INTERFERON-BASED CANCER TREATMENT METHOD AND PHARMACEUTICAL COMPOSITION

Information

  • Patent Application
  • 20230270824
  • Publication Number
    20230270824
  • Date Filed
    April 14, 2023
    a year ago
  • Date Published
    August 31, 2023
    8 months ago
Abstract
The present invention relates to the field of biomedicine. Disclosed herein is interferon-based method and pharmaceutical combination for treating cancer. In particular, the present invention relates to an interferon-based method for treating cancer, comprising i) intermittently administering an interferon-based therapeutic agent, and ii) administering an additional anticancer agent, preferably, Gemcitabine, to a subject. The present invention also relates to a pharmaceutical combination for use in said method.
Description
REFERENCE TO SEQUENCE LISTING

The instant application contains a Sequence Listing conforming to the rules of WIPO Standard ST.26 and is hereby incorporated by reference in its entirety. The Sequence Listing has been filed as an electronic document via Patent Center, is entitled NP2022TC1126.xml, was created on Apr. 13, 2023, and is 9,420 bytes in size and is hereby incorporated by reference.


TECHNICAL FIELD

The present invention relates to the field of biomedicine. Disclosed herein is interferon-based method and pharmaceutical combination for treating cancer. In particular, the present invention relates to an interferon-based method for treating cancer, comprising i) intermittently administering an interferon-based therapeutic agent, and ii) administering an additional anticancer agent, preferably, Gemcitabine, to a subject. The present invention also relates to a pharmaceutical combination for use in said method.


BACKGROUND

Interferons (IFN) are active proteins with multiple functions, and cytokines produced by monocytes and lymphocytes. With various biological activities, such as broad-spectrum anti-viral activity, influence on cell growth, differentiation, and regulation of immune functions, they are at present the most important anti-viral infection and anti-tumor biological products.


At present, interferons are categorized into Type I, Type II, and Type III interferons. The Type I interferons include IFN-α, IFN-β, and the like. The IFN-α interferons are mainly generated by monocytes-macrophages. In addition, B cells and fibroblasts may also synthesize the IFN-α interferons. The IFN-β interferons are mainly generated by fibroblasts. Both IFN-α and IFN-β interferons bind to the same receptors which are widely distributed in for example monocyte-macrophages, polymorphonuclear leukocytes, B cells, T cells, platelets, epithelial cells, endothelial cells and tumor cells. It is known that there are more than 23 subtypes of IFN α. There is only one subtype of IFN β. The Type II interferons, or γ interferons, are mainly generated by activated T cells (including Th0, Th1 cells and almost all CD8+ T cells) and NK cells, and belong to the so-called lymphokines. The IFN-γ may exist in the form of linking to an extracellular matrix, so it may control the cell growth by bystander effect. They may be distributed on the surface of almost all cells except mature red blood cells. There is only one subtype of IFN-γ. The Type III interferons mainly refer to the interferon 2.


Interferons, interferon mutants and interferon derivatives have been approved to be widely used in various treatments. Interferons and mutants that have been approved for human clinical treatment include interferon α 2a, interferon α 2b, interferon α 1b, compound interferon (Infergen), and interferon mutants (such as Novaferon), Interferon β, Interferon γ (1b), and the like. Interferon derivatives include Peginterferon α 2a, Peginterferon α 2b, integrated interferon and the like.


In addition, in recent years, with the discovery and elucidation of signal pathways such as TLRs, RLRs, and STING, a series of agonists that act on the above signal pathways to generate interferons have been discovered and elucidated, the use of which has also become a new direction for future interferon-based treatments. At present, this type of agonists has been used in the treatment of HBV and related tumors.


The Type I IFN is the first batch of immunotherapy drugs approved by FDA for clinical therapy of cancers. It provided the best curative effect for hematological system tumors, followed by for lymphatic system tumors, and the worst curative effect for solid tumors, such as malignant melanoma, ovarian tumor, colorectal cancer, etc., where the curative effect is no more than 20%. In addition, the combined application of interferon with chemotherapy, radiotherapy and immunotherapy is at present a common anti-tumor therapy with interferons. For example, combined application of interferon with 5-fluorouracil for treating liver cancer and colorectal cancer, combined application of interferon with cisplatin, carboplatin and the like for treating lung cancers. Although these studies have achieved some results, the overall effect is not so satisfactory, and the prognosis is poor. In addition, interferon is generally administered by continuous application of large doses in clinical practice, which inevitably causes problems such as drug-related toxicity, and poor compliance and tolerance of patients.


In summary, for application of interferon or combined application of interferon with anti-cancer or radiotherapy and other treatment methods, some progress has been made in the clinical application, but in general, such methods provide undesirable efficacy, poor prognosis, and large toxic side effects.


Therefore, it is still desirable in the art for improvement of interferon-based therapies to achieve better therapeutic effects.


SUMMARY OF THE INVENTION

In one aspect, the present invention provides a method for treating cancer in a subject, comprising

    • i) intermittently administering an interferon-based therapeutic agent for a plurality of consecutive treatment courses; and
    • ii) administering an additional anticancer agent,
    • to the subject.


In some embodiments, the interferon-based therapeutic agent comprises an interferon or a mutant or derivative thereof, or comprises a nucleic acid molecule encoding an interferon or a mutant or a derivative thereof, or comprises a substance promoting the generation of an endogenous interferon.


In some embodiments, the interferon is a Type I, Type II or Type III interferon, such as interferon α, interferon β, interferon γ or interferon λ, preferably interferon α.


In some embodiments, the interferon-based therapeutic agent comprises interferon α 2a, interferon α 2b, interferon α 1b, interferon λ, or a mutant or derivative thereof.


In some embodiments, the interferon or the mutant or derivative thereof is PEGylated.


In some embodiments, the interferon-based therapeutic agent is selected from the group consisting of P1101, Pegberon, Pegasys, Pegintron, Infergen, Novaferon, INTRONA, Roferon-A, Hapgen, PEGINFER and Peginterferon λ.


In some embodiments, the interferon-based therapeutic agent comprises an agonist of the TLRs, RLRs, and STINGs signaling pathways.


In some embodiments, the interferon-based therapeutic agent is selected from the group consisting of GS-9620, GS-9688, RO7020531, RO6864018, TQ-A3334, JNJ-4964, SB9200, MIW815, DMXAA, MK-1454, and diABZI.


In some embodiments, in the consecutive treatment course, the interferon-based therapeutic agent is administered such that during substantially the entire course, the concentration of neopterin in the subject is higher than the concentration of neopterin before the first administration, for example approximately 110%, approximately 120%, approximately 130%, approximately 140%, approximately 150%, approximately 200%, approximately 250% or higher of the neopterin concentration before the first administration.


In some embodiments, the duration of the consecutive treatment course is the time period from the first administration to the last administration, plus about 5 in vivo half-lives of the therapeutic agent.


In some embodiments, the duration of each of the plurality of consecutive treatment courses is from about 1 week to about 24 weeks, preferably from about 1 week to about 12 weeks, further preferably from about 1 week to about 8 weeks, and yet further preferably about 2 weeks to about 6 weeks.


In some embodiments, the duration of each of the consecutive treatment courses is about 1 week to about 12 weeks, and the interval between the consecutive treatment courses is about 1 week to about 12 weeks.


In some embodiments, the interval between the consecutive treatment courses is from about 1 week to about 24 weeks, preferably from about 1 week to about 12 weeks, further preferably from about 1 week to about 8 weeks, and yet further preferably about 2 weeks to about 6 weeks.


In some embodiments, the duration of each of the consecutive treatment courses is about 1 week to about 8 weeks, and the interval between the consecutive treatment courses is about 1 week to about 8 weeks.


In some embodiments, the duration of each of the consecutive treatment courses is about 2 weeks to about 6 weeks, and the interval between the consecutive treatment courses is about 2 week to about 6 weeks.


In some embodiments, the interferon-based therapeutic agent is administered for 2-25 or more consecutive treatment courses.


In some embodiments, the durations of the plurality of consecutive treatment courses are substantially the same.


In some embodiments, the intervals between the consecutive treatment courses are substantially the same.


In some embodiments, the cancer is selected from leukemia (such as acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic myeloid leukemia (CML), chronic lymphocytic leukemia, polycapillary leukemia), liver cancer, lung cancer, colorectal cancer, skin cancer, stomach cancer, breast cancer, prostate cancer, non-Hodgkin's lymphoma, melanoma, multiple myeloma, laryngeal papilloma, follicular lymphoma, AIDS-related Kaposi's sarcoma and renal cell carcinoma, preferably liver cancer, lung cancer, breast cancer, colorectal cancer or melanoma.


In some embodiments, the administration of the interferon-based therapeutic agent does not overlap with the administration of the additional anticancer agent.


In some embodiments, the additional anticancer agent is administered between the plurality of consecutive treatment courses.


In some embodiments, the administration of the interferon-based therapeutic agent overlaps with the administration of the additional anticancer agent.


In some embodiments, the additional anticancer agent is administered during and between plurality of consecutive treatment courses.


In some embodiments, the additional anticancer agent is administered according to its conventional scheme.


In some embodiments, the anticancer agent is

    • i) a chemotherapeutic agent, such as an alkylating agent an alkylating agent: Nimustine, Carmustine, Lomustine, Cyclophosphamide, Ifosfamide, glyciphosphoramide, semustine; an antimetabolite: deoxyfluoguanosine, doxifluguanidine, 5-fluorouracil, mercaptopurine, thioguanine, cytarabine, fluguanosine, tegafur, Gemcitabine, carmofur, hydroxyurea, methotrexate, UFT, Ancitabine, capecitabine; an anti-tumor antibiotic: actinomycin D, doxorubicin, daunorubicin, Epirubicin, mitomycin, pelomycin, pingyangmycin, pirarubicin; a chemotherapeutic anti-tumor animal and plant ingredient: irinotecan, harringtonine, hydroxycamptothecin, Vinorelbine, paclitaxel, albumin paclitaxel, taxotere, topotecan, vincristine, vindesine, Vindesine, vinblastine, teniposide, etoposide, elemene; such as anti-tumor drug hormones: Atamestane, Anastrozole, Aminoglutethimide, Letrozole, Formestane, Metasterone, Tamoxifen; a chemotherapeutic miscellaneous agent: asparaginase, carboplatin, cisplatin, Dacarbazine, Oxaliplatin, Loxadine, Eloxatin, Mitoxantrone, or Procarbazine; or
    • ii) an immune checkpoint inhibitor such as an inhibitor of PD-1, PD-L1, CTLA4, for example, an antibody selected from: Nivolumab, Pembrolizumab, Atezolizumab, Durvalumab, Avelumab; or
    • iii) a small molecule targeting drug, such as imatinib, gefitinib, bortezomib, erlotinib, sorafenib, lenalidomide, Sunitinib, dasatinib, nilotinib, lapatinib, pazopanib, everolimus, vandetanib, crizotinib, verofinib, ruxolitinib, axitinib, vismodegib, carfilzomib, regorafenib, bosutinib, tofacitinib, carbotinib, panatinib, pomalidomide, trametinib, dabrafenib, Afatinib, Icotinib, Ibrutinib, Ceritinib, Idelaris, Apatinib, Pabuccilib, Levatinib, Axitinib, Icotinib, Apatinib, sonidegib, cobimetinib, osimertinib, alectinib, ixazomib; or
    • iv) a tumor-associated antigen-specific antibody such as Rittman, Herceptin;
    • preferably, the anticancer agent is selected from oxaliplatin, epirubicin, paclitaxel and gemcitabine, and more preferably is gemcitabine.


In one aspect, the present invention provides a pharmaceutical combination for use in treating cancer in a subject, comprising an interferon-based therapeutic agent and an anticancer agent.


In some embodiments, the interferon-based therapeutic agent comprises an interferon or a mutant or derivative thereof, or comprises a nucleic acid molecule encoding an interferon or a mutant or a derivative thereof, or comprises a substance promoting the generation of an endogenous interferon.


In some embodiments, the interferon is a Type I, Type II or Type III interferon, such as interferon α, interferon β, interferon γ or interferon λ, preferably interferon α.


In some embodiments, the interferon-based therapeutic agent comprises interferon α 2a, interferon α 2b, interferon α 1b, interferon λ, or a mutant or derivative thereof.


In some embodiments, the interferon or the mutant or derivative thereof is PEGylated.


In some embodiments, the interferon-based therapeutic agent is selected from the group consisting of P1101, Pegberon, Pegasys, Pegintron, Infergen, Novaferon, INTRONA, Roferon-A, Hapgen, PEGINFER and Peginterferon λ.


In some embodiments, the interferon-based therapeutic agent comprises an agonist of the TLRs, RLRs, and STINGs signaling pathways.


In some embodiments, the interferon-based therapeutic agent is selected from the group consisting of GS-9620, GS-9688, R07020531, R06864018, TQ-A3334, JNJ-4964, SB9200, MIW815, DMXAA, MK-1454, and diABZI.


In some embodiments, the anticancer agent is

    • i) a chemotherapeutic agent, such as an alkylating agent an alkylating agent: Nimustine, Carmustine, Lomustine, Cyclophosphamide, Ifosfamide, glyciphosphoramide, semustine; an antimetabolite: deoxyfluoguanosine, doxifluguanidine, 5-fluorouracil, mercaptopurine, thioguanine, cytarabine, fluguanosine, tegafur, Gemcitabine, carmofur, hydroxyurea, methotrexate, UFT, Ancitabine, capecitabine; an anti-tumor antibiotic: actinomycin D, doxorubicin, daunorubicin, Epirubicin, mitomycin, pelomycin, pingyangmycin, pirarubicin; a chemotherapeutic anti-tumor animal and plant ingredient: irinotecan, harringtonine, hydroxycamptothecin, Vinorelbine, paclitaxel, albumin paclitaxel, taxotere, topotecan, vincristine, vindesine, Vindesine, vinblastine, teniposide, etoposide, elemene; such as anti-tumor drug hormones: Atamestane, Anastrozole, Aminoglutethimide, Letrozole, Formestane, Metasterone, Tamoxifen; a chemotherapeutic miscellaneous agent: asparaginase, carboplatin, cisplatin, Dacarbazine, Oxaliplatin, Loxadine, Eloxatin, Mitoxantrone, or Procarbazine; or
    • ii) an immune checkpoint inhibitor such as an inhibitor of PD-1, PD-L1, CTLA4, for example, an antibody selected from: Nivolumab, Pembrolizumab, Atezolizumab, Durvalumab, Avelumab; or
    • iii) a small molecule targeting drug, such as imatinib, gefitinib, bortezomib, erlotinib, sorafenib, lenalidomide, Sunitinib, dasatinib, nilotinib, lapatinib, pazopanib, everolimus, vandetanib, crizotinib, verofinib, ruxolitinib, axitinib, vismodegib, carfilzomib, regorafenib, bosutinib, tofacitinib, carbotinib, panatinib, pomalidomide, trametinib, dabrafenib, Afatinib, Icotinib, Ibrutinib, Ceritinib, Idelaris, Apatinib, Pabuccilib, Levatinib, Axitinib, Icotinib, Apatinib, sonidegib, cobimetinib, osimertinib, alectinib, ixazomib; or
    • iv) a tumor-associated antigen-specific antibody such as Rittman, Herceptin;
    • preferably, the anticancer agent is selected from oxaliplatin, epirubicin, paclitaxel and gemcitabine, and more preferably is gemcitabine.


In some embodiments, the pharmaceutical combination is for use in treating cancer in a subject through the method of the present invention.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows the SP Sepharose Fast Flow elution profile of mIFN-α4 fermentation supernatant.



FIG. 2 shows the non-reduced SDS-PAGE (14% separation gel, silver staining) electrophoresis results of SP Sepharose Fast Flow purified target mIFN-α4.



FIG. 3 shows the Q Sepharose Fast Flow purification elution profile of glycosyl-removed mIFN-α4.



FIG. 4 shows the non-reduced SDS-PAGE (14% separation gel, silver staining) electrophoresis results of Q Sepharose Fast Flow purified raw solution of glycosyl-removed mIFN-α4.



FIG. 5 shows the SP Sepharose Fast Flow purification elution profile of PEG-mIFN-α4.



FIG. 6 shows the non-reduced SDS-PAGE (silver staining) electrophoresis results of PEG-mIFN-α4 raw solution.



FIG. 7 shows the plasma concentration-time curve for a single subcutaneous injection of 1 μg/mouse of PEG-mIFN-α4 to BALB/c mice.



FIG. 8 shows the survival curve of comparison experiment on the efficacy of continuous and intermittent administration of PEG-mIFN-α4 in the treatment of transplanted liver cancer H22 in mice.



FIG. 9 shows comparison of efficacy of PEG-mIFN-α4 continuous administration, PEG-mIFN-α4 intermittent administration, and PEG-mIFN-α4 intermittent administration combined with gemcitabine in the treatment of transplanted liver cancer H22 in mice: survival curve (A) and mortality curve (B).



FIG. 10 shows comparison of efficacy of PEG-mIFN-α4 continuous administration, PEG-mIFN-α4 intermittent administration, and PEG-mIFN-α4 intermittent administration combined with gemcitabine in the treatment of transplanted lung cancer LLC in mice: mortality curve. A: tumor inoculation to Day 45; B: tumor inoculation to Day 64.



FIG. 11 shows comparison of efficacy of PEG-mIFN-α4 continuous administration, PEG-mIFN-α4 intermittent administration, and PEG-mIFN-α4 intermittent administration combined with gemcitabine in the treatment of transplanted colorectal cancer CT26 in mice: survival curve (A) and mortality curve (B).



FIG. 12 shows comparison of efficacy of PEG-mIFN-α4 continuous administration, PEG-mIFN-α4 intermittent administration, and PEG-mIFN-α4 intermittent administration combined with gemcitabine in the treatment of transplanted melanoma B16 in mice: mortality curve.



FIG. 13 shows comparison of efficacy of PEG-mIFN-α4 continuous administration, PEG-mIFN-α4 intermittent administration, and PEG-mIFN-α4 intermittent administration combined with gemcitabine in the treatment of transplanted breast cancer 4T1 in mice: mortality curve.



FIG. 14 shows the survival curve of intermittent administration of interferon combined with administration of anticancer agent Epirubicin in the treatment of transplanted liver cancer H22 in mice.



FIG. 15 shows the survival curve of intermittent administration of interferon combined with administration of anticancer agent Oxaliplatin in the treatment of transplanted liver cancer H22 in mice.



FIG. 16 shows the survival curve of intermittent administration of interferon combined with administration of anticancer agent Paclitaxel in the treatment of transplanted liver cancer H22 in mice.





DETAILED DESCRIPTION OF THE INVENTION

The inventors unexpectedly discovered that compared to continuous administration of interferon in a fixed course of treatment, significantly better therapeutic effects may be achieved through intermittent administration of interferon over multiple courses of treatment, for example, significantly better treatment effects with substantially same doses, with substantially same times of administration, or over a substantially same treatment period.


Without being limited by any theory, it is believed that continuous administration of interferon for a long time may cause the consumption of immune cells which are difficult to recover. The efficacy of interferon depends on the immune system. Therefore, an interferon-based therapy may achieve good therapeutic effects such as high tumor suppression in the early stage of treatment when immune cells are still sufficient. However, in the later stage of treatment, due to depletion of immune cells caused by long-term administration of the interferon, the therapeutic effect may be substantially reduced, and cannot be improved even if the interferon is continued to be administered. This problem may be avoided by intermittent administration of interferon. For example, good therapeutic efficacy may still be achieved if after administering an interferon for a period to achieve a certain therapeutic efficacy, administration of the interferon is suspended before partial immune suppression and exhaustion of immune cells for a period to allow the immune cells to recover as soon as possible, and then the administration of the interferon is resumed. On the basis of intermittent administration of interferon, administration of additional anticancer agent can further improve the therapeutical efficacy.


Therefore, in one aspect, the present invention provides a method for treating cancer in a subject, including

    • i) intermittently administering an interferon-based therapeutic agent for a plurality of consecutive treatment courses; and
    • ii) administering an additional anticancer agent,
    • to the subject.


The “interferon” may be a human interferon, for example, a Type I, II or III interferon, such as interferon α, interferon 13, interferon γ or interferon λ, preferably interferon α.


As used herein, the “interferon-based therapeutic agent” refers to a therapeutic agent capable of generating at least part of the effects of a natural interferon.


For example, the “interferon-based therapeutic agents” may include a nature isolated or a recombinantly generated interferon, such as a Type I interferon, preferably an interferon α. A suitable interferon α includes but is not limited to interferon α 2a, interferon α 2b or interferon α 1b.


The “interferon-based therapeutic agent” may also include an interferon mutant, such as Infergen (a recombinant integrated interferon).


The “interferon-based therapeutic agent” may also include an interferon derivative, such as PEGylated interferon or a mutant thereof, an albuminated interferon or a mutant thereof, and another protein and organic-modified interferon and a mutant thereof, and the like. Examples of the PEGylated modified interferon or the mutant thereof include, but are not limited to, peginterferon α 2a (e.g. Pegasys, 40 Kd bi-branched UPEG-NHS modified), peginterferon α 2b (e.g., Pegintron, 12Kd linear PEG-SC modified), peginterferon α 2b (e.g., Pegberon, Y-type 40 Kd PEG modified), cultured interferon α-2 (e.g., PEGINFER), Peginterferon λ, P1101, and the like.


In some embodiments, the “interferon-based therapeutic agent” includes a long-acting interferon, such as a PEGylated interferon or a mutant thereof. In some embodiments, the “interferon-based therapeutic agent” includes a short-acting interferon.


In some embodiments, the “interferon-based therapeutic agent” includes multiple types of interferons or mutants or derivatives thereof.


The term “interferon-based therapeutic agent” covers various therapeutic agents comprising interferons or mutants or derivatives thereof that have been approved for marketing. In some embodiments, the “interferon-based therapeutic agent” is Pegberon (Y-type 40Kd PEG modified, peginterferon α 2b, Amoytop). In some embodiments, the “interferon-based therapeutic agent” is Pegasys (peginterferon α 2a, Roche). In some embodiments, the “interferon-based therapeutic agent” is Pegintron (peginterferon α 2b injection, Schering-Plough). In some embodiments, the “interferon-based therapeutic agent” is Infergen (recombinant integrated interferon, Amgen, USA). In some embodiments, the “interferon-based therapeutic agent” is Intron A (recombinant human interferon α 2b, Schering-Plough). In some embodiments, the “interferon-based therapeutic agent” is Roferon-A (Interferon α 2a, Roche). In some embodiments, the “interferon-based therapeutic agent” is Hapgen (recombinant human interferon α 1b, Beijing Sanyuan Jiyin Engineering Co., Ltd.) In some embodiments, the “interferon-based therapeutic agent” is PEGINFER (PEGlated-integrated interferon α-2 injection, Beijing Kawin Technology Co., Ltd.). In some embodiments, the “interferon-based therapeutic agent” is peginterferon λ(Nanogen Pharmaceutical biotechbology).


In some preferred embodiments, the interferon-based therapeutic agent includes interferon α 2b. In some preferred embodiments, the interferon-based therapeutic agent includes polyethylene glycol modified interferon α 2b. In some preferred embodiments, the interferon-based therapeutic agent is Pegberon.


In some embodiments, the “interferon-based therapeutic agent” includes an interferon or a mutant or derivative thereof, where the interferon or the mutant or derivative thereof includes an amino acid sequence of any one of SEQ ID NOs: 1-5, or the interferon or the mutant or derivative thereof includes an amino acid sequence having at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to any one of SEQ ID NOs: 1-5.


In some embodiments, the “interferon-based therapeutic agent” also includes a nucleic acid molecule encoding an interferon or a mutant or derivative thereof, such as a recombinant nucleic acid expression vector. Suitable expression vectors, especially those suitable for therapeutic applications, may be easily determined by those skilled in the art.


The “interferon-based therapeutic agent” may also include a substance capable of promoting generation of endogenous interferons, such as agonists of the TLRs, RLRs, and STINGS signaling pathways. Examples of substances capable of promoting the generation of endogenous interferons include but are not limited to GS-9620, GS-9688, RO7020531, RO6864018, TQ-A3334, JNJ-4964, SB9200, MIW815, DMXAA, MK-1454, diABZI, an d the like[1]-[34].


As used herein, the term “consecutive treatment course” means that during the course of treatment, the administration of the therapeutic agent allows to continuously maintain the in vivo effective concentration of (exogenous or endogenous) interferon (for example, the effective blood concentration) within a patient, or to continuously maintain the in vivo concentration (for example, blood concentration) of neopterin (NPT), a main pharmacodynamic marker of interferons, within a patient to be higher than the concentration (initial concentration or baseline concentration) when the therapeutic agent is not administered. Since there is a good correlation between neopterin, a main pharmacodynamic marker of interferons, and the administration of interferons, it is particularly preferable to use the change in the in vivo concentration of neopterin (such as blood concentration) as the indicator of a “consecutive course of treatment”. For example, a “consecutive course of treatment” may be defined as a course of treatment during which the administration of an interferon-based therapeutic agent for one or more times may keep the concentration of neopterin in a subject during substantially the entire course of treatment higher than the concentration of neopterin before the first administration (baseline concentration), for example approximately 110%, approximately 120%, approximately 130%, approximately 140%, approximately 150%, approximately 200%, approximately 250% or higher of the neopterin concentration before the first administration. The in vivo concentration of neopterin may be determined by methods known in the art.


In the consecutive treatment course, the administration scheme of the interferon-based therapeutic agent is generally determined by the characteristics of the selected therapeutic agent, such as its half-life in vivo. For example, in the consecutive treatment course, an long-acting interferon (with an in vivo half-life of generally 30-120 hours) may be administered about once a week, about once every two weeks, or may be administered once in a month or even a longer period in the case of increased dosage; and a short-acting interferon (with an in vivo half-life of generally 2-5 hours) may be administered once a day or once every two days or for three times a week, or may be administered once a week in the case of an increased dosage (for example, 9-36 MIU or higher), or may be administered for multiple times in a day, in the case of a reduced dosage. In the consecutive treatment course, the number of administrations of the interferon-based therapeutic agent is not particularly limited, as long as the above definition of the “consecutive treatment course” is met. Those skilled in the art may determine the consecutive treatment course based on the in vivo concentration (such as the blood concentration) of a pharmacodynamic marker such as neopterin of the interferon-based therapeutic agent.


In some embodiments, in the “consecutive treatment course”, the “interferon-based therapeutic agent” may be administered in its conventional administration scheme. For example, Infergen (recombinant integrated interferon), interferon α 2b (such as Intron A), interferon α 2a (such as Roferon-A), interferon α 1b (such as Hapgen) may be administered once a day or once every two days or for three times a week in the dosage range of 3-18 MIU. Peginterferon α 2a (e.g., Pegasys), peginterferon α 2b (e.g. Pegintron or Pegberon), PEGlated-integrated interferon α-2 (such as PEGINFER) or peginterferon λ may be administered once a week in the dosage range of 45-270 μg. P1101 may be administered in about 400 μg once every two weeks. The albuminated interferon α 2b may be administered at about 900 to about 1800 μg once every two weeks, or about 1200 μg once every 4 weeks.


The duration of each one of the plurality of consecutive courses should allow the therapeutic agent to achieve a certain therapeutic effect, but should avoid excessive consumption of immune cells. The consumption of immune cells in the treatment course is usually identified by changes in treatment indicators. For example, when the relevant treatment indicator shows the worsened efficacy of the therapeutic agent, it may indicate excessive consumption of immune cells.


In some embodiments, duration of each of the plurality of consecutive treatment courses is at least about 1 week.


In some embodiments, duration of each of the plurality of consecutive treatment courses is up to about 24 weeks.


In some embodiments, the duration of each of the plurality of consecutive treatment courses is about 1 week to about 24 weeks, for example, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 9 weeks, about 10 weeks, about 11 weeks, about 12 weeks, about 13 weeks, about 14 weeks, about 15 weeks, about 16 weeks, about 17 weeks, about 18 weeks, about 19 weeks, about 20 weeks, about 21 weeks, about 22 weeks, about 23 weeks, or about 24 weeks.


In some preferred embodiments, the duration of each of the plurality of consecutive treatment courses is about 1 week to about 12 weeks. In some further preferred embodiments, the duration of each of the plurality of consecutive treatment courses is about 1 week to about 8 weeks. In some further preferred embodiments, the duration of each of the plurality of consecutive treatment courses is about 2 weeks to about 6 weeks. In some further preferred embodiments, the duration of each of the plurality of consecutive treatment courses is about 2 weeks.


In some embodiments, the end point of each consecutive treatment course (that is, the starting point of the interval between consecutive courses of treatment) may be the time of the last administration of the interferon-based therapeutic agent in the consecutive treatment course plus about 5 in vivo half-lives of the therapeutic agent. That is, the duration of the consecutive treatment course is the time period from the first administration to the last administration, plus about 5 in vivo half-lives of the therapeutic agent. It is believed that after 5 half-lives, the therapeutic agent will no longer generate a substantial therapeutic effect.


In the method of the present invention, the interval between the plurality of consecutive treatment courses may depend on the regeneration cycle of immune cells. The duration of the interval should allow the immune cells in the patient that have been reduced due to the treatment to be restored to a level that can effectively implement the treatment. It is generally believed that it takes about 1-2 weeks for the immune cells to proliferate. Therefore, the shortest interval between the plurality of consecutive treatment courses can be about 1 week.


In some embodiments, the interval between the plurality of consecutive treatment courses is at least about 1 week apart.


In some embodiments, the interval between the plurality of consecutive treatment courses is up to about 24 weeks.


In some embodiments, the interval between the plurality of consecutive treatment courses is about 1 week to about 24 weeks, for example, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 9 weeks, about 10 weeks, about 11 weeks, about 12 weeks, about 13 weeks, about 14 weeks, about 15 weeks, about 16 weeks, about 17 weeks, about 18 weeks, about 19 weeks, about 20 weeks, about 21 weeks, about 22 weeks, about 23 weeks, or about 24 weeks.


In some preferred embodiments, the interval between the plurality of consecutive treatment courses is about 1 week to about 12 weeks. In some more preferred embodiments, the interval between the plurality of consecutive treatment courses is are about 1 week to about 8 weeks. In some further preferred embodiments, the interval between the plurality of consecutive treatment courses is about 2 weeks to about 6 weeks.


In some embodiments, the duration of each of the consecutive treatment courses is about 1 week to about 24 weeks, and the interval between the treatment courses is about 1 week to about 24 weeks.


In some embodiments, the duration of each of the consecutive treatment courses is about 1 week to about 12 weeks, and the interval between the treatment courses is about 1 week to about 12 weeks.


In some embodiments, the duration of each of the consecutive treatment courses is about 1 week to about 8 weeks, and the interval between the treatment courses is about 1 week to about 8 weeks.


In some embodiments, the duration of each of the consecutive treatment courses is about 2 week to about 6 weeks, and the interval between the treatment courses is about 2 week to about 6 weeks.


In some embodiments, the duration of each of the consecutive treatment courses is about 2 weeks, and the interval between the treatment courses is about 2 weeks.


In some embodiments, the “interferon-based therapeutic agent” is administered for at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25 or more consecutive treatment courses.


In some embodiments, the durations of the plurality of consecutive treatment courses are substantially the same.


In some embodiments, the time intervals between the treatment courses are substantially the same.


In some specific embodiments, the interferon-based therapeutic agent is Pegberon, and the duration of each of the consecutive treatment courses is about 5 weeks to about 24 weeks, and the interval between the consecutive treatment courses is about 2 weeks to 8 weeks.


In some embodiments, the cancer includes, but is not limited to, leukemia (such as acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic myeloid leukemia (CML), chronic lymphocytic leukemia, polycapillary leukemia), liver cancer, lung cancer, colorectal cancer, skin cancer, stomach cancer, breast cancer, prostate cancer, non-Hodgkin's lymphoma, melanoma, multiple myeloma, laryngeal papilloma, follicular lymphoma, AIDS-related Kaposi's sarcoma, renal cell carcinoma, and the like. In some embodiments, the disease is myeloproliferative tumor (MPN). In some preferred embodiments, the disease is liver cancer, lung cancer, breast cancer, colorectal cancer or melanoma.


In some embodiments, the additional anticancer agent is a chemotherapeutic agent, including but not limited to, such as an alkylating agent: Nimustine, Carmustine, Lomustine, Cyclophosphamide, Ifosfamide, glyciphosphoramide, semustine; such as a chemotherapeutic antimetabolite: deoxyfluoguanosine, doxifluguanidine, 5-fluorouracil, mercaptopurine, thioguanine, cytarabine, fluguanosine, tegafur, Gemcitabine, carmofur, hydroxyurea, methotrexate, UFT, Ancitabine, capecitabine; such as a chemotherapeutic anti-tumor antibiotic: actinomycin D, doxorubicin, daunorubicin, Epirubicin, mitomycin, pelomycin, pingyangmycin, pirarubicin; such as a chemotherapeutic anti-tumor animal and plant ingredient: irinotecan, harringtonine, hydroxycamptothecin, Vinorelbine, paclitaxel, albumin paclitaxel, taxotere, topotecan, vincristine, vindesine, Vindesine, vinblastine, teniposide, etoposide, elemene; such as anti-tumor drug hormones: Atamestane, Anastrozole, Aminoglutethimide, Letrozole, Formestane, Metasterone, Tamoxifen; such as a chemotherapeutic miscellaneous agent: asparaginase, carboplatin, cisplatin, Dacarbazine, Oxaliplatin, Loxadine, Eloxatin, Mitoxantrone, or Procarbazine.


In some embodiments, the additional anticancer agent is a small molecule targeting drug, including but not limited to imatinib, gefitinib, bortezomib, erlotinib, sorafenib, lenalidomide, Sunitinib, dasatinib, nilotinib, lapatinib, pazopanib, everolimus, vandetanib, crizotinib, verofinib, ruxolitinib, axitinib, vismodegib, carfilzomib, regorafenib, bosutinib, tofacitinib, carbotinib, panatinib, pomalidomide, trametinib, dabrafenib, Afatinib, Icotinib, Ibrutinib, Ceritinib, Idelaris, Apatinib, Pabuccilib, Levatinib, Axitinib, Icotinib, Apatinib, sonidegib, cobimetinib, osimertinib, alectinib, ixazomib.


The additional anticancer agent may also be a tumor-associated antigen-specific antibody such as Rittman, Herceptin and the like.


The additional anticancer agent may also be an immune checkpoint inhibitor, such as an inhibitor of PD1, PDL1, CTLA4, etc., such as a specific antibody. Examples of an immune checkpoint inhibitor include, but are not limited to, Nivolumab, Pembrolizumab, Atezolizumab, Durvalumab, Avelumab and the like.


In some specific embodiments, the chemotherapeutic agent is oxaliplatin. In some specific embodiments, the chemotherapeutic agent is epirubicin. In some specific embodiments, the chemotherapeutic agent is paclitaxel.


In some preferred embodiments, the chemotherapeutic agent is an antimetabolitic chemotherapeutic agent, such as gemcitabine, capecitabine, and ancitabine. In some most preferred embodiments, the chemotherapeutic agent is gemcitabine.


In some embodiments, the administration of the “interferon-based therapeutic agent” does not overlap with the administration of the additional anticancer agent.


In some embodiments, the additional anticancer agent is administered between the plurality of consecutive treatment courses. For example, the additional anticancer agent may be administered for a period of time during the administration interval of the “interferon-based therapeutic agent”, said period of time shorter than or equal to the interval.


In some embodiments, the administration of the “interferon-based therapeutic agent” overlaps with the administration of the additional anticancer agent.


In some embodiments, the additional anticancer agent is administered during and between the plurality of consecutive treatment courses, that is, the additional anticancer agent is administered during the entire treatment period (completely overlapped).


In some embodiments, the additional anticancer agent is administered according to a conventional scheme, for example, its conventional administration scheme for the specific cancer being treated.


In some embodiments, the additional anticancer agent is gemcitabine, and the administration scheme of gemcitabine can be three times every four weeks: once a week for three weeks, and then stop for one week; twice every three weeks: once a week for two weeks, and then stop for one week; or once every two weeks. Exemplary dose of gemcitabine is 1000 mg/m2 surface area.


In some embodiments, the method of the invention results in tumor regression or reduction in tumor volume or prolonged survival of the subject. In particular, the method of the present invention results in: reduction of the number of cancer cells, reduction of tumor volume, inhibition (i.e., slow-down or stop) of the infiltration of cancer cells into peripheral organs, inhibition (i.e., slow-down or stop) of tumor metastasis, inhibition of tumor growth, and/or alleviation of one or more symptoms associated with the cancer.


In one aspect, the present invention provides a pharmaceutical composition comprising an interferon-based therapeutic agent for use in the treatment of cancer in a subject by a method of the present invention. In some embodiments, the pharmaceutical composition also comprises an additional anticancer agent. The interferon-based therapeutic agent and the anticancer agent are as defined above.


In one aspect, the present invention provides the use of an interferon-based therapeutic agent in preparation of a pharmaceutical composition for treating cancer in a subject by a method of the present invention. In some embodiments, the pharmaceutical composition also comprises an additional anticancer agent. The interferon-based therapeutic agent and the anticancer agent are as defined above.


In one aspect, the present invention provides the use of an interferon-based therapeutic agent in preparation of a pharmaceutical composition for treating cancer in a subject in combination with an additional anticancer agent. The interferon-based therapeutic agent and the anticancer agent are as defined above. The interferon-based therapeutic agent and/or the additional anticancer agent may be administered according to the method of the present invention.


In one aspect, the present invention provides the use of an interferon-based therapeutic agent in preparation of a pharmaceutical composition for enhancing the efficacy of an additional anticancer agent. The interferon-based therapeutic agent and the anticancer agent are as defined above. The interferon-based therapeutic agent and/or the additional anticancer agent may be administered according to the method of the present invention.


In one aspect, the present invention provides a pharmaceutical combination comprising an interferon-based therapeutic agent and an additional anticancer agent for use in treating cancer in a subject through the method of the present invention. The interferon-based therapeutic agent and/or the additional anticancer agent may be administered according to the method of the present invention.


In one aspect, the present invention provides a kit including an interferon-based therapeutic agent, an additional anticancer agent, and instructions for use, where the instructions for use provide a description that the interferon-based therapeutic agent and the an additional anticancer agent may be used according to the method of the present invention or be administered according to the method of the present invention to treat cancer in a subject. The interferon-based therapeutic agent and the additional anticancer agent are as defined above.


EXAMPLES

The following examples are only provided for better explaining the present invention and are not intended to limit the present invention in any way.


Example 1. Preparation of Recombinant Mouse Interferon α4 (mIFN-α4)

Interferons are also widely used in cancer treatment, but the effect needs to be improved. In order to study whether the continuous administration of interferon actually causes partial immune suppression and exhaustion of immune cells, which leads to poor subsequent treatment effects, the efficacy of interferon intermittent administration and continuous administration was compared in mice.


A large number of studies on anti-tumor effects were carried out in nude mice (lack of normal thymus) as the host. As the immune status of nude mice is weaker than that of normal mice, it is difficult to reflect the immune response under the normal state. In the anti-tumor model based on normal mice, the effect of interferon on the immune system may be partially realized, which reflects the effect of the anti-tumor therapy of administering the interferon. From the perspective of function realization, interferon should rely on the host's immune system to play its full anti-viral and anti-tumor effects. Because interferon has strong species specificity, when normal mice were used as the research subjects in the examples, the use of murine interferon or derivatives thereof may better reflect its physiological effects and implementation effects. In the research model using the mice of the present invention, PEGylated recombinant mouse interferon α4 (PEG-mIFNα4) was used as a representative of interferon therapeutic drugs and their derivatives.


According to the amino acid sequence of mIFN-α4 (GenBank NP_034634), the cDNA sequence of mIFN-α4 was optimized and designed according to the preferred codons of Pichia pastoris, and GenScript Biotechnology Co., Ltd. was entrusted to synthesize the cDNA. The cDNA encoding mIFN-α4 was recombined into pPIC9K plasmid, transformed into TOP10 competent cells, plated on LB solid medium, and cultured overnight at 37° C. A single clone was picked to inoculate the LB liquid medium, which was then cultured overnight at 37° C. The plasmid was extracted, and double digestion was performed with XhoI and NotI. Positive clones were identified by nucleic acid electrophoresis, and further confirmed by nucleic acid sequencing. The positive clone plasmid was digested and linearized with SalI, electrotransformed into Pichia pastoris GS115, plated on the RD plate, and cultured at 28-30° C. for 3 days. The positive transformants were picked to inoculate YPD liquid medium, which was cultured overnight at 28-30° C., and transfered to BMMY medium at a final concentration of OD600 nm of about 1, and incubated at 28-30° C. for about 24 hours. Methanol was added until the final concentration of methanol of about 1%, and then further cultured at 28-30° C. for about 24 hours. The culture medium was centrifuged to collect the supernatant, and the expression of mIFN-α4 was detected by SDS-PAGE electrophoresis. According to the results of SDS-PAGE electrophoresis, the engineered strains with higher expression and stable expression were selected for subsequent fermentation in a fermenter.


The fermenter was 30 L. Refer to the “Pichia Fermentation Process Guidelines” for fermentation culture and methanol induction, the induction time was about 30 h. The fermentation supernatant was collected by centrifugation, and concentrated by ultrafiltration for 3-5 times in 5 kD hollow fiber membrane tube, and the buffer system was replaced with 20 mM phosphate buffer-20 mM arginine hydrochloride-50 mM sodium chloride buffer solution (pH6.5). SP Sepharose Fast Flow chromatography column (GE Healthcare, column bed Φ38 mm×160 mm) was then loaded, 20 mM phosphate buffer −20 mM arginine hydrochloride (pH6.5) (solution A) was then used to wash about 3 column volumes. The solution A and 20 mM phosphate buffer −20 mM arginine hydrochloride −1M sodium chloride solution (pH6.5) (solution B) were used to perform gradient elution. mIFN-α4 target samples were collected, and sample for non-reducing SDS-PAGE (with the separation gel concentration of 14%, silver staining) were taken. The elution profile is shown in FIG. 1, and the electrophoresis result is shown in FIG. 2.


mIFN-α4 SP Sepharose Fast Flow purification sample was concentrated by ultrafiltration with 5K ultrafiltration membrane package and replaced with 20 mM phosphate buffer-50 mM arginine hydrochloride-10 mM methionine-20 mM sodium chloride (pH 7.0), and then its concentration was adjusted to about 1.0 mg/ml. Glycosidase was added at a mass ratio of mIFN-α4 protein to enzyme of about 20:1, and digestion was performed at 25° C. for about 20 hours to remove glycosyl groups. The digested sample was diluted by about 6 times with 5 mM boric acid buffer-10 mM arginine hydrochloride (pH9.0), and the sample was loaded on Q Sepharose Fast Flow chromatography column (GE Healthcare, column bed Φ50 mm×154 mm). 20 mM boric acid buffer −20 mM arginine hydrochloride-10 mM methionine (pH 9.0) (solution C) was used to wash about 3 column volumes. The solution C and 20 mM boric acid buffer −20 mM arginine hydrochloride-10 mM formazan thionine-0.3M sodium chloride (pH 9.0) (solution D) were used for gradient elution. The target deglycosylated mIFN-α4 sample was collected, and the pH was adjusted to about pH 5.0 with 10% acetic acid. 5K ultrafiltration membrane was used to concentrate by ultrafiltration and the buffer was replaced with 5 mM acetic acid/sodium acetate buffer-50 mM arginine hydrochloride-100 mM sodium chloride (pH 5.0). The resulting sample was the stock solution of deglycosylated mIFN-α4. Samples were taken to be sent for inspection, and the remaining samples were frozen at −70° C. for later use. The elution profile is shown in FIG. 3, and the SDS-PAGE electrophoresis result is shown in FIG. 4.


The bacterial endotoxin content of the deglycosylated mIFN-α4 stock solution was determined by the Limulus reagent method as lower than 60 EU/mg. specific activity was determined as 5.4×108 U/mg by using the commercial mIFN-α4 (R&D, catalog number 12115-1) as the standard, and by using the mouse fibroblast/encephalomyocarditis virus (L929/EMCV) cytopathic inhibition method.


Example 2. Preparation of PEGed Recombinant Mouse Interferon (PEG-mIFN-α4)

The deglycosylated mIFN-α4 stock solution was replaced through ultrafiltration with buffer of 5 mM acetic acid/sodium acetate buffer-50 mM sodium chloride (pH5.0) with 5 kD ultrafiltration membrane package. About 333 ml sample (with deglycosylated mIFNα4 content of about 500 mg) was taken, added with about 22 ml of 0.8M boric acid/sodium hydroxide buffer (pH 9.4), and stirred well. YPEG-NHS was added based on the mass ratio of protein to 40 kD Y-type polyethylene glycol succinimide ester (YPEG-NHS) of about 1:8, which was then stirred quickly, and reacted at room temperature for 10 minutes. Then about 20 ml of 0.2M methionine was added to stop the reaction, and the pH was adjusted to 5.0 with 10% acetic acid. 550 ml pure water was then added, and then 600 ml 20 mM acetic acid/sodium acetate buffer-20 mM arginine hydrochloride-10 mM methionine (pH5.1) (solution E), mixed well. After that, it was loaded on the SP Sepharose Fast Flow chromatography column (GE Healthcare, column bed 050 mm×194 mm), and the solution E was used to wash about 5 column volumes. After that, the solution E and 20 mM acetic acid/sodium acetate buffer-20 mM arginine hydrochloride-10 mM methionine-600 mM Sodium chloride (pH5.1) (solution F) were used for gradient elution, and PEG-mIFN-α4 target samples were collected. The buffer was replaced through ultrafiltration by using the 5 kD ultrafiltration membrane package to 20 mM phosphate buffer-123 mM sodium chloride (pH 6.5). and then concentrated appropriately, added with 0.5% Tween80 to the final concentration of Tween80 being about 0.005%. The resulting sample was the PEG-mIFN-α4 stock solution (PEG-mIFN-α4). Samples were taken and then sent for inspection, the remaining samples were frozen and stored at −70° C. for later use. The elution profile is shown in FIG. 5, and the SDS-PAGE electrophoresis result is shown in FIG. 6.


The bacterial endotoxin content of the PEG-mIFN-α4 stock solution was determined by the Limulus reagent method as lower than 15 EU/mg. Specific activity was determined as 6.1×106 U/mg by using the commercial mIFN-α4 (R&D, catalog number 12115-1) as the standard, and by using the mouse fibroblast/encephalomyocarditis virus (L929/EMCV) cytopathic inhibition method.


Example 3. Pharmacokinetic Study of Subcutaneous Injection of PEG-mIFN-α4 in Healthy BALB/c Mice

In this example, BALB/c mice were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd. The blood concentration of PEG-mIFN-α4 was detected by double-antibody sandwich ELISA. The test kit of the Mouse IFN alpha Platinum ELISA kit (Cat. No. BMS6027/BMS6027TEN, Thermo) was used, but the reporter antibody was replaced with the anti-PEG antibody 3.3 biotin (Institute of Biomedical Sciences, Academia Sinica (IBMS), Taiwan) to avoid interference of endogenous mIFN-α on the measurement result.


6-8 weeks old, SPF grade BALB/c mice (N=60, with fifty-fifty of male and female mice) were injected with a single injection of 1 μg/mouse PEG-mIFN-α4 subcutaneously on the back of the neck. Plasma samples were collected before the injection (0 h) and after the injection at 15 h, 24 h, 36 h, 48 h, 72 h, 96 h, 120 h, 168 h, 216 h, 264 h, 312 h, to determine the blood concentration. Phoenix WinNonlin 6.4 software was used to calculate the pharmacokinetic parameters.


The results of blood concentration determination are shown in Table 1, the blood concentration-administration time curve is shown in FIG. 7, and the results of pharmacokinetic parameters are shown in Table 2. With a single injection of 1 μg/mouse of PEG-mIFN-α4 subcutaneously on the back of the neck of BALB/c mice, plasma concentration peak time (Tmax) was 24 h, peak concentration time (Tmax) was 268 ng/ml, and elimination half-life (T1/2) was 28.3 h.









TABLE 1







Results of blood concentration determination of


BALB/c mice with a single subcutaneous injection


of 1 μg/mouse of PEG-mIFN-α4 (N = 5, ng/ml)











Individual blood concentration (ng/ml)
Mean















Collected at
1
2
3
4
5
(ng/ml)
SD


















0
h
0
0
0
0
0
0.0
0.0


6
h
218.1
165.9
167.8
220.8
200.6
194.6
26.5


15
h
282.7
326.1
194.3
203.8
183.2
238.0
62.9


24
h
341.5
290.8
362.8
185.3
159.7
268.0
91.5


36
h
204.8
263.1
145.4
60.6
61.4
147.1
88.9


48
h
205.8
128.1
133.3
79
91.4
127.5
49.5


72
h
95.5
73.6
40.4
52.9
54.6
63.4
21.5


96
h
41.2
52.6
87.5
20.6
22.2
44.8
27.4


120
h
22.6
26.1
19.1
11.5
10.6
18.0
6.8


168
h
BLQ
BLQ
BLQ
BLQ
BLQ
BLQ
/


216
h
BLQ
BLQ
BLQ
BLQ
BLQ
BLQ
/


264
h
BLQ
BLQ
BLQ
BLQ
BLQ
BLQ
/


312
h
BLQ
BLQ
BLQ
BLQ
BLQ
BLQ
/





Note:


In the pharmacokinetic analysis, for samples with blood concentration lower than the quantification lower limit, the sample before the peak blood concentration was counted as 0, and the sample after the peak was expressed as BLQ.













TABLE 2







Summary of pharmacokinetic parameters of a single subcutaneous


injection of 1 μg/mouse of PEG-mIFN-α4 in BALB/c mice











Parameter
Unit
PEG-mIFNα4















T1/2
hr
28.3



Tmax
hr
24.0



Cmax
ng/ml
268.0



Tlast
hr
120.0



AUClast
hr*ng/ml
13289.3



AUCINFobs
hr*ng/ml
14023.2



Vz_Fobs
ml
2.91



Cl_Fobs
ml/hr
0.07










Example 4. Comparative Study on the Treatment Efficacy of PEG-mIFN-α4 Intermittent Administration and Continuous Administration for Transplanted Liver Cancer H22 in Mice

In this example, BALB/c mice were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd., and the mouse liver cancer H22 cell line was purchased from the China Center for Type Culture Collection (CCTCC).


6-8 weeks old, 18-22 g SPF grade BALB/c mice were subcutaneously inoculated with H22 cells (0.5×106 cells/mL, 0.2 ml/mouse) under the armpit of the right forelimb. Randomized grouping was made on the day of tumor inoculation. Experimental groups included PEG-mIFN-α4 continuous administration group (N=15, 7 males and 8 females), PEG-mIFN-α4 intermittent administration group (N=15, 7 males and 8 females) and normal saline control group (N=10, 5 females and 5 males) and so on.


PEG-mIFN-α4 was administered by subcutaneous injection at the back of the neck, at a dosage of 1 microgram per animal. The PEG-mIFN-α4 intermittent administration group was administered once every 48 hours for 3 times, then suspended for 144 hours, repeated for 4 rounds. The PEG-mIFN-α4 continuous administration group was administered once every 48 hours, continuously administered for 21 times. The normal saline control group was given an equal volume of normal saline.


All groups were subjected to survival analysis to compare differences between groups. SAS 9.4 and office2010 software were used for statistical analysis, and the statistical tests were all two-sided tests.


The survival curve is shown in FIG. 8, and the statistical comparison results are shown in Table 3. On Day 40 after tumor inoculation, the median survival time of the normal saline control group was 19.5 days, and that of the PEG-mIFN-α4 continuous administration group was 38 days. The survival rate of the PEG-mIFN-α4 intermittent administration group was 93.3%, while the median survival time cannot be calculated yet. The survival curves of the PEG-mIFN-α4 continuous administration group and the intermittent administration group were significantly different from the normal saline control group. The difference between the PEG-mIFN-α4 continuous administration group and the intermittent administration group was also significant (P=0.0035). The survival period of the PEG-mIFN-α4 intermittent administration group was significantly longer than that of the continuous administration group. For the treatment of transplanted liver cancer H22 in mice, intermittent administration of PEG-mIFN-α4 shows significantly better curative effect than continuous administration.









TABLE 3







Survival analysis results of comparative experiment on the efficacy


of continuous and intermittent administration of PEG-mIFN-α4 in the


treatment of transplanted liver cancer H22 in mice











Number
Median survival




of cases
period
Survival curve



(Female +
(Time after tumor
comparison











Group
male)
inoculation, days)
P value1
P value2





Normal saline
5 + 5
19.5
/
/


control group


PEG-mIFN-α4
7 + 8
Survival rate during
<0.0001*
/


intermittent

the observation period


administration

was 93.3%.


group


PEG-mIFN-α4
7 + 8
38.0
 0.0137*
0.0035*


continuous


administration


group





Note:



1Compared with the “normal saline control group”.




2Compared with “PEG-mIFN-α4 intermittent administration group”.



3. *Statistically significant difference.






Example 5. Intermittent Administration of Interferon Combined with Gemcitabine to Treat Liver Cancer H22 in Mice

The inventors studied the antitumor effect of intermittent administration of PEGylated recombinant mouse interferon α (PEG-mIFN-α4) combined with gemcitabine on mouse liver cancer H22, and explored the efficacy of combined administration of interferon and gemcitabine. The inventors surprisingly found that by intermittently administering PEG-mIFN-α4 and simultaneously administering gemcitabine, significantly better results may be obtained in the treatment of H22 tumors, compared to administering gemcitabine alone or intermittently administering PEG-mIFN-α4 alone.


In this example, a comparative study on the efficacy of PEG-mIFN-α4 intermittent administration (once every 48 hours for three consecutive times, then drug withdrawal for 144 hours; for 8 rounds), PEG-mIFN-α4 continuous administration (once every 48 hours for 42 consecutive times), PEG-mIFN-α4 in combination with gemcitabine (PEG-mIFN-α4: once every 48 hours for three consecutive times, then drug withdrawal for 240 hours, 6 rounds; gemcitabine: once a week) on H22 transplanted liver cancer in mice was performed.


In this example, BALB/c mice were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd., and the H22 mouse liver cancer cell line was purchased from China Center for Type Culture Collection (CCTCC).


6-8 weeks old, 18-22 g SPF grade BALB/c mice were subcutaneously inoculated with hepatocellular carcinoma H22 tumor cells (1×106 cells/mL, 0.2 ml/mouse) in the right forelimb axillary. On the day when the tumor was inoculated, they were randomly divided into PEG-mIFN-α4 continuous administration group (N=24, female 12, male 12), PEG-mIFN-α4 intermittent administration group (N=24, female 12, male 12), the normal saline control group (N=24, female 12, male 12), the gemcitabine only group (N=24, female 12, male 12), and PEG-mIFN-α4 combined with gemcitabine group (N=24, female 12, male 12).


The PEG-mIFN-α4 was injected subcutaneously on the back of the neck, 1 μg/mouse. Gemcitabine was injected intraperitoneally, 60 mg/kg. The administration was started on the day after grouping.


For the PEG-mIFN-α4 intermittent administration group, PEG-mIFN-α4 was administered once every 48 hours for three consecutive times, then drug withdrawal for 144 hours; for 8 rounds. For the PEG-mIFN-α4 continuous administration group, PEG-mIFN-α4 was administered once every 48 hours. For the PEG-mIFN-α4 combined with gemcitabine group, PEG-mIFN-α4 was administered once every 48 hours for three consecutive times, then drug withdrawal for 240 hours; for 6 rounds; and gemcitabine was administered once a week. For the gemcitabine only group, gemcitabine was administered once a week. For the saline control group, equal volume of saline was administered.


Survival analysis was performed for all groups to compare the differences between groups. SAS 9.4 and office2010 software were used for statistical analysis, and the statistical tests were all two-sided tests.


The survival curve is shown in FIG. 9, and the statistical comparison results are shown in Table 4. 108 days after tumor inoculation, the median survival period of the PEG-mIFN-α4 continuous administration group was 37.0 days, and that of the PEG-mIFN-α4 intermittent administration group was 64.0 days, both of which were greater than the 17.5 days of the normal saline control group. There was a significant difference between the survival curves of the PEG-mIFN-α4 continuous and intermittent administration groups (P=0.0245), and the survival period of PEG-mIFN-α4 intermittent administration group was significantly longer than that of PEG-mIFN-α4 continuous administration group. For the treatment of transplanted liver cancer H22 in mice, PEG-mIFN-α4 intermittent administration showed significantly improved effect than continuous administration.


The median survival period of gemcitabine only group was 58.5 days, and the survival rate at the end of the observation period (108 days after tumor inoculation) in the PEG-mIFN-α4 intermittent administration combined with gemcitabine group was 75.0%. The survival curves of the two groups were significantly different (P<0.001). The survival period of the PEG-mIFN-α4 intermittent administration combined with gemcitabine group was significantly longer than that of the gemcitabine only group.


The median survival period of PEG-mIFN-α4 intermittent administration group was 64.0 days, and the survival rate at the end of the observation period (108 days after tumor inoculation) in the PEG-mIFN-α4 intermittent administration combined with gemcitabine group was 75.0%. The survival curves of the two groups were significantly different (P<0.001). The survival period of the PEG-mIFN-α4 intermittent administration combined with gemcitabine group was significantly longer than that of the PEG-mIFN-α4 intermittent administration group.


The results showed that for the treatment of H22 transplanted liver cancer in mice, intermittent administration of pegylated mouse interferon α was better than continuous administration of pegylated mouse interferon α; the anticancer activity of intermittent administration of pegylated mouse interferon α combined with gemcitabine was significantly better than that of gemcitabine alone and pegylated mouse interferon α alone.









TABLE 4







Comparison of efficacy of PEG-mIFN-α4 continuous administration,


PEG-mIFN-α4 intermittent administration, and PEG-


mIFN-α4 intermittent administration combined with


gemcitabine in the treatment of transplanted liver cancer


H22 in mice: survival analysis results











Number of
Mean survival




cases
period
Inter-group



(Female +
(days post tumor
comparison











Group
male)
inoculation)
P value1
P value2





Normal saline
12 + 12
17.5
/
/


control


PEG-mIFN-α4
12 + 12
64.0
/
<0.0001*


intermittent


administration


PEG-mIFN-α4
12 + 12
37.0
0.0245*
/


continuous


administration


Gemcitabine
12 + 12
58.5
/
<0.0001*


PEG-mIFN-α4
12 + 12
75% within the
/
/


intermittent

observation


administration +

period


Gemcitabine





Note:



1compared with “PEG-mIFN-α4 intermittent administration group”;




2compared with “PEG-mIFN-α4 intermittent administration + Gemcitabine group”; and



3. *statistically different.






Example 6. Intermittent Administration of Interferon Combined with Gemcitabine to Treat Lung Cancer in Mice

The inventors studied the antitumor effect of intermittent administration of PEGylated recombinant mouse interferon α (PEG-mIFN-α4) combined with gemcitabine on mouse lung cancer LLC, and explored the efficacy of combined administration of interferon and gemcitabine. The inventors found that by intermittently administering PEG-mIFN-α4 and administering gemcitabine, significantly better results may be obtained in the treatment of lung cancer LLC, compared to administering gemcitabine alone or intermittently administering PEG-mIFN-α4 alone.


In this example, a comparative study on the efficacy of PEG-mIFN-α4 intermittent administration (once every 48 hours for four consecutive times, then drug withdrawal for 192 hours; for 5 rounds), PEG-mIFN-α4 continuous administration (once every 48 hours for 32 consecutive times), PEG-mIFN-α4 in combination with gemcitabine (PEG-mIFN-α4: once every 48 hours for four consecutive times, then drug withdrawal for 360 hours, 4 rounds; gemcitabine: once a week) on transplanted liver cancer LLC in mice was performed.


In this example, C57BL/6N mice were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd., and the mouse lung cancer LLC cell line was purchased from Beijing Union Cell Resource Center, China.


6-8 weeks old, 18-22 g SPF grade BALB/c mice were subcutaneously inoculated with lung cancer LLC cells (1×106 cells/mL, 0.2 ml/mouse) in the right forelimb axillary. On the day when the tumor was inoculated, they were randomly divided into PEG-mIFN-α4 continuous administration group (N=26, female 13, male 13), PEG-mIFN-α4 intermittent administration group (N=26, female 13, male 13), the normal saline control group (N=26, female 13, male 13), the gemcitabine only group (N=26, female 13, male 13), and PEG-mIFN-α4 combined with gemcitabine group (N=26, female 13, male 13).


The PEG-mIFN-α4 was injected subcutaneously on the back of the neck, 1 μg/mouse. The administration was started on the day after grouping. For the PEG-mIFN-α4 intermittent administration group, PEG-mIFN-α4 was administered once every 48 hours for 4 consecutive times, then drug withdrawal for 192 hours; for 8 rounds. For the PEG-mIFN-α4 continuous administration group, PEG-mIFN-α4 was administered once every 48 hours. For the PEG-mIFN-α4 combined with gemcitabine group, PEG-mIFN-α4 was administered once every 48 hours for 4 consecutive times, then drug withdrawal for 360 hours; for 4 rounds; and gemcitabine was administered once a week. For the gemcitabine only group, gemcitabine was administered once a week. For the saline control group, equal volume of saline was administered. Gemcitabine was injected intraperitoneally, 60 mg/kg.


SAS9.4 software was used for comparing the difference in mortality, and the statistical tests were all two-sided tests.


The mortality is shown in FIG. 10, and the statistical comparison results are shown in Table 5. 45 days after tumor inoculation, the mortality of the PEG-mIFN-α4 continuous administration group was 100%, and that of the PEG-mIFN-α4 intermittent administration group was 34.6%, and the difference was significant (P<0.001). The mortality of PEG-mIFN-α4 intermittent administration group was significantly lower than that of PEG-mIFN-α4 continuous administration group. For the treatment of transplanted lung cancer LLC in mice, PEG-mIFN-α4 intermittent administration showed significantly improved effect than continuous administration.


64 days after tumor inoculation, the mortality of gemcitabine only group was 80.8%, and that of the PEG-mIFN-α4 intermittent administration combined with gemcitabine group was 53.8%, and the difference was significant (P=0.0385). The mortality of PEG-mIFN-α4 intermittent administration combined with gemcitabine group was significantly lower than that of gemcitabine only group.


64 days after tumor inoculation, the mortality of PEG-mIFN-α4 intermittent administration only group was 84.6%, and that of the PEG-mIFN-α4 intermittent administration combined with gemcitabine group was 53.8%, and the difference was significant (P=0.0162). The mortality of PEG-mIFN-α4 intermittent administration combined with gemcitabine group was significantly lower than that of PEG-mIFN-α4 intermittent administration only group.


The results showed that for the treatment of transplanted lung cancer LLC in mice, intermittent administration of pegylated mouse interferon α was better than continuous administration of pegylated mouse interferon α; the anticancer activity of intermittent administration of pegylated mouse interferon α combined with gemcitabine was significantly better than that of gemcitabine alone and pegylated mouse interferon α alone.









TABLE 5







Comparison of efficacy of PEG-mIFN-α4 continuous administration,


PEG-mIFN-α4 intermittent administration, and PEG-mIFN-


α4 intermittent administration combined with gemcitabine


in the treatment of transplanted lung cancer LLC in mice:


mortality analysis results (Day 64 post tumor inoculation)










Number of












cases

Inter-group



(Female +

comparison











Group
male)
Mortality
P value1
P value2





Normal saline
13 + 13
 100%
/
/


control


PEG-mIFN-α4
13 + 13
84.6%
/
0.0162*


intermittent


administration


PEG-mIFN-α4
13 + 13
 100%
<0.0001*
/


continuous


administration


Gemcitabine
13 + 13
80.8%
/
0.0385*


PEG-mIFN-α4
13 + 13
53.8%
/
/


intermittent


administration +


Gemcitabine





Note:



1compared with “PEG-mIFN-α4 intermittent administration group” on Day 45 post tumor inoculation;




2compared with “PEG-mIFN-α4 intermittent administration + Gemcitabine group” on Day 64 post tumor inoculation; and



3. *statistically different.






Example 7. Intermittent Administration of Interferon Combined with Gemcitabine to Treat Colorectal Cancer

The inventors studied the antitumor effect of intermittent administration of PEGylated recombinant mouse interferon α (PEG-mIFN-α4) combined with gemcitabine on mouse colorectal cancer CT26, and explored the efficacy of combined administration of interferon and gemcitabine. The inventors surprisingly found that by intermittently administering PEG-mIFN-α4 and simultaneously administering gemcitabine, significantly better results may be obtained in the treatment of mouse colorectal cancer CT26, compared to administering gemcitabine alone or intermittently administering PEG-mIFN-α4 alone.


In this example, a comparative study on the efficacy of PEG-mIFN-α4 intermittent administration (once every 48 hours for three consecutive times, then drug withdrawal for 144 hours; for 11 rounds), PEG-mIFN-α4 continuous administration (once every 48 hours for 55 consecutive times), PEG-mIFN-α4 in combination with gemcitabine (PEG-mIFN-α4: once every 48 hours for three consecutive times, then drug withdrawal for 240 hours, 8 rounds; gemcitabine: once a week) on transplanted colorectal cancer CT26 in mice was performed.


In this example, BALB/c mice were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd., and CT26 mouse colorectal cancer cell line was purchased from the Center for Cell Resources, Shanghai Academy of Biological Sciences, Chinese Academy of Sciences. The anticancer agent is Gemcitabine (Jiangsu HAOSEN Pharmaceutical).


6-8 weeks old, 18-22 g SPF grade BALB/c mice were subcutaneously inoculated with colorectal cancer CT26 tumor cells (1×106 cells/mL, 0.2 ml/mouse) in the right forelimb axillary. On the day when the tumor was inoculated, they were randomly divided into PEG-mIFN-α4 continuous administration group (N=24, female 12, male 12), PEG-mIFN-α4 intermittent administration group (N=24, female 12, male 12), the normal saline control group (N=24, female 12, male 12), the gemcitabine only group (N=24, female 12, male 12), and PEG-mIFN-α4 combined with gemcitabine group (N=24, female 12, male 12).


The PEG-mIFN-α4 was injected subcutaneously on the back of the neck, 1 μg/mouse. Gemcitabine was injected intraperitoneally, 60 mg/kg. The administration was started on the day after grouping. For the PEG-mIFN-α4 intermittent administration group, PEG-mIFN-α4 was administered once every 48 hours for three consecutive times, then drug withdrawal for 144 hours; for 11 rounds. For the PEG-mIFN-α4 continuous administration group, PEG-mIFN-α4 was administered once every 48 hours. For the PEG-mIFN-α4 combined with gemcitabine group, PEG-mIFN-α4 was administered once every 48 hours for three consecutive times, then drug withdrawal for 240 hours; for 8 rounds; and gemcitabine was administered once a week. For the gemcitabine only group, gemcitabine was administered once a week. For the saline control group, equal volume of saline was administered.


Survival analysis was performed for all groups to compare the differences between groups. SAS 9.4 and office2010 software were used for statistical analysis, and the statistical tests were all two-sided tests.


The survival curve is shown in FIG. 11, and the statistical comparison results are shown in Table 6. 110 days after tumor inoculation, the median survival period of the PEG-mIFN-α4 continuous administration group was 53.5 days, and that of the PEG-mIFN-α4 intermittent administration group was 89.0 days, both of which were greater than the 49.5 days of the normal saline control group. There was a significant difference between the survival curves of the PEG-mIFN-α4 continuous and intermittent administration groups (P=0.0150), and the survival period of PEG-mIFN-α4 intermittent administration group was significantly longer than that of PEG-mIFN-α4 continuous administration group. For the treatment of transplanted colorectal cancer CT26 in mice, PEG-mIFN-α4 intermittent administration showed significantly improved effect than continuous administration.


The median survival period of gemcitabine only group was 89.0 days, and the survival rate at the end of the observation period (110 days after tumor inoculation) in the PEG-mIFN-α4 intermittent administration combined with gemcitabine group was 75.0%. The survival curves of the two groups were significantly different (P=0.0102). The survival period of the PEG-mIFN-α4 intermittent administration combined with gemcitabine group was significantly longer than that of the gemcitabine only group.


The median survival period of PEG-mIFN-α4 intermittent administration group was 89.0 days, and the survival rate at the end of the observation period (110 days after tumor inoculation) in the PEG-mIFN-α4 intermittent administration combined with gemcitabine group was 75.0%. The survival curves of the two groups were significantly different (P=0.0048). The survival period of the PEG-mIFN-α4 intermittent administration combined with gemcitabine group was significantly longer than that of the PEG-mIFN-α4 intermittent administration group.


The results showed that for the treatment of transplanted colorectal cancer CT26 in mice, intermittent administration of pegylated mouse interferon α was better than continuous administration of pegylated mouse interferon α; the anticancer activity of intermittent administration of pegylated mouse interferon α combined with gemcitabine was significantly better than that of gemcitabine alone and pegylated mouse interferon α alone.









TABLE 6







Comparison of efficacy of PEG-mIFN-α4 continuous administration,


PEG-mIFN-α4 intermittent administration, and PEG-


mIFN-α4 intermittent administration combined with


gemcitabine in the treatment of transplanted colorectal


cancer CT26 in mice: survival analysis results











Number of
Mean survival




cases
period
Inter-group



(Female +
(days post tumor
comparison











Group
male)
inoculation)
P value1
P value2





Normal saline
12 + 12
49.5
/
/


control


PEG-mIFN-α4
12 + 12
89.0
/
0.004801*


intermittent


administration


PEG-mIFN-α4
12 + 12
53.5
0.0150*
/


continuous


administration


Gemcitabine
12 + 12
99.0
/
0.0102* 


PEG-mIFN-α4
12 + 12
75% within the
/
/


intermittent

observation


administration

period


+ Gemcitabine





Note:



1compared with “PEG-mIFN-α4 intermittent administration group”;




2compared with “PEG-mIFN-α4 intermittent administration + Gemcitabine group”; and



3. *statistically different.






Example 8. Intermittent Administration of Interferon Combined with Gemcitabine to Treat Melanoma in Mice

The inventors studied the antitumor effect of intermittent administration of PEGylated recombinant mouse interferon α (PEG-mIFN-α4) combined with gemcitabine on mouse melanoma B16, and explored the efficacy of combined administration of interferon and gemcitabine. The inventors found that by intermittently administering PEG-mIFN-α4 and administering gemcitabine, significantly better results may be obtained in the treatment of mouse melanoma B16, compared to administering gemcitabine alone or intermittently administering PEG-mIFN-α4 alone.


In this example, a comparative study on the efficacy of PEG-mIFN-α4 intermittent administration (once every 48 hours for 3 consecutive times, then drug withdrawal for 240 hours; for 4 rounds), PEG-mIFN-α4 continuous administration (once every 48 hours for 24 consecutive times), PEG-mIFN-α4 in combination with gemcitabine (PEG-mIFN-α4: once every 48 hours for 3 consecutive times, then drug withdrawal for 240 hours, 4 rounds; gemcitabine: once a week) on transplanted melanoma B16 in mice was performed.


6-8 weeks old, 18-22 g SPF grade C57BL/6N mice were subcutaneously inoculated with B16 cells (1×105 cells/mL, 0.2 ml/mouse) in the right forelimb axillary. On the day when the tumor was inoculated, they were randomly divided into PEG-mIFN-α4 continuous administration group (N=28, female 14, male 14), PEG-mIFN-α4 intermittent administration group (N=28, female 14, male 14), the normal saline control group (N=28, female 14, male 14), the gemcitabine only group (N=28, female 14, male 14), and PEG-mIFN-α4 combined with gemcitabine group (N=28, female 14, male 14).


PEG-mIFN-α4 was injected subcutaneously on the back of the neck, 1 μg/mouse. The administration was started on the day after grouping. For the PEG-mIFN-α4 intermittent administration group, PEG-mIFN-α4 was administered once every 48 hours for 3 consecutive times, then drug withdrawal for 240 hours; for 4 rounds. For the PEG-mIFN-α4 continuous administration group, PEG-mIFN-α4 was administered once every 48 hours. For the PEG-mIFN-α4 combined with gemcitabine group, PEG-mIFN-α4 was administered once every 48 hours for 3 consecutive times, then drug withdrawal for 240 hours; for 4 rounds; and gemcitabine was administered once a week. For the gemcitabine only group, gemcitabine was administered once a week. For the saline control group, equal volume of saline was administered. Gemcitabine was injected intraperitoneally, 60 mg/kg.


SAS9.4 software was used for comparing the difference in mortality, and the statistical tests were all two-sided tests.


The mortality is shown in FIG. 12, and the statistical comparison results are shown in Table 7.


46 days after tumor inoculation, the mortality of the PEG-mIFN-α4 continuous administration group was 100%, and that of the PEG-mIFN-α4 intermittent administration group was 39.3%, and the difference was significant (P<0.001). The mortality of PEG-mIFN-α4 intermittent administration group was significantly lower than that of PEG-mIFN-α4 continuous administration group. For the treatment of transplanted melanoma B16 in mice, PEG-mIFN-α4 intermittent administration showed significantly improved effect than continuous administration.


48 days after tumor inoculation, the mortality of gemcitabine only group was 89.3%, and that of the PEG-mIFN-α4 intermittent administration combined with gemcitabine group was 21.4%, and the difference was significant (P<0.0001). The mortality of PEG-mIFN-α4 intermittent administration combined with gemcitabine group was significantly lower than that of gemcitabine only group.


48 days after tumor inoculation, the mortality of PEG-mIFN-α4 intermittent administration only group was 46.4%, and that of the PEG-mIFN-α4 intermittent administration combined with gemcitabine group was 21.4%, and the difference was significant (P=0.0482). The mortality of PEG-mIFN-α4 intermittent administration combined with gemcitabine group was significantly lower than that of PEG-mIFN-α4 intermittent administration only group.


The results showed that for the treatment of transplanted melanoma B16 in mice, intermittent administration of pegylated mouse interferon α was better than continuous administration of pegylated mouse interferon α; the anticancer activity of intermittent administration of pegylated mouse interferon α combined with gemcitabine was significantly better than that of gemcitabine alone and pegylated mouse interferon α alone.









TABLE 7







Comparison of efficacy of PEG-mIFN-α4 continuous administration,


PEG-mIFN-α4 intermittent administration, and PEG-mIFN-α4


intermittent administration combined with gemcitabine in the treatment


of transplanted melanoma B16 in mice: mortality analysis results










Number of












cases

Inter-group



(Female +

comparison











Group
male)
Mortality
P value1
P value2





Normal saline
14 + 14
92.9%
/
/


control


PEG-mIFN-α4
14 + 14
46.4%
/
 0.0482*


intermittent


administration


PEG-mIFN-α4
14 + 14
 100%
<0.0001*
/


continuous


administration


Gemcitabine
14 + 14
89.3%
/
<0.0001*


PEG-mIFN-α4
14 + 14
21.4%
/
/


intermittent


administration +


Gemcitabine





Note:



1compared with “PEG-mIFN-α4 intermittent administration group” on Day 46 post tumor inoculation;




2compared with “PEG-mIFN-α4 intermittent administration + Gemcitabine group” on Day 48 post tumor inoculation; and



3. *statistically different.






Example 9. Intermittent Administration of Interferon Combined with Gemcitabine to Treat Breast Cancer in Mice

The inventors studied the antitumor effect of intermittent administration of PEGylated recombinant mouse interferon α (PEG-mIFN-α4) combined with gemcitabine on mouse breast cancer 4T1, and explored the efficacy of combined administration of interferon and gemcitabine. The inventors found that by intermittently administering PEG-mIFN-α4 and administering gemcitabine, significantly better results may be obtained in the treatment of mouse breast cancer 4T1, compared to administering gemcitabine alone or intermittently administering PEG-mIFN-α4 alone.


In this example, a comparative study on the efficacy of PEG-mIFN-α4 intermittent administration (once every 48 hours for 3 consecutive times, then drug withdrawal for 144 hours; for 5 rounds), PEG-mIFN-α4 continuous administration (once every 48 hours for 25 consecutive times), PEG-mIFN-α4 in combination with gemcitabine (PEG-mIFN-α4: once every 48 hours for 3 consecutive times, then drug withdrawal for 240 hours, 4 rounds; gemcitabine: once a week) on transplanted breast cancer 4T1 in mice was performed.


In this example, BALB/c mice were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd., and the 4T1 mouse breast cancer cell line was purchased from China Center for Type Culture Collection (CCTCC).


6-8 weeks old, 18-22 g SPF grade BALB/c mice were subcutaneously inoculated with 4T1 cells (1×106 cells/mL, 0.2 ml/mouse) in the right forelimb axillary. On the day when the tumor was inoculated, they were randomly divided into PEG-mIFN-α4 continuous administration group (N=24, female 12, male 12), PEG-mIFN-α4 intermittent administration group (N=24, female 12, male 12), the normal saline control group (N=24, female 12, male 12), the gemcitabine only group (N=24, female 12, male 12), and PEG-mIFN-α4 combined with gemcitabine group (N=24, female 12, male 12).


PEG-mIFN-α4 was injected subcutaneously on the back of the neck, 1 μg/mouse. The administration was started on the day after grouping. For the PEG-mIFN-α4 intermittent administration group, PEG-mIFN-α4 was administered once every 48 hours for 3 consecutive times, then drug withdrawal for 144 hours; for 5 rounds. For the PEG-mIFN-α4 continuous administration group, PEG-mIFN-α4 was administered once every 48 hours. For the PEG-mIFN-α4 combined with gemcitabine group, PEG-mIFN-α4 was administered once every 48 hours for 3 consecutive times, then drug withdrawal for 240 hours; for 4 rounds; and gemcitabine was administered once a week. For the gemcitabine only group, gemcitabine was administered once a week. For the saline control group, equal volume of saline was administered. Gemcitabine was injected intraperitoneally, 60 mg/kg.


SAS9.4 software was used for comparing the difference in mortality, and the statistical tests were all two-sided tests.


The mortality is shown in FIG. 13, and the statistical comparison results are shown in Table 8.


53 days after tumor inoculation, the mortality of the PEG-mIFN-α4 continuous administration group was 87.5%, and that of the PEG-mIFN-α4 intermittent administration group was 58.3%, and the difference was significant (P=0.0230), and both were lower than that of the saline control group (100%). The mortality of PEG-mIFN-α4 intermittent administration group was significantly lower than that of PEG-mIFN-α4 continuous administration group. For the treatment of transplanted breast cancer 4T1 in mice, PEG-mIFN-α4 intermittent administration showed significantly improved effect than continuous administration.


53 days after tumor inoculation, the mortality of gemcitabine only group was 87.5%, and that of the PEG-mIFN-α4 intermittent administration combined with gemcitabine group was 4.2%, and the difference was significant (P<0.0001). The mortality of PEG-mIFN-α4 intermittent administration combined with gemcitabine group was significantly lower than that of gemcitabine only group.


53 days after tumor inoculation, the mortality of PEG-mIFN-α4 intermittent administration only group was 58.3%, and that of the PEG-mIFN-α4 intermittent administration combined with gemcitabine group was 4.2%, and the difference was significant (P<0.0001). The mortality of PEG-mIFN-α4 intermittent administration combined with gemcitabine group was significantly lower than that of PEG-mIFN-α4 intermittent administration only group.


The results showed that for the treatment of transplanted breast cancer 4T1 in mice, intermittent administration of pegylated mouse interferon α was better than continuous administration of pegylated mouse interferon α; the anticancer activity of intermittent administration of pegylated mouse interferon α combined with gemcitabine was significantly better than that of gemcitabine alone and pegylated mouse interferon α alone.









TABLE 8







Comparison of efficacy of PEG-mIFN-α4 continuous


administration, PEG-mIFN-α4 intermittent administration,


and PEG-mIFN-α4 intermittent administration combined


with gemcitabine in the treatment of transplanted breast


cancer 4T1 in mice: mortality analysis results










Number of












cases

Inter-group



(Female +

comparison











Group
male)
Mortality
P value1
P value2





Normal saline
12 + 12
 100%
/
/


control


PEG-mIFN-α4
12 + 12
87.5%
/
/


intermittent


administration


PEG-mIFN-α4
12 + 12
58.3%
0.0230*
<0.0001*


continuous


administration


Gemcitabine
12 + 12
87.5%
/
<0.0001*


PEG-mIFN-α4
12 + 12
 4.3%
/
/


intermittent


administration +


Gemcitabine





Note:



1compared with “PEG-mIFN-α4 intermittent administration group”;




2compared with “PEG-mIFN-α4 intermittent administration + Gemcitabine group”; and



3. *statistically different.






Example 10. Study on the Treatment of Transplanted Liver Cancer H22 in Mice by Intermittent Administration of Interferon and Anticancer Agent Epirubicin

The anticancer agent used in this example is the antitumor antibiotic epirubicin among chemotherapeutics, and the effect of intermittent administration of interferon combined with anticancer agent epirubicin in the treatment of transplanted liver cancer H22 in mice was investigated.


In this example, BALB/c mice were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd., and the mouse liver cancer H22 cell line was purchased from the China Center for Type Culture Collection (CCTCC). The manufacturer of epirubicin was Beijing Union Pharmaceutical Factory.


6-8 weeks old, 18-22 g SPF grade BALB/c mice were subcutaneously inoculated with liver cancer H22 tumor cells (1×106 cells/mL, 0.2 ml/mouse) in the right forelimb axillary. 3 days after tumor inoculation, they were randomly divided into groups, with 28 mice in each group (fifty-fifty female and male mice), including normal saline control group, epirubicin only group, and PEG-mIFN-α4 combined with epirubicin group.


The administration was started on the day after grouping, once a week. PEG-mIFN-α4 was injected subcutaneously on the back of the neck, and epirubicin was injected intraperitoneally, in a dose of 3.5 mg/kg each time. The normal saline control group was administered an equal volume of normal saline.


All groups were subjected to survival curve analysis, and the median survival time was compared. SAS 9.4 and office2010 software were used for statistical analysis. Statistical tests were all two-sided tests.


The survival curve is shown in FIG. 14, and the median survival time is shown in Table 9. The median survival time of the saline group was 13.5 days, the median survival time of the epirubicin group was 15 days, and the survival rate of the PEG-mIFN-α4 combined with epirubicin group during the observation period was 57.1%. The survival curve of PEG-mIFN-α4 combined with epirubicin group was significantly different from that of normal saline group (P<0.0001). It was showed that the survival time of tumor-bearing mice may be significantly prolonged (P<0.0001) by intermittent administration of PEG-mIFN-α4 combined with epirubicin, compared with administration of epirubicin alone.


This example suggests that in the treatment of transplanted liver cancer H22 in mice, the effect of intermittent administration of PEG-mIFN-α4 in combination with epirubicin was significantly better than that of epirubicin alone.









TABLE 9







Survival analysis results of Example 10











Number of
Median survival




cases
period
Survival curve



(Female +
(Time after tumor
comparison











Group
male)
inoculation, days)
P value1
P value2





Normal saline
14 + 14
13.5
/



(0.2 mL/w)


Epirubicin
14 + 14
15.0
0.4598


(3.5 mg/kg/w)


Epirubicin +
14 + 14
Survival rate during
<0.0001*
<0.0001*


PEG-mIFN-α4

the observation




period was 57.1%.





Note:


P value1 was compared with “normal saline (0.2 ml/w)”;


P value2 was compared with “epirubicin (3.5 mg/kg/w)”; and


*refers to a statistical difference.






Example 11. Study on the Treatment of Transplanted Liver Cancer H22 in Mice by Intermittent Administration of Interferon in Combination with Anticancer Agent Oxaliplatin

The anticancer agent used in this example is the chemotherapeutic agent oxaliplatin, and the effect of intermittent administration of interferon combined with anticancer agent oxaliplatin in the treatment of transplanted liver cancer H22 in mice was investigated.


In this example, BALB/c mice were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd., and the H22 mouse liver cancer cell line was purchased from the China Center for Type Culture Collection (CCTCC). The manufacturer of oxaliplatin was Qilu Pharmaceutical (Hainan) Co., Ltd.


6-8 weeks old, 18-22 g SPF grade BALB/c mice were subcutaneously inoculated with liver cancer H22 tumor cells (1×106 cells/mL, 0.2 ml/mouse) in the right forelimb axillary. 3 days after tumor inoculation, they were randomly divided into groups, with 28 mice in each group (fifty-fifty female and male mice), including normal saline control group, oxaliplatin only group, and PEG-mIFN-α4 combined with oxaliplatin group.


The administration was started on the day after grouping, once a week. PEG-mIFN-α4 was injected subcutaneously on the back of the neck, and oxaliplatin was injected intraperitoneally, in a dose of 10 mg/kg each time. The normal saline control group was administered an equal volume of normal saline.


All groups were subjected to survival curve analysis, and the median survival time was compared. SAS 9.4 and office2010 software were used for statistical analysis. Statistical tests were all two-sided tests.


The survival curve is shown in FIG. 15, and the median survival time is shown in Table 10. The median survival time of the normal saline group was 11 days, the median survival time of the oxaliplatin group was 12.5 days, and the median survival time of the PEG-mIFN-α4 combined with oxaliplatin group was 31 days. The survival curve of the PEG-mIFN-α4 combined with oxaliplatin group was significantly different from that of the normal saline group (P=0.0001), and also significantly different from the oxaliplatin group alone (P=0.0014). Intermittent administration of PEG-mIFN-α4 combined with oxaliplatin may significantly prolong the survival time of tumor-bearing mice.


This example suggests that in the treatment of transplanted liver cancer H22 in mice, the effect of intermittent administration of PEG-mIFN-α4 in combination with oxaliplatin was significantly better than that of oxaliplatin alone.









TABLE 10







Survival analysis results of Example 13











Number of
Median survival




cases
period
Survival curve



(Female +
(Time after tumor
comparison











Group
male)
inoculation, days)
P value1
P value2





Normal saline
14 + 14
11.0
/
/


(0.2 mL/w)


Oxaliplatin
14 + 14
12.5
0.5112 
/


(10 mg/kg/w)


Oxaliplatin +
14 + 14
31.0
0.0001*
0.0014*


PEG-mIFN-α4





Note:


P value1 was compared with “normal saline (0.2 ml/w)”;


P value2 was compared with “oxaliplatin (10 mg/kg/w)”; and


*refers to a statistical difference.






Example 12. Study on the Treatment of Transplanted Liver Cancer H22 in Mice by Intermittent Administration of Interferon and Anticancer Agent Paclitaxel

The anticancer agent used in this example is the chemotherapeutic agent paclitaxel, and the effect of intermittent administration of interferon combined with anticancer agent paclitaxel in the treatment of transplanted liver cancer H22 in mice was investigated.


In this example, BALB/c mice were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd., and the H22 mouse liver cancer cell line was purchased from the China Center for Type Culture Collection (CCTCC). The manufacturer of paclitaxel was Qilu Pharmaceutical (Hainan) Co., Ltd.


6-8 weeks old, 18-22 g SPF grade BALB/c mice were subcutaneously inoculated with liver cancer H22 tumor cells (1×106 cells/mL, 0.2 ml/mouse) in the right forelimb axillary. 3 days after tumor inoculation, they were randomly divided into groups, with 28 mice in each group (fifty-fifty female and male mice), including normal saline control group, paclitaxel only group, and PEG-mIFN-α4 combined with paclitaxel group.


The administration was started on the day after grouping, once a week. PEG-mIFN-α4 was injected subcutaneously on the back of the neck, in a dose of 1 mg/mouse each time, and oxaliplatin was injected intraperitoneally, in a dose of 10 mg/kg each time.


All groups were subjected to survival curve analysis, and the median survival time was compared. SAS 9.4 and office2010 software were used for statistical analysis. Statistical tests were all two-sided tests.


The survival curve is shown in FIG. 16, and the median survival time is shown in Table 11. The median survival time of the normal saline group was 15 days, and that of the paclitaxel group was 17 days. During the observation period, the survival rate of the PEG-mIFN-α4 combined with paclitaxel group was 82.1%. The survival curve of the PEG-mIFN-α4 combined with paclitaxel group was significantly different from that of the normal saline group (P=0.0001), and also significantly different from the paclitaxel group alone (P<0.0001). Intermittent administration of PEG-mIFN-α4 combined with paclitaxel may significantly prolong the survival time of tumor-bearing mice.


This example suggests that in the treatment of transplanted liver cancer H22 in mice, the effect of intermittent administration of PEG-mIFN-α4 in combination with paclitaxel was significantly better than that of paclitaxel alone.









TABLE 11







Survival analysis results of Example 14











Number of
Median survival




cases
period
Survival curve



(Female +
(Time after tumor
comparison











Group
male)
inoculation, days)
P value1
P value2





Normal saline
14 + 14
15.0
/
/


(0.2 mL/w)


Paclitaxel
14 + 14
17.0
0.3147 
/


(10 mg/kg/w)


Paclitaxel +
14 + 14
Survival rate during
<0.0001*
<0.0001*


PEG-mIFN-α4

the observation




period was 82.1%.





Note:


P value1 was compared with “normal saline (0.2 ml/w)”;


P value2 was compared with “paclitaxel (10 mg/kg/w)”; and


*refers to a statistical difference.






The above results of the present invention suggest that by intermittently administering PEG-mIFN-α4 in combination with a chemotherapeutic agent such as gemcitabine, paclitaxel, oxaliplatin, epirubicin, etc., it is possible to obtain a significantly better cancer treatment effect than administering the chemotherapeutic agent alone.


The above results suggest that continuous administration of interferon for long time will cause immunosuppression and depletion of immune cells, which is difficult to recover. The efficacy of interferon depends on the immune system. Therefore, an interferon-based therapy may achieve good therapeutic effects such as high tumor suppression in the early stage of treatment when immune cells are still sufficient. However, in the later stage of treatment, due to depletion of immune cells or immunosuppression caused by long-term administration of interferon, the therapeutic effect may be substantially reduced, and high efficacy may not be maintained even if interferon is continuely administered. This problem may be avoided by intermittent administration of interferons. For example, after administering interferon for a period of time to achieve a certain effect and before partial immune suppression and immune cell exhaustion, the administration of interferon can be suspended for a period of time so that immune cells can recover as soon as possible, and then re-administration of interferon can still achieve better therapeutic efficacy. By intermittently administering the interferon therapeutic agent in reasonable combination with another drug, better therapeutic effects may be achieved.












Sequences and description thereof















>SEQ ID NO: 1 is the amino acid sequence of interferon a 2a


(e.g., Roferon-A or Pegasys)


CDLPQ THSLG SRRTL MLLAQ MRKIS LFSCL KDRHD FGFPQ EEFGN


QFQKA ETIPV LHEMI QQIFN LFSTK DSSAA WDETL LDKFY TELYQ QLNDL


EACVI QGVGV TEPLM KEDSI LAVRK YFQRI TLYLK EKKYS PCAWE


VVRAE IMRSF SLSTN LQESL RSKE





>SEQ ID NO: 2 is the amino acid sequence of interferon a 2b


(e.g., INTRONA or PEG-INTRON or Pegberon)


CDLPQ THSLG SRRTL MLLAQ MRRIS LFSCL KDRHD FGFPQ EEFGN


QFQKA ETIPV LHEMI QQIFN LFSTK DSSAA WDETL LDKFY TELYQ QLNDL


EACVI QGVGV TETPL MKEDS ILAVR KYFQR ITLYL KEKKY SPCAW EVVRA


EIMRS FSLST NLQES LRSKE .





>SEQ ID NO: 3 is the amino acid sequence of a recombinant


integrated interferon (e.g., Infergen)


MCDLPQTHSL GNRRALILLA QMRRISPFSC LKDRHDFGFP


QEEFDGNQFQ KAQAISVLHE MIQQTFNLFS TKDSSAAWDE SLLEKFYTEL


YQQLNDLEAC VIQEVGVEET PLMNVDSILA VKKYFQRITL YLTEKKYSPC


AWEVVRAEIM RSFSLSTNLQ ERLRRKE .





>SEQ ID NO: 4 is the amino acid sequence of integrated


interferon a-2 (e.g., PEGINFER)


GSGGGCDLPQTHSLGNRRALILLAQMRRISPFSCLKDRHDFGFPQEEFDG


NQFQKAQAISVLHEMIQQTFNLFSTKDSSAAWDESLLEKFYTELYQQLNDLEA


CVIQEVGVEETPLMNVDSILAVRKYFQRITLYLTEKKYSPCAWEVVRAEIMR


SFSLSTNLQERLRRKD





>SEQ ID NO: 5 is the amino acid sequence of interferon 2


MGPVP TSKPT TTGKG CHIGR FKSLS PQELA SFKKA RDALE ESLKL


KNWSC SSPVF PGNWD LRLLQ VRERP VALEA ELALT LKVLE AAAGP


ALEDV LDOPL HTLHH ILSQL QACIQ PQPTA GPRPR GRLHH WLHRL


QEAPK KESAG CLEAS VTFNL FRLLT RDLKY VADGN LCLRT STHPE ST





>SEQ ID NO: 6 is the amino acid sequence of peginterferon


murine a 4


CDLPHTYNLGNKRALTVLEEMRRLPPLSCLKDRKDFGFPLEKVDNQQIQ


KAQAILVLRDLTQQILNLFTSKDLSATWNATLLDSFCNDLHQQLNDLKACVM


QEPPLTQEDSLLAVRTYFHRITVYLRKKKHSLCAWEVIRAEVWRALSSSTNLL


ARLSEEKE









REFERENCES



  • [1] Gisslinger H, et. al. Ropeginterferon alfa-2b, a novel IFNα-2b, induces high response rates with low toxicity in patients with polycythemia vera. Blood. 2015 Oct. 8; 126(15):1762-9. doi: 10.1182/blood-2015-04-637280. Epub 2015 Aug. 10.

  • [2] Gane E J, et. al. The oral toll-like receptor-7 agonist GS-9620 in patients with chronic hepatitis B virus infection. J Hepatol. 2015 August; 63(2):320-8.

  • doi: 10.1016/j.jhep.2015.02.037. Epub 2015 Feb. 27.

  • [3] Janssen HLA, et. al. Safety, efficacy and pharmacodynamics of vesatolimod (GS-9620) in virally suppressed patients with chronic hepatitis B. J Hepatol. 2018 March; 68(3):431-440. doi: 10.1016/j.jhep.2017.10.027. Epub 2017 Dec. 11.

  • [4] Agarwal K, et. al. Safety and efficacy of vesatolimod (GS-9620) in patients with chronic hepatitis B who are not currently on antiviral treatment. Journal of Viral Hepatitis [2018 Aug. 25(11):1331-1340.

  • [5] Lopatin U, et. al. Safety, pharmacokinetics and pharmacodynamics of GS-9620, an oral Toll-like receptor 7 agonist. Antivir Ther. 2013; 18(3):409-18. doi: 10.3851/IMP2548. Epub 2013 Feb. 15.

  • [6] Edward J. Gane, et. al. TLR7 agonist R07020531 triggers immune activation after multiple doses in chronic hepatitis B patients Reported by Jules Levin. AASLD 2018 Nov. 9-13 SF

  • [7] Ebrahim M, et. al. Are RIG-1 and MDAS Expressions Associated with Chronic HBV Infection? Viral Immunol. 2015 November; 28(9):504-8. doi: 10.1089/vim.2015.0056. Epub 2015 Oct. 20.

  • [8] Ma Z, et. al. Contribution of Toll-like receptors to the control of hepatitis B virus infection by initiating antiviral innate responses and promoting specific adaptive immune responses. Cell Mol Immunol. 2015 May; 12(3):273-82. doi: 10.1038/cmi.2014.112. Epub 2014 Nov. 24.

  • [9] Jones M, et. al. SB 9200, a novel agonist of innate immunity, shows potent antiviral activity against resistant HCV variants. J Med Virol. 2017 September; 89(9):1620-1628. doi: 10.1002/jmv.24809. Epub 2017 May 23.

  • [10] Xu C L, et. al. Upregulation of toll-like receptor 4 on T cells in PBMCs is associated with disease aggravation of HBV-related acute-on-chronic liver failure. J Huazhong Univ Sci Technolog Med Sci. 2015 December; 35(6):910-915. doi: 10.1007/s11596-015-1527-x. Epub 2015 Dec. 16.

  • [11] Kato H, et. al. RIG-I-Like Receptors and Type I Interferonopathies. J Interferon Cytokine Res. 2017 May; 37(5):207-213. doi: 10.1089/jir.2016.0095.

  • [12] Li L, et. al. Anti-HBV response to toll-like receptor 7 agonist GS-9620 is associated with intrahepatic aggregates of T cells and B cells. J Hepatol. 2018 May; 68(5):912-921. doi: 10.1016/j.jhep.2017.12.008. Epub 2017 Dec. 14.

  • [13] Harrington, et. al. LBA15Preliminary results of the first-in-human (FIH) study of MK-1454, an agonist of stimulator of interferon genes (STING), as monotherapy or in combination with pembrolizumab (pembro) in patients with advanced solid tumors or lymphomas. Annals of Oncology. 2018 Oct. 29(8):712-712. Meeting Abstract.

  • [14] Cemerski, Saso, et. al. Preclinical characterization of a novel STING agonist, MK-1454. JOURNAL FOR IMMUNOTHERAPY OF CANCER. 2017 Nov. 5(2):16. Meeting Abstract.

  • [15] Ramanjulu J M, et. al. Design of amidobenzimidazole STING receptor agonists with systemic activity. Nature. 2018 December; 564(7736):439-443. doi: 10.1038/s41586-018-0705-y. Epub 2018 Nov. 7.

  • [16] Safety and Efficacy of MIW815 (ADU-S100)+/−Ipilimumab in Patients With Advanced/Metastatic Solid Tumors or Lymphomas.

  • https://www.aduro.com/file.cfm/13/docs/SITC18_MIW815X2101_SITC %20Poster(e_version) 8 Nov. 2018.pdf

  • [17] Lara P N Jr, et. al. Randomized phase III placebo-controlled trial of carboplatin and paclitaxel with or without the vascular disrupting agent vadimezan (ASA404) in advanced non-small-cell lung cancer. J Clin Oncol. 2011 Aug. 1; 29(22):2965-71. doi: 10.1200/JCO.2011.35.0660. Epub 2011 Jun. 27.

  • [18] Yuen, M. F, et. al. Dose response and safety of the daily, oral RIG-I agonist Inarigivir (SB 9200) in treatment naive patients with chronic hepatitis B: results from the 25 mg and 50 mg cohorts in the ACHIEVE trial. JOURNAL OF HEPATOLOGY. 2018 April 68(1): 5509-S510.DOI: 10.1016/S0168-8278(18)31267-4

  • [19] Ma Z, et. al. Interaction between Hepatitis B Virus and Toll-Like Receptors: Current Status and Potential Therapeutic Use for Chronic Hepatitis B. Vaccines (Basel). 2018 Jan. 16; 6(1). pii: E6. doi: 10.3390/vaccines6010006.

  • [20] Walsh, R., et. al. Effects of SB9200 (Inarigivir) therapy on immune responses in patients with chronic hepatitis B. JOURNAL OF HEPATOLOGY. 2018 April 68(1): S89-S89. DOI: 10.1016/S0168-8278(18)30396-9

  • [21] Niu C, et. al. Toll-like receptor 7 agonist GS-9620 induces prolonged inhibition of HBV via a type I interferon-dependent mechanism. J Hepatol. 2018 May; 68(5):922-931. doi: 10.1016/j.jhep.2017.12.007. Epub 2017 Dec. 13.

  • [22] Yuen, M F, et. al. Ascending dose cohort study of inarigivir—A novel RIG I agonist in chronic HBV patients: Final results of the ACHIEVE trial. JOURNAL OF HEPATOLOGY 2019 April 70(1): E47-E48.DOI: 10.1016/S0618-8278(19)30084-2

  • [23] Bourquin C, et. al. Harnessing the immune system to fight cancer with Toll-like receptor and RIG-I-like receptor agonists. Pharmacol Res. 2019 Mar. 2. pii: S1043-6618(18)32066-8. doi: 10.1016/j.phrs.2019.03.001.

  • [24] Zeng Y, et. al. Toll-like receptors, long non-coding RNA NEAT1, and RIG-I expression are associated with HBeAg-positive chronic hepatitis B patients in the active phase. J Clin Lab Anal. 2019 June; 33(5):e22886. doi: 10.1002/jcla.22886. Epub 2019 Mar. 29.

  • [25] Wang Jiawen, et al. Research progress of Toll-like receptors and their agonists. Advances in Physiological Science, 2018, Vol. 49, No. 4, 289-292.

  • [26] Tao Zeyu, et al. Research progress of drugs for the treatment of chronic hepatitis B virus infection. Foreign Medicine and Antibiotics Volume, July 2018, Vol. 39, No. 4: 308-319.

  • [27] Xu Qun, et al. Research progress of immunomodulators targeting Toll-like receptors. Advances in Pharmacy, 2016, 40(1): 42-55.

  • [28] Liu Qiuming, et al. New Advances in Anti-HBV Drug Research, Journal of Virology. September 2016, Vol. 32, Issue 5: 650-658.

  • [29] Mu Na, et al. Expression levels of pattern recognition receptors TLR3, RIG-I and MDAS in peripheral blood of patients with chronic hepatitis B. Chinese Journal of Immunology. 2016-05:715-719.

  • [30] Heinz Gisslinger, et. al. Ropeginterferon alfa-2b, a novel IFNα-2b, induces high response rates with low toxicity in patients with polycythemia vera. Blood. 2015 Oct. 8; 126(15): 1762-1769. Prepublished online 2015 Aug. 10.

  • [31] Monkarsh S P, et. al. Positional isomers of monopegylated interferon alpha-2a: isolation, characterization, and biological activity. Anal Biochem. 1997 May 1; 247(2):434-40.

  • [32] Grace M, et. al. Structural and biologic characterization of pegylated recombinant IFN-alpha2b. J Interferon Cytokine Res. 2001 December; 21(12):1103-15.

  • [33] Foser S, et. al. Isolation, structural characterization, and antiviral activity of positional isomers of monopegylated interferon alpha-2a (PEGASYS). Protein Expr Purif. 2003 July; 30(1):78-87.

  • [34] Nanogen Pharmaceutical biotechbology. PEG-INTERFERON LAMBDA 1 CONJUGATES. WO2013/028233 A1. 28.02.2013.

  • [35] Nanogen Pharmaceutical biotechbology. PEG-INTERFERON LAMBDA 1 CONJUGATES. U.S. Pat. No. 8,454,947 B1. Jun. 4, 2013.

  • https://worldwide.espacenet.com/searchResults?submitted=true&locale=en_EP& DB=EPODOC&ST=advanced&TI=&AB=peg-interferon+lambda&PN=&AP=&PR=&PD=&PA=&IN=&CPC=&IC=

  • [36] Wang Junzhi. Biotechnology drug research and development and quality control. 2nd edition. Science Press, 2007: 540.

  • [37] Interferon alpha-4 precursor [Mus musculus].https://www.ncbi.nlm.nih.gov/protein/NP_034634.

  • [38] Pichia Fermentation Process Guidelines, Thermo Fisher Scientific Inc.https://www.thermofisher.com/document-connect/document-connect.html?url=https%3A %2F%2Fassets.thermofisher.com%2FTFS-Assets%2FLSG%2Fmanuals%2Fpichiaferm_prot.pdf&title=UGljaGlhIEZ1cmllbnRhdGlvbiBHdW1kZWxpbmVz.


Claims
  • 1. A method for treating cancer in a subject, comprising i) intermittently administering an interferon-based therapeutic agent for a plurality of consecutive treatment courses; andii) administering an additional anticancer agent,to the subject.
  • 2. The method according to claim 1, wherein the interferon-based therapeutic agent comprises an interferon or a mutant or derivative thereof, or comprises a nucleic acid molecule encoding an interferon or a mutant or a derivative thereof, or comprises a substance promoting the generation of an endogenous interferon.
  • 3. The method according to claim 1, wherein the interferon is a Type I, Type II or Type III interferon, such as interferon α, interferon β, interferon γ or interferon λ, preferably interferon α.
  • 4. The method according to claim 1, wherein the interferon-based therapeutic agent comprises interferon α 2a, interferon α 2b, interferon α 1b, interferon λ, or a mutant or derivative thereof.
  • 5. The method according to claim 1, wherein the interferon or the mutant or derivative thereof is PEGylated.
  • 6. The method according to claim 1, wherein the interferon-based therapeutic agent is selected from the group consisting of P1101, Pegberon, Pegasys, Pegintron, Infergen, Novaferon, INTRONA, Roferon-A, Hapgen, PEGINFER and Peginterferon λ.
  • 7. The method according to claim 1, wherein the interferon-based therapeutic agent comprises an agonist of the TLRs, RLRs, and STINGs signaling pathways.
  • 8. The method according to claim 7, wherein the interferon-based therapeutic agent is selected from the group consisting of GS-9620, GS-9688, RO7020531, RO6864018, TQ-A3334, JNJ-4964, SB9200, MIW815, DMXAA, MK-1454, and diABZI.
  • 9. The method according to claim 1, wherein in the consecutive treatment course, the interferon-based therapeutic agent is administered such that during substantially the entire course, the concentration of neopterin in the subject is higher than the concentration of neopterin before the first administration, for example approximately 110%, approximately 120%, approximately 130%, approximately 140%, approximately 150%, approximately 200%, approximately 250% or higher of the neopterin concentration before the first administration.
  • 10. The method according to claim 1, wherein the duration of the consecutive treatment course is the time period from the first administration to the last administration, plus about 5 in vivo half-lives of the therapeutic agent.
  • 11. The method according to claim 1, wherein the duration of each of the plurality of consecutive treatment courses is from about 1 week to about 24 weeks, preferably from about 1 week to about 12 weeks, further preferably from about 1 week to about 8 weeks, and yet further preferably about 2 weeks to about 6 weeks.
  • 12. The method according to claim 1, wherein the duration of each of the consecutive treatment courses is about 1 week to about 12 weeks, and the interval between the consecutive treatment courses is about 1 week to about 12 weeks.
  • 13. The method according to claim 1, wherein the interval between the consecutive treatment courses is from about 1 week to about 24 weeks, preferably from about 1 week to about 12 weeks, further preferably from about 1 week to about 8 weeks, and yet further preferably about 2 weeks to about 6 weeks.
  • 14. The method according to claim 1, wherein the duration of each of the consecutive treatment courses is about 1 week to about 8 weeks, and the interval between the consecutive treatment courses is about 1 week to about 8 weeks.
  • 15. The method according to claim 1, wherein the duration of each of the consecutive treatment courses is about 2 weeks to about 6 weeks, and the interval between the consecutive treatment courses is about 2 week to about 6 weeks.
  • 16. The method according to claim 1, wherein the interferon-based therapeutic agent is administered for 2-25 or more consecutive treatment courses.
  • 17. The method according to claim 1, wherein the durations of the plurality of consecutive treatment courses are substantially the same.
  • 18. The method according to claim 1, wherein the intervals between the consecutive treatment courses are substantially the same.
  • 19. The method according to claim 1, wherein the cancer is selected from leukemia (such as acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic myeloid leukemia (CIVIL), chronic lymphocytic leukemia, polycapillary leukemia), liver cancer, lung cancer, colorectal cancer, skin cancer, stomach cancer, breast cancer, prostate cancer, non-Hodgkin's lymphoma, melanoma, multiple myeloma, laryngeal papilloma, follicular lymphoma, AIDS-related Kaposi's sarcoma and renal cell carcinoma, preferably liver cancer, lung cancer, breast cancer, colorectal cancer or melanoma.
  • 20. The method according to claim 1, wherein the administration of the interferon-based therapeutic agent does not overlap with the administration of the additional anticancer agent.
  • 21. The method according to claim 20, wherein the additional anticancer agent is administered between the plurality of consecutive treatment courses.
  • 22. The method according to claim 1, wherein the administration of the interferon-based therapeutic agent overlaps with the administration of the additional anticancer agent.
  • 23. The method according to claim 22, wherein the additional anticancer agent is administered during and between plurality of consecutive treatment courses.
  • 24. The method according to claim 1, wherein the additional anticancer agent is administered according to its conventional scheme.
  • 25. The method according to claim 1, wherein the anticancer agent is i) a chemotherapeutic agent, such as an alkylating agent an alkylating agent: Nimustine, Carmustine, Lomustine, Cyclophosphamide, Ifosfamide, glyciphosphoramide, semustine; an antimetabolite: deoxyfluoguanosine, doxifluguanidine, 5-fluorouracil, mercaptopurine, thioguanine, cytarabine, fluguanosine, tegafur, Gemcitabine, carmofur, hydroxyurea, methotrexate, UFT, Ancitabine, capecitabine; an anti-tumor antibiotic: actinomycin D, doxorubicin, daunorubicin, Epirubicin, mitomycin, pelomycin, pingyangmycin, pirarubicin; a chemotherapeutic anti-tumor animal and plant ingredient: irinotecan, harringtonine, hydroxycamptothecin, Vinorelbine, paclitaxel, albumin paclitaxel, taxotere, topotecan, vincristine, vindesine, Vindesine, vinblastine, teniposide, etoposide, elemene; such as anti-tumor drug hormones: Atamestane, Anastrozole, Aminoglutethimide, Letrozole, Formestane, Metasterone, Tamoxifen; a chemotherapeutic miscellaneous agent: asparaginase, carboplatin, cisplatin, Dacarbazine, Oxaliplatin, Loxadine, Eloxatin, Mitoxantrone, or Procarbazine; orii) an immune checkpoint inhibitor such as an inhibitor of PD-1, PD-L1, CTLA4, for example, an antibody selected from: Nivolumab, Pembrolizumab, Atezolizumab, Durvalumab, Avelumab; oriii) a small molecule targeting drug, such as imatinib, gefitinib, bortezomib, erlotinib, sorafenib, lenalidomide, Sunitinib, dasatinib, nilotinib, lapatinib, pazopanib, everolimus, vandetanib, crizotinib, verofinib, ruxolitinib, axitinib, vismodegib, carfilzomib, regorafenib, bosutinib, tofacitinib, carbotinib, panatinib, pomalidomide, trametinib, dabrafenib, Afatinib, Icotinib, Ibrutinib, Ceritinib, Idelaris, Apatinib, Pabuccilib, Levatinib, Axitinib, Icotinib, Apatinib, sonidegib, cobimetinib, osimertinib, alectinib, ixazomib; oriv) a tumor-associated antigen-specific antibody such as Rituxan, Herceptin;preferably, the anticancer agent is selected from oxaliplatin, epirubicin, paclitaxel and gemcitabine, and more preferably is gemcitabine.
  • 26-35. (canceled)
Priority Claims (1)
Number Date Country Kind
201910660749.1 Jul 2019 CN national
CROSS-REFERENCE TO RELATED APPLICATIONS

The present invention is a continuation of PCT/CN2021/073191, filed Jan. 22, 2021, which is a continuation-in-part of PCT/CN2020/103613, filed Jul. 22, 2020, which claims the benefit of priority of CN 201910660749.1, filed Jul. 22, 2019, the disclosure of each of which application is incorporated herein by reference in its entirety for all purposes.

Continuations (1)
Number Date Country
Parent PCT/CN2021/073191 Jan 2021 US
Child 18099664 US
Continuation in Parts (1)
Number Date Country
Parent PCT/CN2020/103613 Jul 2020 US
Child PCT/CN2021/073191 US