JP2015051970A - Composition for producing adjuvant for cancer patients receiving chemotherapy - Google Patents

Composition for producing adjuvant for cancer patients receiving chemotherapy Download PDF

Info

Publication number
JP2015051970A
JP2015051970A JP2014180452A JP2014180452A JP2015051970A JP 2015051970 A JP2015051970 A JP 2015051970A JP 2014180452 A JP2014180452 A JP 2014180452A JP 2014180452 A JP2014180452 A JP 2014180452A JP 2015051970 A JP2015051970 A JP 2015051970A
Authority
JP
Japan
Prior art keywords
extract
composition
chemotherapy
cancer
composition according
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
JP2014180452A
Other languages
Japanese (ja)
Inventor
クン−ミン ルー
Kung-Ming Lu
クン−ミン ルー
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Microbio Co Ltd
Original Assignee
Microbio Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Microbio Co Ltd filed Critical Microbio Co Ltd
Publication of JP2015051970A publication Critical patent/JP2015051970A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/48Fabaceae or Leguminosae (Pea or Legume family); Caesalpiniaceae; Mimosaceae; Papilionaceae
    • 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
    • A61K35/748Cyanobacteria, i.e. blue-green bacteria or blue-green algae, e.g. spirulina
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/02Algae
    • A61K36/05Chlorophycota or chlorophyta (green algae), e.g. Chlorella
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/06Fungi, e.g. yeasts
    • A61K36/07Basidiomycota, e.g. Cryptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/82Theaceae (Tea family), e.g. camellia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/87Vitaceae or Ampelidaceae (Vine or Grape family), e.g. wine grapes, muscadine or peppervine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/88Liliopsida (monocotyledons)
    • A61K36/906Zingiberaceae (Ginger family)
    • A61K36/9066Curcuma, e.g. common turmeric, East Indian arrowroot or mango ginger
    • 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
    • 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
    • A23V2200/00Function of food ingredients
    • A23V2200/30Foods, ingredients or supplements having a functional effect on health
    • A23V2200/308Foods, ingredients or supplements having a functional effect on health having an effect on cancer prevention
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2236/00Isolation or extraction methods of medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicine
    • A61K2236/10Preparation or pretreatment of starting material
    • A61K2236/19Preparation or pretreatment of starting material involving fermentation using yeast, bacteria or both; enzymatic treatment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2236/00Isolation or extraction methods of medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicine
    • A61K2236/30Extraction of the material

Abstract

PROBLEM TO BE SOLVED: To provide a composition for producing adjuvant for cancer patients receiving chemotherapy.SOLUTION: A composition for producing an adjuvant for cancer patients receiving chemotherapy of the invention mainly contains soybean fermentation extract, green tea extract, spirulina, turmeric extract, Antrodia Cinnamomea mycelium, and grape seed extract. The composition provides development of colon polyps and inhibition of canceration, increased survival ratio of cancer patients, particularly increased efficacy of chemotherapy comprising increased disease-free survival rate, and/or a reduction effect of side effects caused by chemotherapy. And the composition of the invention also provides anti-inflammatory activity which is effective in production reduction of inflammatory mediators, such as prostaglandin E2 (PGE2), nitric oxide (NO), tumor necrosis factor α (TNF-α), and interleukin-6 (IL-6).

Description

本願は、その全ての開示内容が参照によって本願に組み込まれる、2014年9月6日に出願された台湾特許出願第102132310号の利益を主張するものである。   This application claims the benefit of Taiwan Patent Application No. 102132310, filed on September 6, 2014, the entire disclosure of which is incorporated herein by reference.

本発明は化学療法のためのアジュバントを製造するための組成物に関し、特に、本発明は大腸ポリープの増殖とがん化の阻害に有効であり、化学療法の有効性と化学療法に対するコンプライアンスを高め、化学療法に起因する副作用を減少する、化学療法を受けているがん患者向けのアジュバントを製造するための組成物に関する。   The present invention relates to a composition for producing an adjuvant for chemotherapy, and in particular, the present invention is effective in inhibiting the growth and canceration of colorectal polyps and increasing the effectiveness of chemotherapy and compliance with chemotherapy. The present invention relates to a composition for producing an adjuvant for cancer patients undergoing chemotherapy, which reduces side effects caused by chemotherapy.

大豆発酵抽出物、緑茶、スピルリナ、ウコン、ベニクスノキタケ、グレープシードはいずれも健康に対する自然の宝物であり、数々の生化学的試験においてこれら素材のそれぞれに体に健康的な効果があることが示されている。ウコンに含まれるクルクミンの主な薬理活性は、腫瘍の成長を阻害することであり、ベニクスノキタケに豊富に含まれる多糖は免疫機能を強化し、腫瘍細胞を殺すことができる。単一成分を製剤化した製品はさまざまなものが市販されているが、がん治療における化学療法による副作用を減少することに対する6成分の複方の効果について、関連の研究はない。   Soybean fermented extract, green tea, spirulina, turmeric, venix mushroom, and grape seed are all natural treasures for health, and each of these ingredients has health benefits in many biochemical tests. It is shown. The main pharmacological activity of curcumin contained in turmeric is to inhibit tumor growth, and the polysaccharides abundantly contained in Benicus mushrooms can enhance immune function and kill tumor cells. A variety of single-component products are commercially available, but there are no related studies on the combined effects of the six components on reducing the side effects of chemotherapy in cancer treatment.

結腸直腸がんを例とすると、結腸直腸がんには結腸がんと直腸がんが含まれ、世界で最も一般的な悪性腫瘍の1つである。何が結腸直腸がんを引き起こすのかは完全に明らかになってはいないが、結腸直腸がんは食生活に高い関連性があると一般的に考えられている。例えば、加工食品や動物性脂肪、たんぱく質が豊富な食品の摂取増加、及び繊維が豊富な食品の摂取不足の両方が、現代世界の疾患である結腸直腸がんの発生の増加につながる。幸い、結腸直腸がんの発生は多いが、病変は比較的容易に見つけることができ、結腸直腸がんの進行には比較的長い時間を要する。ほとんどの早期結腸直腸がんはポリープから約5〜10年で進行するため、定期検査で大腸ポリープが早期段階で発見されれば、結腸直腸がんは一般に容易に制御できる。しかしながら、結腸直腸がんの早期段階における症状は顕著でなく、特異度が低いため、患者と医師がそれらの症状を看過しやすく、早期の発見と治療の機会を逃しやすい。   Taking colorectal cancer as an example, colorectal cancer includes colon cancer and rectal cancer, and is one of the most common malignant tumors in the world. Although it is not completely clear what causes colorectal cancer, colorectal cancer is generally thought to be highly related to diet. For example, both increased consumption of processed foods, animal fats, protein-rich foods, and insufficient intake of fiber-rich foods lead to an increase in the incidence of colorectal cancer, a modern world disease. Fortunately, although colorectal cancer occurs frequently, lesions are relatively easy to find and colorectal cancer takes a relatively long time to progress. Because most early colorectal cancers progress about 5 to 10 years after polyps, colorectal cancers are generally easily controlled if colon polyps are detected at an early stage on routine examination. However, symptoms in the early stages of colorectal cancer are not noticeable and have low specificity, making it easier for patients and physicians to overlook those symptoms and miss opportunities for early detection and treatment.

通常、結腸直腸がんは手術、放射線治療、化学療法の3種類の治療法が最も一般的である。中でも、腫瘍に対する理解とともに、現在結腸直腸がんの化学療法ではフルオロウラシル(5−FU)ベースの併用化学療法を主として用い、がんの再発と転移の減少及び生存率の向上が図られている。   Colorectal cancer is usually the most common three types of treatment: surgery, radiation therapy, and chemotherapy. Above all, along with understanding of tumors, currently chemotherapy of colorectal cancer mainly uses combination chemotherapy based on fluorouracil (5-FU) to reduce cancer recurrence and metastasis and improve survival rate.

FOLFOX4はステージII及びIIIと転移性の結腸直腸がん治療に現在使用されている化学療法レジメンであり、フルオロウラシル(5−FU)、ロイコボリン、オキサリプラチンを含む薬剤から構成されている。しかしながら、これらの薬剤には発熱、嘔気、嘔吐、下痢、骨髄抑制、手足抹消部のしびれ、血行不良を含む毒性の副作用がある。薬剤に起因する副作用の症状によって、患者は往々にして生活品質の低下と生命を脅かす合併症に苦しんでいる。   FOLFOX4 is a chemotherapy regimen currently used for the treatment of stage II and III and metastatic colorectal cancer and consists of drugs including fluorouracil (5-FU), leucovorin and oxaliplatin. However, these drugs have toxic side effects including fever, nausea, vomiting, diarrhea, bone marrow suppression, numbness in the extremities of the limbs, and poor circulation. Patients often suffer from poor quality of life and life-threatening complications due to the symptoms of side effects caused by drugs.

したがって、不快感を減少し、生活の質を高めるために、化学療法を受けるがん患者向けのアジュバントを見つけることが必要とされている。   Therefore, there is a need to find adjuvants for cancer patients who receive chemotherapy to reduce discomfort and improve quality of life.

さらに、慢性炎症や免疫系の活性化が腫瘍の進行に関連していることが知られている。非ステロイド性抗炎症薬(NSAID)の使用とがん進行のリスク緩和間の関連性など、慢性的な免疫活性化の関与を示唆する証拠がいくつか示されている。腫瘍形成において炎症反応のメディエーターが重要な役割を果たしており、がんの治療的介入の標的となる。化学療法を受けるときの炎症反応を調節する必要がある。   Furthermore, it is known that chronic inflammation and immune system activation are related to tumor progression. There is some evidence to suggest involvement of chronic immune activation, including an association between the use of non-steroidal anti-inflammatory drugs (NSAIDs) and risk mitigation of cancer progression. Mediators of inflammatory responses play an important role in tumorigenesis and are targets for therapeutic intervention for cancer. There is a need to regulate the inflammatory response when receiving chemotherapy.

前述を踏まえ、本発明の目的は、化学療法を受けるがん患者向けのアジュバントを製造するための組成物を提供することにある。   In view of the foregoing, an object of the present invention is to provide a composition for producing an adjuvant for cancer patients undergoing chemotherapy.

本発明の別の目的は、抗炎症剤を製造するための組成物を提供することにある。   Another object of the present invention is to provide a composition for producing an anti-inflammatory agent.

特に、本発明の組成物は、大豆発酵抽出物、緑茶抽出物、スピルリナ、ウコン抽出物、ベニクスノキタケ菌糸体、グレープシード抽出物を含む。   In particular, the composition of the present invention includes a fermented soybean extract, a green tea extract, a spirulina, a turmeric extract, a venomus mycelia mycelium, a grape seed extract.

本発明の一実施形態によると、前記組成物は大豆発酵抽出物、緑茶抽出物、スピルリナ、ウコン抽出物、ベニクスノキタケ菌糸体、グレープシード抽出物を重量比12〜30:4:1〜2:1〜2:1〜2:1〜2で含む。   According to an embodiment of the present invention, the composition comprises a fermented soybean extract, a green tea extract, a spirulina, a turmeric extract, a venix mushroom mycelium, and a grape seed extract in a weight ratio of 12-30: 4: 1-2. : 1-2: 1-2: 1-2.

本発明の一実施形態によると、前記がん患者は結腸直腸がん患者である。   According to one embodiment of the invention, the cancer patient is a colorectal cancer patient.

本発明の一実施形態によると、前記がん患者は、5−フルオロウラシル、ロイコボリン、オキサリプラチン及びそれらの任意の組み合わせから構成される群より選択された化学療法薬剤を投与する化学療法を受ける。   According to one embodiment of the invention, the cancer patient receives chemotherapy that administers a chemotherapeutic agent selected from the group consisting of 5-fluorouracil, leucovorin, oxaliplatin and any combination thereof.

本発明の一実施形態によると、本発明の前記組成物は、経口投与される。   According to one embodiment of the invention, the composition of the invention is administered orally.

一部の実施形態において、本発明の前記組成物は、大腸ポリープの増殖とがん化の阻害、及び腫瘍の大きさの縮小または生存期間の延長(特に無増悪生存期間)化学療法へのコンプライアンス向上、化学療法によって引き起こされる副作用の減少を含む化学療法の有効性向上に有効である。   In some embodiments, the composition of the invention inhibits colonic polyp growth and canceration, and reduces tumor size or prolongs survival (especially progression-free survival), compliance with chemotherapy It is effective for improving the effectiveness of chemotherapy including improvement and reduction of side effects caused by chemotherapy.

特に、本発明の前記組成物は、プロスタグランジンE2(PGE2)、一酸化窒素(NO)、腫瘍壊死因子α(TNF−α)、インターロイキン−6(IL−6)の産生減少において有効である。   In particular, the composition of the present invention is effective in reducing the production of prostaglandin E2 (PGE2), nitric oxide (NO), tumor necrosis factor α (TNF-α), and interleukin-6 (IL-6). is there.

これら及びその他の本発明の特徴は、以下の好ましい実施形態と図面による説明からより明らかになるであろう。それでも、本発明で開示される新規的概念の要旨と範囲を逸脱しない変更や修飾が可能であろう。   These and other features of the present invention will become more apparent from the following preferred embodiments and description with reference to the drawings. Nevertheless, changes and modifications may be made without departing from the spirit and scope of the novel concepts disclosed herein.

本発明の前述の概要、及び以下の詳細な説明は、添付の図面を組み合わせるとより理解しやすいであろう。本発明の例示を目的として、図面に現在好ましい実施形態を示す。しかしながら、本発明は示された正確な配置と手段に限定されないことに理解が必要である。   The foregoing summary, as well as the following detailed description of the present invention, will be better understood when taken in conjunction with the accompanying drawings. For the purpose of illustrating the invention, there are shown in the drawings embodiments which are presently preferred. It should be understood, however, that the invention is not limited to the precise arrangements and instrumentalities shown.

ラットの結腸における異常陰窩巣(ACF)を示すグラフである。(A)MB−6(B1=17.3mg/ラット/日、B2=34.6mg/ラット/日、B3=69.2mg/ラット/日)治療ラットはC1(1,2−ジメチルヒドラジン(DMH)対照)ラットと比較してACF合計数/結腸(40.6〜51.0%、*P<.05)の大幅な減少を示した。(B)ACF当たりの陰窩数が1〜3陰窩、4〜6陰窩、7陰窩以上に分類された。MB−6は化学誘発ACFの形成を大幅に抑制し、7陰窩以上の群において最高の阻害率(81.7%、*P<.05対C1群)が観察された。1 is a graph showing an abnormal crypt nest (ACF) in a rat colon. (A) MB-6 (B1 = 17.3 mg / rat / day, B2 = 34.6 mg / rat / day, B3 = 69.2 mg / rat / day) treated rats were treated with C1 (1,2-dimethylhydrazine (DMH ) Control) Showed a significant reduction in total ACF / colon (40.6-51.0%, * P <0.05) compared to rats. (B) The number of crypts per ACF was classified into 1-3 crypts, 4-6 crypts, 7 crypts or more. MB-6 significantly suppressed the formation of chemically induced ACF, and the highest inhibition rate (81.7%, * P <0.05 versus C1 group) was observed in the group with 7 or more crypts. MB−6とロイコボリン/5−フルオロウラシルの化学療法で治療した結腸腫瘍CT−26−VD保有BALB/cマウスの生存状況を示すグラフである。生存期間の中央値は、27.5日間(腫瘍のみ)、30.5日間(NC)、34.5日間(PSK)、35日間(MB−6−L)、39日間(MB−6−M)、39日間(MB−6−H)であった。NCは陰性対照、PSKは多糖K、MB−6−Lは0.625g/kgのMB−6、MB−6−Mは1.25g/kg、MB−6−Hは2.5g/kgのMB−6を示す。It is a graph which shows the survival condition of the colon tumor CT-26-VD carrying | support BALB / c mouse | mouth treated with the chemotherapy of MB-6 and leucovorin / 5-fluorouracil. Median survival was 27.5 days (tumor only), 30.5 days (NC), 34.5 days (PSK), 35 days (MB-6-L), 39 days (MB-6-M) ), 39 days (MB-6-H). NC is negative control, PSK is polysaccharide K, MB-6-L is 0.625 g / kg MB-6, MB-6-M is 1.25 g / kg, MB-6-H is 2.5 g / kg MB-6 is shown. 全例(intention to treat、ITT)数中の無増悪生存(PFS)が大幅に延長されたことを示すグラフである。It is a graph which shows that progression free survival (PFS) in the number of all cases (intention to treat, ITT) was extended significantly.

例外が特筆されない限り、ここで使われるすべての技術的及び科学的用語は、本発明が属する技術において通常の技能を有する者に一般的に理解されているものと同じ意味を持つ。   Unless otherwise noted, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.

本明細書において使用される単数形には、文脈上明らかに単数である場合を除いて、複数の指示対象も含まれる。即ち、例えば「(ある)部材(component)」と言う場合には、こうした部材及び当業者に公知のその等価物が複数ある場合も含まれる。   As used herein, the singular forms also include a plurality of referents unless the context clearly singular. That is, for example, reference to “a component” includes a case where there are a plurality of such members and their equivalents known to those skilled in the art.

本発明において、生物学的活性薬剤または組成物の投与に関連して使用される用語「治療有効量」とは、未治療の対照群と比較して、治療対象において意図される生物学的効果を生じるに足る生物学的活性薬剤または組成物の用量を指す。具体的に、本発明の組成物の有効量とは、大腸ポリープの増殖及び(または)がん化を阻害する、生存期間、特に無増悪生存期間の延長を含め化学療法の有効性を高める、化学療法へのコンプライアンスを高める、及び(または)化学療法により生じる副作用を減少する量を指す。量は投与経路と、年齢、体重、症状、治療効果、投与法、治療時間を含む治療の条件に基づいて調整できる。例えば、特定の実施形態において、本発明で使用される組成物の経口用量は1日10〜1,000mg/kgであり、具体的には1日10〜800mg/kg、より具体的には1日10〜500mg/kg、さらに具体的には1日10〜250mg/kg、より一層具体的には1日10〜150mg/kgである。   In the present invention, the term “therapeutically effective amount” as used in connection with administration of a biologically active agent or composition refers to the biological effect intended in the treated subject as compared to an untreated control group. Refers to the dose of biologically active agent or composition sufficient to produce Specifically, an effective amount of the composition of the present invention increases the effectiveness of chemotherapy, including prolonging survival, particularly progression-free survival, inhibiting colonic polyp growth and / or canceration, Refers to an amount that increases compliance with chemotherapy and / or reduces side effects caused by chemotherapy. The amount can be adjusted based on the route of administration and the conditions of treatment including age, weight, symptoms, therapeutic effect, dosage regimen, treatment time. For example, in certain embodiments, the oral dose of the composition used in the present invention is 10 to 1,000 mg / kg per day, specifically 10 to 800 mg / kg per day, more specifically 1 10 to 500 mg / kg per day, more specifically 10 to 250 mg / kg per day, more specifically 10 to 150 mg / kg per day.

本発明において使用される用語「個体」または「被験者」とは、ヒト及びコンパニオンアニマル(犬、猫など)を含むヒト以外の動物、家畜(牛、羊、豚、馬など)、または実験動物(ラット、マウス、モルモットなど)を含む。   As used herein, the term “individual” or “subject” refers to non-human animals, including humans and companion animals (dogs, cats, etc.), livestock (cattle, sheep, pigs, horses, etc.), or laboratory animals ( Rat, mouse, guinea pig, etc.).

本発明に基づき、本発明の組成物は化学療法向けのアジュバントとして使用することができる。一実施形態において、本発明の組成物の治療有効量は薬学的に許容可能な担体とともに、送達及び吸収の目的に適した形態の医薬組成物に調製することができる。投与モードにより、本発明の医薬組成物は、好ましくは、約10%〜約100%、または約20%〜約100%、または約40%〜約100%、または約60%〜約100%の重量の活性成分を含み、そのうち、前記重量%は組成物全体の重量に基づいて算出される。   In accordance with the present invention, the composition of the present invention can be used as an adjuvant for chemotherapy. In one embodiment, a therapeutically effective amount of a composition of the present invention can be prepared with a pharmaceutically acceptable carrier into a pharmaceutical composition in a form suitable for delivery and absorption purposes. Depending on the mode of administration, the pharmaceutical composition of the present invention is preferably about 10% to about 100%, or about 20% to about 100%, or about 40% to about 100%, or about 60% to about 100%. Containing the active ingredient by weight, of which the weight percentage is calculated based on the weight of the entire composition.

ここで使用される用語「担体」または「薬学的に許容可能な担体」とは、医薬業界における当業者の知るところにあるものを含む、医薬品用の希釈剤または賦形剤などを指す。   The term “carrier” or “pharmaceutically acceptable carrier” as used herein refers to diluents or excipients for pharmaceuticals, including those known to those skilled in the pharmaceutical arts.

本発明は大豆発酵抽出物、緑茶、スピルリナ、ウコン抽出物、ベニクスノキタケ菌糸体、グレープシード抽出物を主に含む組成物を提供し、前記組成物は、化学療法を受けるがん患者向けのアジュバントの製造に使用することができる。   The present invention provides a composition mainly comprising a soybean fermented extract, green tea, spirulina, turmeric extract, venix mushroom mycelium, grape seed extract, said composition for cancer patients undergoing chemotherapy It can be used for the production of an adjuvant.

具体的に、本発明で使用される大豆発酵抽出物は、水性大豆抽出物と少なくとも1種の乳酸菌及び選択的に少なくとも1種の酵母の発酵によって製造された抽出物を指す。一実施形態において、前記少なくとも1種の乳酸菌はラクトバシラス種であり、前記少なくとも1種の酵母はサッカロミケス種である。特定の実施形態において、前記発酵はラクトバシラスの異種培養を用いて実施され、例えば、5または10、15、20、25、30株のラクトバシラスの培養と、好ましくは少なくとも1種の酵母が、前記ラクトバシラスの異種培養に添加される。前記発酵に使用可能なラクトバシラスの株は、Lactobacillus acidophilus CCRC(台湾食品工業発展研究所生物資源保存及び研究センター、Bioresource Collection and Research Center at Food Industry Research and Development)10695及び(または)14026、14064、14065、14079、Lactobacillus delbrueckii bulgaricus CCRC 10696及び(または)14007、14009、14010、14069、14071、14098、16054、Lactobacillus lactis lactis CCRC 10791及び(または)12267、12306、12312、12315、12323、14016、14015、14117、Lactobacillus kefir CCRC 14011及び(または)Lactobacillus kefiranofaciens CCRC 16059を含むがこれらに限らない。前記発酵に使用可能な酵母株は、Saccharomyces cerevisiae CCRC 20577及び(または)20578、20581、21494、21550、21797、21805、22138、22234、22337、22731、22728、及び(または)Candida kefyr CCRC 21269及び(または)21742、22057を含むがこれらに限らない。具体的に、前記発酵の後には滅菌、ろ過、濃縮、凍結乾燥またはこれらの任意の組み合わせなどの工程が続く。好ましくは、前記発酵の後に例えば加熱による滅菌が行われ、さらに選択的にろ過及び濃縮が行われる。より好ましくは、前記大豆発酵抽出物を例えば凍結乾燥により乾燥させて、粉末状の大豆発酵抽出物を得る。特定の実施形態において、本発明の大豆発酵抽出物は、(a)水性大豆抽出物を少なくとも1種の乳酸菌及び少なくとも1種の酵母と発酵させて発酵液を形成する工程、(b)前記発酵液を滅菌する工程、(c)滅菌済みの前記発酵液をろ過してろ液を収集する工程、(d)前記ろ液を濃縮し、濃縮大豆発酵抽出物を得る工程を含むプロセスによって製造される。具体的な実施形態において、大豆は蒸留水と混合され(例えば、約1:10の割合で)、この混合物が約100℃で約30分間加熱された後ろ過される。これにより水性大豆抽出物が得られ、さらに少なくとも1種の乳酸菌と少なくとも1種の酵母で適切な培地において発酵され、36〜43℃で45〜50時間培養される。発酵した抽出物が滅菌、ろ過、(例えばろ液中の約95%の水含量を除去して)濃縮され、濃縮された形態の大豆発酵抽出物が得られる。特定の実施形態において、前記大豆発酵抽出物の具体的な比重は1.136g/mlで、71.49%の水分、5.15%の灰、0.16%の粗脂肪、5.45%の粗タンパク質、0.15%の粗繊維、及び炭水化物を含む。また、0.004mg/100gのビタミンB1、0.12mg/100gのビタミンB2、2.17mg/100gの鉄、113.55mg/100gのカルシウム、379.19mg/100gのリンなど複数のビタミンとミネラルも含むことができる。本発明で使用される大豆発酵抽出物は、米国特許第6855350号及び第6733801号に従って調製され、これらの文献はその全体を参照として本明細書に組み入れる。   Specifically, the fermented soybean extract used in the present invention refers to an extract produced by fermentation of an aqueous soy extract and at least one lactic acid bacterium and optionally at least one yeast. In one embodiment, the at least one lactic acid bacterium is a Lactobacillus species and the at least one yeast is a Saccharomyces species. In a particular embodiment, the fermentation is carried out using a Lactobacillus heterogeneous culture, for example a culture of 5 or 10, 15, 20, 25, 30 strains of Lactobacillus and preferably at least one yeast is said Lactobacillus. Added to different cultures. The Lactobacillus strains that can be used for the fermentation are Lactobacillus acidophilus CCRC (Taiwan Food Industry Development Laboratory Bioresource Conservation and Research Center, Bioresource Collection and Research Center at Food Research and Development 64 and 14065, 10695). 14079, Lactobacillus delbrueckii bulgaricus CCRC 10696 and / or 14007, 14009, 14010, 14069, 14071, 14098, 16054, Lactobacillus lactis lactis CCRC 10791 and / or 12267, 12306, 12312, 12315, 12323, 14016, 14015, 14117, Lactobacillus kefir CCRC 14011 and / or Lactobacillus kefiranofaciens CCRC 16059. Yeast strains that can be used for the fermentation include Saccharomyces cerevisiae CCRC 20577 and / or 20578, 20581, 21494, 21550, 21597, 21805, 21138, 22234, 22337, 22731, 27728, and / or Candida kefyr CCRC 21269 and ( Or) including, but not limited to, 21742, 22057. Specifically, the fermentation is followed by steps such as sterilization, filtration, concentration, lyophilization or any combination thereof. Preferably, sterilization, for example, by heating is performed after the fermentation, and filtration and concentration are selectively performed. More preferably, the soybean fermented extract is dried by freeze drying, for example, to obtain a powdered fermented soybean extract. In a specific embodiment, the soybean fermented extract of the present invention comprises (a) fermenting an aqueous soybean extract with at least one lactic acid bacterium and at least one yeast to form a fermentation liquor, (b) the fermentation Manufactured by a process comprising the steps of: sterilizing a liquid; (c) collecting the filtrate by filtering the sterilized fermentation broth; and (d) concentrating the filtrate to obtain a concentrated soybean fermented extract. . In a specific embodiment, soy is mixed with distilled water (eg, in a ratio of about 1:10) and the mixture is heated at about 100 ° C. for about 30 minutes and then filtered. This yields an aqueous soy extract, which is further fermented in a suitable medium with at least one lactic acid bacterium and at least one yeast and cultured at 36-43 ° C. for 45-50 hours. The fermented extract is sterilized, filtered, and concentrated (eg, removing about 95% water content in the filtrate) to obtain a concentrated form of soy fermented extract. In a specific embodiment, the specific gravity of the soy fermentation extract is 1.136 g / ml, 71.49% moisture, 5.15% ash, 0.16% crude fat, 5.45% Of crude protein, 0.15% crude fiber, and carbohydrates. Also, multiple vitamins and minerals such as 0.004 mg / 100 g vitamin B1, 0.12 mg / 100 g vitamin B2, 2.17 mg / 100 g iron, 113.55 mg / 100 g calcium, 379.19 mg / 100 g phosphorus Can be included. The soybean fermented extract used in the present invention is prepared according to US Pat. Nos. 6,855,350 and 6,733,801, which are incorporated herein by reference in their entirety.

さらに、前記緑茶抽出物、前記ウコン抽出物、前記グレープシード抽出物は、水とエタノールでの抽出を含む一般的な標準手順で作成される。スピルリナは収穫後乾燥させて粉末または錠剤にすることで得られる。前記ベニクスノキタケ菌糸体は一般的な液体発酵の後、乾燥、濃縮及び(または)粉末に研磨することで得られる。これら成分はすべてメーカーから市販されており、購入可能である。   Furthermore, the green tea extract, the turmeric extract, and the grape seed extract are made by a standard procedure that includes extraction with water and ethanol. Spirulina is obtained by drying after harvesting into a powder or tablet. The above-mentioned venix mushroom mycelium can be obtained by drying, concentrating and / or polishing to a powder after general liquid fermentation. All of these components are commercially available from the manufacturer and can be purchased.

本発明のいくつかの実施形態に基づき、本発明の組成物は、大豆発酵抽出物、緑茶抽出物、スピルリナ、ウコン抽出物、ベニクスノキタケ菌糸体、グレープシード抽出物を含み、それらの重量比は12〜30:4:1〜2:1〜2:1〜2:1〜2であり、好ましくは12〜20:4:1〜2:1〜2:1〜2:1〜2である。本発明の具体的な実施形態に基づくと、大豆発酵抽出物、緑茶抽出物、スピルリナ、ウコン抽出物、ベニクスノキタケ菌糸体、グレープシード抽出物の重量比は12:4:2:1:1:1である。本発明の別の具体的な実施形態に基づくと、大豆発酵抽出物、緑茶抽出物、スピルリナ、ウコン抽出物、ベニクスノキタケ菌糸体、グレープシード抽出物の重量比は12:4:1:1:2:1である。上述の重量比下で、前記組成物の各成分の重量割合は異なる状況に基づいて適切に調整することができる。   In accordance with some embodiments of the present invention, the composition of the present invention comprises a fermented soy extract, green tea extract, spirulina, turmeric extract, venix mushroom mycelium, grape seed extract, and their weight ratio Is 12 to 30: 4: 1 to 2: 1 to 2: 1 to 2: 1 to 2, preferably 12 to 20: 4: 1 to 2: 1 to 2: 1 to 2: 1 to 2. . According to a specific embodiment of the present invention, the weight ratio of soybean fermented extract, green tea extract, spirulina, turmeric extract, venix mushroom mycelium, grape seed extract is 12: 4: 2: 1: 1. : 1. According to another specific embodiment of the present invention, the weight ratio of soy fermentation extract, green tea extract, spirulina, turmeric extract, venix mushroom mycelium, grape seed extract is 12: 4: 1: 1. : 2: 1. Under the above weight ratio, the weight ratio of each component of the composition can be appropriately adjusted based on different situations.

本発明の組成物は食品または医薬品として製造することができる。食品タイプの製品は従来の賦形剤または充填剤を用いた任意の従来の技法により調製することができ、必要に応じて添加物を混合することができる。また、医薬品は本発明の組成物の治療有効量と薬学的に許容可能な担体を用い、一般に薬学的技法または方法で調製することができる。本発明の組成物の投与には、経口投与または注射が含まれ、さらに薬学的に許容可能な担体、希釈剤及び(または)賦形剤を含んでもよい。上述の担体は、溶剤、分散媒質、塗膜、抗菌剤、抗真菌薬、等張剤、吸収遅延剤等とすることができる。前記希釈剤は無菌注射用水(BWFI)、リン酸緩衝液、リンガー溶液、またはグルコース溶液等とすることができる。前記賦形剤は、炭酸カルシウム、炭酸ナトリウム、リン酸カルシウム、リン酸ナトリウム、乳糖、でんぷん、ゼラチン、アルギン酸、ステアリン酸、ステアリン酸マグネシウム等とすることができる。   The composition of this invention can be manufactured as a foodstuff or a pharmaceutical. Food-type products can be prepared by any conventional technique using conventional excipients or fillers, and additives can be mixed as needed. In addition, pharmaceuticals can generally be prepared by pharmaceutical techniques or methods using a therapeutically effective amount of the composition of the present invention and a pharmaceutically acceptable carrier. Administration of the composition of the present invention includes oral administration or injection, and may further comprise a pharmaceutically acceptable carrier, diluent and / or excipient. The carriers described above can be solvents, dispersion media, coatings, antibacterial agents, antifungal agents, isotonic agents, absorption delaying agents, and the like. The diluent can be sterile water for injection (BWFI), phosphate buffer, Ringer's solution, glucose solution, or the like. The excipient may be calcium carbonate, sodium carbonate, calcium phosphate, sodium phosphate, lactose, starch, gelatin, alginic acid, stearic acid, magnesium stearate and the like.

一部の特定の実施形態において、本発明の組成物は大腸ポリープの増殖及び(または)がん化を阻害する効果を備えている。具体的に、実施形態に示されるように、本発明の組成物は1,2−ジメチルヒドラジン(DMH)により誘発される異常陰窩巣(ACF)を阻害することができ、かつ大腸ポリープの増殖及び(または)がん化を阻害する効果も備えている。   In some specific embodiments, the compositions of the invention have the effect of inhibiting colonic polyp growth and / or canceration. Specifically, as shown in the embodiments, the composition of the present invention can inhibit abnormal crypt foci (ACF) induced by 1,2-dimethylhydrazine (DMH) and proliferate colon polyps. And / or has an effect of inhibiting canceration.

一部の特定の実施形態において、本発明の組成物は、がん患者の生存、特に無増悪生存期間の延長を含む、化学療法の有効性を向上する効果を備えている。実施形態に示されるように、本発明の組成物と化学療法を受けている被験者は、本発明の組成物なしで化学療法を受けている被験者より寿命が延長され、特に無増悪生存期間が延長された。   In some specific embodiments, the compositions of the invention have the effect of improving the effectiveness of chemotherapy, including prolonging survival of cancer patients, particularly progression-free survival. As shown in the embodiments, subjects receiving chemotherapy with the composition of the present invention have a longer life than subjects receiving chemotherapy without the composition of the present invention, in particular, progression-free survival. It was done.

一部の特定の実施形態において、本発明の組成物は化学療法へのコンプライアンス向上の効果を備えている。実施形態に示されるように、本発明の組成物と化学療法を受けている被験者は、本発明の組成物なしで化学療法を受けている被験者より、大幅に高い化学療法へのコンプライアンスを示した。   In some specific embodiments, the compositions of the invention have the effect of improving compliance with chemotherapy. As shown in the embodiments, subjects receiving chemotherapy with the composition of the present invention showed significantly higher chemotherapy compliance than subjects receiving chemotherapy without the composition of the present invention. .

一部の特定の実施形態において、本発明の組成物は化学療法により引き起こされる副作用を減少する効果を備えている。実施形態に示されるように、本発明の組成物をがんの化学療法プロセス中に与えると、前記組成物が化学療法の副作用、特に、腎損傷の減少と好中球減少症の改善が可能であることが分かった。   In some specific embodiments, the compositions of the invention have the effect of reducing the side effects caused by chemotherapy. As shown in the embodiment, when the composition of the present invention is given during the cancer chemotherapy process, the composition can reduce the side effects of chemotherapy, especially kidney damage and neutropenia It turns out that.

加えて、本発明に基づき、本明細書で説明する組成物は、抗炎症剤として使用することができる。具体的に、本発明の組成物は、プロスタグランジンE2(PGE2)、一酸化窒素(NO)、腫瘍壊死因子α(TNF−α)、インターロイキン−6(IL−6)など、マクロファージ等の炎症細胞中の炎症メディエーター産生を効果的に減少する。   In addition, based on the present invention, the compositions described herein can be used as anti-inflammatory agents. Specifically, the composition of the present invention includes prostaglandin E2 (PGE2), nitric oxide (NO), tumor necrosis factor α (TNF-α), interleukin-6 (IL-6), macrophages and the like. Effectively reduces inflammatory mediator production in inflammatory cells.

本発明は、前立腺がん、乳がん、子宮がん、血液がん、卵巣がん、子宮内膜がん、子宮頸がん、結腸直腸がん、精巣がん、リンパ腫、横紋筋肉腫、神経芽細胞腫、膵臓がん、肺がん、脳腫瘍、皮膚がん、胃がん、口腔がん、肝臓がん、喉頭がん、胆嚢がん、甲状腺がん、肝臓がん、腎臓がん、上咽頭がんを含む、がんを患う患者に適用できる。本発明の一実施形態によると、前記患者は結腸直腸がん患者である。本発明の実施形態に基づき、前記化学療法は、5−フルオロウラシル、ロイコボリン、オキサリプラチンを含む薬剤を使用した治療である。   The present invention includes prostate cancer, breast cancer, uterine cancer, blood cancer, ovarian cancer, endometrial cancer, cervical cancer, colorectal cancer, testicular cancer, lymphoma, rhabdomyosarcoma, nerve Blastoma, pancreatic cancer, lung cancer, brain cancer, skin cancer, stomach cancer, oral cancer, liver cancer, laryngeal cancer, gallbladder cancer, thyroid cancer, liver cancer, kidney cancer, nasopharyngeal cancer Applicable to patients with cancer, including According to one embodiment of the invention, the patient is a colorectal cancer patient. According to an embodiment of the present invention, the chemotherapy is treatment using a drug comprising 5-fluorouracil, leucovorin, oxaliplatin.

本発明は以下の実施形態によってさらに明らかになる。以下の実施形態は限定ではなく、例示を目的として提供されている。当業者であれば、本発明の開示を踏まえ、開示された具体的な実施形態において多くの変更が可能であり本発明の要旨と範囲から逸脱することなく、同じまたは類似の結果を得ることができる点に理解が必要である。   The present invention will be further clarified by the following embodiments. The following embodiments are provided for purposes of illustration and not limitation. Those skilled in the art will be able to make many changes in the disclosed specific embodiments based on the disclosure of the present invention and obtain the same or similar results without departing from the spirit and scope of the present invention. It is necessary to understand what can be done.

1,2−ジメチルヒドラジン(DMH)により誘発される異常陰窩巣(ACF)形成の阻害 Inhibition of abnormal crypt nest (ACF) formation induced by 1,2-dimethylhydrazine (DMH)

1.1 材料および方法 1.1 Materials and methods

動物実験が国立台湾大学動物実験委員会(Institutional Animal Care and Use Committee of National Taiwan University)により許可され、すべての動物が実験動物のケアと使用に関する標準指針に従って処理された。約6週齢のF344オスラットが国立台湾大学医学部実験動物センター(Laboratory Animal Center, College of Medicine, National Taiwan University)より購入された。   Animal experiments were approved by the National Taiwan University Animal Experiment Committee and the National Committee of National Taiwan University, and all animals were processed according to standard guidelines for the care and use of laboratory animals. Approximately 6 weeks old F344 male rats were purchased from the Laboratory Animal Center of the National Taiwan University School of Medicine (College of Medicine, National Taiwan University).

オスF344ラットは重量別に8匹ずつ無作為にグループ分けされた。ラットは取得1週間後から重量パーセントで17.7%のたんぱく質、64.9%の炭水化物、5.2%の脂肪を含むHarlan AIN−76齧歯類用精製飼料(Harlan Laboratories、米国インディアナ州インディアナポリス)(Harlan AIN−76A精製飼料データシート、Harlan Laboratories.参照元:http://www.harlan.com/download.axd/1131a6e1617348b9a99c8d2af933f796.pdf?d=170481)が与えられた。ラットはステンレス製のケージ内に飼料と蒸留水を自由に摂取できる状態で保たれた。動物の部屋は23±1℃、湿度50±10%、12時間ごとの昼夜サイクルで維持された。飼料の摂取と重量が毎日記録された。   Male F344 rats were randomly grouped by 8 by weight. Rats began using Harlan AIN-76 rodent refined diet (Harlan Laboratories, Indiana, USA) containing 17.7% protein, 64.9% carbohydrate, 5.2% fat by weight from one week after acquisition. (Police) (Harlan AIN-76A purified feed data sheet, Harlan Laboratories. Source: http://www.harlan.com/download.axd/1131a6e1617348b9a99c8d2af933f796.pdf=170). Rats were kept in a stainless steel cage with free access to food and distilled water. The animal room was maintained at 23 ± 1 ° C., 50 ± 10% humidity, with a 12 hour day / night cycle. Feed intake and weight were recorded daily.

本発明の組成物(MB−6)には、例えば大豆発酵抽出物(米国特許第6855350号及び第6733801号の方法に従って調製)、緑茶抽出物(Camellia sinensis O.)、スピルリナアルジー(Arthrospira platensis)、ウコン抽出物(Curcuma longa L.)、ベニクスノキタケ菌糸体、グレープシード抽出物(Vitis vinifera)の6つの成分が含まれる。表1に特定の実施形態における重量比12:4:2:1:1:1のMB−6の成分を示す。   The composition (MB-6) of the present invention includes, for example, soybean fermentation extract (prepared according to the methods of US Pat. Nos. 6,855,350 and 6,733,801), green tea extract (Camellia sinensis O.), Spirulina algi (Arthrospira platensis) , Turmeric extract (Curcuma longa L.), Benix nokitake mycelium, grape seed extract (Vitis vinifera). Table 1 shows the components of MB-6 in a specific embodiment in a weight ratio of 12: 4: 2: 1: 1: 1.

Figure 2015051970
Figure 2015051970

動物は、1,2−ジメチルヒドラジン(DMH)注射を受けた対照群C1、生理食塩水注射を受けた対照群C2、本発明の6成分製剤の低用量(17.3mg/ラット/日)を受けた実験群B1、本発明の6成分製剤の中用量(34.6mg/ラット/日)を受けた実験群B2、本発明の6成分製剤MB−6の高用量(69.2mg/ラット/日)を受けた実験群B3にグループ分けされた。DMHを生理食塩水(0.9%NaCl溶液)に溶解して2%溶液が作成され、pH6.7に調整された後等分され、−20℃で必要時まで保管された。DMHが腹腔内(IP)注射で毎週20mg/kgの用量で4週間投与された(体重1kgにつき2%溶液1mlの注射に相当)。対照群には生理食塩水の注射が行われた(体重1kgにつき1mlの生理食塩水)。表2と表3に示す。   The animals received a control group C1 that received 1,2-dimethylhydrazine (DMH) injection, a control group C2 that received saline injection, and a low dose (17.3 mg / rat / day) of the six-component formulation of the present invention. Experimental group B1 received, experimental group B2 received medium dose (34.6 mg / rat / day) of the six component formulation of the present invention, high dose (69.2 mg / rat / day) of the six component formulation MB-6 of the present invention Group) was received in the experimental group B3. DMH was dissolved in physiological saline (0.9% NaCl solution) to make a 2% solution, adjusted to pH 6.7, aliquoted, and stored at -20 ° C until needed. DMH was administered by intraperitoneal (IP) injection at a dose of 20 mg / kg weekly for 4 weeks (corresponding to 1 ml injection of 2% solution per kg body weight). The control group was injected with saline (1 ml saline per kg body weight). Tables 2 and 3 show.

Figure 2015051970
Figure 2015051970

Figure 2015051970
Figure 2015051970

15週目に動物は1晩絶食され、翌日二酸化炭素吸引により安楽死させられた。その後結腸が取り除かれて長さと重量が測定され、異常陰窩巣(ACF)の数と各ACFを構成する陰窩の数がカウントされた。簡単に言うと、結腸は長手方向の軸に沿って切られ、粘膜内層を上向きに開いて広げられ、手前、中間、後方の3部分に切断された。各部分が10%の緩衝ホルマリンを入れたペトリ皿内のフィルタ紙2枚の間に置かれ、24時間以上固定された。ACF定量化のため、固定された結腸組織が0.2%メチレンブルー(PBSを溶剤とする)で染色され、光学顕微鏡40〜100倍の拡大率でACF数が評価された。陰窩数を判定するため、ACF当たりの陰窩数がさらに1〜3陰窩、4〜6陰窩、7陰窩以上に分類された。   At 15 weeks, the animals were fasted overnight and euthanized by carbon dioxide aspiration the next day. The colon was then removed, length and weight were measured, and the number of abnormal crypt foci (ACF) and the number of crypts that make up each ACF were counted. Briefly, the colon was cut along the longitudinal axis, widened with the mucosal lining open upward, and cut into three parts, front, middle and posterior. Each part was placed between two sheets of filter paper in a Petri dish containing 10% buffered formalin and fixed for over 24 hours. For ACF quantification, fixed colon tissue was stained with 0.2% methylene blue (PBS as solvent), and ACF numbers were evaluated at 40-100 times magnification using a light microscope. In order to determine the number of crypts, the number of crypts per ACF was further classified into 1-3 crypts, 4-6 crypts, 7 crypts or more.

SASソフトウェアを使用して実験データが一元配置分散分析法(One−way ANOVA)により分析され、スチューデントのT検定で群間の違いの比較が行われた。p<0.05の結果は有意差を示している。   Experimental data were analyzed by one-way analysis of variance (One-way ANOVA) using SAS software, and differences between groups were compared with Student's T-test. Results with p <0.05 indicate a significant difference.

1.2 結果 1.2 Results

合計50匹のF344ラットがC1(DMH対照群)、C2(標準対照群)、B1(MB−6−L)、B2(MB−6−M)、B3(MB−6−H)の群に無作為に割り当てられた。経時的に5群の体重の間に大きな差はなかった。結腸当たりのACF数を図1Aに示す。MB−6治療ラットは、C1(DMH対照群)のラットと比較して、結腸当たりの合計ACF数において有意な減少を示した(40.6〜51.0%、P<.05)。ACF当たりの陰窩数がさらに1〜3陰窩(小)、4〜6陰窩(中)、7陰窩以上(大)に分類された。MB−6は化学誘発ACFの形成を有意に抑制し、かつ7陰窩以上の群において最高の阻害率(81.7%、図1B)が観察された。表4と表5も併せて参照する。   A total of 50 F344 rats were assigned to C1 (DMH control group), C2 (standard control group), B1 (MB-6-L), B2 (MB-6-M), and B3 (MB-6-H) groups. Randomly assigned. There was no significant difference between the weights of the five groups over time. The number of ACFs per colon is shown in FIG. 1A. MB-6 treated rats showed a significant decrease in total ACF counts per colon (40.6-51.0%, P <0.05) compared to C1 (DMH control group) rats. The number of crypts per ACF was further classified into 1 to 3 crypts (small), 4 to 6 crypts (medium), and 7 or more crypts (large). MB-6 significantly suppressed the formation of chemically induced ACF, and the highest inhibition rate (81.7%, FIG. 1B) was observed in the group with 7 or more crypts. Refer also to Table 4 and Table 5.

Figure 2015051970
Figure 2015051970

Figure 2015051970
Figure 2015051970

まとめると、動物実験では本発明の6成分の調剤が1,2−ジメチルヒドラジンにより誘発される異常陰窩巣の数を効果的に減少し、特に大きい異常陰窩巣の発生を減少することが示された。   In summary, in animal experiments, the six-component formulation of the present invention can effectively reduce the number of abnormal crypt foci induced by 1,2-dimethylhydrazine, especially the occurrence of large abnormal crypt foci. Indicated.

1,2−ジメチルヒドラジン(DMH)により誘発される結腸直腸がん形成の阻害 Inhibition of colorectal cancer formation induced by 1,2-dimethylhydrazine (DMH)

2.1 材料および方法 2.1 Materials and methods

購入した6週齢のF344オスラットが取得1週間後に無作為にグループ分けされた。各群30匹とした。動物は、1,2−ジメチルヒドラジン(DMH)注射を受けた対照群C1、生理食塩水注射を受けた対照群C2、本発明の6成分製剤の中用量(34.6mg/ラット/日)を受けた実験群B2にグループ分けされた。DMHを生理食塩水(0.9%NaCl溶液)に溶解して2%溶液が作成され、pH6.7に調整された後等分され、−20℃で必要時まで保管された。DMHが腹腔内(IP)注射で毎週30mg/kgの用量で8週間投与された(体重1kgにつき2%溶液1.5mlの注射に相当)。対照群には生理食塩水の注射が行われた(体重1kgにつき1.5mlの生理食塩水)。表6と表7に示す。   Purchased 6 week old F344 male rats were randomly grouped one week after acquisition. There were 30 animals in each group. The animals received a control group C1 that received 1,2-dimethylhydrazine (DMH) injection, a control group C2 that received saline injection, and a medium dose (34.6 mg / rat / day) of the six-component formulation of the present invention. Grouped into experimental group B2 received. DMH was dissolved in physiological saline (0.9% NaCl solution) to make a 2% solution, adjusted to pH 6.7, aliquoted, and stored at -20 ° C until needed. DMH was administered by intraperitoneal (IP) injection at a dose of 30 mg / kg weekly for 8 weeks (equivalent to 1.5 ml injection of 2% solution per kg body weight). The control group was injected with saline (1.5 ml saline per kg body weight). Tables 6 and 7 show.

Figure 2015051970
Figure 2015051970

Figure 2015051970
Figure 2015051970

32週の実験期間の終了時に、実験動物は1晩絶食され、翌日エーテル麻酔により安楽死させられ、解剖された。動物の結腸が取り除かれて長さと重量が測定され、続く腫瘍分析のために保存された。   At the end of the 32 week experimental period, the experimental animals were fasted overnight, euthanized the next day by ether anesthesia, and dissected. The animal's colon was removed and length and weight were measured and stored for subsequent tumor analysis.

結腸、盲腸、小腸、胃が取り出され、これら臓器の長手方向の軸に沿って完全に切開された。リン酸緩衝生理食塩水での洗浄後、これら臓器はフィルタ紙上に粘膜を上向きにして平らに置かれた。試料上の腫瘍が検査され、腫瘍の位置、数、大きさが記録された。各腫瘍の長さ(L)、幅(W)、深さ(D)がバーニヤで測定され、各腫瘍の体積(V)が公式:V=(L*W*D*π)/6で算出された。試料の写真が撮影され、保存された。   The colon, cecum, small intestine, and stomach were removed and completely incised along the longitudinal axis of these organs. After washing with phosphate buffered saline, the organs were placed flat on the filter paper with the mucosa facing up. The tumor on the sample was examined and the location, number and size of the tumor was recorded. The length (L), width (W), and depth (D) of each tumor is measured by vernier, and the volume (V) of each tumor is calculated by the formula: V = (L * W * D * π) / 6 It was done. A photograph of the sample was taken and stored.

SASソフトウェアを使用して実験データが一元配置分散分析法(One−way ANOVA)により分析され、スチューデントのT検定で群間の違いの比較が行われた。p<0.05の結果は有意差を示している。   Experimental data were analyzed by one-way analysis of variance (One-way ANOVA) using SAS software, and differences between groups were compared with Student's T-test. Results with p <0.05 indicate a significant difference.

2.2 結果 2.2 Results

表8に示すように、腫瘍数について、本発明の6成分調剤は1,2−ジメチルヒドラジンにより誘発される腫瘍形成を効果的に阻害し、阻害率は38.4%で、統計的有意差があった(p<0.05)。   As shown in Table 8, with regard to the number of tumors, the 6-component preparation of the present invention effectively inhibited tumor formation induced by 1,2-dimethylhydrazine, and the inhibition rate was 38.4%, a statistically significant difference. (P <0.05).

Figure 2015051970
Figure 2015051970

さらに、表9に示すように、腫瘍はその体積に基づいて、0〜100mm(小型)、100〜500mm(中型)、500mm超(大型)の3つのグループに分類された。本発明の6成分調剤は1,2−ジメチルヒドラジンにより誘発される腫瘍を阻害でき、特に、小型(0〜100mm)の腫瘍を阻害率49.7%で有意に阻害した(p<0.001)。 Furthermore, as shown in Table 9, the tumor on the basis of the volume, 0 to 100 mM 3 (small), 100 to 500 mm 3 (medium size) were divided into three groups of 500 mm 3 greater (large). The six-component preparation of the present invention can inhibit tumors induced by 1,2-dimethylhydrazine, and in particular, small (0-100 mm 3 ) tumors were significantly inhibited with an inhibition rate of 49.7% (p <0. 0). 001).

Figure 2015051970
Figure 2015051970

マウスにおける結腸直腸がんの体積減少と生存期間の延長 Volume reduction and survival of colorectal cancer in mice

3.1 材料および方法 3.1 Materials and methods

オスBALB/cマウスが台湾国立実験動物センター(National Laboratory Animal Center)で購入され、21±2℃、12時間ごとの昼夜サイクルで維持された。AIN−76精製飼料と蒸留水が不断給与された。   Male BALB / c mice were purchased at the National Laboratory Animal Center of Taiwan and maintained at 21 ± 2 ° C. with a 12 hour day / night cycle. AIN-76 refined feed and distilled water were constantly fed.

マウスが12匹ずつ7つの群に無作為に分けられ、1ケージに3匹ずつ入れられた。マウス結腸がん細胞株CT−26−VDが5%のCOを含むインキュベータ内で10%ウシ胎児血清を含む完全なダルベッコ改変イーグル培地(DMEM、GIBCO、Cat#11995)で37℃で培養された。マウス結腸がんモデルの確立については先に説明されている(Witek M et al.、Int J Radiat Oncol Biol Phys 2014;88:1188〜95)。簡潔には、マウスの重量を測定し、体重に基づいた適切な用量のペントバルビタールナトリウム(Somnotol、カナダ、10μL/g、6.5mg/ml)で麻酔した。100μLのCT−26−VD細胞懸濁液(2×10細胞/mL、2×10細胞/マウス)がマウスの脾臓に注射され(0日目)、動物は回復まで低体温症から保護された。治療されたマウスには3つの異なる用量のMB−6(0.625g/kg(MB−6−L)、1.25g/kg(MB−6−M)、2.5g/kg(MB−6−H))が経口投与された。その後マウスは腫瘍細胞注射後3日目と6日目にLV/5−FU(ロイコボリン/5−フルオロウラシル)で治療された。陰性対照(NC)と陽性群マウスには被験物質MB−6の代わりとしてそれぞれ滅菌ミリQ水とPSK(多糖K、0.325g/kg)が投与された。各群の概要を表10に示す。 Mice were randomly divided into 7 groups of 12 mice and 3 mice were placed in a cage. The mouse colon cancer cell line CT-26-VD was cultured at 37 ° C. in complete Dulbecco's modified Eagle medium (DMEM, GIBCO, Cat # 11995) containing 10% fetal calf serum in an incubator containing 5% CO 2. It was. The establishment of a mouse colon cancer model has been described previously (Witek M et al., Int J Radiate Oncol Biol Phys 2014; 88: 1188-95). Briefly, mice were weighed and anesthetized with an appropriate dose of pentobarbital sodium based on body weight (Somnotol, Canada, 10 μL / g, 6.5 mg / ml). 100 μL of CT-26-VD cell suspension (2 × 10 5 cells / mL, 2 × 10 4 cells / mouse) was injected into the spleen of mice (day 0) and the animals were protected from hypothermia until recovery It was done. Treated mice had three different doses of MB-6 (0.625 g / kg (MB-6-L), 1.25 g / kg (MB-6-M), 2.5 g / kg (MB-6 -H)) was administered orally. The mice were then treated with LV / 5-FU (leucovorin / 5-fluorouracil) on days 3 and 6 after tumor cell injection. Sterile milliQ water and PSK (polysaccharide K, 0.325 g / kg) were administered to the negative control (NC) and positive group mice, respectively, instead of the test substance MB-6. A summary of each group is shown in Table 10.

Figure 2015051970
NC、陰性対照;PSK、多糖K;MB−6−L、0.625g/kg MB−6;MB−6−M、1.25g/kg;MB−6−H、2.5g/kg MB−6;LV/5−FU、ロイコボリン/5−フルオロウラシル。
Figure 2015051970
NC, negative control; PSK, polysaccharide K; MB-6-L, 0.625 g / kg MB-6; MB-6-M, 1.25 g / kg; MB-6-H, 2.5 g / kg MB- 6; LV / 5-FU, leucovorin / 5-fluorouracil.

各マウスの体重が週1回監視された。15日目にマウスにルシフェリン(150mg/kg IP)を注射し、3%イソフルランの吸入により麻酔した後、非侵襲性IVISイメージングシステム100シリーズ(Xenogen、カリフォルニア州)を装備した暗い箱の中に入れた。腫瘍成長を判定するためにシステムソフトウェアを使用して生体発光が定量化された。各マウスの寿命が55日まで記録された。研究終了後、すべての動物が二酸化炭素吸引により安楽死された。   The body weight of each mouse was monitored once a week. On day 15, mice were injected with luciferin (150 mg / kg IP) and anesthetized by inhalation of 3% isoflurane, then placed in a dark box equipped with a non-invasive IVIS imaging system 100 series (Xenogen, CA). It was. Bioluminescence was quantified using system software to determine tumor growth. The lifetime of each mouse was recorded up to 55 days. At the end of the study, all animals were euthanized by carbon dioxide inhalation.

3.2 結果 3.2 Results

LV/5−FU化学療法群と比較して、MB−6はCT−26−VD保有BALB/cマウスの飼料摂取及び体重に影響しなかった。生体発光定量化の結果、1.25または2.5g/kgのMB−6投与はLV/5−FU化学療法による治療の有効性を有意に高めることが示された(データ未提示)。生存期間の中央値は27.5日(腫瘍のみ)、30.5日(NC)、34.5日(PSK)、35日(MB−6−L)、39日(MB−6−M)、39日(MB−6−H)であった(図2)。0.625〜2.5g/kgのMB−6の投与は化学療法を受けた腫瘍保有マウスの寿命を有意に延長したが、そのうちMB−6−MはNC群と比較して30%以上寿命を延長した。   Compared to the LV / 5-FU chemotherapy group, MB-6 did not affect feed intake and body weight of CT-26-VD bearing BALB / c mice. Bioluminescence quantification showed that 1.25 or 2.5 g / kg MB-6 administration significantly increased the efficacy of treatment with LV / 5-FU chemotherapy (data not shown). Median survival was 27.5 days (tumor only), 30.5 days (NC), 34.5 days (PSK), 35 days (MB-6-L), 39 days (MB-6-M) 39 days (MB-6-H) (FIG. 2). Administration of 0.625-2.5 g / kg MB-6 significantly prolonged the life of tumor-bearing mice that received chemotherapy, of which MB-6-M was 30% longer than the NC group Was extended.

ヒト臨床試験 Human clinical trials

4.1 材料および方法 4.1 Materials and methods

4.1.1 適格患者 4.1.1 Eligible patients

この研究は比較群あり、多施設共同、並行群間比較、二重盲検、ランダム化、プラセボ対照臨床研究であり、FOLFOX4(5−FU+LV+オキサリプラチン)との組み合わせで、mCRC患者におけるMB−6の有効性と安全性をプラセボと比較して評価した。この臨床研究は、研究に参加した6病院の治験審査委員会により承認され、すべての患者から書面によるインフォームドコンセントが提供された。   This study is a comparative group, a multicenter, parallel-group comparison, double-blind, randomized, placebo-controlled clinical study, MB-6 in mCRC patients in combination with FOLFOX4 (5-FU + LV + oxaliplatin) Efficacy and safety were evaluated in comparison with placebo. The clinical study was approved by the study review board of the six hospitals participating in the study, and all patients provided written informed consent.

20歳以上で、組織学的にCRC及び(または)転移ガンの臨床所見が確認され、コンピュータ断層撮影(CT)または磁気共鳴映像法(MRI)のいずれかにより1つ以上の測定可能な病変を有し、Eastern Cooperative Oncology 群(ECOG)パフォーマンスステータスが2以下で、適切な骨髄予備能(ヘモグロビン9g/dL以上、絶対好中球数が1.5×109/L以上、血小板が100×109/L以上)、及び肝腎機能(総ビリルビンが1.25×以下、クレアチニンが1.25以下、アラニンアミノトランスフェラーゼ(ALT)またはアスパラギン酸アミノ酸転移酵素(AST)が2.5×正常値上限未満)を備えた患者が適格とされた。   At 20 years of age or older, histologically confirmed clinical findings of CRC and / or metastatic cancer with one or more measurable lesions by either computed tomography (CT) or magnetic resonance imaging (MRI) Has an Eastern Cooperative Oncology Group (ECOG) performance status of 2 or less, suitable bone marrow reserve (hemoglobin 9 g / dL or more, absolute neutrophil count 1.5 x 109 / L or more, platelet 100 x 109 / L L or more), and liver and kidney function (total bilirubin is 1.25 × or less, creatinine is 1.25 or less, alanine aminotransferase (ALT) or aspartate aminotransferase (AST) is 2.5 × less than the upper limit of normal value) Prepared patients were eligible.

妊娠中または授乳中の患者、適切な避妊方法を実施していない患者、中枢神経系転移ガンの所見がある患者、全身性抗微生物治療が必要な活動性感染症を有する患者、現在慢性的下痢及びその他重篤な共存症(例:狭心症、心筋梗塞、鬱血性心不全、てんかん、または治験医師の判断でその他重大な医療条件)がある患者、2つ目の原発性悪性腫瘍の病歴がある患者(適切に治療された皮膚の基底細胞がんまたは子宮頸部上皮内がんを除く)、またはその他抗がん治療を同時に受けている患者、または過去4週間以内に別の新薬で治療を受けた患者は除外された。   Patients who are pregnant or breastfeeding, have not used appropriate contraceptives, have evidence of CNS metastatic cancer, have active infections that require systemic antimicrobial therapy, currently chronic diarrhea And other serious comorbidities (eg, angina pectoris, myocardial infarction, congestive heart failure, epilepsy, or other serious medical condition at the discretion of the investigator), a history of the second primary malignancy One patient (excluding basal cell carcinoma or cervical intraepithelial carcinoma of the skin that has been appropriately treated) or other concurrent anticancer treatment or treatment with another new drug within the last 4 weeks Patients who received were excluded.

4.1.2 治療プロトコル 4.1.2 Treatment protocol

患者はMB−6と併用FOLFOX4(de Gramont A et al.、J Clin Oncol 2000;18:2938〜47;Goldberg RM et al.、J Clin Oncol 2004;22:23〜30)で治療された。MB−6の用量は各320mgの6カプセルが1日3回食事とともに投与された。プラセボ群は不活性成分のみの同じ数のカプセルを摂取した。FOLFOX4化学療法レジメンが、LV(200mg/m)とオキサリプラチン(85mg/m)の2時間の点滴、続いて400mg/mの5−FUボーラス投与の22時間の点滴と1日目600mg/m、及び2時間のLV200mg/m点滴、続いて400mg/m5−FUボーラス投与の22時間の点滴と2日目600mg/mとして行われた。FOLFOX4レジメンは2週間ごとに合計16週間繰り返された。 Patients were treated with MB-6 and combined FOLFOX4 (de Gramont A et al., J Clin Oncol 2000; 18: 2938-47; Goldberg RM et al., J Clin Oncol 2004; 22: 23-30). MB-6 was administered in 6 capsules of 320 mg each 3 times a day with meals. The placebo group took the same number of capsules with only inactive ingredients. A FOLFOX4 chemotherapy regimen was administered with a 2-hour infusion of LV (200 mg / m 2 ) and oxaliplatin (85 mg / m 2 ), followed by a 22-hour infusion of 400 mg / m 2 of 5-FU bolus and 600 mg on day 1. / m 2, and 2 hours of LV200mg / m 2 infusion, were subsequently performed as infusion and day 2 600 mg / m 2 22 hours of 400mg / m 2 5-FU bolus. The FOLFOX4 regimen was repeated every 2 weeks for a total of 16 weeks.

4.1.3 経過観察評価 4.1.3 Follow-up evaluation

ランダム化の前に2週間ごとにバイタルサインと体重が測定された。血清がん胎児性抗原(CEA)値と血液学的及び生化学的パラメータ(赤血球(RBC)数、ヘマトクリット値、白血球(WBC)、血小板数、絶対好中球数、白血球百分率、総ビリルビン、クレアチニン、ALT及びAST)の測定が4週間ごとに実施された。腫瘍評価は8週間ごとに実施された。薬剤コンプライアンスは毎回の検査訪問時に記録された。   Vital signs and body weights were measured every 2 weeks prior to randomization. Serum carcinoembryonic antigen (CEA) value and hematological and biochemical parameters (red blood cell (RBC) count, hematocrit value, white blood cell (WBC), platelet count, absolute neutrophil count, white blood cell percentage, total bilirubin, creatinine , ALT and AST) measurements were taken every 4 weeks. Tumor assessment was performed every 8 weeks. Drug compliance was recorded at each test visit.

4.1.4 主要エンドポイント 4.1.4 Major endpoints

この研究の主要な有効性エンドポイントが、先行研究(Eisenhauer EA et al.、Eur J Cancer 2009;45:228〜47)に従い、MB−6に対する最良総合効果(RECIST 1.1基準による完全奏効(CR)+部分奏効(PR))の算出とプラセボを受けた患者の結果との比較により評価された。   The primary efficacy endpoint of this study is the best overall effect on MB-6 (complete response according to RECIST 1.1 criteria (Eisenhauer EA et al., Eur J Cancer 2009; 45: 228-47)). (CR) + partial response (PR)) and was compared with the results of patients receiving placebo.

4.1.5 二次エンドポイント 4.1.5 Secondary endpoint

無増悪生存率(PFS)がランダム化の日から疾患の増悪が観察された日までの期間として評価された。あらゆる死因による死亡が増悪事象と見なされた。疾患の増悪または生存経過観察が記録されていない患者は最後の客観的腫瘍評価の日に打ち切られた。事後ベースライン腫瘍評価または生存経過観察が行われていない場合、増悪は治験薬の開始日に打ち切られた。   Progression free survival (PFS) was evaluated as the period from the date of randomization to the date on which disease progression was observed. Death from any cause of death was considered an exacerbation event. Patients with no disease progression or survival follow-up were censored on the last objective tumor assessment day. Exacerbations were censored on the start of study drug if no post-baseline tumor assessment or survival follow-up was performed.

治験薬の開始時または開始後に生じた有害事象(AE)は、MedDRAコーディング辞典(Program CTE. Common terminology criteria for adverse events(CTCAE).Version 3.0. National Cancer Institute 2006)により器官別大分類と基本語に従って分類された。AEの期間、重篤度、治験薬との関連性、対応措置、結果が記録された。事象が重篤な有害事象(SAE)として分類されるか否かも記録された。   Adverse events (AEs) that occurred at or after the start of study drug were determined by the MedDRA coding dictionary (Program CTE. Common terminology for advance events (CTCAE). Version 3.0. National Cancer Institute, another classification by Organ University 2006). Classified according to basic terms. The duration of AE, severity, relevance to study drug, response measures, and results were recorded. Whether the event was classified as a serious adverse event (SAE) was also recorded.

4.2 結果 4.2 Results

4.2.1 患者層 4.2.1 Patient demographics

合計72名の転移性結腸直腸がんを患う患者が採用された。被験者は二重盲検法でランダム化され、ランダム置換ブロックサイズを使用してMB−6またはプラセボのいずれかを1:1の割合で受けた。34名の患者がMB−6群に、38名がプラセボ群に、それぞれランダム化された。合計60名の患者(MB−6群29名とプラセボ群31名)が16週間の治験を完了した(83.3%)。全72名の患者がITT(intent−to−treat)人数に含められた。2群間の患者層とベースライン特性に違いはなかった(p>.05)。   A total of 72 patients with metastatic colorectal cancer were recruited. Subjects were randomized in a double-blind manner and received either MB-6 or placebo in a 1: 1 ratio using a random replacement block size. Thirty-four patients were randomized to the MB-6 group and 38 were randomized to the placebo group. A total of 60 patients (29 in the MB-6 group and 31 in the placebo group) completed the 16-week trial (83.3%). A total of 72 patients were included in the ITT (intent-to-treat) number. There was no difference in patient demographics and baseline characteristics between the two groups (p> .05).

4.2.2 主要評価項目 4.2.2 Main evaluation items

最良総合効果はMB−6群で41.2%、プラセボ群で39.5%であり、差は有意でなかった(P=1.000)。RECIST 1.1の基準による最後の測定での腫瘍縮小効果(CR+PR)は、MB−6群が61.8%、プラセボ群が63.2%であった。   The best overall effect was 41.2% in the MB-6 group and 39.5% in the placebo group, and the difference was not significant (P = 1.000). The tumor reduction effect (CR + PR) in the final measurement according to the RECIST 1.1 criteria was 61.8% in the MB-6 group and 63.2% in the placebo group.

4.2.3 副次評価項目 4.2.3 Secondary evaluation items

16週間の治験期間中にMB−6群の患者で疾患の増悪を示した者はいなかったが、一方プラセボ群では6名の患者が増悪を示した。MB−6群の患者はプラセボ群の患者よりも有意に低い疾患増悪率を示した(0.0%対15.8%、P=.026)。MB−6群に死亡者はいなかったが、16週間の治験期間中にプラセボ群では2名が死亡した。さらに、8週間の投薬中止後、本発明の6成分製剤で治療を受けた患者における無増悪生存率は有意に高まった(p=0.0056)。表11に示す。   None of the patients in the MB-6 group showed an exacerbation of disease during the 16-week study period, while 6 patients in the placebo group showed an exacerbation. Patients in the MB-6 group showed a significantly lower disease progression rate than those in the placebo group (0.0% vs 15.8%, P = 0.026). There were no deaths in the MB-6 group, but two died in the placebo group during the 16-week study period. Furthermore, progression-free survival in patients treated with the six-component formulation of the present invention was significantly increased after 8 weeks of discontinuation of medication (p = 0.0056). Table 11 shows.

Figure 2015051970
Figure 2015051970

さらに、MB−6群ではベースラインから有意な体重減少はなかったが、一方でプラセボ群では治験の4週目、6週目、8週目で有意な体重減少が示された(P<.05)。しかし、治験終了時2群間の体重変化の違いは有意ではなかった(P=.114)。加えて、血清CEA値にはFOLFOX4化学療法で8週目に有意な減少があり、減少はMB−6の存在下でも同様であった。   Furthermore, there was no significant weight loss from baseline in the MB-6 group, while the placebo group showed significant weight loss at the 4th, 6th and 8th weeks of the trial (P <. 05). However, the difference in weight change between the two groups at the end of the trial was not significant (P = .114). In addition, there was a significant decrease in serum CEA levels at week 8 with FOLFOX4 chemotherapy, and the decrease was similar in the presence of MB-6.

また、MB−6のコンプライアンスは93.8±11.6%で、プラセボでは94.1±11.4%であった(P=.919)。興味深いことに、MB−6群はプラセボ群と比較してFOLFOX4化学療法に有意に高いコンプライアンスを示した(それぞれ94.4±7.8%対89.0±13.2%、P=.037)。表12に示す。   The compliance of MB-6 was 93.8 ± 11.6%, and that of placebo was 94.1 ± 11.4% (P = .919). Interestingly, the MB-6 group showed significantly higher compliance with FOLOX4 chemotherapy compared to the placebo group (94.4 ± 7.8% vs. 89.0 ± 13.2%, respectively, P = 0.037). ). Table 12 shows.

Figure 2015051970
Figure 2015051970

表12に示すように、本発明の組成物(MB−6)は被験者の化学療法への許容度を高めた(p=0.0370)。   As shown in Table 12, the composition of the present invention (MB-6) increased the subject's tolerance to chemotherapy (p = 0.0370).

4.2.3 安全性と副作用の減少 4.2.3 Reduced safety and side effects

AE(副作用)の重篤度がCTCAE 3.0にしたがい、1=軽度、2=中程度、3=重篤、4=生命を脅かす、5=死亡に分類された。一般に、プラセボ群はMB−6群よりもグレード4以上のAE発生率が有意に高かった(それぞれ28.9%対2.9%、P=.004)。合計19名の患者(MB−6群6名、プラセボ群13名)から28の重篤な有害事象(SAE)が報告された。特に、プラセボ群はMB−6群よりも血清クレアチニン増加の発生率も有意に高く(29%対5.9%、P=.014)、MB−6群が化学療法により引き起こされる腎臓への副作用を減少したことを示している。表13に示す。   According to CTCAE 3.0, the severity of AE (side effects) was classified as 1 = mild, 2 = moderate, 3 = severe, 4 = life threatening, 5 = death. In general, the placebo group had a significantly higher incidence of Grade 4 or higher AEs than the MB-6 group (28.9% vs. 2.9%, respectively, P = .004). A total of 19 patients (6 in the MB-6 group, 13 in the placebo group) reported 28 serious adverse events (SAE). In particular, the placebo group had a significantly higher incidence of increased serum creatinine than the MB-6 group (29% vs. 5.9%, P = .014), and the MB-6 group had side effects on the kidneys caused by chemotherapy Indicates a decrease. Table 13 shows.

Figure 2015051970
Figure 2015051970

さらに、本発明の6成分製剤(MB−6)は化学療法により引き起こされる好中球減少症、特により重篤なものを高いレベルで改善することができる(レベル4以上、p=0.0302)。表14に示す。   Furthermore, the six-component preparation (MB-6) of the present invention can improve neutropenia caused by chemotherapy, particularly severe ones, at a high level (level 4 or higher, p = 0.0302). ). Table 14 shows.

Figure 2015051970
Figure 2015051970

消炎の研究 Study of extinguishing

マクロファージは炎症反応において中核的な役割を果たす。LPSによる活性化後、NF−κB経路とMAPキナーゼ経路を含む下流のシグナル伝達経路の活性化を生じる。これらシグナル伝達経路は、NO、PG、炎症性サイトカインを含むさまざまな炎症性メディエーターの発現を誘発する。したがって、RAW 264.7マウスマクロファージ様細胞は、消炎作用のスクリーニングに優れたモデルを提供する。   Macrophages play a central role in the inflammatory response. After activation by LPS, it results in activation of downstream signaling pathways including the NF-κB pathway and the MAP kinase pathway. These signaling pathways induce the expression of various inflammatory mediators including NO, PG and inflammatory cytokines. Thus, RAW 264.7 mouse macrophage-like cells provide an excellent model for screening anti-inflammatory effects.

5.1 材料および方法 5.1 Materials and methods

5.1.1 細胞培養と試料処理 5.1.1 Cell culture and sample processing

RAW 264.7細胞が10%のFBSを添加したDMEM培地で、加湿された5%CO空気中において37℃で培養された。RAW 264.7細胞は0.1〜1×105細胞/ウェルの密度で96ウェルプレートに播種された。その後細胞は0.0625〜1mg/mlの濃度で試験試料(tested samples)(MB−6)と、または陽性対照(デキサメタゾンまたはセレブレックス)とインキュベーションされた後、LPS 100ng/mlで18時間活性化された。培養期間の終わりに、細胞生存性が比色定量MTTアッセイで判定された。細胞培養上清が保存され、−20℃で亜硝酸塩、PGE2、TNF−α、IL−6レベルの分析用に凍結保存された。 RAW 264.7 cells were cultured in DMEM medium supplemented with 10% FBS at 37 ° C. in humidified 5% CO 2 air. RAW 264.7 cells were seeded in 96-well plates at a density of 0.1-1 × 10 5 cells / well. Cells are then incubated with test samples (MB-6) at a concentration of 0.0625 to 1 mg / ml or with a positive control (dexamethasone or Celebrex) and then activated with 100 ng / ml LPS for 18 hours. It was done. At the end of the culture period, cell viability was determined by a colorimetric MTT assay. Cell culture supernatants were stored and stored frozen at −20 ° C. for analysis of nitrite, PGE2, TNF-α, IL-6 levels.

5.1.2 亜硝酸塩の判定 5.1.2 Determination of nitrite

培地中の亜硝酸塩のレベルがGriess試薬システム(Promega、米国ウィスコンシン州マディソン)を使用して判定され、NOレベルを反映すると推測された。簡潔には、50μlの細胞培養培地が50μLのスルファニルアミド溶液と混合され、室温で10分間インキュベーションされた後、50μLのNED溶液が添加され、さらに10分間室温でインキュベーションされた。マイクロプレートリーダー(BioTek、米国バーモント州ウィヌースキ)を使用して520nmで吸収度が測定された。すべての実験で新鮮な培地がブランクとして使用された。試料中の亜硝酸塩のレベルが標準亜硝酸ナトリウム曲線から読み取られた。   The level of nitrite in the medium was determined using the Griess reagent system (Promega, Madison, Wis., USA) and assumed to reflect NO levels. Briefly, 50 μl of cell culture medium was mixed with 50 μL of sulfanilamide solution and incubated for 10 minutes at room temperature, then 50 μL of NED solution was added and incubated for an additional 10 minutes at room temperature. Absorbance was measured at 520 nm using a microplate reader (BioTek, Winooski, Vermont, USA). Fresh media was used as a blank for all experiments. The level of nitrite in the sample was read from a standard sodium nitrite curve.

5.1.3 PGE2、TNF−α、IL−6アッセイ 5.1.3 PGE2, TNF-α, IL-6 assay

培地中のPGE2、TNF−α、IL−6のレベルがEIAキット(R&D Systems、米国ミネソタ州ミネアポリス)を使用して定量化された。   The levels of PGE2, TNF-α, IL-6 in the medium were quantified using an EIA kit (R & D Systems, Minneapolis, Minn., USA).

5.2 結果 5.2 Results

5.2.1 LPS誘発NO及びPGE2産生と細胞生存に対するMB−6の効果 5.2.1 Effects of MB-6 on LPS-induced NO and PGE2 production and cell survival

RAW 264.7細胞におけるLPS誘発NO及びPGE2産生に対するMB−6の効果を評価するため、培地が採取され、亜硝酸塩とPGE2のレベルが測定された。MB−6は用量依存的にLPS誘発NO産生を有意に阻害した。さらに、MB−6での治療はLPS誘発PGE2産生も減少した。セレブレックス、COX−2阻害薬がPGE2産生の陽性対照として使用された。さらに、RAW 264.7細胞でMTTアッセイを使用してMB−6の細胞傷害効果が評価され、MB−6は使用された濃度(0.0625〜1mg/ml)では細胞生存性に影響しなかった。   To evaluate the effect of MB-6 on LPS-induced NO and PGE2 production in RAW 264.7 cells, media was collected and nitrite and PGE2 levels were measured. MB-6 significantly inhibited LPS-induced NO production in a dose-dependent manner. Furthermore, treatment with MB-6 also reduced LPS-induced PGE2 production. Celebrex, a COX-2 inhibitor, was used as a positive control for PGE2 production. Furthermore, the cytotoxic effect of MB-6 was assessed using the MTT assay in RAW 264.7 cells, and MB-6 did not affect cell viability at the concentration used (0.0625-1 mg / ml). It was.

5.2.2 LPS誘発TNF−α及びIL−6放出に対するMB−6の効果 5.2.2 Effect of MB-6 on LPS-induced TNF-α and IL-6 release

TNF−α及びIL−6放出などのLPS誘発サイトカインに対するMB−6の効果がさらに酵素免疫測定法(ELISA)によって検査された。MB−6での細胞の事前処理がIL−6とTNF−αの産生を有意に減少したことが分かった。結果を表15に示す。   The effects of MB-6 on LPS-induced cytokines such as TNF-α and IL-6 release were further examined by enzyme linked immunosorbent assay (ELISA). It was found that pre-treatment of cells with MB-6 significantly reduced IL-6 and TNF-α production. The results are shown in Table 15.

Figure 2015051970
Figure 2015051970

まとめ Summary

この研究の結果は、MB−6が結腸がんを患う患者において化学療法の有効性を高めることを確認した。臨床前の生体内試験ではMB−6がLV/5−FUで治療を受けた結腸がん保有BALB/cマウスの寿命を延長できることが示された。臨床研究の結果によると、MB−6の投与を受けた患者は16週間の治験期間にわたりプラセボ群と比較してPFSが有意に高く、8週目にMB−6群とプラセボ群で同様の血清CEA値低下が見られたことから、MB−6の添加はFOLFOX4の有効性を損なわないことが示された。さらに、MB−6群では有意に高いFOLFOX4のコンプライアンスが観察され、より高い許容度が示された。また、MB−6群はグレード4以上のAE発生が有意に低かった。   The results of this study confirmed that MB-6 increases the effectiveness of chemotherapy in patients with colon cancer. Preclinical in vivo studies have shown that MB-6 can extend the life span of colon cancer bearing BALB / c mice treated with LV / 5-FU. Results from clinical studies show that patients who received MB-6 had significantly higher PFS compared to the placebo group over the 16-week study period, and similar serum in the MB-6 and placebo groups at 8 weeks. A decrease in CEA value was observed, indicating that the addition of MB-6 does not impair the effectiveness of FOLFOX4. Furthermore, significantly higher FOLFOX4 compliance was observed in the MB-6 group, indicating higher tolerance. In the MB-6 group, the occurrence of grade 4 or higher AE was significantly low.

MB−6群の患者はPFS中央値が有意に高く、疾患の進行がより遅かったため、MB−6がFOLFOX4の治療を受けるmCRC患者の全体的生存品質を向上したことが示唆される。これらのデータは、MB−6が化学療法を受けた腫瘍保有マウスの寿命を30%以上延長したことを示す臨床前試験と一貫している。   Patients in the MB-6 group had significantly higher median PFS and slower disease progression, suggesting that MB-6 improved the overall survival quality of mCRC patients treated with FOLFOX4. These data are consistent with preclinical studies showing that MB-6 extended the life span of tumor-bearing mice receiving chemotherapy by more than 30%.

MB−6群とプラセボ群間の最良総合効果(CR+PR)において有意な差はなかったが、データはFOLFOX4の腫瘍縮小効果が58.5%であることを示した先行研究(Tournigand C et al.、J Clin Oncol 2006;24:394〜400)と一貫している。これは、MB−6がmCRC患者におけるFOLFOX4の有効性を損なわなかったことを示唆している。また、研究終了時、2群間でCEA値と免疫機能においても有意な差はなく、MB−6が化学療法レジメンに悪影響を生じないことが確認されている。つまり、MB−6はこれら転移性結腸直腸がん(mCRC)患者におけるFOLFOX4の細胞傷害効果に干渉しなかった。さらに、本研究のデータではMB−6が消炎活性を提供できることも示されており、これはMB−6で治療したがんの進行がより遅いという結果を支持している。   Although there was no significant difference in the best overall effect (CR + PR) between the MB-6 group and the placebo group, the data showed that a previous study (Tourignand C et al.) Showed that the tumor reduction effect of FOLFOX4 was 58.5%. , J Clin Oncol 2006; 24: 394-400). This suggests that MB-6 did not impair the efficacy of FOLFOX4 in mCRC patients. Also, at the end of the study, there was no significant difference in CEA levels and immune function between the two groups, confirming that MB-6 does not adversely affect the chemotherapy regimen. That is, MB-6 did not interfere with the cytotoxic effect of FOLFOX4 in these patients with metastatic colorectal cancer (mCRC). Furthermore, the data in this study also show that MB-6 can provide anti-inflammatory activity, supporting the results of slower progression of cancer treated with MB-6.

まとめると、臨床前体内研究ではMB−6がLV/5−FUで治療された結腸がん保有Balb/cマウスの寿命を延長できることが示された。臨床研究の結果によると、MB−6の投与を受けた患者は16週間の治験期間中プラセボ群と比較してPFSが有意に高く、かつグレード4以上のAE発生率が有意に低いことが示された。   In summary, preclinical studies have shown that MB-6 can extend the life span of colon cancer bearing Balb / c mice treated with LV / 5-FU. Results of clinical studies show that patients receiving MB-6 have significantly higher PFS and a significantly lower grade 4 or higher AE incidence compared to the placebo group during the 16-week study period It was done.

本発明が属する技術分野において通常の知識を有する者が、更なる例示の必要なく、ここに開示された説明に基づき本発明を最大範囲まで拡大利用することが可能であろう。したがって、本発明の説明、実施例、請求の範囲は本発明の範囲を制限するものではなく例示的なものであると理解されるべきである。   A person having ordinary knowledge in the technical field to which the present invention belongs will be able to expand the present invention to the maximum extent based on the description disclosed herein without the need for further illustration. Accordingly, it is to be understood that the description, examples and claims of the invention are exemplary rather than limiting of the scope of the invention.

なし None

Claims (10)

化学療法による患者のがん治療におけるアジュバント製造のための組成物の使用であって、そのうち、前記組成物が大豆発酵抽出物、緑茶抽出物、スピルリナ、ウコン抽出物、ベニクスノキタケ菌糸体、グレープシード抽出物を含むことを特徴とする、化学療法による患者のがん治療におけるアジュバント製造のための組成物の使用。 Use of a composition for the manufacture of an adjuvant in the treatment of cancer in patients with chemotherapy, wherein said composition is a soy fermentation extract, a green tea extract, a spirulina, a turmeric extract, a venix octopus mycelium, a grape Use of a composition for the manufacture of an adjuvant in the treatment of cancer in patients with chemotherapy, characterized in that it comprises a seed extract. 前記大豆発酵抽出物が、水性大豆抽出物と少なくとも1種の乳酸菌及び選択的に少なくとも1種の酵母の発酵により作られたことを特徴とする、請求項1に記載の組成物の使用。 Use of the composition according to claim 1, characterized in that the fermented soy extract is made by fermentation of an aqueous soy extract and at least one lactic acid bacterium and optionally at least one yeast. 前記大豆発酵抽出物が、(a)水性大豆抽出物を少なくとも1種の乳酸菌と、少なくとも1種の酵母と発酵させて発酵液を形成する工程と、(b)前記発酵液を滅菌する工程と、(c)前記滅菌済みの発酵液をろ過してろ液を取得する工程と、(d)前記ろ液を濃縮して濃縮した大豆発酵抽出物を得る工程と、を含むプロセスにより作成されることを特徴とする、請求項1に記載の組成物の使用。 The fermented soybean extract (a) fermenting the aqueous soy extract with at least one lactic acid bacterium and at least one yeast to form a fermented liquid; and (b) sterilizing the fermented liquid. And (c) a step of obtaining a filtrate by filtering the sterilized fermentation broth, and (d) obtaining a concentrated soybean fermentation extract by concentrating the filtrate. Use of the composition according to claim 1, characterized in that 前記大豆発酵抽出物、前記緑茶抽出物、前記スピルリナ、前記ウコン抽出物、前記ベニクスノキタケ菌糸体、前記グレープシード抽出物が、前記組成物中に12〜30:4:1〜2:1〜2:1〜2:1〜2の重量比で存在することを特徴とする、請求項1に記載の組成物の使用。 The soybean fermented extract, the green tea extract, the spirulina, the turmeric extract, the venix mushroom mycelium, and the grape seed extract are 12-30: 4: 1 to 2: 1 in the composition. Use of the composition according to claim 1, characterized in that it is present in a weight ratio of 2: 1 to 2: 1 to 2. 前記がんが結腸直腸がんであることを特徴とする、請求項1に記載の組成物の使用。 Use of the composition according to claim 1, characterized in that the cancer is colorectal cancer. 前記化学療法が、5−フルオロウラシル、ロイコボリン、オキサリプラチン及びそれらの任意の組み合わせから構成される群より選択された化学療法薬剤を投与して実施されることを特徴とする、請求項1に記載の組成物の使用。 2. The chemotherapy according to claim 1, wherein the chemotherapy is performed by administering a chemotherapeutic agent selected from the group consisting of 5-fluorouracil, leucovorin, oxaliplatin and any combination thereof. Use of the composition. 前記組成物が、大腸ポリープの増殖またはがん化の阻害、化学療法の有効性向上、化学療法へのコンプライアンス向上、または化学療法により引き起こされる副作用の減少に有効な量であることを特徴とする、請求項1に記載の組成物の使用。 The composition is in an amount effective to inhibit the growth or canceration of colorectal polyps, improve the effectiveness of chemotherapy, improve compliance with chemotherapy, or reduce side effects caused by chemotherapy. Use of the composition according to claim 1. 前記組成物が生存率を高める効果を備え、特に、無増悪生存率を高めることを特徴とする、請求項1に記載の組成物の使用。 Use of a composition according to claim 1, characterized in that the composition has the effect of increasing survival, in particular increasing progression-free survival. 抗炎症剤の製造における、請求項1乃至4のいずれかに記載の組成物の使用。 Use of the composition according to any one of claims 1 to 4 in the manufacture of an anti-inflammatory agent. 前記組成物が、プロスタグランジンE2(PGE2)、一酸化窒素(NO)、腫瘍壊死因子α(TNF−α)またはインターロイキン−6(IL−6)の産生を減少するために有効な量で存在することを特徴とする、請求項9に記載の組成物の使用。
In an amount effective to reduce the production of prostaglandin E2 (PGE2), nitric oxide (NO), tumor necrosis factor α (TNF-α) or interleukin-6 (IL-6). Use of the composition according to claim 9, characterized in that it is present.
JP2014180452A 2013-09-06 2014-09-04 Composition for producing adjuvant for cancer patients receiving chemotherapy Pending JP2015051970A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
TW102132310 2013-09-06
TW102132310 2013-09-06

Publications (1)

Publication Number Publication Date
JP2015051970A true JP2015051970A (en) 2015-03-19

Family

ID=52701272

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2014180452A Pending JP2015051970A (en) 2013-09-06 2014-09-04 Composition for producing adjuvant for cancer patients receiving chemotherapy

Country Status (5)

Country Link
JP (1) JP2015051970A (en)
KR (1) KR20150028754A (en)
CN (1) CN104415298A (en)
HK (1) HK1207820A1 (en)
TW (1) TWI673060B (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3254686A1 (en) * 2016-06-06 2017-12-13 Charsire Biotechnology Corp. Soybean seed extract, method for producing the same and uses thereof
US20170360866A1 (en) * 2016-06-15 2017-12-21 Dai-Ming Kuo Pharmaceutical composition for decreasing the side effects of cancer drug, and manufacturing method and uses thereof
JP2017226641A (en) * 2016-06-23 2017-12-28 台灣利得生物科技股▲ふん▼有限公司 Antrodia cinnameme active compositions
JP2019510041A (en) * 2016-03-30 2019-04-11 マイクロバイオ カンパニー, リミテッド Combined cancer therapy of fermented products with immune checkpoint modulator and symbiotic microbiota
US10543243B2 (en) 2016-06-06 2020-01-28 Charsire Biotechnology Corp. Soybeam seed extract, method for producing the same and uses thereof

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3737395A4 (en) 2018-01-09 2021-09-29 Microbio Co., Ltd. Method of activating tumor-infiltrating lymphocytes (tils)
TWI658831B (en) * 2018-02-02 2019-05-11 神農真菌生技有限公司 Uses of Antrodia cinnamomea extract
CN109954053A (en) * 2019-04-23 2019-07-02 辽宁抗风竤生物医药科技有限责任公司 One kind having treatment breast cancer food therapeutic composition and preparation method thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2006022038A (en) * 2004-07-08 2006-01-26 Microbio Co Ltd Method for producing soybean fermentation extract
US20070082073A1 (en) * 2005-05-18 2007-04-12 University Of South Florida Catechin Adjuvants
US20080260695A1 (en) * 2007-04-20 2008-10-23 Microbio Company Ltd. Composition for prevention and/or treatment of cancer
WO2011036904A1 (en) * 2009-09-28 2011-03-31 国立大学法人鳥取大学 Agent for preventing adverse side effects of carcinostatic agent
JP2013529899A (en) * 2010-05-14 2013-07-25 株式会社ジーシーエイチアンドピー Process for producing novel processed carrots or processed carrot extracts with increased ginsenoside ingredients

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI252759B (en) * 2002-07-05 2006-04-11 Microbio Co Ltd Method of using fermented glycine max (L.) extract for enhancing natural killer cell activity
JP2005232032A (en) * 2004-02-17 2005-09-02 Kikkoman Corp Preventive and remedy for septicemia
CN102145159B (en) * 2011-03-31 2012-08-29 青岛大学 Ginger and elecampane composition, preparation method of ginger and elecampane composition and use of ginger and elecampane composition for preparing toxicity-reducing efficacy-improving medicine in cancer radiotherapy and chemotherapy

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2006022038A (en) * 2004-07-08 2006-01-26 Microbio Co Ltd Method for producing soybean fermentation extract
US20070082073A1 (en) * 2005-05-18 2007-04-12 University Of South Florida Catechin Adjuvants
US20080260695A1 (en) * 2007-04-20 2008-10-23 Microbio Company Ltd. Composition for prevention and/or treatment of cancer
WO2011036904A1 (en) * 2009-09-28 2011-03-31 国立大学法人鳥取大学 Agent for preventing adverse side effects of carcinostatic agent
JP2013529899A (en) * 2010-05-14 2013-07-25 株式会社ジーシーエイチアンドピー Process for producing novel processed carrots or processed carrot extracts with increased ginsenoside ingredients

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SUGANUMA, M. ET AL.: "New cancer treatment strategy using combination of green tea catechins and anticancer drugs", CANCER SCI., vol. 102巻, JPN6018020087, 2011, pages pp. 317-323. *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2019510041A (en) * 2016-03-30 2019-04-11 マイクロバイオ カンパニー, リミテッド Combined cancer therapy of fermented products with immune checkpoint modulator and symbiotic microbiota
JP2022058740A (en) * 2016-03-30 2022-04-12 マイクロバイオ カンパニー, リミテッド Combined cancer therapy of immune checkpoint modulator and fermented product by symbiotic microbial flora
JP7442555B2 (en) 2016-03-30 2024-03-04 マイクロバイオ カンパニー, リミテッド Combination cancer therapy of fermentation products with immune checkpoint modulators and commensal microbiota
EP3254686A1 (en) * 2016-06-06 2017-12-13 Charsire Biotechnology Corp. Soybean seed extract, method for producing the same and uses thereof
US10543243B2 (en) 2016-06-06 2020-01-28 Charsire Biotechnology Corp. Soybeam seed extract, method for producing the same and uses thereof
US20170360866A1 (en) * 2016-06-15 2017-12-21 Dai-Ming Kuo Pharmaceutical composition for decreasing the side effects of cancer drug, and manufacturing method and uses thereof
CN107510813A (en) * 2016-06-15 2017-12-26 郭道明 Pharmaceutical composition for reducing side effects of cancer treatment drugs, preparation method and application thereof
US11065293B2 (en) * 2016-06-15 2021-07-20 Dai-Ming Kuo Pharmaceutical composition for decreasing the side effects of cancer drug, and manufacturing method and uses thereof
JP2017226641A (en) * 2016-06-23 2017-12-28 台灣利得生物科技股▲ふん▼有限公司 Antrodia cinnameme active compositions
JP2019147849A (en) * 2016-06-23 2019-09-05 台灣利得生物科技股▲ふん▼有限公司 Production method of antrodia cinnameme activity composition

Also Published As

Publication number Publication date
KR20150028754A (en) 2015-03-16
TWI673060B (en) 2019-10-01
CN104415298A (en) 2015-03-18
HK1207820A1 (en) 2016-02-12
TW201605461A (en) 2016-02-16

Similar Documents

Publication Publication Date Title
TWI673060B (en) Composition for manufacturing adjuvant for cancer patient receiving chemotherapy
JP6051261B2 (en) Bacterial composition for treating cancer
Chen et al. Effectiveness of a novel herbal agent MB-6 as a potential adjunct to 5-fluoracil–based chemotherapy in colorectal cancer
CN109419814B (en) Application of paradisella gordonii to inhibition of fatty liver disease
CN110711182A (en) Surface-modified microorganism, and preparation method and application thereof
JP6209579B2 (en) Pharmaceutical composition that is regarded as a supplementary medicine
CN115414390A (en) Probiotic compound preparation with functions of improving intestinal micro-ecological performance and enhancing treatment effect of tumor immune checkpoint inhibitor and application
US9901602B2 (en) Ejaculum of animals as medicinal material and uses thereof in medicaments for treatment of diseases such as tumors, depression, etc
Kawashima et al. Influence of an elemental diet on 5-fluorouracil-induced morphological changes in the mouse salivary gland and colon
JP2018070568A (en) Nonalcoholic fatty liver disease treating or preventing agent and food for preventing nonalcoholic fatty liver disease
KR20200143668A (en) Use of ginsenoside M1 to prepare a medicament for treating oral cancer
US20130171263A1 (en) Snake Powder Extract For Treatment Of Cancer
JP2006282634A (en) Preventing or treating agent for systemic inflammation response syndrome
TWI310686B (en)
TW201932132A (en) Use of cyanobacterial biomass in treating hepatitis B virus infection
TWI813220B (en) Composition comprising Lactobacillus paracasei and its use for inhibiting nasopharyngeal carcinoma via pyroptosis and cell cycle arrest
JP2019131475A (en) Cancer metastasis inhibitor
CN116549418B (en) Nanoparticle for resisting postoperative infection and recurrence of gastric cancer as well as preparation method and application thereof
TWI635867B (en) Use of herbal composition in preparation drug for inhibiting tumor cell metastasis
Hung Probiotic Complex Feed Additive against LL/2 Lung Cancer in Mice
US20060240117A1 (en) Snake powder extract for treatment of cancer
Miles et al. TLR9 Monotherapy in Immune-Competent Mice Suppresses Orthotopic Prostate Tumor Development
JP6588742B2 (en) Immune function enhancer
TW202409268A (en) Novel lactic acid bacteria and compositions, food and drinks and pharmaceuticals containing lactic acid bacteria
CN114053311A (en) Probiotic composition for increasing effect of chemotherapeutic drug gemcitabine on pancreatic cancer and application thereof

Legal Events

Date Code Title Description
A621 Written request for application examination

Free format text: JAPANESE INTERMEDIATE CODE: A621

Effective date: 20170831

A977 Report on retrieval

Free format text: JAPANESE INTERMEDIATE CODE: A971007

Effective date: 20180521

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20180605

A601 Written request for extension of time

Free format text: JAPANESE INTERMEDIATE CODE: A601

Effective date: 20180904

A601 Written request for extension of time

Free format text: JAPANESE INTERMEDIATE CODE: A601

Effective date: 20181030

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20181205

A02 Decision of refusal

Free format text: JAPANESE INTERMEDIATE CODE: A02

Effective date: 20190108