amount for administering the recombinant interferon three times a day.
95. The spray device of claim 88, wherein the recombinant interferon is administered to the subject by spraying the recombinant interferon directly to the subject's nasal cavity or throat.
US15/910,165 2001-02-28 2018-03-02 Recombinant super-compound interferon and uses thereof Abandoned US20180258151A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/910,165 US20180258151A1 (en) 2001-02-28 2018-03-02 Recombinant super-compound interferon and uses thereof

Applications Claiming Priority (13)

Application Number Priority Date Filing Date Title
CN01104367.9 2001-02-28
CNB011043679A CN1245215C (en) 2001-02-28 2001-02-28 Recombination high efficiency composite interferon used as hepatitis B surface antigen and e antigen inhibitor
PCT/CN2002/000128 WO2002080958A1 (en) 2001-02-28 2002-02-28 Recombination super compound interferon used as hepatitis b surface antigen and e antigen inhibitor
US10/650,365 US7364724B2 (en) 2001-02-28 2003-08-28 Recombinant super-compound interferon
IN279MU2004 2004-03-05
IN279/MUM/2004 2004-03-05
IN280/MUM/2004 2004-03-05
IN280MU2004 2004-03-05
US10/927,975 US20050079579A1 (en) 2001-02-28 2004-08-26 Uses of spatial configuration to modulate protein function
US11/077,813 US20060035327A1 (en) 2001-02-28 2005-03-10 Recombinant super-compound interferon and uses thereof
US12/246,153 US8551469B2 (en) 2001-02-28 2008-10-06 Treatment of tumors and viral diseases with recombinant interferon alpha
US14/019,519 US9944686B2 (en) 2001-02-28 2013-09-05 Treatment of tumors with recombinant interferon alpha
US15/910,165 US20180258151A1 (en) 2001-02-28 2018-03-02 Recombinant super-compound interferon and uses thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/019,519 Continuation US9944686B2 (en) 2001-02-28 2013-09-05 Treatment of tumors with recombinant interferon alpha

Publications (1)

Publication Number Publication Date
US20180258151A1 true US20180258151A1 (en) 2018-09-13

Family

ID=35800443

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/246,153 Expired - Lifetime US8551469B2 (en) 2001-02-28 2008-10-06 Treatment of tumors and viral diseases with recombinant interferon alpha
US14/019,519 Expired - Lifetime US9944686B2 (en) 2001-02-28 2013-09-05 Treatment of tumors with recombinant interferon alpha
US15/910,165 Abandoned US20180258151A1 (en) 2001-02-28 2018-03-02 Recombinant super-compound interferon and uses thereof

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US12/246,153 Expired - Lifetime US8551469B2 (en) 2001-02-28 2008-10-06 Treatment of tumors and viral diseases with recombinant interferon alpha
US14/019,519 Expired - Lifetime US9944686B2 (en) 2001-02-28 2013-09-05 Treatment of tumors with recombinant interferon alpha

Country Status (1)

Country Link
US (3) US8551469B2 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI718086B (en) * 2013-01-07 2021-02-11 英屬維爾京群島商遠東超級實驗室有限公司 Methods and compositions for treatment of bone, skin, subcutaneous, mucosal and/or submucosal cancer by percutaneous and/or transmucosal administration of interferon

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4897471A (en) * 1982-05-06 1990-01-30 Amgen Consensus human leukocyte interferon
US5372808A (en) * 1990-10-17 1994-12-13 Amgen Inc. Methods and compositions for the treatment of diseases with consensus interferon while reducing side effect

Family Cites Families (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4672108A (en) * 1981-12-07 1987-06-09 Hoffmann-La Roche Inc. Crystalline human leukocyte interferon
US4681930A (en) * 1983-09-20 1987-07-21 Hoffmann-La Roche Inc. Immune interferon and a method for its extraction and purification
US5441734A (en) * 1993-02-25 1995-08-15 Schering Corporation Metal-interferon-alpha crystals
TW426523B (en) 1995-04-06 2001-03-21 Hoffmann La Roche Interferon solution
US5480640A (en) * 1995-05-02 1996-01-02 Schering Corporation Alpha interferon for treating prostate cancer
US5972331A (en) * 1995-12-22 1999-10-26 Schering Corporation Crystalline interferon alpha for pulmonary delivery and method for producing the same
US5980884A (en) 1996-02-05 1999-11-09 Amgen, Inc. Methods for retreatment of patients afflicted with Hepatitis C using consensus interferon
US6114145A (en) * 1997-12-05 2000-09-05 Human Genome Sciences, Inc. Synferon, a synthetic interferon
CN1062565C (en) 1998-06-29 2001-02-28 深圳九先生物工程有限公司 Recombination human alpha type composite interferon, prepn. method and use therefor
US6833256B1 (en) 1999-06-22 2004-12-21 University Of Maryland Interferon tau mutants and methods for making them
KR100399156B1 (en) 1999-11-19 2003-09-26 주식회사 엘지생명과학 Liquid Formulation of α-Interferon
DE10037111A1 (en) 2000-07-27 2002-02-07 Boehringer Ingelheim Int Production of a recombinant protein in a prokaryotic host cell
US20020043262A1 (en) 2000-08-22 2002-04-18 Alan Langford Spray device
EP1355939A2 (en) 2000-11-03 2003-10-29 Pestka Biomedical Laboratories, Inc. Interferons, uses and compositions related thereto
CN1245215C (en) 2001-02-28 2006-03-15 四川省生物工程研究中心 Recombination high efficiency composite interferon used as hepatitis B surface antigen and e antigen inhibitor
FR2823764B1 (en) 2001-04-24 2003-12-12 Genodyssee NOVEL POLYNUCLEOTIDES AND POLYPEPTIDES OF THE IFN ALPHA-17 GENE
HUP0501070A2 (en) * 2001-11-14 2006-04-28 Biocryst Pharmaceuticals Nucleosides preparation thereof and use as inhibitors of rna viral polymerases
PL2325202T3 (en) 2003-08-28 2015-03-31 Superlab Far East Ltd Uses of interferons with altered spatial structure
US7585647B2 (en) 2003-08-28 2009-09-08 Guangwen Wei Nucleic acid encoding recombinant interferon
ES2528218T3 (en) 2003-08-28 2015-02-05 Superlab Far East Limited Uses of interferons with modified spatial structure
WO2005067963A1 (en) 2003-12-23 2005-07-28 Intermune, Inc. Use of polyethylene glycol-modified interferon-alpha in therapeutic dosing regimens
MY167668A (en) 2005-03-09 2018-09-21 Superlab Far East Ltd Uses of recombinant super-compound interferons
JP2008541769A (en) 2005-06-03 2008-11-27 アンブレツクス・インコーポレイテツド Improved human interferon molecules and their use

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4897471A (en) * 1982-05-06 1990-01-30 Amgen Consensus human leukocyte interferon
US5372808A (en) * 1990-10-17 1994-12-13 Amgen Inc. Methods and compositions for the treatment of diseases with consensus interferon while reducing side effect

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Sung Archives of Disease in Childhood, 1993, 69 440-442 *

Also Published As

Publication number Publication date
US8551469B2 (en) 2013-10-08
US20140050698A1 (en) 2014-02-20
US20090123417A1 (en) 2009-05-14
US9944686B2 (en) 2018-04-17

Similar Documents

Publication Publication Date Title
US8114395B2 (en) Treatment of viral diseases with recombinant interferon α
US20080124302A1 (en) Uses of Recombinant Super-Compound Interferons
US20150174206A1 (en) Uses of interferons with altered spatial structure
US8287852B2 (en) Treatment of viral diseases with recombinant interferon α
EP2325202B1 (en) Uses of interferons with altered spatial structure
US20060035327A1 (en) Recombinant super-compound interferon and uses thereof
US20180258151A1 (en) Recombinant super-compound interferon and uses thereof
CN101001644B (en) Uses of spatial configuration to modulate protein function
AU2011202683B2 (en) Uses of interferons with altered spatial structure

Legal Events

Date Code Title Description
AS Assignment

Owner name: SUPERLAB FAR EAST LIMITED, VIRGIN ISLANDS, BRITISH

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:WEI, GUANGWEN;REEL/FRAME:045813/0996

Effective date: 20130819

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

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