WO2023284069A1 - 一种细菌在制备免疫检查点抑制剂的增效剂中的应用 - Google Patents

一种细菌在制备免疫检查点抑制剂的增效剂中的应用 Download PDF

Info

Publication number
WO2023284069A1
WO2023284069A1 PCT/CN2021/113916 CN2021113916W WO2023284069A1 WO 2023284069 A1 WO2023284069 A1 WO 2023284069A1 CN 2021113916 W CN2021113916 W CN 2021113916W WO 2023284069 A1 WO2023284069 A1 WO 2023284069A1
Authority
WO
WIPO (PCT)
Prior art keywords
bacteria
bacterium
cancer
alistipes
tumor
Prior art date
Application number
PCT/CN2021/113916
Other languages
English (en)
French (fr)
Inventor
徐瑞华
赵霞
Original Assignee
中山大学肿瘤防治中心(中山大学附属肿瘤医院、中山大学肿瘤研究所)
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 中山大学肿瘤防治中心(中山大学附属肿瘤医院、中山大学肿瘤研究所) filed Critical 中山大学肿瘤防治中心(中山大学附属肿瘤医院、中山大学肿瘤研究所)
Publication of WO2023284069A1 publication Critical patent/WO2023284069A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23KFODDER
    • A23K10/00Animal feeding-stuffs
    • A23K10/10Animal feeding-stuffs obtained by microbiological or biochemical processes
    • A23K10/16Addition of microorganisms or extracts thereof, e.g. single-cell proteins, to feeding-stuff compositions
    • A23K10/18Addition of microorganisms or extracts thereof, e.g. single-cell proteins, to feeding-stuff compositions of live microorganisms
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L2/00Non-alcoholic beverages; Dry compositions or concentrates therefor; Their preparation
    • A23L2/52Adding ingredients
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/135Bacteria or derivatives thereof, e.g. probiotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention relates to the technical field of cancer treatment, in particular to the application of a bacterium in the preparation of synergists of immune checkpoint inhibitors.
  • Malignant tumors are one of the biggest problems facing the world at present. Among the various lethal factors that cause death every year, malignant tumors rank first. Conventional treatment options, such as surgery, radiotherapy and chemotherapy, are difficult to completely remove tumor cells, and the recurrence rate is high, and the treatment methods such as radiotherapy and chemotherapy are not targeted, so they can kill tumor cells while also killing normal cells. The killing effect causes great harm to the patient and seriously affects the quality of life of the patient.
  • the target of tumor immunotherapy is not tumor cells and tissues, but the body's own immune system.
  • the body's immune system By regulating and activating the body's immune system, it relies on its own immune mechanism to eliminate tumor cells, including tumor cells that have metastasized to distant places. , has little toxic and side effects on normal cells, and greatly improves the prognosis and quality of life of patients with advanced malignant tumors.
  • immune checkpoint inhibitors immune checkpoint inhibitors
  • ICIs immune checkpoint inhibitors
  • tumor cells such as PD-L1/PD-1 and CTLA-4/B7-1
  • ICIs therapy can block the immune suppression pathway hijacked by tumor cells (such as PD-L1/PD-1 and CTLA-4/B7-1 ) to reactivate T cells and reactivate the body's immune system to recognize and eliminate tumor cells, which has the advantages of good curative effect and long-lasting response, creating a new era of tumor treatment.
  • the application of ICIs therapy in a variety of tumor types due to the lack of treatment options has triggered a revolution in cancer treatment and has become a new milestone in cancer treatment.
  • the intestine is the largest lymphoid organ in the human body, where more than 70% of T cells exist, and it is the residence of the vast majority of memory T cells.
  • the intestinal flora residing in the intestinal tract promotes the development of the human immune system through the interaction with the intestinal immune organs, balances and establishes the immune tone of the human body.
  • both bacteria and tumors are "non-self", so the human body uses the same set of defense mechanisms in the immune response to bacteria and tumors, and the immune response stimulated by intestinal endogenous bacteria has both antitumor potential.
  • Fecal microbiota transplant overcomes to resistance anti–PD-1 therapy in melanoma patients[J].Science,2021,371(6529):595-602.), and has no toxic and side effects, which proves the synergistic effect of endogenous intestinal microbes on ICIs therapy.
  • the composition of fecal microbiota transplantation is complex, and it is the whole gut microbiome that plays a role in fecal microbiota transplantation, and the intestinal microbiota is greatly affected by the environment, diet and lifestyle, etc., and the inability to obtain a continuous and stable source of donor fecal microbiota limits the Clinical application of fecal microbiota transplantation ICIs combined with immunotherapy.
  • the purpose of the present invention is to overcome the above-mentioned deficiencies of the prior art and provide the application of bacteria in the preparation of synergists for immune checkpoint inhibitors.
  • the present invention relates to a combination therapy for synergizing the curative effect of immune checkpoint inhibitors, suitable for One or more tumors, and live whole-cell Alistipes finegoldii suitable for use in the method wherein cancer immunotherapy involves immune checkpoint inhibitor therapy for the purpose of enhancing the antitumor efficacy of cancer immunotherapy , prolong the overall survival time of cancer patients, improve the response rate of cancer immunotherapy populations, and expand the beneficiary tumor patient population of cancer immunotherapy.
  • the treatment method given by the present invention has obvious improvements for the shortcomings of conventional therapy: large toxic and side effects, easy transfer and recurrence, short duration, short survival period, and poor quality of life; for the shortcomings of immune checkpoint monotherapy: The range of tumor types is limited and the drug response population is small, which has been significantly improved; the disadvantages of combined radiotherapy and chemotherapy for immune checkpoints: large toxic and side effects and drug response population is small, which has been significantly improved.
  • the treatment method given by the present invention has a good therapeutic effect on the following patients: treatment of inoperable tumor patients with no available targeted drugs, radiotherapy, chemotherapy, etc.; treatment of single-drug immune checkpoint inhibitors that are ineffective or drug-resistant ( Primary, adaptive and acquired drug resistance) tumor patients; treatment of immune checkpoint inhibitors combined with radiotherapy, chemotherapy, targeted therapy ineffective or resistant (primary, adaptive and acquired drug resistance) tumor patients .
  • the application is as an active ingredient in the preparation of synergists of immune checkpoint inhibitors.
  • the application is an application as a synergist of an immune checkpoint inhibitor to improve the effect of the immune checkpoint inhibitor in treating tumors.
  • the bacterial strain belongs to the genus Alistipes, that is, the 16S rDNA sequence of the bacterial strain has at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, or 100% agreement.
  • the bacteria belonging to the genus Alistipes is a combination of one or more species or strains of bacteria belonging to the genus Alistipes.
  • the bacteria belonging to the genus Alistipes is Alistipes finegoldii.
  • the 16S rDNA sequence contained in the Alistipes finegoldii has at least 99% identity with the 16S rDNA sequence of Alistipes finegoldii.
  • the 16S rDNA sequence contained in the Alistipes finegoldii has at least 99.5% or 100% identity with the 16S rDNA sequence of Alistipes finegoldii DSM17242 (Alistipes finegoldii DSM17242) .
  • the bacterial strain of Alistipes finegoldii is the genome database of the National Center for Biotechnology Information (NCBI) (https://www.ncbi.nlm.nih. gov/genome/browse/#!/prokaryotes/11196/) ⁇ (Strain name of sub species classification) ⁇ :Alistipes finegoldii DSM 17242 ⁇ Alistipes finegoldii D53t1_180928_D3 ⁇ Alistipes finegoldii 2789STDY5834947 ⁇ Alistipes finegoldii 1001713B170207_170306_H2 ⁇ Alistipes finegoldii DFI.2.31 ⁇ Alistipes finegoldii BIOML-A1 ⁇ Alistipes finegoldii DFI.2.16 ⁇ Alistipes finegoldii DFI.2.10 ⁇ Alistipes finegoldii aa_0143 ⁇ Alistipes finegoldii 2789STDY560
  • Bacillus fennerii strain is one or a combination of several of the following strains:
  • the preservation number is CCUG46020 Bacillus phentrichum
  • the 16S rDNA sequence contained in the Alistipes finegoldii has at least 99% identity with the 16S rDNA sequence of Alistipes finegoldii, and the Alistipes finegoldii
  • the 16S rDNA sequence of Alistipes finegoldii is shown in SEQ ID NO:1.
  • the Alistipes finegoldii bacterium is a strain of Alistipes finegoldii deposited in DSM with accession number 17242.
  • the bacteria include one or more of live bacteria, derivatives of bacteria, or metabolites of bacteria.
  • the bacterium belonging to the genus Alistipes and the immune checkpoint inhibitor are administered simultaneously or separately.
  • said bacterial metabolites include all molecules produced or modified by said bacteria as a result of growth, survival, retention, transport or presence of said bacteria during their preparation and storage and during transit in the gastrointestinal tract of mammals.
  • said bacterial metabolites include all organic acids, inorganic acids, bases, proteins and peptides, enzymes and coenzymes, amino acids and nucleic acids, carbohydrates, lipids, glycoproteins, lipoproteins, glycolipids, vitamins, all biological Active compounds, metabolites with inorganic components and all small molecules, e.g. nitrogen- or sulfurous-containing molecules.
  • the bacterial derivatives include bacterial components, genetic material and related components, examples of which include bacterial cell membranes, pili, flagella, LPS, nucleic acid substances and other components derived from bacteria.
  • the live bacteria are whole bacteria.
  • the live bacteria are whole-bodied bacteria.
  • the treatment of the tumor is that the tumor shrinks or stabilizes, the overall survival time is prolonged, the progression-free survival period is prolonged, and the quality of life is improved.
  • the tumor is adenoma, malignant tumor and adenocarcinoma, wherein tumors are classified according to tissue origin or cell name, including: adrenocortical carcinoma, bladder urothelial carcinoma, breast cancer, pancreatic cancer, cervical cancer, cholangiocarcinoma, Colon cancer, colorectal cancer, diffuse large B-cell lymphoma, glioma multiforme, glioma, head and neck cancer, chromophobe renal cell carcinoma, mixed renal cancer, kidney cancer, leukemia, lymphoma, brain cancer , liver cancer, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma, ovarian cancer, pancreatic cancer, pheochromocytoma, paraganglioma, prostate cancer, rectal adenocarcinoma, sarcoma, skin melanoma, gastric cancer, esophageal cancer, testicular cancer Cancer, thyroid carcinoma
  • the tumor is colorectal cancer or melanoma.
  • the tumor is a malignant tumor, a metastatic tumor or a non-metastatic tumor.
  • the tumors herein include malignant, metastatic and non-metastatic types; including any stage of cancer (clinical stage I, II, III or IV, malignant tumor TNM classification T1-4, N0-4, or M0- 1, histological grade G1, G2, G3 or G4, etc.)
  • the immune checkpoint inhibitor is a combination of one or more blockers acting on T cell negative co-stimulatory (co-inhibitory) molecules and/or their respective ligands.
  • T cell negative co-stimulatory (co-inhibitory) molecules and/or their respective ligands are selected from CTLA-4, PD-1, PD-L1, PD-L2, B7-1, B7-2, B7 -H3, B7-H4, B7-H6, A2AR, IDO, TIM-3, BTLA, VISTA, TIGIT, LAG-3, CD40, KIR, CEACAM1, GARP, PS, CSF1R, CD94/NKG2A, TDO, TNFR, DcR3 .
  • the blocker of the ligand of the T cell negative co-stimulatory (co-inhibitory) molecule is selected from nivolumab (nivolumab, PD-1 monoclonal antibody), ipilimumab (ipilimumab, CTLA -4 monoclonal antibody), pembrolizumab (pembrolizumab, PD-1 monoclonal antibody), atezolizumab (azetolizumab, PD-L1 monoclonal antibody), atezolizumab (atezolizumab, PD-L1 monoclonal antibody), card Camrelizumab (camrelizumab, PD-L1 monoclonal antibody), tislelizumab (tislelizumab, BGB-A317), durvalumab (durvalumab, PD-L1 monoclonal antibody), tremelimumab (tremelimumab, CTLA-4 monoclonal antibody),
  • the immune checkpoint inhibitor is an inhibitor acting on the PD-1/PD-L1 signaling pathway and/or PD-1/PD-L2 signaling pathway, wherein PD-1 refers to programmed cell death Protein 1, also known as CD279, PD-L1 (B7-H1 or CD274) and PD-L2 (B7-DC or CD273) are ligands for PD-1.
  • PD-1 refers to programmed cell death Protein 1, also known as CD279
  • PD-L1 B7-H1 or CD274
  • PD-L2 B7-DC or CD273
  • the inhibitor of the PD-1/PD-L1 signaling pathway or PD-1/PD-L2 signaling pathway is selected from nivolumab (nivolumab, PD-1 monoclonal antibody), pembrolizumab Monoclonal antibody (pembrolizumab, PD-1 monoclonal antibody), atezolizumab (azetolizumab, PD-L1 monoclonal antibody), atezolizumab (atezolizumab, PD-L1 monoclonal antibody), camrelizumab (camrelizumab, PD-L1 monoclonal antibody) -L1 monoclonal antibody), tislelizumab (tislelizumab, BGB-A317), durvalumab (PD-L1 monoclonal antibody), spartalizumab (PD-1 monoclonal antibody) ), avelumab (avelumab, PD-L1 monoclonal antibody), sint
  • the immune checkpoint inhibitor is an inhibitor acting on the CTLA-4/B7-1 signaling pathway and/or the CTLA-4/B7-2 signaling pathway, wherein CTLA-4 refers to cytotoxic T lymphocyte Cytoprotein 4, also known as CD152, B7-1 (CD80) and B7-2 (CD86), are ligands for CTLA-4.
  • CTLA-4 refers to cytotoxic T lymphocyte Cytoprotein 4, also known as CD152, B7-1 (CD80) and B7-2 (CD86), are ligands for CTLA-4.
  • ipilimumab CTLA-4 monoclonal antibody
  • tremelimumab tremelimumab, CTLA-4 monoclonal antibody
  • MGD019 PD-1 and CTLA-4 monoclonal antibody
  • the immune checkpoint inhibitor is an inhibitor acting on the PD-1/PD-L1 signaling pathway and/or PD-1/PD-L2 signaling pathway and/or acting on CTLA Inhibitors of -4/B7-1 signaling pathway and/or CTLA-4/B7-2 signaling pathway.
  • the immune checkpoint inhibitor is PD-1 monoclonal antibody or CTLA-4 monoclonal antibody.
  • chemotherapy, immunotherapy or radiation therapy is performed simultaneously, separately or sequentially with the administration of P. fennerii.
  • the subject of administration of said application is a human being, which is an infant, child, adolescent, adult or elderly.
  • non-human primate being a mammal (e.g. dog, cat, ferret, horse, rabbit, guinea pig, gerbil, hamster, chinchilla) , rats, mice); birds; reptiles; fish; amphibians; arthropods or livestock animals (such as cattle, pigs, sheep, goats, alpacas, donkeys, camels, buffaloes or minks)
  • mammal e.g. dog, cat, ferret, horse, rabbit, guinea pig, gerbil, hamster, chinchilla
  • rats mice
  • reptiles fish
  • amphibians arthropods or livestock animals (such as cattle, pigs, sheep, goats, alpacas, donkeys, camels, buffaloes or minks)
  • the bacteria are contained in between 10 5 and 10 12 CFU (colony forming units), or between 10 7 and 10 11 CFU, or between 10 8 and 10 11 CFU, or A dose of between 10 9 and 10 11 CFU, or between 10 10 and 10 11 CFU is administered, more preferably a dose of between 10 9 and 10 11 CFU comprising the bacteria.
  • CFU colony forming units
  • a dose of between 10 9 and 10 11 CFU, or between 10 10 and 10 11 CFU is administered, more preferably a dose of between 10 9 and 10 11 CFU comprising the bacteria.
  • a kit for tumor treatment which includes one or more of the immune checkpoint inhibitors, and the bacteria described above, and optionally includes a container;
  • the immune checkpoint inhibitors consists of one or more of the immune checkpoint inhibitors and the bacteria, and optionally includes a container.
  • a bacterium comprising a 16S rDNA sequence that is at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% identical to the 16S rDNA sequence of Alistipes , at least 99.5% or 100% agreement.
  • a bacterium characterized in that the 16S rDNA sequence it comprises has at least 99% identity with the 16S rDNA sequence of Alistipes finegoldii.
  • a bacterium comprising a 16S rDNA sequence that is at least 99.5% or 100% identical to the 16S rDNA sequence of any of the following strains:
  • the preservation number is CCUG46020 Bacillus phentrichum
  • DSM 17242 The 16S rDNA sequence of Alistipes finegoldii DSM17242 is shown in SEQ ID NO: 1.
  • pharmaceutically acceptable carrier refers to a carrier that does not cause significant irritation to the organism and does not interfere with the biological activity and properties of the administered compound.
  • auxiliary material refers to solvent, diluent or other excipients, dispersant, surfactant.
  • composition comprising the bacteria described in any one and a pharmaceutically acceptable carrier.
  • composition comprising the bacterium of any one formulated for use as a medicament.
  • composition characterized in that it comprises the bacteria described in any of the above formulated for use as a medical food.
  • composition characterized in that it comprises a mixture of two or more bacterial strains described in any one of the above, and optionally further comprises a pharmaceutically acceptable carrier.
  • a composition characterized in that it comprises an effective amount of a mixture of two or more of any one of the bacteria for tumor treatment, and optionally also comprises a pharmaceutically acceptable carrier.
  • a composition characterized in that it comprises an effective amount of one of two or more live bacteria, bacterial derivatives, or bacterial metabolites of any of the bacteria described in any one of the two or more species used for tumor treatment or several compositions, and also optionally include a pharmaceutically acceptable carrier.
  • a food, beverage, food supplement, probiotic or health food comprising the bacteria described in any one, formulated as a preparation for enteral nutrition
  • a food, beverage, food supplement, probiotic or health food comprising a mixture of two or more of any of the bacteria described in any one, formulated as a preparation for enteral nutrition.
  • the bacterium, preparation, composition, food, beverage, food supplement, probiotic or health food described in any one is characterized in that the subject of administration is human.
  • said human is an infant, child, adolescent, adult or elderly.
  • any one of the bacteria, preparation, composition, food, beverage, food supplement, probiotic or health food comprising an effective amount of the bacteria, in the treatment of tumors.
  • Any one of the bacterium, preparation, composition, food, beverage, food supplement, probiotic or health food, comprising an effective amount of the bacterium, and its application in tumor treatment is characterized in that the tumor treatment Administration of any one of the bacteria, preparations, compositions, foods, beverages, food supplements, probiotics or health foods.
  • a composition comprising an effective amount of any one of the bacteria or composition for treating cancer, and optionally further comprising a pharmaceutically acceptable carrier.
  • a composition comprising an effective amount of any one of two or more bacteria or the composition for treating cancer, and optionally further comprising a pharmaceutically acceptable carrier.
  • a composition comprising Alistipes finegoldii and an immune checkpoint inhibitor.
  • the immune checkpoint inhibitor is PD-1 monoclonal antibody ( ⁇ PD-1) and/or CTLA-4 monoclonal antibody ( ⁇ CTLA4).
  • the present invention also claims the use of the composition in the preparation of medicaments for treating tumors.
  • the present invention also claims a medicine, including the composition, specifically, a pharmaceutical composition whose active ingredients are Alistipes finegoldii and immune checkpoint inhibitors for treating tumors,
  • the use of P. fennerii as an active ingredient in the preparation of a synergist of an immune checkpoint inhibitor, and the synergist can improve the effect of the immune checkpoint inhibitor in treating tumors.
  • the P. fennerii strain is one or a combination of several of the following strains:
  • the present invention proposes an active whole-cell Alistipes finegoldii for treating tumor patients, which can enhance the curative effect of immune checkpoint inhibitors.
  • the present invention proposes a combination therapy: the immune checkpoint inhibitory treatment will be performed simultaneously, separately or sequentially with the administration of P. fennerii, thereby increasing the therapeutic effect of immune checkpoint inhibition.
  • the sequence of administering the combined therapy is: administering the Alistipes finegoldii while, before and/or after the immune checkpoint inhibitory therapy.
  • the Alistipes finegoldii described in the present invention may comprise an effective amount of Alistipes finegoldii usually dispersed in a pharmaceutically or pharmacologically acceptable carrier.
  • pharmaceutically or pharmacologically acceptable refers to molecular entities and compositions that do not produce adverse, allergic or other adverse reactions when administered to an animal (eg, a human, if appropriate).
  • pharmacologically acceptable carriers as described herein are borate buffer or sterile saline solution.
  • the synergistic immune checkpoint inhibitors described in the present invention can be used in patients refractory to immune checkpoint inhibitors, wherein the patients refractory to immune checkpoint inhibitors show innate innate resistance to the treatment of immune checkpoint inhibitors Sexual (primary) resistance, manifested as a lack of response or an insufficient response to the checkpoint inhibitor therapy for at least about 8 weeks or 12 weeks from the first dose.
  • the synergistic immune checkpoint inhibitors described in the present invention can be used in patients refractory to immune checkpoint inhibitors, wherein the patients refractory to immune checkpoint inhibitors show acquired resistance to the treatment of the checkpoint inhibitors (Secondary) resistance, manifested as an initial response to said checkpoint therapy, but one or more tumors subsequently recur and develop.
  • Therapeutic effect of the combination therapy described in the present invention is assessed, the enhanced therapeutic effect is measured by increased overall survival time.
  • Therapeutic effect of the combination therapy described in the present invention is assessed with enhanced therapeutic effect as measured by improved overall response rate and/or enhanced quality of life.
  • the safety assessment of the combination therapy described in the present invention is measured by whether it induces diarrhea or enteritis.
  • the present invention has the following beneficial effects:
  • the anti-tumor immune protection response generated by bacterial stimulation can significantly enhance the efficacy of immune checkpoint inhibitors on many kinds of tumors, and the safety is better , prolong the overall survival time of cancer patients, improve the response rate of cancer immunotherapy populations, and expand the population of tumor patients who benefit from cancer immunotherapy (immunotherapy checkpoint inhibitors).
  • Fig. 1 is the flowchart of embodiment 1.
  • Figure 2 is a photo of the tumor (day 21).
  • Figure 3 is the curve of tumor volume change.
  • Figure 4 is a statistical chart of tumor weight.
  • Figure 5 is a photo of the mouse anus (day 21)
  • Fig. 6 is a HE staining image of mouse intestinal tissue (day 21).
  • Figure 7 shows the infiltration of immune cells in tumor tissue (day 21).
  • Figure 8 is the survival curve of mice.
  • Fig. 9 shows the distribution and relative abundance of Bacillus fennerii in different populations.
  • test methods used in the following examples are conventional methods unless otherwise specified; the materials and reagents used are commercially available reagents and materials unless otherwise specified.
  • Example 1 The therapeutic effect of P. fennerii combined with immune checkpoint inhibitors on tumors
  • Tumor cell lines mouse melanoma cell line (B16-OVA, ATCC), mouse intestinal cancer cell line (MC38, ATCC)
  • Alistipes finegoldii (Alistipes finegoldii, DSM No.: 17242, Type strain, its 16S rDNA sequence is shown in SEQ ID NO: 1), referred to as Af, commercially purchased from DSMZ German National Culture Depository Center, (official website of DSMZ: http://www.dsmz.de).
  • Bacterial culture medium liquid DSMZ104 medium, the formula mainly includes peptone, yeast extract, beef extract and glucose, etc., commercially purchased from DSMZ German National Culture Collection Center.
  • Immune checkpoint inhibitor PD-1 monoclonal antibody ( ⁇ PD-1), clone number G4C2, the reagent was presented by Shanghai Junshi Biomedical Technology Co., Ltd.
  • Antibiotic combination metronidazole 100mg/kg, vancomycin 50mg/kg, penicillin sodium 100mg/kg, neomycin sulfate 100mg/kg
  • Bacterial culture inoculate Bacillus fennerii in DSMZ104 liquid medium, culture in an anaerobic box at 37°C for 18 hours, and centrifuge to a concentration of 1 ⁇ 10 10 CFU/ml.
  • mice in each group were intragastrically administered antibiotic combinations to eliminate the intestinal flora.
  • IgG or ⁇ PD-1 were injected intraperitoneally on the 5th, 8th, and 11th days, respectively, 200 ⁇ g/rat.
  • the Af live bacterial liquid was administered to the bacteria preparation by intragastric administration, 100 ⁇ l/cattle, 1 ⁇ 10 9 CFU/cattle.
  • the tumor size was measured on the 5th, 8th, 11th, 14th, 17th, and 21st day, and the tumor volume was calculated.
  • mice On the 21st day, the mice were euthanized, the tumor tissues were taken out, photographed and weighed, and the intestinal tissues were taken for HE staining to confirm the enteritis:
  • FIG. 2 shows tumors on day 21.
  • Fig. 3 is the curve of tumor volume change and
  • Fig. 4 is the statistical graph of tumor weight.
  • the single-drug immune checkpoint inhibitor group ⁇ PD-1
  • both the combination therapy group ⁇ PD-1+Af
  • p ⁇ 0.01 tumor reduction demonstrated that P. fennerii has enhanced ⁇ PD-1 anti-tumor effect.
  • Tumor cell lines mouse melanoma cell line (B16-OVA, ATCC), mouse intestinal cancer cell line (MC38, ATCC)
  • Alistipes finegoldii (Alistipes finegoldii, DSM No.: 17242, Type strain, its 16S rDNA sequence is shown in SEQ ID NO: 1), referred to as Af, commercially purchased from DSMZ German National Culture collection center.
  • Bacterial culture medium liquid DSMZ104 medium, the formula mainly includes peptone, yeast extract, beef extract and glucose, etc., commercially purchased from DSMZ German National Culture Collection.
  • Immune checkpoint inhibitor PD-1 monoclonal antibody ( ⁇ PD-1), clone number G4C2, the reagent was presented by Shanghai Junshi Biomedical Technology Co., Ltd.
  • Antibiotic combination metronidazole 100mg/kg, vancomycin 50mg/kg, penicillin sodium 100mg/kg, neomycin sulfate 100mg/kg
  • Bacterial culture inoculate Bacillus fennerii in DSMZ104 liquid medium, culture in an anaerobic box at 37°C for 18 hours, and centrifuge to a concentration of 1 ⁇ 10 10 CFU/ml.
  • mice in each group were intragastrically administered antibiotic combinations to eliminate the intestinal flora.
  • IgG or ⁇ PD-1 or ⁇ CTLA4 was injected intraperitoneally every 3 days, 200 ⁇ g per mouse.
  • the tumor size was measured every 3 days, and the tumor volume was calculated.
  • Figure 8 is the survival curve of mice, in the MC38 intestinal cancer and B16-OVA melanoma mouse models, relative to the single-drug immune checkpoint inhibitor group ( ⁇ PD-1), Af high (high), medium (medium) And low (low) dose combination therapy group ( ⁇ PD-1+Af) can significantly increase the survival time of tumor-bearing mice.
  • ⁇ PD-1 single-drug immune checkpoint inhibitor group
  • Af high (high), medium (medium) And low (low) dose combination therapy group
  • ⁇ PD-1+Af low
  • ⁇ CTLA4 exhibited a promoting effect on the efficacy of ⁇ CTLA4 immune checkpoint inhibitors: ⁇ CTLA4 combined with high (high), medium (medium) and low (low) doses of Af could significantly increase Survival time of tumor-bearing mice.
  • the synergistic effect of Af on ⁇ CTLA4 was not observed due to the strong pharmacodynamic effect of ⁇ CTLA4 immune checkpoint inhibitors.
  • Example 3 Alistipes finegoldii is human endogenous intestinal commensal bacteria
  • Metagenome sequencing technology can achieve the accuracy of identification of human gut bacteria level.
  • 9 different types of populations include: 1) healthy adults; 2) patients with colorectal adenoma; 3) patients with colorectal cancer; 4) patients after surgery for colorectal cancer; 5) patients with atherosclerotic disease; NSCLC patients before immune checkpoint inhibitors (ICIs); 7) NSCLC patients after receiving immune checkpoint inhibitors (ICIs); 8) kidney cancer before receiving immune checkpoint inhibitors (ICIs) Patient gut microbiota metagenomic data; 9) Kidney cancer patients after receiving immune checkpoint inhibitors (ICIs).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Polymers & Plastics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Food Science & Technology (AREA)
  • Mycology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Molecular Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Nutrition Science (AREA)
  • Organic Chemistry (AREA)
  • Physiology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Animal Husbandry (AREA)
  • Zoology (AREA)
  • Endocrinology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Oncology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

一种细菌在制备免疫检查点抑制剂的增效剂中的应用。通过使用人体内源性肠道细菌单菌口服制剂,联合免疫检查点抑制剂,由内源性肠道菌刺激产生的抗肿瘤免疫保护反应,显著增强免疫检查点抑制剂对多个瘤种的药效,且安全性更好,延长癌症患者整体存活时间,提高癌症免疫治疗人群的响应率,扩大癌症免疫治疗的受益肿瘤患者人群。

Description

一种细菌在制备免疫检查点抑制剂的增效剂中的应用 技术领域
本发明涉及癌症治疗技术领域,具体地,涉及一种细菌在制备免疫检查点抑制剂的增效剂中的应用。
背景技术
恶性肿瘤是目前全球面临的最大问题之一,在每年引发死亡的各类致死因素中,恶性肿瘤位居首位。常规治疗方案,如手术治疗,放疗和化疗,很难将肿瘤细胞完全清除,复发率高,且放化疗等治疗手段因靶向性不强,在杀死肿瘤细胞的同时对正常的细胞也具有杀伤作用,对患者造成很大的伤害,严重影响患者生活质量。
相比于常规疗法,肿瘤免疫疗法针对的靶标不是肿瘤细胞和组织,而是人体自身的免疫系统,通过调节和激活人体免疫系统,依靠自身免疫机清除肿瘤细胞,包括转移至远处的肿瘤细胞,对正常细胞毒副作用小,极大改善晚期恶性肿瘤患者的预后和生活质量。
目前肿瘤免疫疗法多种多样,其中免疫检查点抑制剂(immune checkpoint inhibitors,ICIs)疗法通过阻断被肿瘤细胞劫持的免疫抑制通路(如PD-L1/PD-1和CTLA-4/B7-1)来重新活化T细胞,重新激活机体的免疫系统对肿瘤细胞的识别与清除能力,具有疗效好,反应持久的优势,开创了肿瘤治疗的新时代。ICIs疗法在因缺乏治疗选择的多种肿瘤类型的应用中引发了癌症治疗的变革,成为癌症治疗的新里程碑。
目前,接受ICIs治疗的患者在治疗中断仍后可长期缓解,为某些患者增加了治愈希望。尤其在黑色素瘤患者中,患者可以实现完全缓解,即所有可见肿瘤转移灶完全消失。ICIs单药在患者中响应率低的问题成为其目前发展和应用的最大瓶颈,能够在单药ICIs治疗中到达完全缓解的患者仅占有20%~40%,亟需开发增效免疫检查点抑制剂功效的联合策略。目前主流的联合策略包括手术、化疗、放疗、靶向治疗等治疗手段与ICIs的联合疗法,可产生协同作用,增强ICIs的持久性和疗效,克服ICIs的响应率低的问题。尽管这些联合疗法可在一定程度上提高ICIs的疗效和响应率,但通常毒性也会随之增加。
在众多联合疗法中,通过操纵内源性肠道微生物辅助ICIs治疗的多项研究成为肿瘤治疗领域中的里程碑事件。肠道是人体内最大的淋巴器官,存在超过70%的T细胞,是绝多数记忆T细胞的居住地。寄居于肠道的肠道菌群通过与肠道免疫器官的相互作用促进人体免疫系统的发育,平衡并奠定人体的免疫基调。对于人体的防御系统和免疫系统来说,细 菌和肿瘤均为“非己”,因此人体在对细菌和肿瘤的免疫反应中使用了同一套防御机制,肠道内源性细菌刺激的免疫反应同时具有抗肿瘤的潜能。基于这一肠道菌群的抗肿瘤免疫交叉理论及某些肠道细菌自身分泌的的溶瘤酶,细菌治疗肿瘤已有数百年的历史。值得注意的是,由肠道内源性细菌诱导产生的抗肿瘤效应来源于机体自身免疫反应,相对于放化疗的联合疗法,具有较高的安全性和较小的毒副作用。
近期两项临床研究报道,接受ICIs治疗响应(达到部分缓解或完全缓解)患者的粪菌移植可使ICIs无响应的难治性转移性黑色素瘤患者达到部分或完全缓解(Baruch E N,Youngster I,Ben-Betzalel G,et al.Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients[J].Science,371.以及Davar D,Dzutsev A K,Mcc Ulloch J A,et al.Fecal microbiota transplant overcomes resistance to anti–PD-1 therapy in melanoma patients[J].Science,2021,371(6529):595-602.),且无毒副作用,力证了内源性肠道微生物对ICIs疗法的增效作用。粪菌移植成分复杂,在粪菌移植中发挥功效的是肠道微生物整体,且肠道菌群受环境,饮食及生活方式等的影响很大,无法获取持续稳定的供体粪菌来源限制了粪菌移植ICIs联合免疫疗法在临床中的应用。因此鉴定可标准化量化生产的成分单一的抗肿瘤肠道菌成为解决这一问题的关键。在一项最新研究中(Mager LF,Burkhard R,Pett N,Cooke NCA,Brown K,Ramay H,Paik S,Stagg J,Groves RA,Gallo M,Lewis IA,Geuking MB,McCoy KD.Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy.Science.2020 Sep 18;369(6510):1481-1489.),研究人员从ICIs治疗的肠癌小鼠模型中分离出三种细菌:假长双歧杆菌(Bifidobacterium pseudolongum)、约氏乳杆菌(Lactobacillus johnsonii)和欧陆森氏菌(Olsenella species)。这3种特殊细菌分别联合免疫检查点抑制剂可在4种小鼠癌症模型中增强ICIs的疗效。进一步研究发现,其中假长双歧杆菌可通过其代谢产物肌苷来发挥ICIs的增效作用。
发明内容
本发明的目的是为了克服现有技术的上述不足,提供细菌在制备免疫检查点抑制剂的增效剂中的应用,本发明涉及一种增效免疫检查点抑制剂疗效的联合疗法,适用于一种或多种肿瘤,以及适用于所述方法的活性全细胞芬氏别样杆菌(Alistipes finegoldii),其中癌症免疫治疗涉及免疫检查点抑制剂疗法,目的是增强癌症免疫治疗的抗肿瘤药效,延长癌症患者整体存活时间,提高癌症免疫治疗人群的响应率,扩大癌症免疫治疗的受益肿瘤患者人群。
本发明给出的治疗方法,针对常规疗法的缺点:毒副作用大、易转移复发、持续时间短、生存期短、和生活质量差,有明显的改善;针对免疫检查点单药疗法的缺点:作用瘤 种范围有限和药物响应人群少,有明显的改善;针对免疫检查点联合放化疗疗法的缺点:毒副作用大和药物响应人群少,有明显的改善。
本发明给出的治疗方法针对以下患者都有很好的治疗效果:治疗不可手术,无可用靶向药物,放疗,化疗等无效的肿瘤患者;治疗单药免疫检查点抑制剂无效或耐药(原发性、适应性和获得性耐药)的肿瘤患者;治疗免疫检查点抑制剂联合放疗、化疗、靶向治疗无效或耐药(原发性、适应性和获得性耐药)的肿瘤患者。
为了实现上述目的,本发明是通过以下方案予以实现的:
一种细菌在肿瘤治疗上的应用。
优选地,所述应用为作为活性成分在制备免疫检查点抑制剂的增效剂中的应用。
优选地,所述应用为作为免疫检查点抑制剂的增效剂的应用,提高免疫检查点抑制剂治疗肿瘤的效果。
优选地,所述细菌菌种属于别样杆菌属(Alistipes),即,所述细菌菌种的16S rDNA序列与别样杆菌属(Alistipes)的16S rDNA序列具有至少85%,至少90%,至少95%,至少96%,至少97%,至少98%,至少99%,至少99.5%或100%的一致性。
优选地,所述属于别样杆菌属(Alistipes)的细菌为一个或多个属于别样杆菌属(Alistipes)的细菌的菌种或菌株的组合。
更优选地,所述属于别样杆菌属(Alistipes)细菌为芬氏别样杆菌(Alistipes finegoldii)。
进一步优选地,所述芬氏别样杆菌(Alistipes finegoldii),其包含的16S rDNA序列,其与芬氏别样杆菌菌种(Alistipes finegoldii)的16S rDNA序列具有至少99%的一致性。
进一步优选地,所述芬氏别样杆菌(Alistipes finegoldii),其包含的16S rDNA序列,与芬氏别样杆菌菌株DSM17242(Alistipes finegoldii DSM17242)的16S rDNA序列具有至少99.5%或100%的一致性。
再进一步优选地,所述芬氏别样杆菌(Alistipes finegoldii)的一个或多个芬氏别样杆菌(Alistipes finegoldii)菌株的组合。
再进一步更优选地,所述芬氏别样杆菌(Alistipes finegoldii)的菌株为美国国家生物技术中心(National Center for Biotechnology Information,简称NCBI)基因组数据库(https://www.ncbi.nlm.nih.gov/genome/browse/#!/prokaryotes/11196/)中亚种分类的菌株名称(Strain name of sub species classification)为:Alistipes finegoldii DSM 17242、Alistipes finegoldii D53t1_180928_D3、Alistipes finegoldii 2789STDY5834947、Alistipes finegoldii 1001713B170207_170306_H2、Alistipes finegoldii DFI.2.31、Alistipes finegoldii BIOML-A1、 Alistipes finegoldii DFI.2.16、Alistipes finegoldii DFI.2.10、Alistipes finegoldii aa_0143、Alistipes finegoldii 2789STDY5608890、Alistipes finegoldii MGBC116453、Alistipes finegoldii COPD076、或Alistipes finegoldii UBG195中的一个或几个的组合。
再进一步优选地,所述芬氏别样杆菌菌株为以下菌株的中的一个或几个的组合:
保藏于德国DSM微生物和细胞培养物保藏中心,保藏号为DSM 17242的芬氏别样杆菌(NCBI:txid679935,https://www.ncbi.nlm.nih.gov/Taxonomy/Browser/wwwtax.cgi?id=679935);
保藏于日本JCM菌种保藏中心(Deutsche Sammlung von Mikroorganismen und Zellkulturen=German collection of microorganisms and cell cultures),保藏号为JCM16770的芬氏别样杆菌;
保藏于韩国KCTC菌种保藏中心(Korean Collection for Type Cultures),保藏号为KCTC 15236的芬氏别样杆菌;
保藏于芬兰赫尔辛基厌氧菌参考实验室(Anaerobe Reference Laboratory,Helsinki Collection,National Public health Institute,Helsinki,Finland),保藏号为AHN 2437的芬氏别样杆菌;
保藏于瑞典CCUG菌种保藏中心(Culture Collection University of Gothenburg),保藏号为CCUG46020的芬氏别样杆菌;
保藏于法国CIP菌种保藏中心(Collection de L'Institut Pasteur of Institut Pasteur),保藏号为CIP107999的芬氏别样杆菌。
保藏于中国广东省微生物菌种保藏中心,保藏号为GDMCC 1.2324的芬氏别样杆菌。
优选地,所述芬氏别样杆菌(Alistipes finegoldii),其包含的16S rDNA序列,其与芬氏别样杆菌菌种(Alistipes finegoldii)的16S rDNA序列具有至少99%的一致性,所述芬氏别样杆菌菌种(Alistipes finegoldii)的16S rDNA序列如SEQ ID NO:1所示。
最优选地,所述芬氏别样杆菌细菌为保藏于DSM保藏号17242的芬氏别样杆菌(Alistipes finegoldii)菌株。
优选地,所述细菌,包括细菌活菌、细菌衍生物、或细菌代谢产物中的一种或几种。
更优选地,同时或分别施用属于别样杆菌属(Alistipes)的细菌和免疫检查点抑制剂。
优选地,所述细菌代谢物包括细菌制备和储存过程中以及在哺乳动物胃肠道转运过程中作为细菌生长、生存、留滞、转运或存在的结果由所述细菌产生或修饰的所有分子。
更优选地,所述细菌代谢物包括所有有机酸、无机酸、碱、蛋白质和肽、酶和辅酶、 氨基酸和核酸、碳水化合物、脂质、糖蛋白、脂蛋白、糖脂、维生素、所有生物活性化合物、含有无机成分的代谢物以及所有小分子,例如含氮分子或含亚硫酸分子。
优选地,所述细菌衍生物,包括细菌组成成分和遗传物质及其相关成分,其实例包括细菌细胞膜,菌毛,鞭毛,LPS,核酸物质等来源于细菌的成分。
优选地,所述细菌活菌为完整细菌。
更优选地,所述细菌活菌为整活力菌。
优选地,述治疗肿瘤为肿瘤缩小或稳定、总生存时间延长、无恶化生存期延长、生活质量改善。
优选地,所述肿瘤为腺瘤、恶性肿瘤和腺癌,其中肿瘤按照组织来源或细胞名称分类,包括:肾上腺皮质癌、膀胱尿路上皮癌、乳腺癌、胰腺癌、宫颈癌、胆管癌、结肠癌、结直肠癌、弥漫性大B细胞淋巴瘤、多形成性胶质细胞瘤、胶质瘤、头颈癌、肾嫌色细胞癌、混合肾癌、肾癌、白血病、淋巴瘤、脑癌、肝癌、肺腺癌、肺鳞癌、间皮瘤、卵巢癌、胰腺癌、嗜铬细胞瘤、副神经节瘤、前列腺癌、直肠腺癌、肉瘤、皮肤黑色素瘤、胃癌、食管癌、睾丸癌、甲状腺癌、胸腺癌、子宫内膜癌、子宫肉瘤、葡萄膜黑色素瘤和软组织肉瘤中的一种或几种。
在本发明的一个具体的实施例中,肿瘤为结直肠癌或黑色素瘤。
优选地,所述肿瘤为恶性肿瘤、转移性肿瘤或非转移性肿瘤。
优选地,在此所述肿瘤包括恶性、转移性和非转移性类型;包括癌症的任何阶段(临床分期I、II、III或IV,恶性肿瘤TNM分类T1-4,N0-4、或M0-1,组织学等级G1、G2、G3或G4等)
更优选地,所述免疫检查点抑制剂为作用于T细胞负性共刺激(共抑制)分子和/或它们各自的配体的阻断剂的一种或多种的组合。
进一步优选地,T细胞负性共刺激(共抑制)分子和/或它们各自的配体选自CTLA-4、PD-1、PD-L1、PD-L2、B7-1、B7-2、B7-H3、B7-H4、B7-H6、A2AR、IDO、TIM-3、BTLA、VISTA、TIGIT、LAG-3、CD40、KIR、CEACAM1、GARP、PS、CSF1R、CD94/NKG2A、TDO、TNFR、DcR3。
再进一步优选地,所述T细胞负性共刺激(共抑制)分子的配体的阻断剂选自纳武利尤单抗(nivolumab,PD-1单抗)、伊匹单抗(ipilimumab,CTLA-4单抗)、帕博利珠单抗(pembrolizumab,PD-1单抗)、阿托珠单抗(azetolizumab,PD-L1单抗)、阿特朱单抗(atezolizumab,PD-L1单抗)、卡瑞珠丽单抗(camrelizumab,PD-L1单抗)、替雷丽珠单 抗(tislelizumab,BGB-A317)、度伐利尤单抗(durvalumab,PD-L1单抗)、曲美木单抗(tremelimumab,CTLA-4单抗)、斯巴达珠单抗(spartalizumab,PD-1单抗)、阿维鲁单抗(avelumab,PD-L1单抗)、信迪利单抗(sintilimab,PD-1单抗)、特瑞普利单抗(toripalimab,PD-1单抗)、西米普利单抗(cemiplimab,PD-1单抗)、MGA012(retifanlimab,PD-1单抗)、MGD013(tebotelimab,PD-1/LAG-3双抗)、MGD019(PD-1/CTLA-4双抗)、恩比利珠单抗(enoblituzumab,B7-H3单抗)、MGD009(B7-H3单抗)、MGC018(B7-H3单抗)、MEDI0680(PD-1单抗)、PDR001(PD-1单抗)FAZ053(PD-L1单抗)、TSR022(TIM-3单抗体)、MBG453(TIM-3单抗)、relatlimab(BMS986016,LAG-3单抗)、LAG525(LAG-3单抗)、IMP321(LAG-3单抗)、REGN3767(LAG-3单抗)、培西达替尼(pexidartinib,CSF-1R单抗)、LY3022855(CSF-1R单抗)、FPA008(CSF-1R单抗)、BLZ945(CSF-1R单抗)、GDC0919(navoximod,IDO单抗)、艾帕斯塔(epacadostat,IDO单抗)、indoximid(IDO单抗)、BMS986205(IDO单抗)、CPI‐444(A2AR单抗)、MEDI9447(oleclumab,CD73单抗)、PBF509(A2AR单抗)、利瑞路单抗(lirilumab,KIR单抗)或其中任意几种的组合;优选地,其中所述阻断剂选自纳武利尤单抗、帕博利珠单抗、特瑞普利单抗、信迪利单抗、西米普利单抗或其中任意几种的组合。
更进一步优选地,所述免疫检查点抑制剂为作用于PD-1/PD-L1信号通路和/或PD-1/PD-L2信号通路的抑制剂,其中PD-1是指程序性细胞死亡蛋白1,又称CD279,PD-L1(B7-H1或CD274)和PD-L2(B7-DC或CD273)是PD-1的配体。
再更进一步优选地,所述PD-1/PD-L1信号通路或PD-1/PD-L2信号通路的抑制剂选自纳武利尤单抗(nivolumab,PD-1单抗)、帕博利珠单抗(pembrolizumab,PD-1单抗)、阿托珠单抗(azetolizumab,PD-L1单抗)、阿特朱单抗(atezolizumab,PD-L1单抗)、卡瑞珠丽单抗(camrelizumab,PD-L1单抗)、替雷丽珠单抗(tislelizumab,BGB-A317)、度伐利尤单抗(durvalumab,PD-L1单抗)、斯巴达珠单抗(spartalizumab,PD-1单抗)、阿维鲁单抗(avelumab,PD-L1单抗)、信迪利单抗(sintilimab,PD-1单抗)、特瑞普利单抗(toripalimab,PD-1单抗)、西米普利单抗(cemiplimab,PD-1单抗)、MGA012(retifanlimab,PD-1单抗)、MGD013(tebotelimab,PD-1/LAG-3双抗)、MGD019(PD-1/CTLA-4双抗)、MEDI0680(PD-1单抗)、PDR001(PD-1单抗)、FAZ053(PD-L1单抗)或其中任意几种的组合。
更进一步优选地,所述免疫检查点抑制剂为作用于CTLA-4/B7-1信号通路和/或CTLA-4/B7-2信号通路的抑制剂,其中CTLA-4是指胞毒性T淋巴细胞蛋白4,又称CD152,B7-1(CD80)和B7-2(CD86)是CTLA-4的配体。
再更进一步优选地,其可选自伊匹单抗(ipilimumab,CTLA-4单抗)、曲美木单抗(tremelimumab,CTLA-4单抗)、MGD019(PD-1和CTLA-4双抗)或其中任意几种的组合。
作为本发明的一个具体的实施例,所述免疫检查点抑制剂为作用于PD-1/PD-L1信号通路和/或PD-1/PD-L2信号通路的抑制剂和/或作用于CTLA-4/B7-1信号通路和/或CTLA-4/B7-2信号通路的抑制剂。
具体地,所述免疫检查点抑制剂为PD-1单克隆抗体或CTLA-4单克隆抗体。
优选地,在与芬氏别样杆菌的施用同时、分别或顺序进行化学治疗,免疫治疗或放射治疗。
优选地,所述应用的施用的对象是人类,所述人类是婴幼儿,儿童,青少年,成年人或老人。
优选地,其中所述应用施用的对象是非人类灵长类动物,所述非人类灵长类动物是哺乳动物(例如狗、猫、雪貂、马、兔、豚鼠、沙鼠、仓鼠、灰鼠、大鼠、小鼠);鸟类;爬行动物;鱼;两栖动物;节肢动物或家畜动物(例如牛、猪、羊、山羊、羊驼、驴、骆驼、水牛或水貂)
优选地,将所述细菌以包含所述细菌的10 5至10 12个CFU(菌落形成单位)之间,或以10 7至10 11CFU之间、或10 8至10 11CFU之间、或10 9至10 11CFU之间、或10 10至10 11CFU之间的剂量施用,更优选地,以包含所述细菌的10 9至10 11CFU之间的剂量施用。
本发明还要求保护以下内容:
一种用于肿瘤治疗的试剂盒,其包含所述的免疫检查点抑制剂的一种或多种,以及权所述的细菌,并且任选地包含容器;
或由所述的免疫检查点抑制剂的一种或多种,以及所述的细菌组成,并且任选地包含容器。
一种细菌,其包含的16S rDNA序列,与别样杆菌(Alistipes)的16S rDNA序列具有至少85%,至少90%,至少95%,至少96%,至少97%,至少98%,至少99%,至少99.5%或100%的一致性。
一种细菌,其特征在于,其包含的16S rDNA序列,与芬氏别样杆菌(Alistipes finegoldii)的16S rDNA序列具有至少99%的一致性。
一种细菌,其包含的16S rDNA序列,其16S rDNA序列与以下菌株的中的任意一个的16S rDNA序列具有至少99.5%或100%的一致性:
保藏于德国DSM微生物和细胞培养物保藏中心,保藏号为DSM 17242的芬氏别样杆菌(NCBI:txid679935,https://www.ncbi.nlm.nih.gov/Taxonomy/Browser/wwwtax.cgi?id=679935);
保藏于日本JCM菌种保藏中心(Deutsche Sammlung von Mikroorganismen und Zellkulturen=German collection of microorganisms and cell cultures),保藏号为JCM16770的芬氏别样杆菌;
保藏于韩国KCTC菌种保藏中心(Korean Collection for Type Cultures),保藏号为KCTC 15236的芬氏别样杆菌;
保藏于芬兰赫尔辛基厌氧菌参考实验室(Anaerobe Reference Laboratory,Helsinki Collection,National Public health Institute,Helsinki,Finland),保藏号为AHN 2437的芬氏别样杆菌;
保藏于瑞典CCUG菌种保藏中心(Culture Collection University of Gothenburg),保藏号为CCUG46020的芬氏别样杆菌;
保藏于法国CIP菌种保藏中心(Collection de L'Institut Pasteur of Institut Pasteur),保藏号为CIP107999的芬氏别样杆菌。
保藏于中国广东省微生物菌种保藏中心,保藏号为GDMCC 1.2324的芬氏别样杆菌。
其中保藏号为:DSM 17242的芬氏别样杆菌(Alistipes finegoldii DSM17242)的16S rDNA序列如SEQ ID NO:1所示。
一种制剂,其特征在于,将有效量的用于治疗肿瘤的权利要求32到33中任一项所述的细菌中的一种或多种的组合物,加入药学上可接受的载体和辅料制备成冻干粉,片剂、胶囊剂、颗粒剂或针剂。
其中“药学上可接受的载体”是指对有机体不引起明显刺激性和不干扰所给予化合物的生物活性和性质的载体。其中“辅料”是指溶剂、稀释剂或其它赋形剂、分散剂、表面活性剂。
一种组合物,其包含任一项中所述的细菌和药学上可接受的载体。
一种组合物,其包含配制成用作药物的任一项中所述的细菌。
一种组合物,其特征在于,其包含配制为用作医疗食品的任一项中所述的细菌。
一种组合物,其特征在于,其包含两种或更多种任一项中所述的细菌菌株的混合物,并且任选地还包含药学上可接受的载体。
一种组合物,其特征在于,其包含有效量的用于肿瘤治疗的两种或更多种任一项所述 的细菌的混合物,并且还任选地包含药学上可接受的载体。
一种组合物,其特征在于,其包含有效量的用于肿瘤治疗的两种或更多种任一项中所述的细菌的细菌活菌、细菌衍生物、或细菌代谢产物中的一种或几种组合物,并且还任选地包含药学上可接受的载体。
一种食品、饮料、食品补充剂、益生菌或保健食品,其包含任一项中所述的细菌,被配制成制剂用于肠内营养
一种食品、饮料、食品补充剂、益生菌或保健食品,其包含两种或更多种任一项中所述的细菌的混合物,被配制成制剂用于肠内营养。
任一项细菌或组合物,其中所述细菌或组合物被制备成冻干粉,片剂、胶囊剂、颗粒剂或针剂。
任一项所述的细菌、制剂、组合物、食品、饮料、食品补充剂、益生菌或保健食品,其特征在于,其施用对象为人类。
优选地,其中所述人类是婴幼儿、儿童、青少年、成年人或老人。
任一项所述的细菌、制剂、组合物、食品、饮料、食品补充剂、益生菌或保健食品,施用于非人类灵长类动物,所述非人类灵长类动物是哺乳动物(例如狗、猫、雪貂、马、兔、豚鼠、沙鼠、仓鼠、灰鼠、大鼠、小鼠);鸟类;爬行动物;鱼;两栖动物;节肢动物或家畜动物(例如牛、猪、羊、山羊、羊驼、驴、骆驼、水牛或水貂)。
任一项所述的细菌、制剂、组合物、食品、饮料、食品补充剂、益生菌或保健食品,在肿瘤治疗中的应用。
任一项所述的细菌、制剂、组合物、食品、饮料、食品补充剂、益生菌或保健食品,包含有效量的所述细菌,在肿瘤治疗中的应用。
任一项所述的细菌、制剂、组合物、食品、饮料、食品补充剂、益生菌或保健食品,包含有效量的所述细菌,在肿瘤治疗中的应用,其特征在于,所述肿瘤治疗施用任一项所述的细菌、制剂、组合物、食品、饮料、食品补充剂、益生菌或保健食品。
一种组合物,其包含用于治疗癌症的有效量的任一项细菌或组合物,并且任选地还包含药学上可接受的载体。
一种组合物,其包含有效量的用于治疗癌症的两种或更多种中任一项细菌或组合物,并且还任选地包含药学上可接受的载体。
一种组合物,包括芬氏别样杆菌(Alistipes finegoldii)和免疫检查点抑制剂。
优选地,所述免疫检查点抑制剂为PD-1单克隆抗体(αPD-1)和/或CTLA-4单克隆 抗体(αCTLA4)。
本发明还要求保护所述组合物在制备肿瘤治疗的药物中的应用。
本发明还要求保护一种药物,包括所述的组合物,具体地,一种活性成分为芬氏别样杆菌(Alistipes finegoldii)和免疫检查点抑制剂治疗肿瘤的药物组合物,
属于别样杆菌属(Alistipes)的细菌的细菌活菌、细菌衍生物、或细菌代谢产物中的一种或几种在制备免疫检查点抑制剂治疗肿瘤的增效剂中的应用。
优选地,芬氏别样杆菌作为活性成分在制备免疫检查点抑制剂的增效剂中的应用,所述增效剂能够提高免疫检查点抑制剂治疗肿瘤的效果。
更优选地,述芬氏别样杆菌菌株为以下菌株的中的一个或几个的组合:
保藏于德国DSM微生物和细胞培养物保藏中心,保藏号为DSM 17242的芬氏别样杆菌(NCBI:txid679935,https://www.ncbi.nlm.nih.gov/Taxonomy/Browser/wwwtax.cgi?id=679935);
保藏于日本JCM菌种保藏中心(Deutsche Sammlung von Mikroorganismen und Zellkulturen=German collection of microorganisms and cell cultures),保藏号为JCM16770的芬氏别样杆菌;
保藏于韩国KCTC菌种保藏中心(Korean Collection for Type Cultures),保藏号为KCTC 15236的芬氏别样杆菌;
保藏于芬兰赫尔辛基厌氧菌参考实验室(Anaerobe Reference Laboratory,Helsinki Collection,National Public health Institute,Helsinki,Finland),保藏号为AHN 2437的芬氏别样杆菌;
保藏于瑞典CCUG菌种保藏中心(Culture Collection University of Gothenburg),保藏号为CCUG46020的芬氏别样杆菌;
保藏于法国CIP菌种保藏中心(Collection de L'Institut Pasteur of Institut Pasteur),保藏号为CIP107999的芬氏别样杆菌。
保藏于中国广东省微生物菌种保藏中心,保藏号为GDMCC 1.2324的芬氏别样杆菌。
本发明提出一种用于治疗肿瘤患者的活性全细胞芬氏别样杆菌(Alistipes finegoldii),其增效免疫检查点抑制剂疗效。具体地,本发明提出一种联合疗法:将在与芬氏别样杆菌的施用同时、分别或顺序进行免疫检查点抑制治疗,从而增加免疫检查点抑制的治疗效果。
施用芬氏别样杆菌(Alistipes finegoldii)的给药方式为口服。
施用联合疗法顺序为:在免疫检查点抑制治疗同时,之前和/或之后施用所述芬氏别 样杆菌(Alistipes finegoldii)。
所述联合疗法中,根据个体患者对治疗的耐受性,根据需要执行剂量延迟和/或剂量减少以及时间调整。
本发明中所述的芬氏别样杆菌(Alistipes finegoldii)可包含有效量的通常分散于药学上或药理学上可接受的载体中的芬氏别样杆菌(Alistipes finegoldii)。
术语“药学上或药理学上可接受的”是指当施用至动物(例如人,如果合适)时不产生不良反应、过敏反应或其他不利反应的分子实体和组合物。如本文所述的药理学上可接受的载体的具体实例是硼酸盐缓冲液或无菌盐水溶液。
本发明中所述增效免疫检查点抑制剂,可用于免疫检查点抑制剂难治性患者,其中所述免疫检查点抑制剂难治性患者表现出对所述免疫检查点抑制剂治疗的先天性(原发性)抗性,表现为从第一剂开始持续至少约8周或12周对所述检查点抑制剂治疗的应答缺乏或应答不足。
本发明中所述增效免疫检查点抑制剂,可用于免疫检查点抑制剂难治性患者,其中所述免疫检查点抑制剂难治性患者表现出对所述检查点抑制剂治疗的后天性(继发性)抗性,表现为对所述检查点治疗有初始应答,但一种或多种肿瘤随后复发和发展。
本发明中所述的联合疗法的治疗效果评估,通过增加的总生存时间来测量增强的治疗效果。
本发明中所述的联合疗法的治疗效果评估,通过增加的无进展生存期来测量增强的治疗效果。
本发明中所述的联合疗法的治疗效果评估,其中如RECIST 1.1所定义,通过减少或稳定一个或多个所述肿瘤的肿瘤尺寸来测量所述增强的治疗效果,包括目标肿瘤的疾病稳定(SD)、完全缓解(CR)或部分缓解(PR);和/或一种或多种非目标肿瘤的疾病稳定(SD)或完全缓解(CR)。
本发明中所述的联合疗法的治疗效果评估,通过改善的总缓解率和/或提升的生活质量来测量增强的治疗效果。
本发明中所述的联合疗法的安全性评估,通过是否引发腹泻或肠炎来测量其安全性。
与现有技术相比,本发明具有以下有益效果:
本发明通过使用人体共生细菌单菌口服制剂,联合免疫检查点抑制剂,由细菌刺激产生的抗肿瘤免疫保护反应,显著增强免疫检查点抑制剂对多多种肿瘤的药效,且安全性更好,延长癌症患者整体存活时间,提高癌症免疫治疗人群的响应率,扩大癌症免疫治疗(免 疫治疗检查点抑制剂)的受益肿瘤患者人群。
附图说明
图1为实施例1的流程图。
图2为肿瘤照片(第21天)。
图3为肿瘤体积变化曲线。
图4为肿瘤重量统计图。
图5为小鼠肛门照片(第21天)
图6为小鼠肠道组织HE染色图(第21天)。
图7为肿瘤组织免疫细胞浸润(第21天)。
图8为小鼠的生存曲线图。
图9为芬氏别样杆菌在不同人群中的分布及相对丰度。
具体实施方式
下面结合说明书附图及具体实施例对本发明作出进一步地详细阐述,所述实施例只用于解释本发明,并非用于限定本发明的范围。下述实施例中所使用的试验方法如无特殊说明,均为常规方法;所使用的材料、试剂等,如无特殊说明,为可从商业途径得到的试剂和材料。
实施例1芬氏别样杆菌联合免疫检查点抑制剂对肿瘤的治疗作用
一、实验方法
1、实验材料
(1)小鼠品系:6周龄的雌性C57BL/6J小鼠
(2)肿瘤细胞株:鼠黑色素瘤细胞系(B16-OVA,ATCC),鼠肠癌细胞系(MC38,ATCC)
(3)细菌制剂:芬氏别样杆菌(Alistipes finegoldii,DSM No.:17242,Type strain,其16S rDNA序列如SEQ ID NO:1所示),简称为Af,商购自DSMZ德国国家培养物保藏中心,(DSMZ的官方网址:http://www.dsmz.de)。
(4)细菌培养基:液体DSMZ104培养基,配方主要包括含蛋白胨,酵母提取物,牛肉提取物和葡萄糖等,商购自DSMZ德国国家培养物保藏中心。
(5)免疫检查点抑制剂:PD-1单抗(αPD-1),克隆号G4C2,试剂为上海君实生物医药科技股份有限公司赠送。
(6)抗生素组合:甲硝唑100mg/kg,万古霉素50mg/kg,青霉素钠100mg/kg,硫酸 新霉素100mg/kg
2、实验分组
实验分组如表1所示,
表1:
Figure PCTCN2021113916-appb-000001
3、实验步骤(流程如图1所示)
(1)细菌培养:接种芬氏别样杆菌于DSMZ104液体培养基,37℃厌氧箱培养18小时后离心浓度至1×10 10CFU/ml。
(2)皮下接种肿瘤细胞,MC38细胞系1×10 6/只,B16-OVA细胞系5×10 5/只。
(3)第1~3天:对各组小鼠灌胃抗生素组合清除肠道固有菌群。
(4)分别在第5、8、11天腹腔注射IgG或αPD-1,200μg/只。
(5)分别在第5、7、9、11、13、15天,通过灌胃方式进行Af活体菌液进行细菌制剂治疗,100μl/只,1×10 9CFU/只。
(6)分别在第5、8、11、14、17、21天测量肿瘤大小,并计算肿瘤体积。
Figure PCTCN2021113916-appb-000002
(7)在第21天对小鼠实施安乐死,取出肿瘤组织拍照并称重,取肠道组织行HE染色明确肠炎情况:
小鼠肿瘤体积的测量,终末时间点肿瘤的重量测量,免疫组化评估肿瘤组织免疫细胞的浸润进行效果评估;
采用小鼠肛门及肠道组织切片HE染色观测评估芬氏别样杆菌是否会引发肠炎以进行安全性评估;
二、实验结果
图2为第21天的肿瘤。图3为肿瘤体积变化曲线及图4为肿瘤重量统计图,在MC38肠癌和B16-OVA黑色素瘤小鼠模型中,相对于安慰剂治疗组(IgG),单药免疫检查点抑制剂组(αPD-1)显示出明显而显著(p<0.01)的肿瘤缩减,而相对于单药免疫检查点抑制剂组(αPD-1),联合疗法组(αPD-1+Af)均显示出明显而显著(p<0.01)的肿瘤缩减,证明芬氏别样杆菌具有增强αPD-1的抗肿瘤效应。芬氏别样杆菌单菌治疗组小鼠(IgG+Af)与安慰剂治疗组(IgG)肿瘤无差别,说明芬氏别样杆菌发挥抗肿瘤作用依赖于αPD-1。
图5小鼠的肛门照片及图6肠道组织切片HE染色显示,芬氏别样杆菌单菌(IgG+Af)与联合疗法组(αPD-1+Af)小鼠均未发现有肠炎现象,证明芬氏别样杆菌经肠胃给药的安全性。
图7实验终点MC38小鼠模型肿瘤组织免疫组化显示,芬氏别样杆菌单菌治疗组(IgG+Af)能够显著增加肿瘤间质区CD4+辅助T细胞浸润,与单药免疫检查点抑制剂组(αPD-1)相比,联合疗法组(αPD-1+Af)表现出增加的肿瘤间质区CD4+辅助T细胞浸润趋势,但是未达到统计学差异。与安慰剂组(IgG)相比,单药免疫检查点抑制剂组(αPD-1)的小鼠实验终点的CD4+辅助T细胞浸润也只有增加的趋势,未达到统计学差异。这个结果说明,αPD-1单药治疗和联合疗法(αPD-1+Af)对小鼠肿瘤组织免疫细胞浸润的影响可能是短时间的,在实验终点的肿瘤样本中无法检测出显著差异。芬氏别样杆菌单菌治疗(IgG+Af)对小鼠肿瘤组织免疫细胞浸润的影响持续时间较长,在治疗停止一周后仍然具有显著增高的CD4+辅助T细胞浸润,证实了芬氏别样杆菌单菌口服对系统免疫的调节作用。
实施例2不同剂量芬氏别样杆菌联合不同免疫检查点抑制剂对总生存时间的延长
一、实验方法
1、实验材料
(1)小鼠品系:6周龄的雌性C57BL/6J小鼠
(2)肿瘤细胞株:鼠黑色素瘤细胞系(B16-OVA,ATCC),鼠肠癌细胞系(MC38,ATCC)
(3)细菌制剂:芬氏别样杆菌(Alistipes finegoldii,DSM No.:17242,Type strain,其16S rDNA序列如SEQ ID NO:1所示),简称为Af,商购自DSMZ德国国家培养物保藏中心。
(4)细菌培养基:液体DSMZ104培养基,配方主要包括含蛋白胨,酵母提取物, 牛肉提取物和葡萄糖等,商购自DSMZ德国国家培养物保藏中心。
(5)免疫检查点抑制剂:PD-1单抗(αPD-1),克隆号G4C2,试剂为上海君实生物医药科技股份有限公司赠送。CTLA4单抗(αCTLA4),克隆号9D9,商购自美国BioXcell。
(6)抗生素组合:甲硝唑100mg/kg,万古霉素50mg/kg,青霉素钠100mg/kg,硫酸新霉素100mg/kg
2、实验分组
实验分组如表2所示,
表2
Figure PCTCN2021113916-appb-000003
3、实验步骤(流程如图1所示)
(1)细菌培养:接种芬氏别样杆菌于DSMZ104液体培养基,37℃厌氧箱培养18小时后离心浓度至1×10 10CFU/ml。
(2)皮下接种肿瘤细胞,MC38细胞系1×10 6/只,B16-OVA细胞系5×10 5/只。
(3)第1~3天:对各组小鼠灌胃抗生素组合清除肠道固有菌群。
(4)从第5天开始,每3天腹腔注射IgG或αPD-1或αCTLA4,200μg/只。
(5)从第5天开始,每2天通过灌胃方式进行Af活体菌液不同剂量的细菌制剂治疗,其中低剂量组(low)为100μl/只,1×10 9CFU/只,中等剂量组(medium)为200μl/只,2×10 9CFU/只,高剂量组(high)为400μl/只,4×10 9CFU/只。
(6)从第5天开始,每3天测量肿瘤大小,并计算肿瘤体积。
Figure PCTCN2021113916-appb-000004
(7)治疗周期直到小鼠肿瘤生长到伦理体积大小(2000mm 3)实施安乐死或未达到 肿瘤伦理体积大小而自然死亡。
(8)记录每一只小鼠的死亡状态及时间,并绘制生存曲线图。
二、实验结果
图8为小鼠的生存曲线,在MC38肠癌和B16-OVA黑色素瘤小鼠模型中,相对于单药免疫检查点抑制剂组(αPD-1),Af高(high),中(medium)和低(low)剂量的联合疗法组(αPD-1+Af)均能显著增加荷瘤小鼠的生存时间。在B16-OVA黑色素瘤小鼠模型中,Af还呈现出对αCTLA4免疫检查点抑制剂的疗效促进作用:αCTLA4联合高(high),中(medium)和低(low)剂量的Af均能显著增加荷瘤小鼠的生存时间。而在MC38肠癌小鼠模型中,由于αCTLA4免疫检查点抑制剂药效作用太强,未观察到Af对αCTLA4的增效作用。
实施例3芬氏别样杆菌(Alistipes finegoldii)为人类的内源性肠道共生细菌
一、实验方法
分析了四个人类肠道宏基因组数据集,共包含1396个人类粪便样本,涉及9种不同类型的人群。宏基因组测序技术可达到人类肠道菌种水平鉴定的精度。
9种不同类型的人群包括:1)健康成人;2)结直肠腺瘤患者;3)结直肠癌患者;4)结直肠癌术后患者;5)动脉粥样硬化性疾病患者;6)接受免疫检查点抑制剂(ICIs)前的非小细胞肺癌患者;7)接受免疫检查点抑制剂(ICIs)后的非小细胞肺癌患者;8)接受免疫检查点抑制剂(ICIs)前的肾癌患者的肠道菌群宏基因组学数据;9)接受免疫检查点抑制剂(ICIs)后的肾癌患者。
数据集的具体情况如表3所示。
表3人类肠道宏基因组公共数据集:
Figure PCTCN2021113916-appb-000005
二、实验结果
如图9和表4结果显示,芬氏别样杆菌(Alistipes finegoldii)在不同的人群中均有存在,其相对丰度为0.001~0.07%,且在不同的人群芬氏别样杆菌的相对丰度存在差异,其中相对丰度是指某一个菌种占肠道全部细菌菌种的比例。按照人类肠道包含的全部细菌菌种数量约为10 14CFU/ml的估算,芬氏别样杆菌在人类肠道的数量约为10 9CFU/ml~10 10CFU/ml。
表4芬氏别样杆菌在不同人群中的相对丰度
Figure PCTCN2021113916-appb-000006
最后所应当说明的是,以上实施例仅用以说明本发明的技术方案而非对本发明保护范围的限制,对于本领域的普通技术人员来说,在上述说明及思路的基础上还可以做出其它不同形式的变化或变动,这里无需也无法对所有的实施方式予以穷举。凡在本发明的精神和原则之内所作的任何修改、等同替换和改进等,均应包含在本发明权利要求的保护范围 之内。

Claims (55)

  1. 一种细菌在肿瘤治疗上的应用。
  2. 根据权利要求1所述的应用,其特征在于,所述细菌菌种属于别样杆菌属(Alistipes),即,所述细菌菌种的16S rDNA序列与别样杆菌属(Alistipes)的16S rDNA序列具有至少85%,至少90%,至少95%,至少96%,至少97%,至少98%,至少99%,至少99.5%或100%的一致性。
  3. 根据权利要求2所述的应用,其特征在于,所述属于别样杆菌属(Alistipes)的细菌为一个或多个属于别样杆菌属(Alistipes)的细菌的菌种或菌株的组合。
  4. 根据权利要求3所述的应用,其特征在于,所述属于别样杆菌属(Alistipes)的细菌为芬氏别样杆菌(Alistipes finegoldii)。
  5. 根据权利要求3所述的应用,其特征在于,所述芬氏别样杆菌(Alistipes finegoldii),其包含的16S rDNA序列,其与芬氏别样杆菌菌种(Alistipes finegoldii)的16S rDNA序列具有至少99%的一致性。
  6. 根据权利要求5所述的应用,其特征在于,所述芬氏别样杆菌(Alistipes finegoldii),其包含的16S rDNA序列,与芬氏别样杆菌菌株DSM17242(Alistipes finegoldii DSM17242)的16S rDNA序列具有至少99.5%或100%的一致性。
  7. 根据权利要求5或6所述的应用,其特征在于,所述芬氏别样杆菌(Alistipes finegoldii)为一个或多个芬氏别样杆菌(Alistipes finegoldii)菌株的组合。
  8. 根据权利要求7所述的应用,其特征在于,所述芬氏别样杆菌(Alistipes finegoldii)中亚种分类的菌株名称为:Alistipes finegoldii DSM 17242、Alistipes finegoldii D53t1_180928_D3、Alistipes finegoldii 2789STDY5834947、Alistipes finegoldii 1001713B170207_170306_H2、Alistipes finegoldii DFI.2.31、Alistipes finegoldii BIOML-A1、Alistipes finegoldii DFI.2.16、Alistipes finegoldii DFI.2.10、Alistipes finegoldii aa_0143、Alistipes finegoldii 2789STDY5608890、Alistipes finegoldii MGBC116453、Alistipes finegoldii COPD076、或Alistipes finegoldii UBG195中的一个或几个的组合。
  9. 根据权利要求7所述的应用,其特征在于,所述芬氏别样杆菌菌株为以下菌株的中的一个或几个的组合:
    保藏于德国DSM微生物和细胞培养物保藏中心,保藏号为DSM 17242的芬氏别样杆菌;
    保藏于日本JCM菌种保藏中心,保藏号为JCM16770的芬氏别样杆菌;
    保藏于韩国KCTC菌种保藏中心,保藏号为KCTC 15236的芬氏别样杆菌;
    保藏于芬兰赫尔辛基厌氧菌参考实验室,保藏号为AHN 2437的芬氏别样杆菌;
    保藏于瑞典CCUG菌种保藏中心,保藏号为CCUG46020的芬氏别样杆菌;
    保藏于法国CIP菌种保藏中心,保藏号为CIP107999的芬氏别样杆菌。
    保藏于中国广东省微生物菌种保藏中心,保藏号为GDMCC 1.2324的芬氏别样杆菌。
  10. 根据权利要求1所述的应用,其特征在于,所述细菌,包括细菌活菌、细菌衍生物、或细菌代谢产物中的一种或几种。
  11. 根据权利要求10所述的应用,其特征在于,所述细菌活菌为完整细菌。
  12. 根据权利要求10所述的应用,其特征在于,所述细菌活菌为整活力菌。
  13. 根据权利要求10所述的应用,其特征在于,所述细菌代谢物包括细菌制备和储存过程中以及在哺乳动物胃肠道转运过程中作为细菌生长、生存、留滞、转运或存在的结果由所述细菌产生或修饰的所有分子。
  14. 根据权利要求13所述的应用,其特征在于,所述细菌代谢物包括所有有机酸、无机酸、碱、蛋白质和肽、酶和辅酶、氨基酸和核酸、碳水化合物、脂质、糖蛋白、脂蛋白、糖脂、维生素、所有生物活性化合物、含有无机成分的代谢物以及所有小分子,例如含氮分子或含亚硫酸分子。
  15. 根据权利要求10所述的应用,其特征在于,所述细菌衍生物,包括细菌组成成分和遗传物质及其相关成分,其实例包括细菌细胞膜,菌毛,鞭毛,LPS,核酸物质等来源于细菌的成分。
  16. 根据权利要求1到15任一所述的应用,其特征在于,同时或分别施用属于别样杆菌属(Alistipes)的细菌和免疫检查点抑制剂。
  17. 根据权利要求1所述的应用,其特征在于,所述治疗肿瘤为肿瘤缩小或稳定、总生存时间延长、无恶化生存期延长、生活质量改善。
  18. 根据权利要求1所述的应用,其特征在于,所述肿瘤为腺瘤、恶性肿瘤和腺癌,其中肿瘤按照组织来源或细胞名称分类,包括:肾上腺皮质癌、膀胱尿路上皮癌、乳腺癌、胰腺癌、宫颈癌、胆管癌、结肠癌、结直肠癌、弥漫性大B细胞淋巴瘤、多形成性胶质细胞瘤、胶质瘤、头颈癌、肾嫌色细胞癌、混合肾癌、肾癌、白血病、淋巴瘤、脑癌、肝癌、肺腺癌、肺鳞癌、间皮瘤、卵巢癌、胰腺癌、嗜铬细胞瘤、副神经节瘤、前列腺癌、直肠腺癌、肉瘤、皮肤黑色素瘤、胃癌、食管癌、睾丸癌、甲状腺癌、胸腺癌、子宫内膜癌、 子宫肉瘤、葡萄膜黑色素瘤和软组织肉瘤中的一种或几种。
  19. 根据权利要求1所述的应用,其特征在于,所述肿瘤为恶性肿瘤、转移性肿瘤或非转移性肿瘤。
  20. 根据权利要求1所述的应用,其特征在于,在此所述肿瘤包括恶性、转移性和非转移性类型;包括癌症的任何阶段。
  21. 根据权利要求16所述的应用,其特征在于,所述免疫检查点抑制剂为作用于T细胞负性共刺激(共抑制)分子和/或它们各自的配体的阻断剂的一种或多种的组合。
  22. 根据权利要求21所述的应用,其特征在于,T细胞负性共刺激(共抑制)分子和/或它们各自的配体选自CTLA-4、PD-1、PD-L1、PD-L2、B7-1、B7-2、B7-H3、B7-H4、B7-H6、A2AR、IDO、TIM-3、BTLA、VISTA、TIGIT、LAG-3、CD40、KIR、CEACAM1、GARP、PS、CSF1R、CD94/NKG2A、TDO、TNFR、DcR3。
  23. 根据权利要求22所述的应用,其特征在于,所述T细胞负性共刺激(共抑制)分子的配体的阻断剂选自纳武利尤单抗、伊匹单抗、帕博利珠单抗、阿托珠单抗、阿特朱单抗、卡瑞珠丽单抗、替雷丽珠单抗、度伐利尤单抗、曲美木单抗、斯巴达珠单抗、阿维鲁单抗、信迪利单抗、特瑞普利单抗、西米普利单抗、MGA012、MGD013、MGD019、恩比利珠单抗、MGD009、MGC018、MEDI0680、PDR001FAZ053、TSR022、MBG453、relatlimab、LAG525、IMP321、REGN3767、培西达替尼、LY3022855、FPA008、BLZ945、GDC0919、艾帕斯塔、indoximid、BMS986205、CPI‐444、MEDI9447、PBF509、利瑞路单抗或其中任意几种的组合;优选地,其中所述阻断剂选自纳武利尤单抗、帕博利珠单抗、特瑞普利单抗、信迪利单抗、西米普利单抗或其中任意几种的组合。
  24. 根据权利要求21所述的应用,其特征在于,所述免疫检查点抑制剂为作用于PD-1/PD-L1信号通路和/或PD-1/PD-L2信号通路的抑制剂,其中PD-1是指程序性细胞死亡蛋白1,PD-L1(B7-H1或CD274)和PD-L2(B7-DC或CD273)是PD-1的配体。
  25. 根据权利要求24所述的应用,其特征在于,所述PD-1/PD-L1信号通路或PD-1/PD-L2信号通路的抑制剂选自纳武利尤单抗、帕博利珠单抗、阿托珠单抗、阿特朱单抗、卡瑞珠丽单抗、替雷丽珠单抗、度伐利尤单抗、斯巴达珠单抗、阿维鲁单抗、信迪利单抗、特瑞普利单抗、西米普利单抗、MGA012、MGD013、MGD019(PD-1/CTLA-4双抗)、MEDI0680、PDR001、FAZ053或其中任意几种的组合。
  26. 根据权利要求23所述的应用,其特征在于,所述免疫检查点抑制剂为作用于CTLA-4/B7-1信号通路和/或CTLA-4/B7-2信号通路的抑制剂,其中CTLA-4是指胞毒性T 淋巴细胞蛋白4,B7-1(CD80)和B7-2(CD86)是CTLA-4的配体。
  27. 根据权利要求26所述的应用,其特征在于,其可选自伊匹单抗、曲美木单抗、MGD019或其中任意几种的组合。
  28. 根据权利要求16到27任一所述的应用,其特征在于,所述免疫检查点抑制剂为作用于PD-1/PD-L1信号通路和/或PD-1/PD-L2信号通路的抑制剂和/或作用于CTLA-4/B7-1信号通路和/或CTLA-4/B7-2信号通路的抑制剂。
  29. 根据权利要求1所述的应用,其特征在于,在与芬氏别样杆菌的施用同时、分别或顺序进行化学治疗,免疫治疗或放射治疗。
  30. 根据权利要求1所述的应用,其特征在于,所述应用的施用的对象是人类,所述人类是婴幼儿,儿童,青少年,成年人或老人。
  31. 根据权利要求1所述的应用,其特征在于,其中所述应用施用的对象是非人类灵长类动物,所述非人类灵长类动物是哺乳动物。
  32. 根据权利要求1所述的应用,其特征在于,将所述细菌以包含所述细菌的10 5至10 12个CFU之间,或以10 7至10 11CFU之间、或10 8至10 11CFU之间、或10 9至10 11CFU之间、或10 10至10 11CFU之间的剂量施用,更优选地,以包含所述细菌的10 9至10 11CFU之间的剂量施用。
  33. 一种用于肿瘤治疗的试剂盒,其特征在于,其包含权利要求16、21到28中任一项所述的免疫检查点抑制剂的一种或多种,以及权利要求1到15中任一项所述的细菌,并且任选地包含容器;
    或由权利要求16、21到28中任一项所述的免疫检查点抑制剂的一种或多种,以及权利要求1到15中任一项所述的细菌组成,并且任选地包含容器。
  34. 一种细菌,其特征在于,其包含的16S rDNA序列,与别样杆菌(Alistipes)的16S rDNA序列具有至少85%,至少90%,至少95%,至少96%,至少97%,至少98%,至少99%,至少99.5%或100%的一致性。
  35. 一种细菌,其特征在于,其包含的16S rDNA序列,与芬氏别样杆菌(Alistipes finegoldii)的16S rDNA序列具有至少99%的一致性。
  36. 一种细菌,其特征在于,其包含的16S rDNA序列,其16S rDNA序列与以下菌株的中的任意一个的16S rDNA序列具有至少99.5%或100%的一致性:
    保藏于德国DSM微生物和细胞培养物保藏中心,保藏号为DSM 17242的芬氏别样杆菌;
    保藏于日本JCM菌种保藏中心,保藏号为JCM16770的芬氏别样杆菌;
    保藏于韩国KCTC菌种保藏中心,保藏号为KCTC 15236的芬氏别样杆菌;
    保藏于芬兰赫尔辛基厌氧菌参考实验室,保藏号为AHN 2437的芬氏别样杆菌;
    保藏于瑞典CCUG菌种保藏中心,保藏号为CCUG46020的芬氏别样杆菌;
    保藏于法国CIP菌种保藏中心,保藏号为CIP107999的芬氏别样杆菌。
    保藏于中国广东省微生物菌种保藏中心,保藏号为GDMCC 1.2324的芬氏别样杆菌。
  37. 一种制剂,其特征在于,将有效量的用于治疗肿瘤的权利要求34到36中任一项所述的细菌中的一种或多种的组合物,加入药学上可接受的载体和辅料制备成冻干粉,片剂、胶囊剂、颗粒剂或针剂;其中“药学上可接受的载体”是指对有机体不引起明显刺激性和不干扰所给予化合物的生物活性和性质的载体。其中“辅料”是指溶剂、稀释剂或其它赋形剂、分散剂、表面活性剂。
  38. 一种组合物,其特征在于,其包含权利要求34到36中任一项的细菌和药学上可接受的载体。
  39. 一种组合物,其特征在于,其包含配制成用作药物的权利要求34到36中任一项的细菌。
  40. 一种组合物,其特征在于,其包含配制为用作医疗食品的权利要求34到36中任一项的细菌。
  41. 一种组合物,其特征在于,其包含两种或更多种权利要求34到36中任一项所述的细菌的混合物,并且任选地还包含药学上可接受的载体。
  42. 一种组合物,其特征在于,其包含有效量的用于肿瘤治疗的两种或更多种权利要求34到36中任一项所述的细菌的混合物,并且还任选地包含药学上可接受的载体。
  43. 一种组合物,其特征在于,其包含有效量的用于肿瘤治疗的两种或更多种权利要求34到36中任一项所述的细菌的细菌活菌、细菌衍生物、或细菌代谢产物中的一种或几种组合物,并且还任选地包含药学上可接受的载体。
  44. 一种食品、饮料、食品补充剂、益生菌或保健食品,其包含权利要求34到36中任一项所述的细菌,被配制成制剂用于肠内营养。
  45. 一种食品、饮料、食品补充剂、益生菌或保健食品,其包含两种或更多种权利要求34到36中任一项所述的细菌菌株的混合物,被配制成制剂用于肠内营养。
  46. 权利要求34到43中任一项细菌、制剂或组合物,其中所述细菌或组合物被制备成冻干粉,片剂、胶囊剂、颗粒剂或针剂。
  47. 权利要求34至45任一项所述的细菌、制剂、组合物、食品、饮料、食品补充剂、益生菌或保健食品,其特征在于,其施用对象为人类。
  48. 根据权利要求34至45中任一项所述的细菌、制剂、组合物、食品、饮料、食品补充剂、益生菌或保健食品,其特征在于,其中所述人类是婴幼儿、儿童、青少年、成年人或老人。
  49. 权利要求34至45中任一项所述的细菌、组合物、食品、饮料、食品补充剂、益生菌或保健食品,施用于非人类灵长类动物,所述非人类灵长类动物是哺乳动物;鸟类;爬行动物;鱼;两栖动物;节肢动物或家畜动物。
  50. 权利要求34至45中任一项所述的细菌、制剂、组合物、食品、饮料、食品补充剂、益生菌或保健食品,在肿瘤治疗中的应用。
  51. 权利要求34至45中任一项所述的细菌、制剂、组合物、食品、饮料、食品补充剂、益生菌或保健食品,包含有效量的所述细菌,在肿瘤治疗上中的应用。
  52. 权利要求34至45中任一项所述的细菌、制剂、组合物、食品、饮料、食品补充剂、益生菌或保健食品,包含有效量的所述细菌,在肿瘤治疗中的应用,其特征在于,所述肿瘤治疗施用权利要求34至45中任一项所述的细菌、制剂、组合物、食品、饮料、食品补充剂、益生菌或保健食品。
  53. 一种组合物,其包含用于治疗癌症的有效量的权利要求32到33中任一项细菌或组合物,并且任选地还包含药学上可接受的载体。
  54. 一种组合物,其包含有效量的用于治疗癌症的两种或更多种权利要求32到33中任一项细菌或组合物,并且还任选地包含药学上可接受的载体。
  55. 属于别样杆菌属(Alistipes)的细菌的细菌活菌、细菌衍生物、或细菌代谢产物中的一种或几种在制备免疫检查点抑制剂治疗肿瘤的增效剂中的应用。
PCT/CN2021/113916 2021-07-16 2021-08-20 一种细菌在制备免疫检查点抑制剂的增效剂中的应用 WO2023284069A1 (zh)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202110808366.1 2021-07-16
CN202110808366 2021-07-16
CN202110939699.8A CN114159475B (zh) 2021-07-16 2021-08-16 一种细菌在制备免疫检查点抑制剂的增效剂中的应用
CN202110939699.8 2021-08-16

Publications (1)

Publication Number Publication Date
WO2023284069A1 true WO2023284069A1 (zh) 2023-01-19

Family

ID=80476633

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/113916 WO2023284069A1 (zh) 2021-07-16 2021-08-20 一种细菌在制备免疫检查点抑制剂的增效剂中的应用

Country Status (2)

Country Link
CN (2) CN114159475B (zh)
WO (1) WO2023284069A1 (zh)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20230050401A (ko) * 2021-07-16 2023-04-14 선 얏-센 유니버시티 캔서 센터 (캔서 하스피탈 어필리에이티드 투 선 얏-센 유니버시티, 캔서 리서치 인스티튜트 오브 선 얏-센 유니버시티) 면역관문 억제제의 상승제의 제조에서 박테리아의 응용
CN114668782B (zh) * 2022-02-16 2023-07-14 中山大学肿瘤防治中心(中山大学附属肿瘤医院、中山大学肿瘤研究所) 一种非活性全细胞细菌在肿瘤治疗上的应用

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110267651A (zh) * 2016-09-27 2019-09-20 得克萨斯系统大学评议会 通过调节微生物组来增强免疫检查点阻断疗法的方法
US20190365829A1 (en) * 2016-11-18 2019-12-05 Sanford Burnham Prebys Medical Discovery Institute Gut microbiota and treatment of cancer
CN110582291A (zh) * 2016-12-22 2019-12-17 古斯达威罗斯研究所 抗pd1/pd-l1/pd-l2抗体的反应性标志物和功效改善用调节剂
US20200129570A1 (en) * 2017-06-02 2020-04-30 Board Of Regents, The University Of Texas System Specific Bacterial Species and Metabolite That Improves Immune Checkpoint Inhibitor Therapy Efficacy
WO2021091981A1 (en) * 2019-11-04 2021-05-14 Vedanta Biosciences, Inc. Compositions and methods for enhancing immune checkpoint inhibitor therapy

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2017014787A (es) * 2015-05-21 2018-05-01 Yeda Res & Dev Poblaciones bacterianas para promover la salud.
JP2022513623A (ja) * 2018-11-21 2022-02-09 ボード オブ リージェンツ,ザ ユニバーシティ オブ テキサス システム がんを処置するための方法および組成物
CN109966320B (zh) * 2019-04-26 2022-08-05 青岛东海药业有限公司 凝结芽孢杆菌在制备诱导抗肿瘤免疫及免疫检查点抑制剂增敏制剂中的应用
CN110063970A (zh) * 2019-04-30 2019-07-30 上海心脉途医疗科技有限公司 与irAE相关的肠道菌群及irAE的治疗和预防方法
US20220257673A1 (en) * 2019-07-19 2022-08-18 Finch Therapeutics Holdings Llc Methods and products for treatment of gastrointestinal disorders
US20220370553A1 (en) * 2019-07-31 2022-11-24 Sanford Burnham Prebys Medical Discovery Institute Prebiotic-induced anti-tumor immunity
WO2021141828A1 (en) * 2020-01-10 2021-07-15 Assembly Biosciences, Inc. Compositions comprising bacterial species and methods related thereto

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110267651A (zh) * 2016-09-27 2019-09-20 得克萨斯系统大学评议会 通过调节微生物组来增强免疫检查点阻断疗法的方法
US20190365829A1 (en) * 2016-11-18 2019-12-05 Sanford Burnham Prebys Medical Discovery Institute Gut microbiota and treatment of cancer
CN110582291A (zh) * 2016-12-22 2019-12-17 古斯达威罗斯研究所 抗pd1/pd-l1/pd-l2抗体的反应性标志物和功效改善用调节剂
US20200129570A1 (en) * 2017-06-02 2020-04-30 Board Of Regents, The University Of Texas System Specific Bacterial Species and Metabolite That Improves Immune Checkpoint Inhibitor Therapy Efficacy
WO2021091981A1 (en) * 2019-11-04 2021-05-14 Vedanta Biosciences, Inc. Compositions and methods for enhancing immune checkpoint inhibitor therapy

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SUO JIAO-JIAO;WEI HAO;WANG YAN;ZHU JIANG: "Modulatory Target for Tumor Immunotherapy:Gut Microbiota", CHINA CANCER, vol. 29, no. 4, 30 March 2020 (2020-03-30), pages 304 - 308, XP093024833, ISSN: 1004-0242 *

Also Published As

Publication number Publication date
CN115998771A (zh) 2023-04-25
CN114159475A (zh) 2022-03-11
CN114159475B (zh) 2022-09-13

Similar Documents

Publication Publication Date Title
Roy et al. Microbiota: a key orchestrator of cancer therapy
CN110582291A (zh) 抗pd1/pd-l1/pd-l2抗体的反应性标志物和功效改善用调节剂
TW201717975A (zh) 藉由控制共生微生物叢來治療癌症
WO2023284069A1 (zh) 一种细菌在制备免疫检查点抑制剂的增效剂中的应用
WO2023284758A1 (zh) 一种细菌在制备免疫检查点抑制剂的增效剂中的应用
CN109771445B (zh) 酪酸梭菌在制备诱导抗肿瘤免疫及免疫检查点抑制剂增敏制剂中的应用
CN109966320B (zh) 凝结芽孢杆菌在制备诱导抗肿瘤免疫及免疫检查点抑制剂增敏制剂中的应用
CN111249314B (zh) 人体共生菌群在提高肿瘤免疫治疗应答中的作用
CN114452382B (zh) 脆弱拟杆菌荚膜多糖a与pd-1及pd-l1抗体联合治疗呼吸系统肿瘤的应用
CN114540229A (zh) 一种增强免疫检查点抑制剂治疗效应的副干酪乳杆菌株及其应用
CN112618577A (zh) 动物双歧杆菌在提高肿瘤免疫治疗应答中的作用
KR20200111183A (ko) 암을 치료하거나 예방하기 위한 조합 치료법
CN114668782A (zh) 一种非活性全细胞细菌在肿瘤治疗上的应用
JP7040701B2 (ja) 抗がん腫瘍溶解性ウイルス併用療法および優良レスポンダー選択プラットフォーム
Zhou et al. Gut microbiota: A promising milestone in enhancing the efficacy of PD1/PD-L1 blockade therapy
WO2024037273A1 (en) Use of enterococcus lactis in preparation of drug for treating melanoma
JP2023526100A (ja) 治療用細菌組成物
CN112791106B (zh) 药物组合物及其治疗疾病的用途
US20230106313A1 (en) A bacterial composition for the treatment of cancer
TWI817977B (zh) 活化腫瘤浸潤淋巴細胞之方法
TW201934139A (zh) 用於治療或預防癌症之組合療法
Behrouzi et al. The role of microbiota and immune system crosstalk in cancer development and therapy
CN116103196B (zh) 一种活泼瘤胃球菌及其应用
Rawat et al. Microflora impacts immune system and its antitumor function
KR20230121536A (ko) 락토바실러스 플란타룸 균주 및 항암제를 포함하는 병용 요법을 이용한 암 예방 또는 치료용 조성물

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21949844

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE