TW202228772A - Pancreatic cancer quadruplet therapeutic agent combining atezolizumab, tocilizumab, gemcitabine, and nab-paclitaxel - Google Patents

Pancreatic cancer quadruplet therapeutic agent combining atezolizumab, tocilizumab, gemcitabine, and nab-paclitaxel Download PDF

Info

Publication number
TW202228772A
TW202228772A TW110133456A TW110133456A TW202228772A TW 202228772 A TW202228772 A TW 202228772A TW 110133456 A TW110133456 A TW 110133456A TW 110133456 A TW110133456 A TW 110133456A TW 202228772 A TW202228772 A TW 202228772A
Authority
TW
Taiwan
Prior art keywords
antibody
cancer
pancreatic cancer
administration
once
Prior art date
Application number
TW110133456A
Other languages
Chinese (zh)
Inventor
山口京子
寺尾公男
光永修一
哈娜 阿爾葛
瑪莉娜 雷吉納
Original Assignee
日商中外製藥股份有限公司
慕尼黑工業大學
日本國立癌症研究中心
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 日商中外製藥股份有限公司, 慕尼黑工業大學, 日本國立癌症研究中心 filed Critical 日商中外製藥股份有限公司
Publication of TW202228772A publication Critical patent/TW202228772A/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Inorganic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

This disclosure relates to a drug, treatment method, kit, and use, for treating pancreatic cancer in an individual by combining a PD-1/PD-L1 signal inhibitor, an IL-6 inhibitor, and at least one other antitumor agent.

Description

藉由併用阿替利珠單抗、托珠單抗、吉西他濱、白蛋白結合紫杉醇之4劑的胰臟癌治療劑Pancreatic cancer therapeutic agent by concomitant use of 4 doses of atezolizumab, tocilizumab, gemcitabine, and nab-paclitaxel

本發明關於一種特徵在於投予IL-6訊號抑制劑(更具體而言托珠單抗)、吉西他濱、白蛋白結合紫杉醇、及PD-1/PD-L1訊號抑制劑(更具體而言阿替利珠單抗)的用以處置個體之胰臟癌之醫藥、處置方法、套組、用途。更具體而言,本發明關於一種特徵在於投予托珠單抗、吉西他濱、白蛋白結合紫杉醇、及阿替利珠單抗的用以處置個體之胰臟癌之醫藥、處置方法、套組、用途。The present invention relates to a method characterized by the administration of an IL-6 signaling inhibitor (more specifically tocilizumab), gemcitabine, nab-paclitaxel, and a PD-1/PD-L1 signaling inhibitor (more specifically ati Medicines, methods of treatment, kits, uses for treating pancreatic cancer in an individual. More specifically, the present invention relates to a medicament, method of treatment, kit, treatment for pancreatic cancer in an individual characterized by the administration of tocilizumab, gemcitabine, nab-paclitaxel, and atezolizumab. use.

胰臟癌為5年生存率僅7%之難治性癌,日本胰臟癌死亡患者數及罹患者數分別為31,831人、37,233人(2015年),數量幾乎持平,預後不良(非專利文獻7)。胰臟癌存在上皮性腫瘤及非上皮性腫瘤,上皮性腫瘤大體上分成外分泌腫瘤及內分泌腫瘤。外分泌腫瘤分為漿液性囊性腫瘤、黏液性囊性腫瘤、胰臟導管內乳頭狀黏液性腫瘤、浸潤性胰臟導管癌、腺泡細胞腫瘤等類別。根據胰臟癌全國登記調查報告,浸潤性胰臟導管癌約占全部胰臟癌患者之90%,其中管狀腺癌之頻度最高(非專利文獻8)。Pancreatic cancer is a refractory cancer with a 5-year survival rate of only 7%. The number of pancreatic cancer deaths and patients in Japan were 31,831 and 37,233, respectively (2015). ). Epithelial tumors and non-epithelial tumors exist in pancreatic cancer, and epithelial tumors are roughly divided into exocrine tumors and endocrine tumors. Exocrine tumors are classified into serous cystic tumors, mucinous cystic tumors, intraductal papillary mucinous tumors of the pancreas, invasive pancreatic ductal carcinoma, and acinar cell tumors. According to the National Registration Survey Report of Pancreatic Cancer, invasive pancreatic ductal carcinoma accounts for about 90% of all pancreatic cancer patients, and tubular adenocarcinoma has the highest frequency (Non-Patent Document 8).

外科手術切除係唯一可以期待根治胰臟癌之治療法,但一般認為胰臟癌在第一期、第二期時可進行切除。然而,切除率低至10%-20%,且大多數患者於發現時為無法切除之進展期病例,即便能夠成功切除,復發率亦高達80%以上,預後極其不良(非專利文獻9)。Surgical resection is the only treatment that can be expected to cure pancreatic cancer, but it is generally believed that pancreatic cancer can be removed in the first and second stages. However, the resection rate is as low as 10%-20%, and most patients are unresectable advanced cases at the time of discovery. Even if they can be successfully resected, the recurrence rate is as high as 80%, and the prognosis is extremely poor (Non-Patent Document 9).

於胰臟癌診療指南2019年版中,關於針對無法切除之胰臟癌之初次化學療法,根據腫瘤狀況來推薦之方案有所不同(非專利文獻10)。對於遠處轉移性胰臟癌,推薦FOLFIRINOX療法(5-FU・伊立替康・奧沙利鉑・左亞葉酸鹽)或GN療法(鹽酸吉西他濱(GEM)、白蛋白結合紫杉醇),根據各患者之狀態而考慮採用GEM單劑療法、GEM+鹽酸埃羅替尼併用療法、S-1單劑療法、或GEM+S-1併用療法。對於局部進展期胰臟癌,單獨化學療法或化學-放射線療法已在臨床實踐中很普遍,從而期待FOLFIRINOX療法或GN療法之有用性。In the 2019 edition of the guidelines for the diagnosis and treatment of pancreatic cancer, the recommended regimens for primary chemotherapy for unresectable pancreatic cancer vary according to the tumor status (Non-Patent Document 10). For distant metastatic pancreatic cancer, FOLFIRINOX therapy (5-FU, irinotecan, oxaliplatin, levofolinate) or GN therapy (gemcitabine hydrochloride (GEM), nab-paclitaxel) is recommended. Depending on the patient's status, GEM single-dose therapy, GEM+erlotinib hydrochloride combination therapy, S-1 single-agent therapy, or GEM+S-1 combination therapy should be considered. For locally advanced pancreatic cancer, chemotherapy alone or chemo-radiation therapy has become common in clinical practice, and the usefulness of FOLFIRINOX therapy or GN therapy is expected.

二次治療以後之開發亦在推進中,報告有於以具有GEM治療史之轉移性胰臟癌患者作為對象的III期試驗(NAPOLI-1)中,併用伊立替康之奈米脂質體化製劑MM-398與5-FU・亞葉酸之治療法。5-FU與亞葉酸之併用療法中作為主評估項目之總生存期(OS)為4.2個月,相對於此,藉由追加伊立替康之奈米脂質體化製劑而延長至6.1個月[風險比(HR)0.67,95%信賴區間0.49-0.92(p=0.012)],於統計學上有顯著改善(非專利文獻11),日本亦於2020年獲得核准。指南中,於GEM相關方案後提出有包含5-FU・亞葉酸・伊立替康之奈米脂質體化製劑之併用療法的5-FU相關方案,於5-FU相關方案後提出有包含GEM單劑療法或GN療法之GEM相關方案。The development after the second treatment is also in progress, and it was reported in a phase III trial (NAPOLI-1) in patients with metastatic pancreatic cancer with a history of GEM therapy, and the nanoliposome formulation of irinotecan MM was used. -398 and 5-FU・Leucovorin therapy. The overall survival (OS), which was the main evaluation item in the combined therapy of 5-FU and folinic acid, was 4.2 months, while it was extended to 6.1 months by adding the nanoliposome preparation of irinotecan [risk]. ratio (HR) 0.67, 95% confidence interval 0.49-0.92 (p=0.012)], a statistically significant improvement (Non-Patent Document 11), and it was also approved in Japan in 2020. In the guidelines, a 5-FU-related regimen containing a combination therapy of 5-FU, folinic acid, and irinotecan nanoliposome preparations was proposed after the GEM-related regimen, and a single dose of GEM was proposed after the 5-FU-related regimen. GEM-related regimens of therapy or GN therapy.

以胰臟癌為代表之難治性癌的特徵之一在於相較於癌細胞之量,間質之量非常多,且於該間質中伴有大量纖維母細胞增生。作為胰臟癌微環境特徵之一的纖維化被稱為結締組織增生(desmoplasia)。抑制結締組織增生會改善藥劑向腫瘤內之滲透等,而提高現有抗癌劑之效果(非專利文獻13),因此認為有望成為一種新穎之治療概念。另一方面,關於以一種纖維性間質構成成分即玻尿酸(HA)為靶向之胰臟癌之治療方針,於針對轉移性胰臟導管腺癌之玻尿酸融解劑(聚乙二醇化重組人透明質酸酶(PEGPH20:pegylated recombinant human hyaluronidase))與作為標準治療用藥之吉西他濱+白蛋白結合紫杉醇(GEM+nab-PTX)+PEGPH20療法(PAG療法)的隨機化Ⅱ期試驗中顯示出期望結果,還顯示玻尿酸之表現狀態可以為生物標記(HALO 109-202試驗、非專利文獻12)。然而,針對HA高表現之轉移性胰臟導管腺癌之PAG療法之III期試驗(HALO 109-301試驗)的結果與nab-PTX+GEM(AG療法)相比並無改善,若只是利用玻尿酸分解來抑制結締組織增生,則未見增大現行治療效果之作用(非專利文獻3)。One of the characteristics of refractory cancers represented by pancreatic cancer is that the amount of stroma is very large compared to the amount of cancer cells, and a large number of fibroblasts are proliferated in the stroma. Fibrosis, one of the characteristics of the pancreatic cancer microenvironment, is called desmoplasia. Inhibition of connective tissue hyperplasia improves the penetration of drugs into tumors, etc., and enhances the effect of existing anticancer agents (Non-Patent Document 13), so it is expected to be a novel therapeutic concept. On the other hand, regarding the therapeutic policy of pancreatic cancer targeting hyaluronic acid (HA), a fibrous interstitial component, in the hyaluronic acid dissolving agent for metastatic pancreatic ductal adenocarcinoma (PEGylated recombinant human transparent Ronidase (PEGPH20: pegylated recombinant human hyaluronidase) and gemcitabine + nab-paclitaxel (GEM + nab-PTX) + PEGPH20 therapy (PAG therapy) as standard treatment drugs showed the expected results in the randomized phase II trial, and also showed that hyaluronic acid The expression status can be a biomarker (HALO 109-202 test, Non-Patent Document 12). However, the results of the phase III trial (HALO 109-301 trial) of PAG therapy for metastatic pancreatic ductal adenocarcinoma with high HA expression did not improve compared with nab-PTX+GEM (AG therapy). Inhibition of connective tissue hyperplasia does not increase the effect of the current treatment (Non-Patent Document 3).

另一方面,亦報告有不直接以玻尿酸為靶向,而是影響癌細胞之周圍環境以提高化學療法效果的方法。關於白蛋白結合紫杉醇與吉西他濱(GEM)之併用療法之作用機制,揭示有於胰臟癌之小鼠異種移植物模型中,腫瘤微環境會產生變化,而使得GEM向腫瘤內之傳遞增強(專利文獻1)。又,於投予作為糖尿病治療用藥之二甲雙胍之情形時,無論對癌細胞或腫瘤代謝本身之作用如何,均會改變腫瘤微環境,而促進藥劑(抗癌劑或抗纖維化劑)到達標靶,而減輕結締組織增生(專利文獻5)。On the other hand, methods of not directly targeting hyaluronic acid but affecting the surrounding environment of cancer cells to enhance the effect of chemotherapy have also been reported. Regarding the mechanism of action of the combination therapy of nab-paclitaxel and gemcitabine (GEM), it was revealed that in a mouse xenograft model of pancreatic cancer, changes in the tumor microenvironment resulted in enhanced delivery of GEM into the tumor (patent Reference 1). In addition, when metformin is administered as a drug for diabetes treatment, regardless of the effect on cancer cells or tumor metabolism itself, the tumor microenvironment will be changed, and the drug (anti-cancer agent or anti-fibrotic agent) will be promoted to reach the target. , and reduce connective tissue hyperplasia (Patent Document 5).

托珠單抗(Actemra ( 註冊商標 )、TCZ,CAS編號:375823-41-9)為日本國內開發之IgG1亞型之人源化抗人IL-6受體(IL-6R)單株抗體(非專利文獻14)。上述托珠單抗係藉由基因重組技術,將可變區中尤其是與抗原之親和性較高之互補決定區(CDR)設為小鼠型人IL-6R單株抗體,其他部分設為人IgG1,並使用中國倉鼠卵巢(CHO)細胞而創製。托珠單抗係藉由與可溶性IL-6R及膜結合性IL-6R之兩者特異性結合來抑制IL-6之生物學作用而顯示出藥效。於日本國內,作為抗IL-6受體單株抗體之Actemra ( 註冊商標 )點滴靜脈注射用200於2005年4月被核准用於Castleman氏病,於2008年4月被追加核准用於類風濕性關節炎、多關節活動性幼年特發性關節炎、全身型幼年特發性關節炎,於2017年8月被追加核准用於高安氏動脈炎及巨細胞性動脈炎,此後開發工作一直在繼續。 Tocilizumab (Actemra ( registered trademark ) , TCZ, CAS number: 375823-41-9) is a humanized anti-human IL-6 receptor (IL-6R) monoclonal antibody of IgG1 subtype developed in Japan ( Non-patent document 14). The above-mentioned tocilizumab is a mouse-type human IL-6R monoclonal antibody by gene recombination technology, especially the complementarity determining region (CDR) with high affinity to the antigen in the variable region, and the other parts are set as the mouse-type human IL-6R monoclonal antibody. Human IgG1, and created using Chinese Hamster Ovary (CHO) cells. Tocilizumab exhibits efficacy by inhibiting the biological effects of IL-6 by specifically binding to both soluble IL-6R and membrane-bound IL-6R. In Japan, Actemra ( registered trademark ) intravenous injection 200, which is an anti-IL-6 receptor monoclonal antibody, was approved for Castleman's disease in April 2005, and was additionally approved for rheumatoid arthritis in April 2008. Arthritis, polyarticular active juvenile idiopathic arthritis, and systemic juvenile idiopathic arthritis were additionally approved for Takayasu arteritis and giant cell arteritis in August 2017, and development work has been ongoing since then. continue.

另一方面,關於托珠單抗對胰臟癌患者之投予,以15名CRP(C-reactive protein,C反應蛋白)為2.0 mg/dL以上之初次治療前之進展期胰臟癌患者作為對象,實施TCZ+GEM併用療法之多中心、開放標籤、I/II期試驗。關於投予方法,僅計劃投予TCZ 8 mg/kg(Day1、Day15)+GEM 1000 mg/m2(Day1、Day8、Day15),未出現DLT(dose-limiting toxicity,劑量限制性毒性),主要不良事件為血小板減少(三級以上:40%)、嗜中性球減少(三級以上:33%)。CRP為10.6 mg/dL以上之患者群組(8名)之生存期為1.6個月,相對於此,CRP為2.0 mg/dL以上且未達10.6 mg/dL的患者群組為5.8個月。判斷CRP為10.6 mg/dL以上之患者群組難以繼續試驗而中止(非專利文獻15、專利文獻2)。又,關於托珠單抗投予與吉西他濱及白蛋白結合紫杉醇療法之併用,雖揭示有臨床試驗方案,但迄今未報告治療結果(非專利文獻4、5)。On the other hand, regarding the administration of tocilizumab to pancreatic cancer patients, 15 patients with advanced pancreatic cancer before initial treatment with a CRP (C-reactive protein, C-reactive protein) of 2.0 mg/dL or more were used as Subjects, a multicenter, open-label, phase I/II trial of TCZ+GEM combination therapy was performed. Regarding the administration method, only planned administration of TCZ 8 mg/kg (Day1, Day15) + GEM 1000 mg/m2 (Day1, Day8, Day15), no DLT (dose-limiting toxicity, dose-limiting toxicity), major adverse events Thrombocytopenia (grade 3 and above: 40%) and neutropenia (grade 3 and above: 33%). Survival was 1.6 months for the cohort of patients with a CRP of 10.6 mg/dL or higher (8) compared to 5.8 months for the cohort of patients with a CRP of 2.0 mg/dL or higher and less than 10.6 mg/dL. The patient group with a CRP of 10.6 mg/dL or more was judged to be difficult to continue the test and was discontinued (Non-Patent Document 15, Patent Document 2). Furthermore, regarding the combined use of tocilizumab administration with gemcitabine and nab-paclitaxel therapy, although a clinical trial protocol has been disclosed, no treatment results have been reported so far (Non-Patent Documents 4 and 5).

由JAK/STAT訊號傳遞介導之炎症係癌症之化學治療劑抗藥性的原因之一。JAK/STAT3通路因以IL-6家族為代表之複數個細胞激素或生長因子之刺激而活化。於小鼠中,藉由使用抗IL-6R抗體來阻斷IL-6受體,而抑制STAT3活化,使得胰臟導管腺癌之化學療法之效果提昇(非專利文獻2)。Inflammation mediated by JAK/STAT signaling is one of the causes of chemotherapeutic resistance in cancer. The JAK/STAT3 pathway is activated by the stimulation of multiple cytokines or growth factors represented by the IL-6 family. In mice, the use of an anti-IL-6R antibody to block IL-6 receptors inhibits STAT3 activation, resulting in enhanced efficacy of chemotherapy for pancreatic ductal adenocarcinoma (Non-Patent Document 2).

PD-1/PD-L1訊號抑制劑作為免疫檢查點抑制劑被用於治療各類癌症。作為目前可使用者,已知有作為抗PD-1抗體之納武單抗(Opdivo ( 註冊商標 ))及帕博利珠單抗(Keytruda ( 註冊商標 ))、或作為抗PD-L1抗體之度伐魯單抗(Imfinzi ( 註冊商標 ))、阿替利珠單抗(Tecentriq ( 註冊商標 ),CAS編號:1380723-44-3)及阿維魯單抗(Bavencio ( 註冊商標 ))等。 PD-1/PD-L1 signaling inhibitors are used as immune checkpoint inhibitors to treat various cancers. Currently available, nivolumab (Opdivo ( registered trademark ) ) and pembrolizumab (Keytruda ( registered trademark ) ) as anti-PD-1 antibodies, or as anti-PD-L1 antibodies are known Valulumab (Imfinzi ( registered trademark ) ), atezolizumab (Tecentriq ( registered trademark ) , CAS number: 1380723-44-3), and avelumab (Bavencio ( registered trademark ) ) and the like.

阿替利珠單抗為以程序性死亡配體1(PD-L1)為靶向之人源化免疫球蛋白G1(IgG1)單株抗體。認為阿替利珠單抗藉由與PD-L1結合,而抑制PD-L1與PD-1以及PD-L1與B7-1間之相互作用,促進T細胞之再活化,從而產生抗腫瘤效果。於日本國內,在2018年4月以「無法切除之進展期、復發性非小細胞肺癌」作為功效、效果開始售賣,在同年12月追加核准針對「未經化學療法治療之鱗狀細胞癌以外之無法切除之進展期、復發性非小細胞肺癌」之用法、劑量。又,在2019年8月核准用於「進展期小細胞肺癌」。又,在2019年9月被核准擴大適用於乳癌(PD-L1陽性之三陰性乳癌(TNBC)),進而於2020年9月與癌思停(Avastin)之併用療法被追加核准適用於無法切除之肝細胞癌(HCC:hepatocellular carcinoma)。Atezolizumab is a humanized immunoglobulin G1 (IgG1) monoclonal antibody targeting programmed death ligand 1 (PD-L1). It is believed that atezolizumab binds to PD-L1, inhibits the interaction between PD-L1 and PD-1 and between PD-L1 and B7-1, and promotes the reactivation of T cells, thereby producing an anti-tumor effect. In Japan, in April 2018, it was sold with "unresectable advanced, recurrent non-small cell lung cancer" as efficacy and effect, and in December of the same year, it was additionally approved for "squamous cell carcinoma that has not been treated with chemotherapy". Unresectable advanced and recurrent non-small cell lung cancer" usage and dosage. In addition, it was approved for "advanced small cell lung cancer" in August 2019. Also, in September 2019, it was approved for expanded use in breast cancer (PD-L1 positive triple-negative breast cancer (TNBC)), and in September 2020, the combination therapy with Avastin was additionally approved for unresectable treatment. Hepatocellular carcinoma (HCC: hepatocellular carcinoma).

關於用以獲得抗腫瘤效果之PD-1/PD-L1訊號抑制劑與其他藥劑之組合,有多個提案及報告。例如,提出有併用抗PD-L1抗體與吉西他濱(專利文獻3)。藉由對於非小細胞肺癌患者併用抗PD-L1抗體與nab-PTX及卡鉑,而在I期臨床試驗中實現完全緩解(專利文獻4)。於小鼠模型中,提示出藉由併用抗IL-6抗體與抗PD-L1抗體而有可能治療胰臟癌(非專利文獻1)。然而,於進展期胰臟癌I期試驗中,抗PD-1抗體納武單抗+nab-PTX+吉西他濱之併用效果與nab-PTX+吉西他濱為同程度(非專利文獻6)。There are many proposals and reports on the combination of PD-1/PD-L1 signaling inhibitors and other agents to obtain anti-tumor effects. For example, the combined use of an anti-PD-L1 antibody and gemcitabine has been proposed (Patent Document 3). Complete remission was achieved in a phase I clinical trial by using an anti-PD-L1 antibody in combination with nab-PTX and carboplatin in patients with non-small cell lung cancer (Patent Document 4). In a mouse model, it has been suggested that it is possible to treat pancreatic cancer by using an anti-IL-6 antibody in combination with an anti-PD-L1 antibody (Non-Patent Document 1). However, in a phase I trial of advanced pancreatic cancer, the combined effect of the anti-PD-1 antibody nivolumab + nab-PTX + gemcitabine was comparable to that of nab-PTX + gemcitabine (Non-Patent Document 6).

於進展期三陰性乳癌及對含鉑方案無反應之進展期NSCLC之臨床試驗中取得了如下資料,據該資料提示,腫瘤組織內之PD-L1表現上升有助於增強免疫檢查點抑制劑阿替利珠單抗之效果(非專利文獻16、17)。 [先前技術文獻] [專利文獻] The following data were obtained in clinical trials of advanced triple-negative breast cancer and advanced NSCLC that did not respond to platinum-containing regimens. According to the data, the increased expression of PD-L1 in tumor tissue can help enhance immune checkpoint inhibitors. Effects of tislizumab (Non-Patent Documents 16 and 17). [Prior Art Literature] [Patent Literature]

[專利文獻1]日本專利6242213號/WO2011123393/日本專利特表2013-523743/US20120076862 [專利文獻2]日本專利5904552號/WO2011149046/US20130149302 [專利文獻3]日本專利5681638/WO2010077634/日本專利特表2012-511329/US20100203056 [專利文獻4]WO2015095404/日本專利特表2017-501155/US20150210772 [專利文獻5]WO2016140714 [非專利文獻] [Patent Document 1] Japanese Patent No. 6242213/WO2011123393/Japanese Patent Publication No. 2013-523743/US20120076862 [Patent Document 2] Japanese Patent No. 5904552/WO2011149046/US20130149302 [Patent Document 3] Japanese Patent No. 5681638/WO2010077634/Japanese Patent Publication No. 2012-511329/US20100203056 [Patent Document 4] WO2015095404/Japanese Patent Publication No. 2017-501155/US20150210772 [Patent Document 5] WO2016140714 [Non-patent literature]

[非專利文獻1]Thomas A. Mace, et.al, Gut.2018, 67(2), 320-332. [非專利文獻2]Kristen B. Long et.al, Mol Cancer Ther; 2017, 16(9), 1898-1908. [非專利文獻3]ASCO-GI 2020 Abs.638 https://ascopubs.org/doi/abs/10.1200/JCO.2020.38.4_suppl.638 “HALO 109-301: A randomized, double-blind, placebo-controlled, phase 3 study of pegvorhyaluronidase alfa (PEGPH20) + nab-paclitaxel/gemcitabine (AG) in patients (pts) with previously untreated hyaluronan (HA)-high metastatic pancreatic ductal adenocarcinoma (mPDA).” [非專利文獻4]NCT02767557, https://clinicaltrials.gov/ct2/show/NCT02767557 First Posted: May 10, 2016 Herlev Hospital/Celgene “Study of Nab-Paclitaxel and Gemcitabine With or Without Tocilizumab in Pancreatic Cancer Patients (PACTO)” [非專利文獻5]JapicCTI-194753 https://www.clinicaltrials.jp/cti-user/trial/ShowDirect.jsp?japicId=JapicCTI-194753&language=ja 首次登載日:2019/05/13「針對吉西他濱與白蛋白結合紫杉醇無反應之轉移性胰臟癌的托珠單抗與吉西他濱-白蛋白結合紫杉醇併用療法的I期臨床試驗(A Phase I study of tocilizumab plus gemcitabine/nab-paclitaxel in patient with gemcitabine/nab-paclitaxel-refractory metastatic pancreatic cancer)」 [非專利文獻6]Zev A. Wainberg1 et al., Clin Cancer Res. 2020; 26:4814-4822, Published Online First June 18, [PMID: 32554514] https://pubmed.ncbi.nlm.nih.gov/32554514/ [非專利文獻7]國立癌研究中心癌對策資訊中心:全國癌患者監測統計數據(Monitoring of Cancer Incidence in Japan:MCIJ2015) 癌症資訊服務、最新癌症統計 p.304 https://ganjoho.jp/data/reg_stat/statistics/brochure/mcij2015_report.pdf. [非專利文獻8]胰臟學會:胰臟癌全國登記調查報告2007.胰臟 23: 105~123, 2008. [非專利文獻9]Lancet. 2011 Aug 13; 378 (9791): 607-620. [非專利文獻10]胰臟癌診療指南2019年版 http://www.suizou.org/pdf/pancreatic_cancer_cpg-2019.pdf [非專利文獻11]Lancet. 2016 Feb 6; 387 (10018): 545-557. [非專利文獻12]J. Clin. Oncol., 2018 Feb, 1: 36 (4): 359-66. [非專利文獻13]Cancer Cell. 2012 Mar 20; 21 (3): 418-29 [非專利文獻14]Ann Rheum Dis 2000; 59 (suppl I): i21-i27 [非專利文獻15]J Med Diagn Meth 6 (1): 234, 2017. [非專利文獻16]Schmid P, et al. N Engl J Med 2018; 379: 2108-21. [非專利文獻17]Rittmeyer A, et al. Lancet 2017; 389: 255-65 [Non-Patent Document 1] Thomas A. Mace, et.al, Gut. 2018, 67(2), 320-332. [Non-Patent Document 2] Kristen B. Long et.al, Mol Cancer Ther; 2017, 16(9), 1898-1908. [Non-Patent Document 3] ASCO-GI 2020 Abs.638 https://ascopubs.org/doi/abs/10.1200/JCO.2020.38.4_suppl.638 "HALO 109-301: A randomized, double-blind, placebo-controlled, phase 3 study of pegvorhyaluronidase alfa (PEGPH20) + nab-paclitaxel/gemcitabine (AG) in patients (pts) with previously untreated hyaluronan (HA)-high metastatic pancreatic ductal adenocarcinoma (mPDA).” [Non-Patent Document 4] NCT02767557, https://clinicaltrials.gov/ct2/show/NCT02767557 First Posted: May 10, 2016 Herlev Hospital/Celgene “Study of Nab-Paclitaxel and Gemcitabine With or Without Tocilizumab in Pancreatic Cancer Patients (PACTO)” [Non-Patent Document 5] JapicCTI-194753 https://www.clinicaltrials.jp/cti-user/trial/ShowDirect.jsp?japicId=JapicCTI-194753&language=ja Date of first publication: 2019/05/13 "A Phase I study of tocilizumab and gemcitabine-nab-paclitaxel combination therapy for metastatic pancreatic cancer unresponsive to gemcitabine and nab-paclitaxel. tocilizumab plus gemcitabine/nab-paclitaxel in patient with gemcitabine/nab-paclitaxel-refractory metastatic pancreatic cancer)” [Non-Patent Document 6] Zev A. Wainberg1 et al., Clin Cancer Res. 2020; 26:4814-4822, Published Online First June 18, [PMID: 32554514] https://pubmed.ncbi.nlm.nih.gov/32554514/ [Non-Patent Document 7] National Cancer Research Center Cancer Countermeasure Information Center: National Cancer Patient Surveillance Statistics (Monitoring of Cancer Incidence in Japan: MCIJ2015) Cancer Information Service, Latest Cancer Statistics p.304 https://ganjoho.jp/data/reg_stat/statistics/brochure/mcij2015_report.pdf. [Non-Patent Document 8] Pancreatic Society: Pancreatic Cancer National Registration Survey Report 2007. Pancreas 23: 105-123, 2008. [Non-Patent Document 9] Lancet. 2011 Aug 13; 378 (9791): 607-620. [Non-Patent Document 10] Pancreatic Cancer Diagnosis and Treatment Guidelines 2019 Edition http://www.suizou.org/pdf/pancreatic_cancer_cpg-2019.pdf [Non-Patent Literature 11] Lancet. 2016 Feb 6; 387 (10018): 545-557. [Non-Patent Document 12] J. Clin. Oncol., 2018 Feb, 1: 36 (4): 359-66. [Non-Patent Document 13] Cancer Cell. 2012 Mar 20; 21 (3): 418-29 [Non-Patent Document 14] Ann Rheum Dis 2000; 59 (suppl 1): i21-i27 [Non-Patent Document 15] J Med Diagn Meth 6 (1): 234, 2017. [Non-Patent Document 16] Schmid P, et al. N Engl J Med 2018; 379: 2108-21. [Non-Patent Document 17] Rittmeyer A, et al. Lancet 2017; 389: 255-65

[發明所欲解決之問題][Problems to be Solved by Invention]

本發明提供一種用以處置胰臟癌之藥劑之新穎組合。本發明之新穎組合可於如圖12記載之現有療法(包括化學-放射線療法、初次化學療法及二次化學療法)之基礎上追加使用、或代替現有療法來使用。 [解決問題之技術手段] The present invention provides a novel combination of agents for the treatment of pancreatic cancer. The novel combination of the present invention can be used in addition to, or in place of, existing therapies (including chemo-radiotherapy, primary chemotherapy, and secondary chemotherapy) as described in FIG. 12 . [Technical means to solve problems]

本發明具體而言提供一種於使用至少一種化學治療劑之標準療法之基礎上使用IL-6抑制劑及PD-1/PD-L1訊號抑制劑之任一者或兩者的新穎之併用療法。關於可與IL-6抑制劑及/或PD-1/PD-L1訊號抑制劑組合之已知之化學治療劑之代表例,有鹽酸吉西他濱(GEM)單劑、或GEM與白蛋白結合紫杉醇之併用。In particular, the present invention provides a novel concomitant therapy using either or both of an IL-6 inhibitor and a PD-1/PD-L1 signaling inhibitor in addition to standard therapy using at least one chemotherapeutic agent. Representative examples of known chemotherapeutic agents that can be combined with IL-6 inhibitors and/or PD-1/PD-L1 signaling inhibitors include single-dose gemcitabine hydrochloride (GEM), or combined use of GEM and nab-paclitaxel .

本發明者等人經過銳意研究,發現包含化學治療劑、IL-6訊號抑制劑及/或PD-1/PD-L1訊號抑制劑之藥劑之組合會改善目前難以治療之胰臟癌之治療效果,從而完成本發明。例如,本發明者等人發現,藉由對低分子化學治療劑無反應之無法切除・復發性胰臟癌患者併用抗介白素6(IL-6)受體抗體,可以消除對低分子化學治療劑之無反應性,並改善免疫抑制性,從而有效處置胰臟癌。又,本發明者等人發現,藉由利用抗IL-6R抗體阻斷IL-6訊號而抑制結締組織增生。本發明者等人進而發現,藉由該作用,使得低分子藥劑向腫瘤組織內部之吸收增大。又,提示化學治療劑與抗IL-6R抗體之併用不僅誘導低分子藥劑向腫瘤內部之浸潤亢進,還誘導高分子量之抗體製劑或免疫細胞向腫瘤內部之浸潤亢進。又表明,抗IL-6R抗體與抗PD-L1抗體之組合不僅促進T細胞,還促進B細胞之浸潤及活化,而有可能提昇癌症免疫療法之效果。而且表明,藉由併用化學治療劑、抗IL-6R抗體及抗PD-L1抗體,可以發揮單獨或其他組合無法達成之協同抗腫瘤效果。Through intensive research, the inventors of the present invention found that a combination of agents including chemotherapeutic agents, IL-6 signaling inhibitors and/or PD-1/PD-L1 signaling inhibitors can improve the therapeutic effect of currently difficult-to-treat pancreatic cancer , thereby completing the present invention. For example, the inventors of the present invention found that the combination of anti-interleukin-6 (IL-6) receptor antibodies in patients with unresectable or recurrent pancreatic cancer unresponsive to low-molecular-weight chemotherapeutics can eliminate the need for low-molecular-weight chemotherapeutic agents. Anergy to therapeutic agents and improved immunosuppression for effective treatment of pancreatic cancer. Furthermore, the present inventors found that connective tissue hyperplasia is inhibited by blocking IL-6 signaling with an anti-IL-6R antibody. The inventors of the present invention further found that this action increases the absorption of the low molecular weight drug into the tumor tissue. In addition, it was suggested that the combined use of chemotherapeutic agents and anti-IL-6R antibodies induces not only increased infiltration of low-molecular-weight drugs into tumors, but also increased infiltration of high-molecular-weight antibody preparations or immune cells into tumors. It is also shown that the combination of anti-IL-6R antibody and anti-PD-L1 antibody not only promotes the infiltration and activation of T cells, but also promotes the infiltration and activation of B cells, which may improve the effect of cancer immunotherapy. Furthermore, it has been shown that the combined use of chemotherapeutic agents, anti-IL-6R antibodies and anti-PD-L1 antibodies can exert synergistic anti-tumor effects that cannot be achieved alone or in other combinations.

本發明者等人成功取得表明於胰臟癌腫瘤組織中,藉由將標準治療之吉西他濱與抗IL-6R抗體(托珠單抗)及抗PD-L1抗體(阿替利珠單抗)併用投予而使得樹狀細胞(DC)、T細胞、B細胞等CD45陽性免疫細胞向腫瘤內之浸潤亢進的具體證據。本發明者等人並不拘於理論,認為除各種免疫細胞浸潤以外,樹狀細胞會識別特定癌抗原並活化,藉此激活T細胞與B細胞而誘導後天性免疫,從而發揮顯著之抗腫瘤效果。The inventors of the present invention have successfully demonstrated that in pancreatic cancer tumor tissue, by combining standard therapy gemcitabine with anti-IL-6R antibody (tocilizumab) and anti-PD-L1 antibody (atezolizumab) Specific evidence that the infiltration of CD45-positive immune cells, such as dendritic cells (DC), T cells, and B cells, into tumors is enhanced by administration. The inventors of the present invention, without being bound by theory, believe that in addition to the infiltration of various immune cells, dendritic cells recognize and activate specific cancer antigens, thereby activating T cells and B cells to induce acquired immunity, thereby exerting a significant anti-tumor effect .

即,本發明更具體而言提供以下之發明。 (1)一種醫藥組合物,其係含有免疫治療劑(例如CTLA-4抑制劑、PD-1抑制劑、PD-L1抑制劑之類的免疫檢查點抑制劑)作為有效成分者,且與IL-6抑制劑、及至少一種化學治療劑組合使用,以治療胰臟癌。 (1-1)一種醫藥組合物,其係含有IL-6抑制劑作為有效成分者,且與免疫治療劑、及至少一種化學治療劑組合使用,以治療胰臟癌。 (1-2)一種醫藥組合物,其係含有PD-1/PD-L1訊號抑制劑(例如PD-1抑制劑或PD-L1抑制劑)作為有效成分者,且與IL-6抑制劑、及至少一種化學治療劑組合使用,以治療胰臟癌。 (1-3)一種醫藥組合物,其係含有IL-6抑制劑作為有效成分者,且與PD-1/PD-L1訊號抑制劑、及至少一種化學治療劑組合使用,以治療胰臟癌。 That is, the present invention provides the following inventions more specifically. (1) A pharmaceutical composition comprising an immunotherapeutic agent (for example, a CTLA-4 inhibitor, a PD-1 inhibitor, an immune checkpoint inhibitor such as a PD-L1 inhibitor) as an active ingredient, and in combination with IL A -6 inhibitor is used in combination with at least one chemotherapeutic agent to treat pancreatic cancer. (1-1) A pharmaceutical composition containing an IL-6 inhibitor as an active ingredient and used in combination with an immunotherapeutic agent and at least one chemotherapeutic agent to treat pancreatic cancer. (1-2) A pharmaceutical composition comprising a PD-1/PD-L1 signaling inhibitor (such as a PD-1 inhibitor or a PD-L1 inhibitor) as an active ingredient, and an IL-6 inhibitor, and at least one chemotherapeutic agent for the treatment of pancreatic cancer. (1-3) A pharmaceutical composition containing an IL-6 inhibitor as an active ingredient, and used in combination with a PD-1/PD-L1 signaling inhibitor and at least one chemotherapeutic agent, to treat pancreatic cancer .

(2)如上述醫藥組合物,其中上述化學治療劑為嘧啶系、紫杉烷系、鉑系、亞葉酸、喜樹鹼衍生物、分子靶向藥。 (2-1)如上述醫藥組合物,其中上述化學治療劑為嘧啶系、紫杉烷系、鉑系。 (2-2)如上述醫藥組合物,其中上述化學治療劑為選自氟尿嘧啶、替加氟、吉西他濱及卡培他濱中之嘧啶系藥劑、及/或選自由紫杉醇、白蛋白結合紫杉醇及歐洲紫杉醇所組成之群中之紫杉烷系藥劑、及/或選自順鉑及奧沙利鉑中之鉑系藥劑。 (2-3)如上述醫藥組合物,其中上述化學治療劑為吉西他濱或其藥學上容許之鹽。 (2-4)如上述醫藥組合物,其中上述化學治療劑為吉西他濱或其藥學上容許之鹽、及包含白蛋白與紫杉烷之奈米粒子。 (2-5)如上述醫藥組合物,其中上述化學治療劑為鹽酸吉西他濱及白蛋白結合紫杉醇(Abraxane ( 註冊商標 ))。 (2) The aforementioned pharmaceutical composition, wherein the aforementioned chemotherapeutic agent is a pyrimidine-based, taxane-based, platinum-based, folinic acid, camptothecin derivative, or molecular targeted drug. (2-1) The aforementioned pharmaceutical composition, wherein the aforementioned chemotherapeutic agent is a pyrimidine-based, taxane-based, or platinum-based drug. (2-2) The above pharmaceutical composition, wherein the chemotherapeutic agent is a pyrimidine-based agent selected from fluorouracil, tegafur, gemcitabine and capecitabine, and/or selected from paclitaxel, nab-paclitaxel and paclitaxel A taxane-based drug in the group, and/or a platinum-based drug selected from cisplatin and oxaliplatin. (2-3) The above-mentioned pharmaceutical composition, wherein the above-mentioned chemotherapeutic agent is gemcitabine or a pharmaceutically acceptable salt thereof. (2-4) The pharmaceutical composition as described above, wherein the chemotherapeutic agent is gemcitabine or a pharmaceutically acceptable salt thereof, and nanoparticles comprising albumin and taxane. (2-5) The above-mentioned pharmaceutical composition, wherein the above-mentioned chemotherapeutic agents are gemcitabine hydrochloride and nab-paclitaxel (Abraxane ( registered trademark ) ).

(3)如上述醫藥組合物,其中上述IL-6抑制劑為抗IL-6R抗體。 (3-1)如(3)記載之醫藥組合物,其中上述抗IL-6R抗體為托珠單抗、薩特利珠單抗或沙利姆單抗。 (3-2)如(3)記載之醫藥組合物,其中上述抗IL-6R抗體為托珠單抗。 (3-3)如(3)記載之醫藥組合物,其中上述抗IL-6R抗體為薩特利珠單抗。 (3) The aforementioned pharmaceutical composition, wherein the aforementioned IL-6 inhibitor is an anti-IL-6R antibody. (3-1) The pharmaceutical composition according to (3), wherein the anti-IL-6R antibody is tocilizumab, satrilizumab, or salimumab. (3-2) The pharmaceutical composition according to (3), wherein the anti-IL-6R antibody is tocilizumab. (3-3) The pharmaceutical composition according to (3), wherein the anti-IL-6R antibody is satelizumab.

(4)如上述醫藥組合物,其中上述PD-1/PD-L1訊號抑制劑為PD-1拮抗劑或PD-L1拮抗劑。 (4-1)如(4)記載之醫藥組合物,其中上述PD-1/PD-L1訊號抑制劑為PD-1拮抗劑。 (4-2)如(4)記載之醫藥組合物,其中上述PD-1/PD-L1訊號抑制劑為PD-L1拮抗劑。 (4-3)如(4-1)記載之醫藥組合物,其中上述PD-1拮抗劑為抗PD-1抗體。 (4-4)如(4-1)記載之醫藥組合物,其中上述PD-1拮抗劑為AMP-224、BMS-936559、MEDI4736、MSB0010718C、MPDL3280A、納武單抗、AMP-514、帕博利珠單抗(MK-3475)、REGN2810、PDR001、BGB-A317或匹地利珠單抗(CT-011)。 (4) The aforementioned pharmaceutical composition, wherein the aforementioned PD-1/PD-L1 signaling inhibitor is a PD-1 antagonist or a PD-L1 antagonist. (4-1) The pharmaceutical composition according to (4), wherein the PD-1/PD-L1 signaling inhibitor is a PD-1 antagonist. (4-2) The pharmaceutical composition according to (4), wherein the PD-1/PD-L1 signaling inhibitor is a PD-L1 antagonist. (4-3) The pharmaceutical composition according to (4-1), wherein the PD-1 antagonist is an anti-PD-1 antibody. (4-4) The pharmaceutical composition according to (4-1), wherein the PD-1 antagonist is AMP-224, BMS-936559, MEDI4736, MSB0010718C, MPDL3280A, nivolumab, AMP-514, pembrolizumab benzizumab (MK-3475), REGN2810, PDR001, BGB-A317 or pidilizumab (CT-011).

(5)如(4-2)記載之醫藥組合物,其中上述PD-L1拮抗劑為抗PD-L1抗體。 (5-1)如(4-2)記載之醫藥組合物,其中上述PD-L1拮抗劑為度伐魯單抗、阿替利珠單抗、阿維魯單抗、BMS-936559、MSB0010718C、MPDL3280A、或MEDI4736。 (5-2)如(4-2)記載之醫藥組合物,其中上述PD-L1拮抗劑為阿替利珠單抗。 (6)如上述醫藥組合物,其中化學治療劑、IL-6訊號抑制劑及PD-1/PD-L1訊號抑制劑係同時投予、分開投予、或連續投予。 (5) The pharmaceutical composition according to (4-2), wherein the PD-L1 antagonist is an anti-PD-L1 antibody. (5-1) The pharmaceutical composition according to (4-2), wherein the PD-L1 antagonist is durvalumab, atezolizumab, avelumab, BMS-936559, MSB0010718C, MPDL3280A, or MEDI4736. (5-2) The pharmaceutical composition according to (4-2), wherein the PD-L1 antagonist is atezolizumab. (6) The above pharmaceutical composition, wherein the chemotherapeutic agent, the IL-6 signaling inhibitor and the PD-1/PD-L1 signaling inhibitor are administered simultaneously, separately, or continuously.

(7)一種含有阿替利珠單抗之醫藥,其藉由下述方法使用, 該方法包括為了將含有阿替利珠單抗之醫藥與托珠單抗、吉西他濱或其藥學上容許之鹽及白蛋白結合紫杉醇組合來治療人類個體之胰臟癌,而按照如下用法劑量對個體進行投予: (i)托珠單抗之劑量為1次8 mg/kg且間隔4週(Q4W);及阿替利珠單抗之劑量係選自由1次840 mg且間隔2週(Q2W)、1次1200 mg且間隔3週(Q3W)、1次1680 mg且間隔4週(Q4W)所組成之群; (ii)托珠單抗之劑量為1次4 mg/kg且間隔2週(Q2W);及阿替利珠單抗之劑量係選自由1次840 mg且間隔2週(Q2W)、1次1200 mg且間隔3週(Q3W)、1次1680 mg且間隔4週(Q4W)所組成之群; (iii)托珠單抗之劑量為1次8 mg/kg且間隔2週(Q2W);及阿替利珠單抗之劑量係選自由1次840 mg且間隔2週(Q2W)、1次1200 mg且間隔3週(Q3W)、1次1680 mg且間隔4週(Q4W)所組成之群; (iv)托珠單抗之劑量為1次8 mg/kg且間隔4週(Q4W);及阿替利珠單抗之劑量為1次1680 mg且間隔4週(Q4W);及連續3週每週投予一次白蛋白結合紫杉醇與鹽酸吉西他濱,第4週停藥,白蛋白結合紫杉醇之劑量以紫杉醇計1次125 mg/m 2(體表面積)、鹽酸吉西他濱之劑量以吉西他濱計1次1000 mg/m 2(體表面積)。 (7-1)一種含有托珠單抗之醫藥,其藉由下述方法使用: 該方法包括為了將含有托珠單抗之醫藥與阿替利珠單抗、吉西他濱或其藥學上容許之鹽及白蛋白結合紫杉醇組合來治療人類個體之胰臟癌,而按照如下用法劑量對個體進行投予: (i)托珠單抗之劑量為1次8 mg/kg且間隔4週(Q4W);及阿替利珠單抗之劑量係選自由1次840 mg且間隔2週(Q2W)、1次1200 mg且間隔3週(Q3W)、1次1680 mg且間隔4週(Q4W)所組成之群; (ii)托珠單抗之劑量為1次4 mg/kg且間隔4週(Q4W);及阿替利珠單抗之劑量係選自由1次840 mg且間隔2週(Q2W)、1次1200 mg且間隔3週(Q3W)、1次1680 mg且間隔4週(Q4W)所組成之群; (iii)托珠單抗之劑量為1次8 mg/kg且間隔2週(Q2W);及阿替利珠單抗之劑量係選自由1次840 mg且間隔2週(Q2W)、1次1200 mg且間隔3週(Q3W)、1次1680 mg且間隔4週(Q4W)所組成之群; (iv)托珠單抗之劑量為1次8 mg/kg且間隔4週(Q4W);及阿替利珠單抗之劑量為1次1680 mg且間隔4週(Q4W);及連續3週每週投予一次白蛋白結合紫杉醇與鹽酸吉西他濱,第4週停藥,白蛋白結合紫杉醇之劑量以紫杉醇計1次125 mg/m 2(體表面積),鹽酸吉西他濱之劑量以吉西他濱計1次1000 mg/m 2(體表面積)。 (7) A medicine containing atezolizumab, which is used by a method comprising combining the medicine containing atezolizumab with tocilizumab, gemcitabine, or a pharmaceutically acceptable salt thereof and nab-paclitaxel in combination with nab-paclitaxel for the treatment of pancreatic cancer in human subjects, and the subjects are administered at the following dosages: (i) tocilizumab at a dose of 8 mg/kg once at 4-week intervals (Q4W); and atezolizumab were selected from the group consisting of 840 mg once with a 2-week interval (Q2W), 1200 mg once with a 3-week interval (Q3W), and 1680 mg once with a 4-week interval (Q4W). group; (ii) the dose of tocilizumab was 4 mg/kg once with 2-week intervals (Q2W); and the dose of atezolizumab was selected from the group consisting of 840 mg once with 2-week intervals (Q2W), Cohort consisting of 1200 mg once with 3-week interval (Q3W) and 1680 mg once with 4-week interval (Q4W); (iii) Tocilizumab at 8 mg/kg once with 2-week interval (Q2W) ); and the dose of atezolizumab was selected from the group consisting of 840 mg once with a 2-week interval (Q2W), 1200 mg once with a 3-week interval (Q3W), and 1680 mg once with a 4-week interval (Q4W). (iv) Tocilizumab at a dose of 8 mg/kg once every 4 weeks (Q4W); and atezolizumab at a dose of 1680 mg once at a 4-week interval (Q4W); Administer nab-paclitaxel and gemcitabine hydrochloride once a week for 3 consecutive weeks, and discontinue the drug in the 4th week. The dose of nab-paclitaxel is 125 mg/m 2 (body surface area) based on paclitaxel once, and the dose of gemcitabine hydrochloride is calculated as gemcitabine Count 1000 mg/m 2 (body surface area) once. (7-1) A medicine containing tocilizumab, which is used by the following method: The method comprises the steps of combining the medicine containing tocilizumab with atezolizumab, gemcitabine or a pharmaceutically acceptable salt thereof and nab-paclitaxel in combination with nab-paclitaxel for the treatment of pancreatic cancer in human subjects, and the subjects are administered at the following dosages: (i) tocilizumab at a dose of 8 mg/kg once at 4-week intervals (Q4W); and atezolizumab were selected from the group consisting of 840 mg once with a 2-week interval (Q2W), 1200 mg once with a 3-week interval (Q3W), and 1680 mg once with a 4-week interval (Q4W). group; (ii) the dose of tocilizumab was 4 mg/kg once with 4-week intervals (Q4W); and the dose of atezolizumab was selected from the group consisting of 840 mg once with 2-week intervals (Q2W), Cohort consisting of 1200 mg once with 3-week interval (Q3W) and 1680 mg once with 4-week interval (Q4W); (iii) Tocilizumab at 8 mg/kg once with 2-week interval (Q2W) ); and the dose of atezolizumab was selected from the group consisting of 840 mg once with a 2-week interval (Q2W), 1200 mg once with a 3-week interval (Q3W), and 1680 mg once with a 4-week interval (Q4W). (iv) Tocilizumab at a dose of 8 mg/kg once every 4 weeks (Q4W); and atezolizumab at a dose of 1680 mg once at a 4-week interval (Q4W); Administer nab-paclitaxel and gemcitabine hydrochloride once a week for 3 consecutive weeks, and discontinue the drug in the 4th week. The dose of nab-paclitaxel is calculated as paclitaxel at 125 mg/ m2 (body surface area) once, and the dose of gemcitabine hydrochloride is calculated as gemcitabine Count 1000 mg/m 2 (body surface area) once.

(8)一種含有IL-6訊號抑制劑之醫藥,其用於在癌症之處置中與化學治療劑組合投予,以提高該化學治療劑之效果, 同時投予、分開投予、或連續投予化學治療劑與IL-6訊號抑制劑。 (8-1)如(8)記載之醫藥,其中癌症對化學治療劑具有抗藥性。 (8-2)一種用於處置對化學治療劑具有抗藥性之癌症的如(8)記載之醫藥,其中於包含需要進行對化學治療劑具有抗藥性之癌症之處置的個體之群組(患者群體)中,相較於不與IL-6訊號抑制劑組合而投予化學治療劑之情形,提高群組中之緩解率或病情控制率。 (8-3)一種用於處置對化學治療劑具有抗藥性之癌症的與化學治療劑組合而成或與化學治療劑併用投予之含有IL-6訊號抑制劑之醫藥,其中將化學治療劑與IL-6訊號抑制劑同時投予、分開投予、或連續投予, 於包含需要處置對化學治療劑具有抗藥性之癌症的個體之群組中,相較於不與IL-6訊號抑制劑組合而投予化學治療劑之情形,提高群組中之緩解率或病情控制率。 (8-4)一種如上述(8)或(8-1)記載之含有IL-6訊號抑制劑之醫藥,其中 (a)上述包含需要處置對化學治療劑具有抗藥性之癌症的個體之群組包含於化學治療劑投予前未經診斷調查有無抗藥性之個體;或 (b)上述對化學治療劑具有抗藥性之癌症為對化學治療劑具有先天抗藥性之癌症;或 (c)上述對化學治療劑具有抗藥性之癌症為對化學治療劑產生後天抗藥性之癌症;或 (d)上述對化學治療劑具有抗藥性之癌症為投予化學治療劑後復發之癌症。 (8) A medicament containing an IL-6 signaling inhibitor for administration in combination with a chemotherapeutic agent in the treatment of cancer to enhance the effect of the chemotherapeutic agent, The chemotherapeutic agent and the IL-6 signaling inhibitor are administered simultaneously, separately, or sequentially. (8-1) The medicine according to (8), wherein the cancer is resistant to a chemotherapeutic agent. (8-2) A medicine according to (8) for treating a cancer resistant to a chemotherapeutic agent, in a group (patients) comprising an individual requiring treatment of a cancer resistant to a chemotherapeutic agent cohort), the rate of remission or disease control in the cohort was increased compared to the case where the chemotherapeutic agent was administered without combination with an IL-6 signaling inhibitor. (8-3) A medicine containing an IL-6 signaling inhibitor for treating cancer resistant to a chemotherapeutic agent and administered in combination with a chemotherapeutic agent or in combination with a chemotherapeutic agent, wherein the chemotherapeutic agent is administered Simultaneous, separate, or sequential administration with the IL-6 signaling inhibitor, In a cohort comprising individuals in need of treatment of chemotherapeutic-resistant cancers, the rate of remission or disease in the cohort is increased compared to the case where the chemotherapeutic agent is administered without a combination of an IL-6 signaling inhibitor control rate. (8-4) A medicine containing an IL-6 signaling inhibitor as described in (8) or (8-1) above, wherein (a) the aforementioned group comprising individuals in need of treatment of chemotherapeutic-resistant cancers comprises individuals who have not been diagnostically investigated for resistance prior to administration of the chemotherapeutic agent; or (b) the above-mentioned cancers that are resistant to chemotherapeutic agents are cancers that are innately resistant to chemotherapeutic agents; or (c) the above-mentioned cancer that is resistant to a chemotherapeutic agent is a cancer that has acquired resistance to a chemotherapeutic agent; or (d) The above-mentioned cancers that are resistant to chemotherapeutic agents are cancers that recur after administration of the chemotherapeutic agents.

(9)一種含有抗IL-6R抗體與抗PD-L1抗體之醫藥組合物或套組,其用於與化學治療劑組合投予而處置胰臟癌。 (9-1)如(9)記載之醫藥組合物或套組,其中胰臟癌為對化學治療劑具有先天抗藥性、或對化學治療劑產生後天抗藥性之胰臟癌,該醫藥組合物或套組之特徵在於藉由將化學治療劑與抗IL-6R抗體及抗PD-L1抗體組合使用而提高投予群之有效率。 (9-2)一種含有抗IL-6R抗體與抗PD-L1抗體之醫藥組合物或套組,其用於處置需要處置投予化學治療劑後復發之胰臟癌的個體之胰臟癌。 (9) A pharmaceutical composition or kit comprising an anti-IL-6R antibody and an anti-PD-L1 antibody, which is used in combination with a chemotherapeutic agent to treat pancreatic cancer. (9-1) The pharmaceutical composition or set according to (9), wherein the pancreatic cancer is pancreatic cancer that is inherently resistant to a chemotherapeutic agent or has acquired resistance to a chemotherapeutic agent, and the pharmaceutical composition Or the kit is characterized by the use of chemotherapeutic agents in combination with the anti-IL-6R antibody and the anti-PD-L1 antibody to increase the efficiency of the administered population. (9-2) A pharmaceutical composition or kit comprising an anti-IL-6R antibody and an anti-PD-L1 antibody for treating pancreatic cancer in an individual who needs to treat pancreatic cancer that has recurred after administration of a chemotherapeutic agent.

(10)一種含有IL-6抑制劑之組合物,其用於在使用癌症治療劑之個體之癌症之處置中,促進上述癌治療劑向癌組織之滲透。 (10-1)一種用於處置個體之癌的促進上述癌之治療用藥劑向癌組織之滲透之方法,其特徵在於:上述方法包括投予含有IL-6抑制劑之醫藥組合物,上述醫藥組合物係與上述治療用藥劑組合投予。 (10-2)如(10)記載之組合物或如(10-1)記載之方法,其中上述個體為人類。 (10-3)如(10)記載之組合物或如(10-1)記載之方法,其中上述癌症為高度纖維性及/或具有緻密間質之癌症。 (10-4)如(10)記載之組合物或如(10-1)記載之方法,其中上述癌症為胰臟癌或肺癌。 (10-5)如(10)記載之組合物或如(10-1)記載之方法,其中上述癌症之治療用藥劑為分子量400以下之藥劑。 (10-6)如(10)記載之組合物或如(10-1)記載之方法,其中上述癌症之治療用藥劑為分子量400以下之化學治療劑。 (10-7)如(10)記載之組合物或如(10-1)記載之方法,其中上述癌症之治療用藥劑為吉西他濱及其藥學上容許之鹽。 (10-8)如(10)記載之組合物或如(10-1)記載之方法,其中IL-6抑制劑為抗IL-6R抗體、例如托珠單抗。 (10) An IL-6 inhibitor-containing composition for promoting the penetration of the above-mentioned cancer therapeutic agent into cancer tissue in the treatment of cancer in an individual using the cancer therapeutic agent. (10-1) A method for promoting the penetration of the above-mentioned cancer therapeutic agent into cancer tissue for treating cancer in an individual, characterized in that the above-mentioned method comprises administering a pharmaceutical composition containing an IL-6 inhibitor, the above-mentioned pharmaceutical composition The composition is administered in combination with the above-mentioned therapeutic agents. (10-2) The composition according to (10) or the method according to (10-1), wherein the individual is a human. (10-3) The composition according to (10) or the method according to (10-1), wherein the above-mentioned cancer is a cancer with high fibrosis and/or a dense stroma. (10-4) The composition according to (10) or the method according to (10-1), wherein the cancer is pancreatic cancer or lung cancer. (10-5) The composition according to (10) or the method according to (10-1), wherein the drug for the treatment of cancer is a drug with a molecular weight of 400 or less. (10-6) The composition according to (10) or the method according to (10-1), wherein the drug for treating cancer is a chemotherapeutic agent with a molecular weight of 400 or less. (10-7) The composition according to (10) or the method according to (10-1), wherein the drug for the treatment of cancer is gemcitabine and a pharmaceutically acceptable salt thereof. (10-8) The composition according to (10) or the method according to (10-1), wherein the IL-6 inhibitor is an anti-IL-6R antibody such as tocilizumab.

(11)一種含有IL-6抑制劑之組合物,其用於促進個體中CD45陽性細胞向靶組織之浸潤。 (11-1)一種促進個體中CD45陽性細胞向靶組織之浸潤之方法,其中上述方法之特徵在於投予包含IL-6抑制劑之醫藥組合物。 (11-2)如(11)記載之組合物或如(11-1)記載之方法,其中上述個體為人類。 (11-3)如(11)記載之組合物或如(11-1)記載之方法,其中上述靶組織為IL-6過度表現組織。 (11-4)如(11)記載之組合物或如(11-1)記載之方法,其中上述靶組織為膠原蛋白過度產生組織。 (11-5)如(11)記載之組合物或如(11-1)記載之方法,其中上述靶組織為腫瘤組織。 (11-6)如(11-5)記載之組合物或方法,其中上述腫瘤組織為高度纖維性及/或含較密間質之組織。 (11-7)如(11-5)或(11-6)記載之組合物或方法,其中上述腫瘤組織為胰臟癌或肺癌。 (11) A composition comprising an IL-6 inhibitor for use in promoting infiltration of CD45 positive cells into a target tissue in an individual. (11-1) A method of promoting the infiltration of CD45-positive cells into a target tissue in an individual, wherein the above method is characterized by administering a pharmaceutical composition comprising an IL-6 inhibitor. (11-2) The composition according to (11) or the method according to (11-1), wherein the individual is a human. (11-3) The composition according to (11) or the method according to (11-1), wherein the target tissue is an IL-6 overexpressing tissue. (11-4) The composition according to (11) or the method according to (11-1), wherein the target tissue is a collagen overproducing tissue. (11-5) The composition according to (11) or the method according to (11-1), wherein the target tissue is a tumor tissue. (11-6) The composition or method according to (11-5), wherein the tumor tissue is highly fibrous and/or tissue containing relatively dense stroma. (11-7) The composition or method according to (11-5) or (11-6), wherein the tumor tissue is pancreatic cancer or lung cancer.

(12)一種促進免疫細胞(例如CD45陽性免疫細胞)向個體之靶組織之浸潤之方法,其中上述方法之特徵在於將包含IL-6抑制劑之醫藥組合物及PD-1/PD-L1訊號抑制劑組合投予。 (12-1)一種含有IL-6抑制劑之組合物,其與PD-1/PD-L1訊號抑制劑組合投予,以促進個體中T細胞、樹狀細胞及B細胞向靶組織之浸潤。 (12-2)一種含有PD-1/PD-L1訊號抑制劑之組合物,其與IL-6抑制劑組合投予,以促進個體中T細胞、樹狀細胞及B細胞向靶組織之浸潤。 (12-3)如(12)記載之方法、或者如(12-1)或(12-2)記載之組合物,其中上述靶組織為IL-6過度表現組織。 (12-4)如(12)記載之方法、或者如(12-1)或(12-2)記載之組合物,其中上述靶組織為腫瘤組織。 (12-5)如(12-4)記載之方法或組合物,其中上述腫瘤組織為胰臟癌或肺癌。 (12-6)一種PD-1/PD-L1訊號抑制劑與IL-6抑制劑組合而成之醫藥,其用於在癌之治療中,促進免疫細胞(例如CD45陽性免疫細胞)向靶組織之浸潤。 (12-7)如(12-6)記載之醫藥,其中上述免疫細胞為T細胞、樹狀細胞及B細胞。 (12-8)一種PD-1/PD-L1訊號抑制劑與IL-6抑制劑組合而成之醫藥,其用於癌症免疫療法基礎之併用治療。 (12) A method of promoting the infiltration of immune cells (eg, CD45-positive immune cells) into target tissues of an individual, wherein the method is characterized in that a pharmaceutical composition comprising an IL-6 inhibitor and PD-1/PD-L1 signaling are combined Inhibitor combination administration. (12-1) A composition comprising an IL-6 inhibitor administered in combination with a PD-1/PD-L1 signaling inhibitor to promote infiltration of T cells, dendritic cells and B cells into target tissues in an individual . (12-2) A composition comprising a PD-1/PD-L1 signaling inhibitor administered in combination with an IL-6 inhibitor to promote infiltration of T cells, dendritic cells and B cells into target tissues in an individual . (12-3) The method according to (12), or the composition according to (12-1) or (12-2), wherein the target tissue is an IL-6 overexpressing tissue. (12-4) The method according to (12), or the composition according to (12-1) or (12-2), wherein the target tissue is a tumor tissue. (12-5) The method or composition according to (12-4), wherein the tumor tissue is pancreatic cancer or lung cancer. (12-6) A medicine composed of a PD-1/PD-L1 signaling inhibitor and an IL-6 inhibitor, which is used in the treatment of cancer to promote immune cells (such as CD45 positive immune cells) to target tissues of infiltration. (12-7) The medicine according to (12-6), wherein the immune cells are T cells, dendritic cells and B cells. (12-8) A medicine composed of a PD-1/PD-L1 signaling inhibitor and an IL-6 inhibitor, which is used for combined treatment based on cancer immunotherapy.

(13)一種PD-1/PD-L1訊號抑制劑,其用於與至少一種化學治療劑及IL-6訊號抑制劑組合而治療胰臟癌。 (13-1)一種IL-6抑制劑,其用於與至少一種化學治療劑及PD-1/PD-L1訊號抑制劑組合而治療胰臟癌。 (13-2)如(13)記載之PD-1/PD-L1訊號抑制劑或如(13-1)記載之IL-6抑制劑,其中上述化學治療劑為選自氟尿嘧啶、替加氟、吉西他濱及卡培他濱中之嘧啶系藥劑、及/或選自由紫杉醇、白蛋白結合紫杉醇及歐洲紫杉醇所組成之群中之紫杉烷系藥劑、及/或選自順鉑及奧沙利鉑中之鉑系藥劑。 (13-3)如(13)記載之PD-1/PD-L1訊號抑制劑或如(13-1)記載之IL-6抑制劑,其中上述化學治療劑為吉西他濱或其藥學上容許之鹽。 (13-4)如(13)記載之PD-1/PD-L1訊號抑制劑或如(13-1)記載之IL-6抑制劑,其中上述化學治療劑為吉西他濱或其藥學上容許之鹽、及包含白蛋白與紫杉烷之奈米粒子。 (13-5)如上述PD-1/PD-L1訊號抑制劑、或IL-6抑制劑,其中上述IL-6抑制劑為抗IL-6R抗體。 (13-6)如上述PD-1/PD-L1訊號抑制劑、或IL-6抑制劑,其中上述PD-1/PD-L1訊號抑制劑為PD-1拮抗劑(例如抗PD-1抗體)或PD-L1拮抗劑(例如抗PD-L1抗體)。 (13-7)如上述PD-1/PD-L1訊號抑制劑、或IL-6抑制劑,其中化學治療劑、IL-6訊號抑制劑及PD-1/PD-L1訊號抑制劑同時投予、分開投予、或連續投予。 (13) A PD-1/PD-L1 signaling inhibitor for treating pancreatic cancer in combination with at least one chemotherapeutic agent and an IL-6 signaling inhibitor. (13-1) An IL-6 inhibitor for treating pancreatic cancer in combination with at least one chemotherapeutic agent and a PD-1/PD-L1 signaling inhibitor. (13-2) The PD-1/PD-L1 signaling inhibitor according to (13) or the IL-6 inhibitor according to (13-1), wherein the chemotherapeutic agent is selected from fluorouracil, tegafur, A pyrimidine-based agent in gemcitabine and capecitabine, and/or a taxane-based agent selected from the group consisting of paclitaxel, nab-paclitaxel, and paclitaxel, and/or selected from cisplatin and oxaliplatin of platinum-based drugs. (13-3) The PD-1/PD-L1 signaling inhibitor according to (13) or the IL-6 inhibitor according to (13-1), wherein the chemotherapeutic agent is gemcitabine or a pharmaceutically acceptable salt thereof . (13-4) The PD-1/PD-L1 signaling inhibitor according to (13) or the IL-6 inhibitor according to (13-1), wherein the chemotherapeutic agent is gemcitabine or a pharmaceutically acceptable salt thereof , and nanoparticles containing albumin and taxanes. (13-5) The above PD-1/PD-L1 signaling inhibitor or IL-6 inhibitor, wherein the above IL-6 inhibitor is an anti-IL-6R antibody. (13-6) The aforementioned PD-1/PD-L1 signaling inhibitor or IL-6 inhibitor, wherein the aforementioned PD-1/PD-L1 signaling inhibitor is a PD-1 antagonist (eg, an anti-PD-1 antibody) ) or PD-L1 antagonists (eg, anti-PD-L1 antibodies). (13-7) The above PD-1/PD-L1 signaling inhibitor or IL-6 inhibitor, wherein the chemotherapeutic agent, the IL-6 signaling inhibitor and the PD-1/PD-L1 signaling inhibitor are administered simultaneously , separate administration, or continuous administration.

(14)一種PD-1/PD-L1訊號抑制劑之用途,其用於與至少一種化學治療劑及IL-6訊號抑制劑組合而治療或預防胰臟癌、或者製造用於此類治療或預防之醫藥。 (14-1)一種IL-6訊號抑制劑之用途,其用於與至少一種化學治療劑及PD-1/PD-L1訊號抑制劑組合而治療胰臟癌、或者製造用於此類治療或預防之醫藥。 (14-2)如(14)或(14-1)記載之用途,其中上述化學治療劑為選自氟尿嘧啶、替加氟、吉西他濱及卡培他濱中之嘧啶系藥劑、及/或選自由紫杉醇、白蛋白結合紫杉醇及歐洲紫杉醇所組成之群中之紫杉烷系藥劑、及/或選自順鉑及奧沙利鉑中之鉑系藥劑。 (14-3)如(14)或(14-1)記載之用途,其中上述化學治療劑為吉西他濱或其藥學上容許之鹽。 (14-4)如(14)或(14-1)記載之用途,其中上述化學治療劑為吉西他濱或其藥學上容許之鹽、及包含白蛋白與紫杉烷之奈米粒子。 (14-5)如上述用途,其中上述IL-6抑制劑為抗IL-6R抗體。 (14-6)如上述用途,其中上述PD-1/PD-L1訊號抑制劑為PD-1拮抗劑(例如抗PD-1抗體)或PD-L1拮抗劑(例如抗PD-L1抗體)。 (14-7)如上述用途,其中化學治療劑、IL-6訊號抑制劑及PD-1/PD-L1訊號抑制劑係同時投予、分開投予、或連續投予。 (14) Use of a PD-1/PD-L1 signaling inhibitor for the treatment or prevention of pancreatic cancer in combination with at least one chemotherapeutic agent and an IL-6 signaling inhibitor, or for the manufacture of such treatment or Preventive Medicine. (14-1) Use of an IL-6 signaling inhibitor for the treatment of pancreatic cancer in combination with at least one chemotherapeutic agent and a PD-1/PD-L1 signaling inhibitor, or manufacture for such treatment or Preventive Medicine. (14-2) The use according to (14) or (14-1), wherein the chemotherapeutic agent is a pyrimidine-based agent selected from fluorouracil, tegafur, gemcitabine and capecitabine, and/or selected from paclitaxel , a taxane-based drug in the group consisting of albumin-bound paclitaxel and European paclitaxel, and/or a platinum-based drug selected from cisplatin and oxaliplatin. (14-3) The use according to (14) or (14-1), wherein the chemotherapeutic agent is gemcitabine or a pharmaceutically acceptable salt thereof. (14-4) The use according to (14) or (14-1), wherein the chemotherapeutic agent is gemcitabine or a pharmaceutically acceptable salt thereof, and nanoparticles comprising albumin and taxane. (14-5) The above-mentioned use, wherein the above-mentioned IL-6 inhibitor is an anti-IL-6R antibody. (14-6) The use as above, wherein the PD-1/PD-L1 signaling inhibitor is a PD-1 antagonist (eg, anti-PD-1 antibody) or a PD-L1 antagonist (eg, anti-PD-L1 antibody). (14-7) The use as described above, wherein the chemotherapeutic agent, the IL-6 signaling inhibitor and the PD-1/PD-L1 signaling inhibitor are administered simultaneously, separately, or continuously.

(15)一種對確診胰臟癌之人類個體進行治療之方法,其包括按照如下劑量,對個體投予包含托珠單抗、吉西他濱、白蛋白結合紫杉醇及阿替利珠單抗之醫藥: (i)托珠單抗之劑量為1次8 mg/kg且間隔4週(Q4W);及阿替利珠單抗之劑量係選自由1次840 mg且間隔2週(Q2W)、1次1200 mg且間隔3週(Q3W)、1次1680 mg且間隔4週(Q4W)所組成之群; (ii)托珠單抗之劑量為1次4 mg/kg且間隔2週(Q2W);及阿替利珠單抗之劑量係選自由1次840 mg且間隔2週(Q2W)、1次1200 mg且間隔3週(Q3W)、1次1680 mg且間隔4週(Q4W)所組成之群; (iii)托珠單抗之劑量為1次8 mg/kg且間隔2週(Q2W);及阿替利珠單抗之劑量係選自由1次840 mg且間隔2週(Q2W)、1次1200 mg且間隔3週(Q3W)、1次1680 mg且間隔4週(Q4W)所組成之群。 (15-2)一種對確診胰臟癌之人類個體進行治療之方法,其包括 對個體投予托珠單抗、吉西他濱、白蛋白結合紫杉醇及阿替利珠單抗,用法劑量如下: (iv)托珠單抗之劑量為1次8 mg/kg且間隔4週(Q4W);阿替利珠單抗之劑量為1次1680 mg且間隔4週(Q4W);及連續3週每週投予一次白蛋白結合紫杉醇與鹽酸吉西他濱,第4週停藥,白蛋白結合紫杉醇之劑量以紫杉醇計1次125 mg/m 2(體表面積),鹽酸吉西他濱之劑量以吉西他濱計1次1000 mg/m 2(體表面積)。 [發明之效果] (15) A method of treating a human subject diagnosed with pancreatic cancer, comprising administering to the subject a medicament comprising tocilizumab, gemcitabine, nab-paclitaxel, and atezolizumab at the following doses: ( i) The dose of tocilizumab is 8 mg/kg once with a 4-week interval (Q4W); and the dose of atezolizumab is selected from 840 mg once with a 2-week interval (Q2W), 1200 mg once mg at 3-week intervals (Q3W) and 1680 mg once at 4-week intervals (Q4W); (ii) tocilizumab at 4 mg/kg once at 2-week intervals (Q2W); and The dose of atezolizumab was selected from the group consisting of 840 mg once with a 2-week interval (Q2W), 1200 mg once with a 3-week interval (Q3W), and 1680 mg once with a 4-week interval (Q4W) (iii) the dose of tocilizumab is 8 mg/kg once with 2-week intervals (Q2W); and the dose of atezolizumab is selected from the group consisting of 840 mg once with 2-week intervals (Q2W), 1 A group consisting of 1200 mg once with a 3-week interval (Q3W) and 1680 mg once with a 4-week interval (Q4W). (15-2) A method of treating a human subject diagnosed with pancreatic cancer, comprising administering to the subject tocilizumab, gemcitabine, nab-paclitaxel and atezolizumab in the following doses: (iv ) tocilizumab at 8 mg/kg once with 4-week intervals (Q4W); atezolizumab at 1680 mg once with 4-week intervals (Q4W); and weekly for 3 consecutive weeks Administer nab-paclitaxel and gemcitabine hydrochloride once, and stop the drug in the 4th week. The dose of nab-paclitaxel is 125 mg/m 2 (body surface area) calculated on paclitaxel once, and the dose of gemcitabine hydrochloride is 1000 mg/m calculated by gemcitabine once. 2 (body surface area). [Effect of invention]

藉由本發明之醫藥、處置方法、套組、用途,提高現有標準療法之有效性。又,能夠克服現有標準治療對其無效之症例之藥劑抗藥性,再次恢復化學療法之效果。進而,藉由與迄今為止因成效不佳而未經核准之其他治療方案組合使用,能夠擴大療法之選擇範圍。又,可對由於高齡或臟器功能等原因無法實施標準治療法或移植治療而無有效治療方案可選之胰臟癌患者進行處置。The effectiveness of the existing standard therapy can be improved by the medicine, treatment method, kit and use of the present invention. Furthermore, it is possible to overcome the drug resistance of cases in which the existing standard treatment is ineffective, and to restore the effect of chemotherapy again. Furthermore, the choice of therapy can be expanded by combining it with other treatment options that have hitherto been unapproved due to their ineffectiveness. In addition, it is possible to treat pancreatic cancer patients for whom standard therapy or transplantation therapy cannot be performed due to reasons such as advanced age or organ function, and there is no effective treatment option available.

本說明書中使用之一般表達與用語之說明本發明關於一種用以處置個體之胰臟癌之醫藥。又,本發明關於一種個體之胰臟癌之處置方法。又,本發明關於一種用於製造用以處置個體之胰臟癌之醫藥或套組的藥劑之用途。 Description of General Expressions and Phrases Used in the Specification The present invention relates to a medicament for the treatment of pancreatic cancer in an individual. Furthermore, the present invention relates to a method for treating pancreatic cancer in an individual. Furthermore, the present invention relates to the use of a medicament for the manufacture of a medicament or a kit for treating pancreatic cancer in an individual.

「處置」、「進行處置」(treatment、treat或treating)亦稱為「治療」、「進行治療」,意指減少、緩和或減輕某疾病或症狀,亦包括防禦(預防)將來疾病或症狀發作或者抑制疾病進展。期望藉由處置產生之治療效果包括:症狀之緩解、疾病之任意之直接或間接性病理結果之減輕、症狀惡化之進行速度之減緩、疾病狀態之恢復或緩和、預後之改善。「處置」或「治療」不限定於其結果疾病或症狀一定治好或治癒之「處置」或「治療」,亦不限定於獲得期望之治療效果者即有效之「處置」或「治療」。例如所謂癌之處置(或對癌之處置),意指對於患癌、或診斷患癌之對象,為了達成減少癌細胞數、縮小腫瘤尺寸、減緩癌細胞向末梢器官之浸潤速度、或者減緩腫瘤轉移或腫瘤生長速度等至少一個期望之治療效果,而實施給藥、外科手術、放射線療法等。於本發明之一實施形態中,所謂癌之處置係指利用本發明之醫藥、套組、處置方法進行之處置。"Treatment," "treat," or "treating," also known as "treating," "treating," means reducing, alleviating, or alleviating a disease or symptom, and also includes preventing (preventing) future disease or symptom onset or inhibit disease progression. Expected therapeutic effects from treatment include: relief of symptoms, alleviation of any direct or indirect pathological consequences of disease, slowing of progression of symptomatic exacerbations, restoration or alleviation of disease state, and improved prognosis. "Treatment" or "treatment" is not limited to "treatment" or "treatment" that results in a certain cure or cure of the disease or symptom, nor is it limited to "treatment" or "treatment" that is effective if the desired therapeutic effect is obtained. For example, the so-called cancer treatment (or cancer treatment) means that for a subject suffering from cancer or diagnosed with cancer, in order to reduce the number of cancer cells, reduce the size of the tumor, slow the infiltration rate of cancer cells to peripheral organs, or slow down the tumor At least one desired therapeutic effect, such as metastasis or tumor growth rate, is administered, and surgery, radiation therapy, and the like are performed. In one embodiment of the present invention, the so-called cancer treatment refers to treatment using the medicine, set, and treatment method of the present invention.

於本發明之一形態中,胰臟癌為復發或治療抗性胰臟癌。又,於本發明中,胰臟癌可為具有先天抗藥性、或產生後天抗藥性或復發之胰臟癌。又,於本發明中,胰臟癌可為無標準治療且對1個以上之現有化學療法方案不耐受、無反應或復發之胰臟癌。又,胰臟癌可為復發/治療抗性胰臟癌。胰臟癌可為無標準治療且對1個以上之現有化學療法方案不耐受、無反應或復發之胰臟癌。In one aspect of the present invention, the pancreatic cancer is recurrent or treatment-resistant pancreatic cancer. In addition, in the present invention, pancreatic cancer may be a pancreatic cancer with congenital drug resistance, acquired drug resistance, or recurrence. In addition, in the present invention, pancreatic cancer may be pancreatic cancer that has no standard treatment and is intolerant, unresponsive or recurring to one or more existing chemotherapy regimens. Also, the pancreatic cancer may be recurrent/treatment-resistant pancreatic cancer. Pancreatic cancer may be pancreatic cancer that has no standard treatment and is intolerant, unresponsive, or recurring to more than one existing chemotherapy regimen.

用語「抗藥性」、「無反應性」、「抗性」係以相同含義使用,只要為細胞或個體對疾病之處置或治療無應答性(亦稱為敏感性)、且/或產生顯著之應答(例如部分緩解及/或完全緩解)之能力降低之狀態,則無限定。例如,所謂對化學治療劑具有抗藥性之胰臟癌係對使用化學療法之處置完全無應答性、或者無部分緩解或完全緩解等顯著之應答之胰臟癌。即便對於對藥劑具有「抗藥性」或「無反應性」之胰臟癌投予該藥劑,亦不會獲得期望效果,不僅如此,甚至存在進一步進展、轉變成高惡性度胰臟癌之情況。「抗藥性」或「無反應性」可為「先天抗藥性」,亦可為「後天抗藥性」。The terms "drug resistance," "anergy," and "resistance" are used synonymously, so long as a cell or individual is unresponsive (also known as susceptibility) to treatment or treatment of the disease, and/or produces a significant The state of reduced ability to respond (eg, partial remission and/or complete remission) is not limited. For example, a so-called chemotherapeutic-resistant pancreatic cancer is a pancreatic cancer that is completely unresponsive to treatment with chemotherapy, or has no significant response such as partial remission or complete remission. Even if the drug is administered to pancreatic cancer that is "drug-resistant" or "unresponsive" to the drug, the desired effect will not be obtained. Moreover, it may even progress further and turn into a highly malignant pancreatic cancer. "Drug resistance" or "anergy" can be either "innate drug resistance" or "acquired drug resistance".

「先天抗藥性(intrinsic resistance)」一詞係指細胞或個體先天對藥劑無反應性。若細胞或個體對藥劑具有先天抗藥性,則會導致如下狀態:藥劑不發揮效用、或與無抗藥性之情形(即敏感性或應答性之情形)相比難以起效。任何藥物治療之有效率均無法達到100%,必然存在藥劑對其無效之人、無反應者。醫藥品對於哮喘、癌、抑鬱症、糖尿病、消化性潰瘍、高脂血症等之有效率基本上一般約為數十%。The term "intrinsic resistance" refers to the innate unresponsiveness of a cell or individual to an agent. If a cell or individual is innately resistant to an agent, it results in a state in which the agent is ineffective or less effective than would be the case without resistance (ie, sensitivity or responsiveness). The effective rate of any drug treatment cannot reach 100%, and there must be people who are ineffective and unresponsive to the drug. The effectiveness of pharmaceuticals for asthma, cancer, depression, diabetes, peptic ulcer, hyperlipidemia, etc. is generally about tens of percent.

「後天抗藥性(acquired resistance)」一詞係指細胞或個體原本對藥劑具有應答性(或敏感性),但因曝露於該藥劑下而後天獲得之無反應性。後天抗藥性會導致出現如下狀態:最初有效之藥劑於投予過程中失去效用、或與無抗藥性之情形(即應答性之情形)相比難以起效。尤其本發明之醫藥等之後天抗藥性可為於先前處置後產生之抗藥性。例如,抗血管新生治療劑一般用於包括肝癌之多種癌治療,但存在於治療初期有效,若繼續反覆治療則不久就獲得抗藥性之情況。例如,對化學治療劑具有抗藥性之胰臟癌於化學治療劑之存在下已經不會消退、甚至會進一步進展。The term "acquired resistance" refers to a cell or individual that is originally responsive (or sensitive) to an agent, but acquires anergy due to exposure to the agent. Acquired resistance can lead to a state in which an initially effective agent becomes ineffective during administration, or is less effective than a situation without resistance (ie, a responsive situation). In particular, the acquired drug resistance of the medicine of the present invention may be drug resistance developed after previous treatment. For example, anti-angiogenic therapeutic agents are generally used for the treatment of various cancers including liver cancer, but they are effective in the initial stage of treatment, and may acquire resistance soon after repeated treatment is continued. For example, pancreatic cancer that is resistant to chemotherapeutic agents has not regressed or even progressed further in the presence of chemotherapeutic agents.

「復發(recurrence)」一詞係指癌經過治療而暫時治癒或縮小後,再次確認到癌出現或生長。雖然看似治療順利,但藉由手術未完全去除之肉眼無法觀察到之微小癌灶殘存而再次顯現、或藉由藥物療法(抗癌劑治療)或放射線治療暫時縮小之癌再次擴大,引起「復發」。「復發」有時係於與經過治療之癌部相同之位置或其附近出現同一種癌,有時於與經過治療之癌部不同之臟器或器官出現同一種癌。於與經過治療之癌不同之臟器或器官出現同一種癌之情況特指「轉移」。癌細胞自最初產生之部位進入血管或淋巴,再隨著血液或淋巴之流動而轉移至其他臟器或器官,並在此生長。癌大多向淋巴節、肺、肝臟、腦、骨等血液流動豐富之部位發生轉移。因復發出現癌之情形時,患有該癌之個體已接受過藥物療法或放射線治療等先前處置。因此,復發出現之癌可能為對先前處置具有後天抗藥性之癌。The term "recurrence" refers to the re-confirmation of the appearance or growth of cancer after the cancer has been temporarily cured or shrunk by treatment. Although the treatment seems to be successful, the re-emergence of tiny cancerous foci that cannot be seen with the naked eye that was not completely removed by surgery, or the cancer that was temporarily shrunk by drug therapy (anticancer drug treatment) or radiation therapy re-expands, causing " relapse". "Recurrence" sometimes refers to the occurrence of the same type of cancer in the same location as the treated cancerous part or near it, and sometimes the same type of cancer occurs in a different organ or organ from the treated cancerous part. The occurrence of the same type of cancer in a different organ or organ from the treated cancer is specifically referred to as "metastasis". Cancer cells enter blood vessels or lymphatics from the site where they were originally generated, and then move to other organs or organs with the flow of blood or lymphatics, where they grow. Most cancers metastasize to lymph nodes, lungs, liver, brain, bone and other places with rich blood flow. When cancer occurs due to recurrence, the individual with the cancer has received prior treatment such as drug therapy or radiation therapy. Thus, cancers that recur may be cancers that are acquired resistance to previous treatments.

作為用以判別胰臟癌對化學治療劑有無抗藥性之方法,只要為能夠進行判別之方法,則並無限定,例如可於處置前採集之癌組織中檢測具有此種抗藥性之癌所特異性表現之基因、蛋白、組織學特徵性等標記物,以其有無作為基準進行判別。業者可於公知方法中任意選擇用於進行判別之遺傳學或組織學方法。具體之標記物例如可為血中游離(cell-free)DNA或CA19-9(糖鏈抗原(carbohydrate antigen)19-9)、癌組織中之p16或BRCA2基因突變之表現量,或可為癌組織中之表現該等基因或蛋白之細胞數或細胞之比率。又,於採用化學療法之先前處置後未獲得後述藉由對癌之處置期望產生之治療效果、或失去該效果或該效果降低的情形時,亦可判別該癌對化學治療劑具有抗藥性。The method for judging whether pancreatic cancer is resistant to a chemotherapeutic agent is not limited as long as it is a method capable of judging. For example, it is possible to detect the specificity of the cancer having such resistance in the cancer tissue collected before the treatment. The presence or absence of markers of sexual expression, such as genes, proteins, and histological characteristics, is used as a criterion for discrimination. The practitioner can arbitrarily select a genetic or histological method for discrimination among known methods. Specific markers can be, for example, cell-free DNA in blood or the expression of CA19-9 (carbohydrate antigen 19-9), p16 or BRCA2 gene mutation in cancer tissue, or cancer The number or ratio of cells in a tissue expressing the gene or protein. In addition, when the therapeutic effect expected by the treatment of the cancer described later is not obtained, or the effect is lost or reduced after the previous treatment with chemotherapy, the cancer can also be judged to be resistant to the chemotherapeutic agent.

藉由對癌之處置產生之治療效果即有效性可根據本說明書中例示之方法或指標、或者公知之一個或複數個方法或指標進行評估(參照W.A.Weber, J.Nucl.Med.50 : 1S-10S(2009))。例如關於腫瘤生長抑制,若依據美國國立癌研究所(NCI)基準,42%以下之T/C為抗腫瘤活性之最低水平。T/C<10%視為高抗腫瘤活性水平,T/C(%)=接受處置者之腫瘤體積中央值/對照之腫瘤體積中央值×100。The therapeutic effect, that is, the effectiveness, produced by the treatment of cancer can be evaluated according to the methods or indicators exemplified in this specification, or one or a plurality of known methods or indicators (refer to W.A.Weber, J.Nucl.Med.50: 1S -10S (2009)). For example, regarding tumor growth inhibition, according to the National Cancer Institute (NCI) benchmark, T/C below 42% is the lowest level of anti-tumor activity. T/C<10% was regarded as a high level of anti-tumor activity, T/C (%)=median value of tumor volume of treated subjects/median value of tumor volume of control group×100.

於一實施形態中,藉由對癌之處置產生之治療效果即抗癌作用之有效性可根據有關患者、患病器官或患病細胞等之狀態的公知之一個或複數個指標進行評估。作為該指標,例如可列舉「緩解率」,其表示於對患者採用某癌治療法時,實施該治療後,癌細胞縮小或被消滅之患者之比率。又,可列舉「病情控制率(DCR)」,其為作為CR(完全緩解)與PR(部分緩解)之合計的緩解率加上作為腫瘤之大小無變化之狀態的SD(穩定)所得之比率。進而,可列舉:部分緩解(PR)、完全緩解(CR)、總緩解率(ORR)、無進展生存期(PFS)、無病生存期(DFS)、總生存期(OS)、病理學上完全緩解(pCR)、及臨床上完全緩解(cCR)等。In one embodiment, the therapeutic effect produced by the treatment of cancer, that is, the effectiveness of the anti-cancer effect can be evaluated based on one or more known indicators regarding the state of the patient, diseased organ, or diseased cell. As such an index, for example, a "response rate", which indicates the ratio of patients whose cancer cells are shrunk or eliminated after a certain cancer treatment method is administered to a patient, is given. In addition, the "disease control rate (DCR)" is a ratio obtained by adding the remission rate which is the sum of CR (complete remission) and PR (partial remission) to SD (stable) which is a state in which the size of the tumor does not change. . Further, there may be mentioned: partial remission (PR), complete remission (CR), overall remission rate (ORR), progression-free survival (PFS), disease-free survival (DFS), overall survival (OS), pathologically complete remission (pCR), and clinical complete remission (cCR).

ORR係指顯示出癌體積縮小或數量減少並維持最低時限要求之患者之比率,又,ORR可以完全緩解率與部分緩解率之和表示。DCR係指疾病未發生惡化之患者之比率,可以完全緩解率與部分緩解率及疾病穩定率(SD)之和表示。PFS亦稱為「腫瘤進展之前的時間」,表示處置期間及處置之後癌症無進展患者生存之時間長度,包括患者經歷CR或PR之時間、以及經歷SD之時間。DFS係指處置期間及處置之後未復發或無其他疾病患者生存之時間長度。OS係指自治療開始日起對象生存時間。藉由處置延長OS係指例如與無處置之患者群組相比,接受處置之患者群組自試驗開始日起中位生存期之點推測值之延長。上述「群組」係指包含需要進行癌之處置之個體之集合、即「患者群體」。ORR refers to the proportion of patients who show a reduction in the size or number of cancers and maintain a minimum time limit, and ORR can be expressed as the sum of the complete remission rate and the partial remission rate. DCR refers to the proportion of patients whose disease has not deteriorated, and can be expressed as the sum of the complete remission rate, the partial remission rate, and the stable disease rate (SD). PFS, also known as "time to tumor progression," represents the length of time during and after treatment that a cancer progression-free patient survives, including the time a patient experiences CR or PR, and the time to SD. DFS refers to the length of time during and after treatment that a patient survives without recurrence or other disease. OS refers to the time a subject has survived since the start of treatment. Prolongation of OS by treatment refers, for example, to the extension of the point-predicted value of median survival from the start of the trial in the cohort of patients receiving treatment compared to the cohort of patients without treatment. The above-mentioned "group" refers to a collection comprising individuals in need of cancer treatment, that is, a "patient group".

無反應性及/或應答性之評估可使用該領域中之公知方法進行,只要能夠進行所需評估,則無限定。例如,無反應性及/或應答性可藉由活體內或活體外分析於處置(例如使用藥劑處置)下之源自疾病之細胞之生長等而評估。無反應性之獲得、應答性之維持、及/或無反應答者之增加亦可藉由活體內或活體外分析源自疾病之細胞之生長等而評估。於一實施形態中,可藉由IC 50、EC 50之變化、或源自疾病之細胞之生長或減少來表示無反應性。於一實施形態中,該變化可大於約50%、100%及/或200%之任一者。並且,無反應性及/或應答性之變化例如可藉由評估對治療之應答、應答之持續期、及/或無惡化期例如部分緩解及完全緩解而進行活體內評估。無反應性及/或應答性之變化亦可基於個體之群組中之對治療之應答、應答之持續期、及/或無惡化期之變化例如部分緩解及/或完全緩解之數量。 The assessment of anergy and/or responsiveness can be performed using well-known methods in the art and is not limited as long as the desired assessment can be performed. For example, anergy and/or responsiveness can be assessed by in vivo or in vitro analysis of disease-derived cell growth, etc., under treatment (eg, treatment with an agent). The acquisition of anergy, the maintenance of responsiveness, and/or the increase in anergy can also be assessed by in vivo or in vitro analysis of the growth of disease-derived cells, and the like. In one embodiment, anergy may be indicated by a change in IC50 , EC50 , or growth or decrease in cells derived from disease. In one embodiment, the variation can be greater than any of about 50%, 100%, and/or 200%. Also, changes in anergy and/or responsiveness can be assessed in vivo, eg, by assessing response to treatment, duration of response, and/or periods of no exacerbation, eg, partial and complete remission. Changes in anergy and/or responsiveness can also be based on changes in response to treatment, duration of response, and/or exacerbation-free periods, such as the number of partial responses and/or complete responses, in a group of individuals.

於一實施形態中,對於藉由本發明之醫藥、套組、處置方法進行之處置的應答係使用實體癌療效判定基準(RECIST)1.1應答基準所評估之PR、CR、PFS、DFS、ORR、DCR或OS之任一者。In one embodiment, the response to treatment by the medicine, set, and treatment method of the present invention is PR, CR, PFS, DFS, ORR, DCR assessed using the Response Criteria for Solid Cancer (RECIST) 1.1 Response Criteria or any of the OS.

於一實施形態中,可使用特定之效果判定標記物評估對癌之處置之有效性。投予含有包含PD-1/PD-L1訊號抑制劑及白蛋白之奈米粒子之組合物後之個體之血清CA19-9水平與處置前之血清CA19-9水平相比至少降低約50%(至少約60%、70%、80%、90%、95%中之任一者等)。In one embodiment, specific efficacy-determining markers can be used to assess the efficacy of treatment of cancer. Serum CA19-9 levels in subjects following administration of a composition comprising nanoparticles comprising a PD-1/PD-L1 signaling inhibitor and albumin are reduced by at least about 50% compared to pre-treatment serum CA19-9 levels ( at least about any of 60%, 70%, 80%, 90%, 95%, etc.).

本發明之所有實施形態均並非對於全部對象達成期望之治療效果有效,應理解為對於藉由學生T檢驗、卡方檢驗、曼-惠特尼U檢驗、KW(Kruskal-Wallis)檢驗(H檢驗)、JT(Jonckheere-Terpstra)檢驗及Wilcoxon檢驗等本技術領域公知之任意之統計檢驗而確定之統計學上有意義數量之對象,本發明之實施形態之任一實施形態達成期望之治療效果。All embodiments of the present invention are not effective for all subjects to achieve the desired therapeutic effect, and should be understood to be effective for achieving the desired therapeutic effect by Student's T test, chi-square test, Mann-Whitney U test, KW (Kruskal-Wallis) test (H test) ), JT (Jonckheere-Terpstra) test and Wilcoxon test, etc. any statistical test known in the art to determine the statistically significant number of objects, any one of the embodiments of the present invention achieves the desired therapeutic effect.

只要基於用以評估癌之治療效果之本說明書中之例示指標、或公知指標中之至少1個指標而確認有效性,則可判斷該治療有效、或具有抗癌作用。該有效性較佳為關於患者之生存期之延長、或疼痛之減輕等全身症狀之改善,於獲得癌縮小或暫時消失、或者血清CA19-9減少等治療效果之情形時,亦可判斷該治療有效。又,與比較對象相比「更有效」、「有效性較高」、「治療效果較高」或「治療效果提昇」等意指就此處例示指標、或公知指標中之至少1個指標進行比較更有效。As long as the efficacy is confirmed based on at least one of the indicators exemplified in the present specification or known indicators for evaluating the therapeutic effect of cancer, the treatment can be judged to be effective or to have an anticancer effect. The effectiveness is preferably related to the prolongation of the patient's survival period, or the improvement of systemic symptoms such as pain relief, and the treatment can also be judged when the treatment effect such as the reduction or temporary disappearance of cancer, or the reduction of serum CA19-9 is obtained. efficient. Furthermore, "more effective", "higher effectiveness", "higher therapeutic effect" or "improved therapeutic effect" compared to the comparison object means to compare with at least one of the indexes exemplified here or known indexes More effective.

修飾以數值形式定義之參數(例如藥劑之劑量、或採用本說明書中記載之併用療法之處置之時長)時使用的「約」意指參數有時在關於該參數所記述數值之上下10%之範圍內變動。例如「約5 mg」意指「4.5 mg至5.5 mg」。"About" used when modifying a numerically defined parameter (such as the dose of an agent, or the length of treatment with a concomitant therapy described in this specification) means that the parameter is sometimes 10% above or below the value stated for the parameter change within the range. For example "about 5 mg" means "4.5 mg to 5.5 mg".

本發明之組合物或藥劑可以醫藥品之形態投予,可經口或非經口地於全身或局部投予。例如可選擇點滴等靜脈內注射、肌肉內注射、腹腔內注射、皮下注射、栓劑、注腸、經口性腸溶劑等,可根據患者之年齡、症狀而選擇適宜之投予方法。 本發明之組合物或藥劑之投予對象為哺乳動物。哺乳動物較佳為人類。 The composition or medicament of the present invention can be administered in the form of pharmaceuticals, orally or parenterally, systemically or locally. For example, intravenous injection such as drip, intramuscular injection, intraperitoneal injection, subcutaneous injection, suppository, enteral injection, oral enteric solvent, etc. can be selected, and an appropriate administration method can be selected according to the age and symptoms of the patient. The subject to which the composition or medicament of the present invention is administered is a mammal. The mammal is preferably a human.

根據通常進行之方法,可將製劑製成錠劑、顆粒劑、散劑、膠囊劑、乳劑、懸浮劑、糖漿劑、或無菌性溶液、懸浮液劑等注射劑。於將該等有效成分分別製劑化而製成2種以上之製劑之情形時,可同時投予、或者隔開一定時間間隔而分開或連續投予各個製劑。亦可於1天中分別以不同之次數投予該2種以上之製劑。本發明之醫藥可於全身或局部經口投予或非經口投予。於將該等有效成分分別製劑化而製成2種以上之製劑之情形時,亦可以不同路徑投予各個製劑。According to the usual method, the preparation can be prepared into lozenges, granules, powders, capsules, emulsions, suspensions, syrups, or injections such as sterile solutions and suspensions. When these active ingredients are separately formulated to prepare two or more kinds of preparations, the respective preparations may be administered simultaneously, or may be administered separately or continuously at a predetermined time interval. The two or more preparations may be administered at different times in one day. The medicament of the present invention can be administered systemically or locally orally or parenterally. When these active ingredients are separately formulated to prepare two or more formulations, each formulation may be administered by different routes.

於一實施形態中,所謂「同時投予、分開投予或連續投予」係為了達成所期望之併用治療效果,對本發明之化合物或其藥學上容許之鹽、及其他活性成分之量以及投予之相對時間點加以選擇而投予。本發明之化合物可經由各種路徑投予,例如非經口、皮下、靜脈內、關節內、蛛網膜下腔內、肌肉內、腹腔內、局部、及皮內注射,以及經皮、直腸、頰部、口腔黏膜、經鼻、經眼,以及吸入及植入式投予。於一實施形態中,最先投予哪一者均可。於同時投予之情形時,活性成分可包含於同一或不同之醫藥組合物中被投予。輔助療法,即,使用1個有效成分作為一次治療,使用其他有效成分來支援該一次治療之情形亦為本發明之實施形態。In one embodiment, the so-called "simultaneous administration, separate administration or continuous administration" refers to the amount and administration of the compound of the present invention or a pharmaceutically acceptable salt thereof, and other active ingredients in order to achieve the desired combined therapeutic effect. The relative time point of giving is selected and administered. The compounds of the present invention can be administered by various routes, such as parenteral, subcutaneous, intravenous, intraarticular, subarachnoid, intramuscular, intraperitoneal, topical, and intradermal injection, as well as transdermal, rectal, buccal Topical, oral mucosa, nasal, ocular, as well as inhalation and implantable administration. In one embodiment, whichever is administered first may be administered. In the case of simultaneous administration, the active ingredients may be administered in the same or different pharmaceutical compositions. Adjuvant therapy, that is, using one active ingredient as a treatment, and using other active ingredients to support the one treatment is also an embodiment of the present invention.

本發明關於一種用於經改善之醫藥用途之2種以上之藥劑之組合。各藥劑可一次投予或複數次分開投予、或者持續注入投予。2種以上之藥劑可同時、分開、或連續投予。又,2種以上之藥劑可按照特定之劑量及特定之間隔進行投予。於將本發明之醫藥製成不同之複數個製劑、且同時或空開特定間隔進行投予之可能性較高之情形時,可於例如市售之醫藥之隨附說明書或銷售小冊子等文書中記載併用各者之要點。或者亦可製成含有包含各者之製劑之套組或多部件套組(Kit-of-Parts)。套組可含有分別用以保持2種以上之不同之醫藥組合物(其至少1種含有本發明之藥劑作為有效成分)之手段例如容器、分開之小瓶或分開之小包、小袋等。套組尤其適於投予不同之劑形例如經口劑形及非經口劑形、以不同之投予間隔投予各個組合物、或者相對地設定各個組合物之劑量。為了提高適應性,通常可一併提供套組之投予指示。The present invention relates to a combination of two or more medicaments for improved medical use. Each agent can be administered at one time, divided into multiple administrations, or administered by continuous infusion. Two or more drugs may be administered simultaneously, separately, or consecutively. In addition, two or more kinds of drugs can be administered at a specific dose and at a specific interval. In the case where the medicine of the present invention is prepared into a plurality of different preparations and is highly likely to be administered at the same time or at specific intervals, it can be included in documents such as the accompanying instructions or sales brochures of the commercially available medicines. Record and use the main points of each. Alternatively, kits or Kit-of-Parts containing formulations containing each can be made. The kit may contain means such as containers, separate vials or separate packets, pouches, etc., for respectively holding two or more different pharmaceutical compositions (at least one of which contains the agent of the present invention as an active ingredient). The kits are particularly suitable for administering different dosage forms such as oral and parenteral dosage forms, administering each composition at different administration intervals, or setting the doses of each composition relatively. In order to improve adaptability, a set of dosing instructions are usually provided together.

製劑係藉由將所需有效成分與任意選擇之藥理學上容許之載體、賦形劑或穩定劑加以混合製成冷凍乾燥製劑或水溶液之形態進行保存以備用。 作為製劑上容許之材料,例如可列舉:滅菌水或生理鹽水、穩定劑、賦形劑、緩衝劑、防腐劑、界面活性劑、螯合劑(EDTA等)、結合劑等。 The preparations are prepared by mixing the desired active ingredients with any selected pharmacologically acceptable carriers, excipients or stabilizers to prepare freeze-dried preparations or aqueous solutions for future use. Examples of materials acceptable for formulation include sterile water or physiological saline, stabilizers, excipients, buffers, preservatives, surfactants, chelating agents (EDTA, etc.), binding agents, and the like.

作為界面活性劑,可列舉非離子界面活性劑,作為典型示例,例如可列舉:山梨醇酐單辛酸酯、山梨醇酐單月桂酸酯、山梨醇酐單棕櫚酸酯等山梨醇酐脂肪酸酯;甘油單辛酸酯、甘油單肉豆蔻酸酯、甘油單硬脂酸酯等甘油脂肪酸酯等具有HLB6~18者等。Examples of the surfactant include nonionic surfactants, and typical examples include sorbitan fatty acids such as sorbitan monocaprylate, sorbitan monolaurate, and sorbitan monopalmitate. Esters; glycerol fatty acid esters such as glycerol monocaprylate, glycerol monomyristate, and glycerol monostearate, etc. having HLB 6 to 18, and the like.

又,作為界面活性劑,可列舉陰離子界面活性劑,作為典型示例,例如可列舉:乙醯基硫酸鈉、月桂基硫酸鈉、油基硫酸鈉等具有碳原子數10~18之烷基之烷基硫酸鹽;聚氧乙烯月桂基硫酸鈉等環氧乙烷之平均加成莫耳數為2~4且烷基之碳原子數為10~18之聚氧乙烯烷基醚硫酸鹽;月桂基磺基琥珀酸酯鈉等烷基之碳原子數為8~18之烷基磺基琥珀酸酯鹽;天然系之界面活性劑,例如卵磷脂、甘油磷脂;神經鞘磷脂等鞘磷脂;碳原子數12~18之脂肪酸之蔗糖脂肪酸酯等。In addition, as the surfactant, an anionic surfactant can be mentioned, and typical examples include alkanes having an alkyl group having 10 to 18 carbon atoms, such as sodium acetyl sulfate, sodium lauryl sulfate, and sodium oleyl sulfate. Alkyl sulfates; polyoxyethylene alkyl ether sulfates, such as polyoxyethylene sodium lauryl sulfate, with an average added molar number of 2 to 4 ethylene oxide and an alkyl group of 10 to 18 carbon atoms; lauryl Sodium sulfosuccinate and other alkyl sulfosuccinates whose alkyl group has 8 to 18 carbon atoms; natural surfactants, such as lecithin, glycerophospholipid; sphingomyelin such as sphingomyelin; carbon atoms Sucrose fatty acid esters of fatty acids numbered 12 to 18, etc.

作為緩衝劑,可列舉:磷酸、檸檬酸緩衝液、乙酸、蘋果酸、酒石酸、琥珀酸、乳酸、磷酸鉀、葡萄糖酸、辛酸、去氧膽酸、水楊酸、三乙醇胺、反丁烯二酸等其他有機酸等、或者碳酸緩衝液、Tris(三羥甲基胺基甲烷)緩衝液、組胺酸緩衝液、咪唑緩衝液等。Examples of the buffer include: phosphoric acid, citric acid buffer, acetic acid, malic acid, tartaric acid, succinic acid, lactic acid, potassium phosphate, gluconic acid, caprylic acid, deoxycholic acid, salicylic acid, triethanolamine, fumaric acid Acid and other organic acids, etc., or carbonic acid buffer, Tris (tris (tris(hydroxymethyl)aminomethane) buffer, histidine buffer, imidazole buffer, etc.).

又,亦可藉由溶解於溶液製劑領域公知之水性緩衝液而製備溶液製劑。緩衝液之濃度一般為1~500 mM,較佳為5~100 mM,更佳為10~20 mM。In addition, solution preparations can also be prepared by dissolving in aqueous buffers known in the field of solution preparations. The concentration of the buffer is generally 1-500 mM, preferably 5-100 mM, more preferably 10-20 mM.

作為多糖及單糖等糖類或碳水化合物,例如可列舉:葡聚糖、葡萄糖、果糖、乳糖、木糖、甘露糖、麥芽糖、蔗糖、海藻糖、棉子糖等。 作為糖醇,例如可列舉:甘露醇、山梨糖醇、肌醇等。 Examples of saccharides or carbohydrates such as polysaccharides and monosaccharides include glucan, glucose, fructose, lactose, xylose, mannose, maltose, sucrose, trehalose, and raffinose. Examples of sugar alcohols include mannitol, sorbitol, inositol, and the like.

於將製劑製成水溶液之情形時,例如可列舉含有生理鹽水、葡萄糖或其他輔助劑之等張液,例如D-山梨糖醇、D-甘露糖、D-甘露醇、氯化鈉,亦可併用適宜之溶解輔助劑,例如醇(乙醇等)、多元醇(丙二醇、PEG等)、非離子性界面活性劑(聚山梨醇酯80、HCO-50)等。視需要亦可進而含有稀釋劑、溶解輔助劑、pH調整劑、鎮痛劑、含硫還原劑、抗氧化劑等。When the preparation is made into an aqueous solution, for example, an isotonic solution containing physiological saline, glucose or other auxiliary agents, such as D-sorbitol, D-mannose, D-mannitol, and sodium chloride, can be used. Appropriate dissolving aids, such as alcohols (ethanol, etc.), polyols (propylene glycol, PEG, etc.), nonionic surfactants (polysorbate 80, HCO-50), etc., are used together. If necessary, a diluent, a dissolution aid, a pH adjuster, an analgesic, a sulfur-containing reducing agent, an antioxidant, etc. may be further contained.

縮寫之說明於本說明書中可使用以下之縮寫。 GEM或Gem或G:鹽酸吉西他濱; Nab-pac或nab-PTX或N-PTX:紫杉醇(白蛋白懸浮型)或白蛋白結合紫杉醇; GN:鹽酸吉西他濱與白蛋白結合紫杉醇之併用療法; ATZ或Atezo或A:阿替利珠單抗(基因重組); TCZ或T:托珠單抗; IL-6:介白素6;IL-6R:介白素6受體; PD-1:程序性死亡受體-1(Programmed death-1)、程序性細胞死亡蛋白1(programmed cell death protein 1)、CD279、PD1、SLEB2、PDCD1; PD-L1:PD-1配體1(PD-1 Ligand 1)、程序性死亡配體1(programmed death ligand 1)、CD274、B7-H、B7H1、PD-L1、PDCD1L1、PDCD1LG1、B7同源物1(B7 homolog 1)、PDCD1配體1(PDCD1 ligand 1); CTLA4:細胞毒性T淋巴細胞相關蛋白4(Cytotoxic T-Lymphocyte Associated Protein 4)、CD152; Chemo:化學治療劑; 5FU:氟尿嘧啶; IV(靜脈內投予);IM(肌肉內投予);SC(皮下組織內投予); CIT:癌症免疫療法或Cancer immunotherapy; CRP:血中C反應蛋白; HR:風險比; DLT:劑量限制性毒性; RECIST:實體癌療效判定基準;CR(完全緩解);ORR(總緩解率);PFS(無進展生存期);DFS(無病生存期)、OS(總生存期);pCR(病理學上完全緩解);cCR(臨床上完全緩解);RR(緩解率或Response rate);PR(部分緩解或partial response);DCR(病情控制率或Disease control rate);SD(疾病穩定或stable disease);PD(疾病進展或progressive disease); CRF(症例報告書或Case Report Form); α-SMA:α-平滑肌動蛋白(α-smooth muscle actin); IHC:免疫組織化學染色或Immunohistochemistry; HE染色:蘇木精-伊紅染色; MALDI:基質輔助雷射脫附電離或matrix assisted laser desorption/ionization; MALDI-MSI:基於MALDI法之質譜分析成像法(Mass Spectrometry Imaging或MSI); 核磁共振成像法:Magnetic Resonance Imaging或MRI; αCHCA:α-氰基-4-羥基肉桂酸; PK-PD:藥動學(PK)-藥效學(PD); TIL:腫瘤浸潤淋巴細胞或Tumor Infiltrating Lymphocyte; DC:樹狀細胞或Dendritic cell; iRECIST:適用於免疫療法之改良之RECIST v1.1。(用以測試免疫療法之試驗之相關資料管理與資料收集用指南;A guideline for data management and data collection for trials testing immunotherapeutics.: Seymour L, et al. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol 2017; 18: e143-52) Description of Abbreviations The following abbreviations may be used in this specification. GEM or Gem or G: gemcitabine hydrochloride; Nab-pac or nab-PTX or N-PTX: paclitaxel (albumin suspension) or albumin-bound paclitaxel; GN: combination therapy of gemcitabine hydrochloride and albumin-bound paclitaxel; ATZ or Atezo Or A: atezolizumab (gene recombination); TCZ or T: tocilizumab; IL-6: interleukin 6; IL-6R: interleukin 6 receptor; PD-1: programmed death Receptor-1 (Programmed death-1), programmed cell death protein 1 (programmed cell death protein 1), CD279, PD1, SLEB2, PDCD1; PD-L1: PD-1 Ligand 1 , programmed death ligand 1 (programmed death ligand 1), CD274, B7-H, B7H1, PD-L1, PDCD1L1, PDCD1LG1, B7 homolog 1 (B7 homolog 1), PDCD1 ligand 1 (PDCD1 ligand 1) ; CTLA4: Cytotoxic T-Lymphocyte Associated Protein 4, CD152; Chemo: chemotherapeutic agent; 5FU: fluorouracil; IV (intravenous administration); IM (intramuscular administration); SC (intradermal administration); CIT: Cancer immunotherapy or Cancer immunotherapy; CRP: C-reactive protein in blood; HR: Hazard ratio; DLT: Dose-limiting toxicity; RECIST: Response criteria for solid cancer; CR (complete response) ; ORR (overall response rate); PFS (progression-free survival); DFS (disease-free survival), OS (overall survival); pCR (pathological complete response); cCR (clinical complete response); RR (remission) PR (partial response or partial response); DCR (disease control rate or Disease control rate); SD (stable disease or stable disease); PD (progressive disease or progressive disease); CRF (case report or Case Report Form); α-SMA: α-smooth muscle actin; IHC: immunohistochemical staining or Immunohistochemistry; HE staining: hematoxylin-eosin staining; MALDI: matrix-assisted laser desorption Ionization or matrix assisted laser desorption/ioniz ation; MALDI-MSI: Mass Spectrometry Imaging or MSI based on MALDI method; MRI: Magnetic Resonance Imaging or MRI; αCHCA: α-cyano-4-hydroxycinnamic acid; PK-PD: Pharmacokinetics (PK)-Pharmacodynamics (PD); TIL: Tumor Infiltrating Lymphocyte; DC: Dendritic cell or Dendritic cell; iRECIST: Modified RECIST v1.1 for immunotherapy. (A guideline for data management and data collection for trials testing immunotherapeutics.: Seymour L, et al. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics . Lancet Oncol 2017; 18: e143-52)

詳細說明 化學治療劑+ IL-6 抑制劑及 / 或免疫治療劑於一形態中,本發明提供一種為了處置癌症(例如胰臟癌),而在利用至少一種化學治療劑之現有標準療法之基礎上,使用免疫治療劑及/或IL-6抑制劑的新穎之併用療法。所謂「免疫治療劑」係主要藉由激活身體本來具有之免疫力(免疫細胞)以達到治療目的之療法所使用的藥劑。將以治療癌症為目的之免疫療法特別稱為「癌症免疫療法」,大體分為激活會攻擊癌細胞而使免疫應答亢進之免疫細胞的治療;及藉由阻礙免疫應答抑制分子之作用來進行之治療。作為前一種治療法之代表例,可列舉樹狀細胞疫苗療法,作為後一種治療法,可列舉免疫檢查點抑制劑。於一形態中,作為免疫檢查點抑制劑,可列舉:PD-1抑制劑(PD-1拮抗劑)、CTLA-4抑制劑、PD-L1抑制劑(PD-L1拮抗劑)等。例如,作為抗PD-1抗體,可列舉:納武單抗(Opdivo ( 註冊商標 ))及帕博利珠單抗(Keytruda ( 註冊商標 )),或作為抗PD-L1抗體,可列舉:度伐魯單抗(Imfinzi ( 註冊商標 ))、阿替利珠單抗(Tecentriq ( 註冊商標 ))及阿維魯單抗(Bavencio ( 註冊商標 )),作為抗CTLA-4抗體,可列舉:伊匹單抗(Yervoy ( 註冊商標 ))。 Detailing chemotherapeutic agent + IL-6 inhibitor and / or immunotherapeutic agent in one form, the present invention provides a basis for existing standard therapy utilizing at least one chemotherapeutic agent for the treatment of cancer (eg, pancreatic cancer). above, novel concomitant therapy using immunotherapeutics and/or IL-6 inhibitors. The so-called "immunotherapeutic agent" is a drug used for therapy mainly by activating the body's inherent immunity (immune cells) to achieve the purpose of treatment. Immunotherapy for the purpose of treating cancer is particularly referred to as "cancer immunotherapy", and is roughly divided into treatment that activates immune cells that attack cancer cells to increase the immune response; and treatment that inhibits the action of immune response inhibitory molecules. treat. Representative examples of the former treatment method include dendritic cell vaccine therapy, and as the latter treatment method, immune checkpoint inhibitors can be cited. In one form, examples of the immune checkpoint inhibitor include PD-1 inhibitors (PD-1 antagonists), CTLA-4 inhibitors, PD-L1 inhibitors (PD-L1 antagonists), and the like. For example, as the anti-PD-1 antibody, nivolumab (Opdivo ( registered trademark ) ) and pembrolizumab (Keytruda ( registered trademark ) ), or as the anti-PD-L1 antibody, there may be mentioned: duval Lutuzumab (Imfinzi ( registered trademark ) ), atezolizumab (Tecentriq ( registered trademark ) ), and avelumab (Bavencio ( registered trademark ) ), as anti-CTLA-4 antibodies, include: Monoclonal antibody (Yervoy ( registered trademark ) ).

於對象癌症為胰臟癌之情形時,可與IL-6抑制劑及/或免疫治療劑組合之已知之化學治療劑之代表例係鹽酸吉西他濱(GEM)單劑、或GEM與白蛋白結合紫杉醇之併用,但不限於此。業者可根據處置對象之癌症之種類、治療效果、患者之病情、耐受性、以及併用療法之反應性來適當選擇化學治療劑之種類。Representative examples of known chemotherapeutic agents that can be combined with IL-6 inhibitors and/or immunotherapeutics are single doses of gemcitabine hydrochloride (GEM), or GEM and albumin-bound paclitaxel when the subject cancer is pancreatic cancer used together, but not limited to this. The type of chemotherapeutic agent can be appropriately selected by the practitioner according to the type of cancer to be treated, the therapeutic effect, the patient's condition, tolerance, and the responsiveness of concomitant therapy.

作為「化學治療劑」,已有多種藥劑被核准主要作為抗癌劑在臨床上使用,且有更多之化合物處在開發中。作為代表性之化學治療劑,根據其化學結構與作用,分為嘧啶系(氟尿嘧啶、替加氟、吉西他濱、卡培他濱、其他);紫杉烷系(紫杉醇、白蛋白結合紫杉醇、歐洲紫杉醇、其他)、鉑系(順鉑、奧沙利鉑、其他)、亞葉酸、喜樹鹼衍生物、分子靶向藥等。於本發明中,可將該等化學治療劑中之1種或2種或其以上與IL-6抑制劑及/或免疫治療劑組合使用。於一形態中,本發明中使用之化學治療劑為吉西他濱或其藥學上容許之鹽(例如鹽酸吉西他濱)。於另一形態中,本發明中使用之化學治療劑為吉西他濱或其藥學上容許之鹽(例如鹽酸吉西他濱)及包含白蛋白與紫杉烷之奈米粒子(例如白蛋白結合紫杉醇(Abraxane ( 註冊商標 )))。 As "chemotherapeutic agents", various agents have been approved for clinical use mainly as anticancer agents, and many more compounds are under development. Representative chemotherapeutic agents are classified into pyrimidines (fluorouracil, tegafur, gemcitabine, capecitabine, others), taxanes (paclitaxel, nab-paclitaxel, paclitaxel, etc.) according to their chemical structures and actions. , others), platinum series (cisplatin, oxaliplatin, others), folinic acid, camptothecin derivatives, molecular targeted drugs, etc. In the present invention, one or two or more of these chemotherapeutic agents may be used in combination with an IL-6 inhibitor and/or an immunotherapeutic agent. In one form, the chemotherapeutic agent used in the present invention is gemcitabine or a pharmaceutically acceptable salt thereof (eg, gemcitabine hydrochloride). In another form, the chemotherapeutic agents used in the present invention are gemcitabine or a pharmaceutically acceptable salt thereof (such as gemcitabine hydrochloride) and nanoparticles comprising albumin and taxane (such as albumin-bound paclitaxel (Abraxane ( registered)) trademark ) )).

進而,將來可被採用的胰臟癌之標準療法之化學治療劑與IL-6抑制劑及/或免疫治療劑之併用亦包含於本發明之範圍內。關於此種化學治療劑之較佳用法、劑量,詳細資訊可自專家能容易訪問之來源獲取,例如美國FDA、日本醫藥品醫療器械資訊提供主頁或相關學會之公開資料等。Furthermore, the concomitant use of chemotherapeutic agents and IL-6 inhibitors and/or immunotherapeutic agents as standard therapy for pancreatic cancer, which may be adopted in the future, is also included in the scope of the present invention. Detailed information on the preferred usage and dosage of this chemotherapeutic agent can be obtained from sources that can be easily accessed by experts, such as the US FDA, the Japan Pharmaceutical and Medical Device Information Homepage, or the public information of relevant societies.

所謂「IL-6抑制劑」或「IL-6訊號抑制劑」係阻斷IL-6介導之訊號傳遞而抑制IL-6之生物學活性之物質。作為IL-6抑制劑之具體例,可列舉:與IL-6結合之物質、與IL-6受體結合之物質、與gp130結合之物質等。又,作為IL-6抑制劑,可列舉:IL-6訊號下游之JAK-STAT通路或MAP激酶通路之抑制劑,尤其是抑制作為IL-6介導之重要細胞內訊號之STAT3磷酸化的物質,例如AG490、魯索替尼等。IL-6抑制劑並無特別限定,包括:抗IL-6抗體、抗IL-6R抗體、抗gp130抗體、IL-6改型體、可溶性IL-6受體改型體、IL-6部分肽、IL-6受體部分肽、顯示與該等相同之活性之低分子化合物等。The so-called "IL-6 inhibitor" or "IL-6 signaling inhibitor" is a substance that blocks the signal transmission mediated by IL-6 and inhibits the biological activity of IL-6. Specific examples of IL-6 inhibitors include substances that bind to IL-6, substances that bind to IL-6 receptors, and substances that bind to gp130. In addition, as IL-6 inhibitors, there can be mentioned: inhibitors of the JAK-STAT pathway or MAP kinase pathway downstream of IL-6 signaling, especially substances that inhibit the phosphorylation of STAT3, which is an important intracellular signal mediated by IL-6 , such as AG490, Ruxolitinib, etc. IL-6 inhibitor is not particularly limited, including: anti-IL-6 antibody, anti-IL-6R antibody, anti-gp130 antibody, IL-6 modified form, soluble IL-6 receptor modified form, IL-6 partial peptide , IL-6 receptor partial peptides, low molecular compounds showing the same activity as these, etc.

作為IL-6抑制劑之較佳形態,可列舉:IL-6受體抑制劑、尤其是抗IL-6R抗體。作為准許於臨床上使用之抗IL-6R抗體(亦稱為抗IL-6R抗體),可列舉:托珠單抗、薩特利珠單抗或沙利姆單抗,均可用於本發明之併用療法之醫藥、處置方法、套組、用途。Preferable examples of IL-6 inhibitors include IL-6 receptor inhibitors, especially anti-IL-6R antibodies. Examples of anti-IL-6R antibodies (also called anti-IL-6R antibodies) approved for clinical use include tocilizumab, satrilizumab or salimumab, all of which can be used in the present invention. Combination therapy medicine, treatment method, set, use.

於一形態中,抗IL-6R抗體考慮到所需功能,只要為具有與托珠單抗相同之抗原結合特性之抗體即可,例如可為含有包含與托珠單抗之重鏈及輕鏈中之各超可變區(CDR)之胺基酸序列相同之胺基酸序列的各CDR之胺基酸序列之抗體或其抗原結合片段。In one form, the anti-IL-6R antibody may be an antibody having the same antigen-binding properties as tocilizumab in consideration of the desired function, for example, it may contain a heavy chain and a light chain including tocilizumab. An antibody or an antigen-binding fragment thereof of the amino acid sequence of each CDR in which the amino acid sequence of each hypervariable region (CDR) is the same.

於一實施形態中,本醫藥可為包含與托珠單抗之胺基酸序列具有至少80%、85%、90%、95%、98%、99%序列同一性之胺基酸序列的抗體或其抗原結合片段。於一實施形態中,本醫藥可為含有包含與托珠單抗之重鏈及輕鏈中之各CDR之胺基酸序列具有至少80%、85%、90%、95%、98%、99%序列同一性之胺基酸序列的各CDR之抗體或其抗原結合片段。於一實施形態中,本醫藥可為含有包含與托珠單抗之VH區及VL區各自之胺基酸序列具有至少80%、85%、90%、95%、98%、99%序列同一性之胺基酸序列的VH區及VL區之抗體或其抗原結合片段。In one embodiment, the medicine can be an antibody comprising an amino acid sequence having at least 80%, 85%, 90%, 95%, 98%, 99% sequence identity with the amino acid sequence of tocilizumab or antigen-binding fragments thereof. In one embodiment, the medicament may contain at least 80%, 85%, 90%, 95%, 98%, 99% of the amino acid sequence comprising each CDR in the heavy and light chains of tocilizumab. The antibody or antigen-binding fragment thereof to each CDR of the amino acid sequence of % sequence identity. In one embodiment, the medicament may contain at least 80%, 85%, 90%, 95%, 98%, 99% sequence identity with the amino acid sequence of each of the VH and VL regions of tocilizumab Antibodies or antigen-binding fragments thereof in the VH and VL regions of the amino acid sequences of sex.

於另一形態中,本發明提供一種為了進行癌(例如胰臟癌)之處置,而於利用至少一種化學治療劑之現有標準療法之基礎上,使用IL-6抑制劑及免疫檢查點抑制劑(例如PD-1/PD-L1訊號抑制劑)之任一者或兩者的新穎之併用療法。「PD-1/PD-L1訊號抑制劑」藉由阻止PD-1與PD-L1結合,而阻礙所謂免疫檢查點抑制機制,使T細胞再活化,藉此發揮抗腫瘤效果。PD-1/PD-L1訊號抑制劑包括PD-1拮抗劑或PD-L1拮抗劑。作為目前已准許於臨床上使用之PD-1/PD-L1訊號抑制劑,已知複數種抗PD-1抗體或抗PD-L1抗體。作為PD-1拮抗劑,並無特別限定,包括:AMP-224、BMS-936559、MEDI4736、MSB0010718C、MPDL3280A、納武單抗、AMP-514、帕博利珠單抗(MK-3475)、REGN2810、PDR001、BGB-A317及匹地利珠單抗(CT-011)。作為PD-L1拮抗劑,並無特別限定,包括:度伐魯單抗、阿替利珠單抗、阿維魯單抗、BMS-936559、MSB0010718C、MPDL3280A、或MEDI4736。In another aspect, the present invention provides an IL-6 inhibitor and an immune checkpoint inhibitor in addition to existing standard therapy using at least one chemotherapeutic agent for the treatment of cancer (eg, pancreatic cancer). Novel combination therapy of either or both (eg PD-1/PD-L1 signaling inhibitors). "PD-1/PD-L1 signaling inhibitors" block the so-called immune checkpoint inhibition mechanism by preventing the binding of PD-1 and PD-L1, thereby reactivating T cells, thereby exerting anti-tumor effects. PD-1/PD-L1 signaling inhibitors include PD-1 antagonists or PD-L1 antagonists. As PD-1/PD-L1 signaling inhibitors currently approved for clinical use, a plurality of anti-PD-1 antibodies or anti-PD-L1 antibodies are known. The PD-1 antagonist is not particularly limited, and includes: AMP-224, BMS-936559, MEDI4736, MSB0010718C, MPDL3280A, nivolumab, AMP-514, pembrolizumab (MK-3475), REGN2810, PDR001, BGB-A317 and pidilizumab (CT-011). The PD-L1 antagonist is not particularly limited, and includes durvalumab, atezolizumab, avelumab, BMS-936559, MSB0010718C, MPDL3280A, or MEDI4736.

於一形態中,本發明之醫藥係用於處置個體之胰臟癌的含有抗PD-L1抗體、更具體而言阿替利珠單抗之醫藥,本發明之醫藥亦可表示為用於置個體之胰臟癌的與含有其他藥劑之第二醫藥併用之含有阿替利珠單抗之第一醫藥。其他藥劑可為至少1種化學治療劑、及/或托珠單抗。因此,本發明之醫藥亦可表示為用於處置個體之胰臟癌的與其他第二及第三醫藥併用之含有阿替利珠單抗之第一醫藥。本發明之醫藥亦可表示為用於處置個體之胰臟癌的與其他第二、第三及第四醫藥併用之含有阿替利珠單抗之第一醫藥。本發明之醫藥亦可表示為用於處置個體之胰臟癌的與其他第二、第三、第四及第五醫藥併用之含有阿替利珠單抗之第一醫藥。In one form, the medicine of the present invention is a medicine containing an anti-PD-L1 antibody, more specifically atezolizumab, for treating pancreatic cancer in an individual. The medicine of the present invention may also be expressed as a medicine for treating pancreatic cancer. The first drug containing atezolizumab is used in combination with the second drug containing other drugs for pancreatic cancer of the individual. The other agents can be at least one chemotherapeutic agent, and/or tocilizumab. Therefore, the medicine of the present invention can also be expressed as the first medicine containing atezolizumab used in combination with other second and third medicines for treating pancreatic cancer in an individual. The medicine of the present invention can also be represented as the first medicine containing atezolizumab used in combination with other second, third and fourth medicines for treating pancreatic cancer in an individual. The medicine of the present invention can also be represented as the first medicine containing atezolizumab used in combination with other second, third, fourth and fifth medicines for treating pancreatic cancer in an individual.

於一形態中,本發明之醫藥係用於處置個體之胰臟癌的含有抗介白素6受體抗體、更具體而言托珠單抗之醫藥。本發明之醫藥亦可表示為用於處置個體之胰臟癌的與含有其他藥劑之第二醫藥併用之含有托珠單抗之第一醫藥。其他藥劑可為至少1種化學治療劑、及/或阿替利珠單抗。因此,本發明之醫藥亦可表示為用於處置個體之胰臟癌的與其他第二及第三醫藥併用之含有托珠單抗之第一醫藥。本發明之醫藥亦可表示為用於處置個體之胰臟癌的與其他第二、第三及第四醫藥併用之含有托珠單抗之第一醫藥。本發明之醫藥亦可表示為用於處置個體之胰臟癌的與其他第二、第三、第四及第五醫藥併用之含有托珠單抗之第一醫藥。In one form, the medicament of the present invention is a medicament containing an anti-interleukin-6 receptor antibody, more specifically tocilizumab, for treating pancreatic cancer in an individual. The medicine of the present invention can also be represented as a first medicine containing tocilizumab used in combination with a second medicine containing other medicines for treating pancreatic cancer in an individual. The other agents can be at least one chemotherapeutic agent, and/or atezolizumab. Therefore, the medicine of the present invention can also be represented as the first medicine containing tocilizumab used in combination with other second and third medicines for treating pancreatic cancer in an individual. The medicine of the present invention can also be represented as the first medicine containing tocilizumab used in combination with other second, third and fourth medicines for treating pancreatic cancer in an individual. The medicine of the present invention can also be represented as the first medicine containing tocilizumab used in combination with other second, third, fourth and fifth medicines for treating pancreatic cancer in an individual.

於本發明之醫藥、套組、處置方法、用途中,可與托珠單抗及/或阿替利珠單抗組合投予之化學治療劑例如為公知或新穎之抗癌物質,更具體而言,例如可列舉:嘧啶系化合物、紫杉烷系化合物、鉑製劑等,例如包括選自由鹽酸吉西他濱、奧沙利鉑、氟尿嘧啶、替加氟-尿嘧啶複合劑、替加氟-吉莫斯特-奧替拉西鉀複合劑、順鉑、卡培他濱、鹽酸伊立替康、亞葉酸、左亞葉酸鹽、白蛋白結合紫杉醇、埃羅替尼、歐洲紫杉醇、MM-398(伊立替康脂質體製劑)所組成之群中之1種以上之化學治療劑。In the medicine, kit, treatment method and use of the present invention, the chemotherapeutic agents that can be administered in combination with tocilizumab and/or atezolizumab are, for example, known or novel anticancer substances, more specifically For example, pyrimidine-based compounds, taxane-based compounds, platinum preparations, etc., including gemcitabine hydrochloride, oxaliplatin, fluorouracil, tegafur-uracil complex, tegafur-gimos Te-oteracil potassium complex, cisplatin, capecitabine, irinotecan hydrochloride, leucovorin, levofolinate, nab-paclitaxel, erlotinib, paclitaxel, MM-398 One or more chemotherapeutic agents in the group consisting of lipotecan liposome preparations.

例如,於併用托珠單抗及/或阿替利珠單抗之情形時,作為化學治療劑之組合,可例示: ・單獨之吉西他濱、 ・吉西他濱及奧沙利鉑、 ・吉西他濱及替加氟-吉莫斯特-奧替拉西鉀複合劑(亦稱為S-1或TS-1)、 ・單獨之順鉑、 ・吉西他濱及順鉑、 ・FOLFORINOX(氟尿嘧啶、亞葉酸(LV)或左亞葉酸鹽(l-LV)、鹽酸伊立替康及奧沙利鉑)、 ・吉西他濱及卡培他濱、 ・單獨之卡培他濱、 ・單獨之氟尿嘧啶、 ・氟尿嘧啶及亞葉酸、 ・單獨之替加氟-吉莫斯特-奧替拉西鉀複合劑(S-1或TS-1)、 ・吉西他濱及白蛋白結合紫杉醇、 ・吉西他濱及埃羅替尼、 ・GTX(吉西他濱、歐洲紫杉醇及卡培他濱)、 ・OFF(奧沙利鉑、氟尿嘧啶、及亞葉酸(LV)或左亞葉酸鹽(l-LV))、 ・CapeOX(卡培他濱與奧沙利鉑)、 ・FOLFOX(亞葉酸(LV)或左亞葉酸鹽(l-LV)、及氟尿嘧啶與奧沙利鉑)、 ・FOLFIRI(亞葉酸(LV)或左亞葉酸鹽(l-LV)、及氟尿嘧啶與鹽酸伊立替康) ・MM-398(伊立替康脂質體製劑)、氟尿嘧啶及亞葉酸(LV)或左亞葉酸鹽(l-LV), 但不限定於該等。又,該等化學治療劑由於溶解性等原因,有時記載其於藥學上容許之鹽或溶劑合物作為製劑之有效成分,但該等藥劑之投予量通常以活性本體之投予量來規定。 For example, when tocilizumab and/or atezolizumab are used in combination, the combination of chemotherapeutic agents can be exemplified: ・Gemcitabine alone, ・Gemcitabine and oxaliplatin, ・Gemcitabine and tegafur-gemoster-oteracil potassium combination (also known as S-1 or TS-1), ・Cisplatin alone, ・Gemcitabine and cisplatin, ・FOLFORINOX (fluorouracil, folinic acid (LV) or levofolinate (l-LV), irinotecan hydrochloride and oxaliplatin), ・Gemcitabine and capecitabine, ・Capecitabine alone, ・Fluorouracil alone, ・Fluorouracil and folinic acid, ・Single tegafur-gimoster-oteracil potassium complex (S-1 or TS-1), ・Gemcitabine and nab-paclitaxel, ・Gemcitabine and erlotinib, ・GTX (Gemcitabine, Paclitaxel and Capecitabine), ・OFF (oxaliplatin, fluorouracil, and leucovorin (LV) or levofolinate (l-LV)), ・CapeOX (capecitabine and oxaliplatin), ・FOLFOX (folinic acid (LV) or levofolinate (l-LV), and fluorouracil and oxaliplatin), ・FOLFIRI (folinic acid (LV) or levofolinate (l-LV), and fluorouracil and irinotecan hydrochloride) ・MM-398 (liposomal formulation of irinotecan), fluorouracil and leucovorin (LV) or levofolinate (l-LV), But not limited to these. In addition, these chemotherapeutic agents are sometimes described in pharmaceutically acceptable salts or solvates as active ingredients of the preparation due to solubility and other reasons, but the dosage of these drugs is usually based on the dosage of the active substance. Regulation.

IL-6R 抗體、抗 PD-L1 抗體於一形態中,本發明之醫藥係用於處置胰臟癌的含有抗介白素6受體抗體(抗IL-6R抗體)、更具體而言托珠單抗之醫藥,相較於未投予抗IL-6R抗體之情形,於包含需要進行胰臟癌之處置之個體的群組中,投予抗IL-6R抗體使群組中之有效率提高。 In one form, anti- IL-6R antibody and anti- PD-L1 antibody , the medicine of the present invention contains anti-interleukin-6 receptor antibody (anti-IL-6R antibody) for treating pancreatic cancer, more specifically The medicine of zizumab, in a group comprising individuals in need of pancreatic cancer treatment, administration of an anti-IL-6R antibody resulted in greater efficacy in the group compared to the situation where no anti-IL-6R antibody was administered improve.

於一形態中,本發明之醫藥係用於處置無法切除之局部進展期或遠處轉移性胰臟癌的含有抗PD-L1抗體、更具體而言阿替利珠單抗之醫藥,相較於未投予抗PD-L1抗體之情形,於包含需要進行胰臟癌之處置之個體的群組中,投予抗PD-L1抗體使群組中之有效率提高。可為無針對轉移性胰臟癌之全身療法之治療史的個體群組,亦可為有針對轉移性胰臟癌之一次治療之全身療法之前期治療史的個體群組。In one form, the medicament of the present invention is a medicament containing an anti-PD-L1 antibody, more specifically atezolizumab, for the treatment of unresectable locally advanced or distant metastatic pancreatic cancer, compared to In the case where no anti-PD-L1 antibody was administered, in a cohort comprising individuals in need of treatment for pancreatic cancer, administration of an anti-PD-L1 antibody resulted in increased efficacy in the cohort. It can be a group of individuals without a treatment history of systemic therapy for metastatic pancreatic cancer, or it can be a group of individuals with a history of prior treatment with systemic therapy for one treatment of metastatic pancreatic cancer.

於一形態中,本發明之醫藥係用於處置無法切除之局部進展期或遠處轉移性胰臟癌的含有抗IL-6R抗體、更具體而言托珠單抗之醫藥,相較於未投予抗IL-6R抗體之情形,於包含需要進行胰臟癌之處置之個體的群組中,投予抗IL-6R抗體使群組中之有效率提高。可為無針對轉移性胰臟癌之全身療法之治療史的個體群組,亦可為有針對轉移性胰臟癌之一次治療之全身療法之前期治療史的個體群組。In one form, the medicament of the present invention is a medicament containing an anti-IL-6R antibody, more specifically tocilizumab, for the treatment of unresectable locally advanced or distant metastatic pancreatic cancer, compared to unresectable medicaments. In the case of administration of an anti-IL-6R antibody, in a group comprising individuals in need of pancreatic cancer treatment, administration of an anti-IL-6R antibody resulted in an increase in the efficiency in the group. It can be a group of individuals without a treatment history of systemic therapy for metastatic pancreatic cancer, or it can be a group of individuals with a history of prior treatment with systemic therapy for one treatment of metastatic pancreatic cancer.

於一形態中,本發明之醫藥係用於處置復發或治療抗性胰臟癌的含有抗PD-L1抗體、更具體而言阿替利珠單抗之醫藥,相較於未投予抗PD-L1抗體之情形,於包含需要進行胰臟癌之處置之個體的群組中,投予抗PD-L1抗體使群組中之有效率提高。In one form, the medicament of the present invention is a medicament containing an anti-PD-L1 antibody, more specifically atezolizumab, for the treatment of recurrent or treatment-resistant pancreatic cancer, compared to no anti-PD administration - In the case of L1 antibody, in a group comprising individuals in need of pancreatic cancer treatment, administration of an anti-PD-L1 antibody resulted in increased efficacy in the group.

於一形態中,本發明之醫藥係用於處置復發或治療抗性胰臟癌的含有抗IL-6R抗體、更具體而言托珠單抗之醫藥,相較於未投予抗IL-6R抗體之情形,於包含需要進行胰臟癌之處置之個體的群組中,投予抗IL-6R抗體使群組中之有效率提高。In one form, the medicament of the present invention is a medicament containing an anti-IL-6R antibody, more specifically tocilizumab, for the treatment of recurrent or treatment-resistant pancreatic cancer, compared to no administration of anti-IL-6R In the case of antibodies, in a group comprising individuals in need of pancreatic cancer treatment, administration of an anti-IL-6R antibody resulted in increased efficacy in the group.

於一形態中,本發明之醫藥係用於處置無標準治療且對1個以上之現有化學療法方案不耐受、無反應或復發之胰臟癌的含有阿替利珠單抗之醫藥,相較於未投予阿替利珠單抗之情形,於包含需要進行胰臟癌之處置之個體的群組中,投予阿替利珠單抗使群組中之有效率提高。In one form, the medicament of the present invention is a medicament containing atezolizumab for the treatment of pancreatic cancer without standard treatment and intolerant, unresponsive or recurring to one or more existing chemotherapy regimens. In a cohort comprising individuals in need of treatment for pancreatic cancer, administration of atezolizumab resulted in increased efficacy in the cohort compared to the case where atezolizumab was not administered.

於一形態中,本發明之醫藥係用於處置無標準治療且對1個以上之現有化學療法方案不耐受、無反應或復發之胰臟癌的含有托珠單抗之醫藥,相較於未投予托珠單抗之情形,於包含需要進行胰臟癌之處置之個體的群組中,投予托珠單抗使群組中之有效率提高。In one form, the medicament of the present invention is a tocilizumab-containing medicament for treating pancreatic cancer without standard treatment and intolerant, unresponsive or recurring to one or more existing chemotherapy regimens, compared to In the case where tocilizumab was not administered, in a cohort comprising individuals in need of pancreatic cancer treatment, administration of tocilizumab resulted in increased efficacy in the cohort.

於一形態中,本發明之處置方法係用於處置胰臟癌之方法,包括如下步驟:按照本說明書中記載之各實施形態,對個體投予本發明之醫藥。可為無針對轉移性胰臟癌之全身療法之治療史的個體群組,亦可為有針對轉移性胰臟癌之一次治療之全身療法之前期治療史的個體群組。In one aspect, the treatment method of the present invention is a method for treating pancreatic cancer, comprising the steps of administering the medicine of the present invention to an individual according to each embodiment described in this specification. It can be a group of individuals without a treatment history of systemic therapy for metastatic pancreatic cancer, or it can be a group of individuals with a history of prior treatment with systemic therapy for one treatment of metastatic pancreatic cancer.

於一形態中,本發明之處置方法係用於處置復發或治療抗性胰臟癌之方法,包括如下步驟:按照本說明書中記載之各實施形態,對個體投予本發明之醫藥。In one aspect, the treatment method of the present invention is a method for treating recurrent or treatment-resistant pancreatic cancer, comprising the steps of: administering the medicine of the present invention to an individual according to each embodiment described in this specification.

於一形態中,本發明之處置方法係用於處置無標準治療且對1個以上之現有化學療法方案不耐受、無反應或復發之胰臟癌的方法,包括如下步驟:按照本說明書中記載之各實施形態,對個體投予本發明之醫藥。In one form, the treatment method of the present invention is a method for treating pancreatic cancer without standard treatment and intolerant, unresponsive or recurring to one or more existing chemotherapy regimens, comprising the steps of: In each of the described embodiments, the medicine of the present invention is administered to an individual.

於一形態中,本發明之用途係用於製造本發明之醫藥的抗PD-L1抗體、更具體而言阿替利珠單抗之用途。 於一形態中,本發明之用途係用於製造本發明之醫藥的抗IL-6R抗體、更具體而言托珠單抗之用途。 In one form, the use of the present invention is the use of an anti-PD-L1 antibody, more specifically atezolizumab, for the manufacture of the medicament of the present invention. In one form, the use of the present invention is the use of an anti-IL-6R antibody, more specifically tocilizumab, for the manufacture of the medicament of the present invention.

IL-6 抑制劑之作用本發明之一形態係基於新發現之IL-6抑制劑之作用。發現IL-6抑制劑促進靶組織中低分子藥劑之滲透。又,IL-6抑制劑存在抑制靶組織纖維化之可能性。基於該等作用,於使用癌治療劑之個體之癌之處置中,為了促進該治療劑向癌組織之滲透,可使用IL-6抑制劑。因此,較佳為將含有IL-6抑制劑之醫藥組合物與上述治療用藥劑組合投予。上述癌之治療用藥劑較佳為分子量400以下之藥劑。癌之治療用藥劑更佳為分子量400以下之化學治療劑。治療對象之癌可為胰臟癌或肺癌,但不限定於該等。癌之治療用藥劑可為吉西他濱及其藥學上容許之鹽。IL-6抑制劑可為抗IL-6R抗體,例如托珠單抗。 Effects of IL-6 Inhibitors One aspect of the present invention is based on the newly discovered effects of IL-6 inhibitors. IL-6 inhibitors were found to promote the penetration of low molecular weight agents in target tissues. In addition, IL-6 inhibitors have the potential to inhibit target tissue fibrosis. Based on these effects, an IL-6 inhibitor can be used in the treatment of cancer of an individual using a cancer therapeutic agent in order to promote the penetration of the therapeutic agent into the cancer tissue. Therefore, it is preferable to administer the pharmaceutical composition containing an IL-6 inhibitor in combination with the above-mentioned therapeutic agent. The drug for the treatment of cancer is preferably a drug with a molecular weight of 400 or less. The therapeutic agent for cancer is more preferably a chemotherapeutic agent with a molecular weight of 400 or less. The cancer to be treated may be pancreatic cancer or lung cancer, but is not limited to these. The drug for cancer treatment can be gemcitabine and its pharmaceutically acceptable salts. The IL-6 inhibitor can be an anti-IL-6R antibody, such as tocilizumab.

進而,本發明之另一形態亦基於新發現之IL-6抑制劑之作用。藉由使用抗IL-6R抗體之試驗,發現IL-6抑制劑促進免疫細胞(例如CD45陽性細胞)向靶組織之浸潤。上述靶組織可為IL-6過度表現組織。又,上述靶組織可為膠原蛋白過度產生組織。又,上述靶組織可為高度纖維性及/或含較密間質(結締組織增生)狀態之組織。又,上述靶組織可為腫瘤組織。因此,為了促進CD45陽性細胞向腫瘤組織之浸潤,可使用IL-6抑制劑。IL-6抑制劑可為抗IL-6R抗體,例如托珠單抗。腫瘤組織可為胰臟癌或肺癌之腫瘤組織,但不限定於該等。Furthermore, another aspect of the present invention is also based on the effect of the newly discovered IL-6 inhibitor. Through experiments using anti-IL-6R antibodies, IL-6 inhibitors were found to promote the infiltration of immune cells (eg, CD45 positive cells) into target tissues. The aforementioned target tissue may be an IL-6 overexpressing tissue. In addition, the above-mentioned target tissue may be a collagen overproducing tissue. In addition, the target tissue may be a highly fibrous and/or tissue containing a relatively dense stroma (connective tissue hyperplasia). In addition, the above-mentioned target tissue may be a tumor tissue. Therefore, in order to promote the infiltration of CD45-positive cells into tumor tissue, IL-6 inhibitors can be used. The IL-6 inhibitor can be an anti-IL-6R antibody, such as tocilizumab. The tumor tissue may be pancreatic cancer or lung cancer tumor tissue, but is not limited to these.

於本發明中,所謂「IL-6過度表現組織」係指與周圍組織或健康正常人對照組該部位之組織相比,IL-6之表現量較高之組織。作為此種組織,可列舉:慢性風濕病患者之關節腔液中等慢性炎症狀態或炎症局部或腫瘤等。可將血中IL-6濃度或血中CRP濃度比健康正常人之血中濃度平均值高之癌患者定義為體內存在「IL-6過度表現腫瘤組織」,但不限定於此。In the present invention, the so-called "IL-6 overexpressing tissue" refers to a tissue in which the expression level of IL-6 is higher than that in the surrounding tissue or the tissue in the healthy normal control group. Examples of such tissues include chronic inflammatory states such as joint fluid of patients with chronic rheumatism, local inflammation, tumors, and the like. Cancer patients whose blood IL-6 concentration or blood CRP concentration is higher than the average blood concentration of healthy normal subjects can be defined as having "IL-6 overexpressing tumor tissue" in the body, but it is not limited to this.

胰臟癌(PC)間質中會高度產生膠原蛋白,膠原蛋白之蓄積會增大腫瘤間質液壓、減少血流與藥物滲透。於本發明中,所謂「膠原蛋白過度產生組織」係指與周圍組織或健康正常人對照組該部位之組織相比,膠原蛋白之產生量較高之組織。所謂膠原蛋白,可列舉:I型膠原蛋白(C1)、II型膠原蛋白(C2)、III型膠原蛋白(C3)、IⅤ型膠原蛋白(C4)、ⅤI型膠原蛋白(C6)及作為該等之肽原的I型膠原蛋白肽原(Pro-C1),II型膠原蛋白肽原(Pro-C2),III型膠原蛋白肽原(Pro-C3、Pro-C3X)、IⅤ型膠原蛋白肽原(Pro-C4)、ⅤI型膠原蛋白肽原(ProC-6)。可將膠原蛋白之血中濃度比健康正常人之血中濃度平均值高之癌患者定義為體內存在「膠原蛋白過度產生腫瘤組織」,但不限定於此。Collagen is highly produced in the stroma of pancreatic cancer (PC), and the accumulation of collagen increases the fluid pressure in the tumor stroma, reducing blood flow and drug penetration. In the present invention, the so-called "collagen overproducing tissue" refers to a tissue that produces a higher amount of collagen than the surrounding tissue or the tissue of the healthy normal control group. Collagen includes type I collagen (C1), type II collagen (C2), type III collagen (C3), type IV collagen (C4), type VI collagen (C6), and other types of collagen. Pro-collagen peptide I (Pro-C1), pro-collagen peptide type II (Pro-C2), pro-collagen peptide type III (Pro-C3, Pro-C3X), pro-collagen peptide type I V (Pro-C3X) (Pro-C4), VI pro-collagen peptide (ProC-6). Cancer patients whose blood concentration of collagen is higher than the average blood concentration of healthy normal subjects can be defined as having "tumor tissue overproducing collagen" in the body, but it is not limited to this.

本發明中,於自投予GN+抗IL-6R抗體後之患者之胰臟癌肝轉移灶採集之腫瘤檢體中觀察到結締組織增生受到抑制,IL-6訊號下游之因子STAT3之磷酸化得到顯著抑制,由此確認抗IL-6R抗體之效果。根據該結果可確認,相較於GN療法,抗IL-6R抗體+GN療法誘導優異之結締組織增生抑制,發揮抗腫瘤效果。因此,本發明之一形態係基於藉由IL-6抑制劑之作用而改善腫瘤微環境、促進藥劑之滲透。In the present invention, in tumor specimens collected from pancreatic cancer liver metastases of patients after administration of GN+anti-IL-6R antibody, it was observed that connective tissue hyperplasia was inhibited, and phosphorylation of STAT3, a factor downstream of IL-6 signaling, was obtained. Significant inhibition was observed, thereby confirming the effect of the anti-IL-6R antibody. From this result, it was confirmed that anti-IL-6R antibody + GN therapy induces superior inhibition of connective tissue proliferation compared with GN therapy, and exerts an antitumor effect. Therefore, one aspect of the present invention is based on improving the tumor microenvironment and promoting the penetration of drugs through the action of IL-6 inhibitor.

因此,本發明之一形態可為磷酸化STAT3陽性之個體群組。所謂「磷酸化STAT3陽性」係指投予前之腫瘤組織中之細胞(例如可列舉:腫瘤細胞、免疫細胞、纖維母細胞,但不限定於該等)之磷酸化STAT3之陽性面積比率較高,更具體而言,可列舉:1%以上100%以下、5%以上100%以下,但不限定於該等。Therefore, one aspect of the present invention can be a group of individuals positive for phosphorylated STAT3. "Phosphorylated STAT3 positive" means that cells (for example, tumor cells, immune cells, and fibroblasts, but not limited to these) in the tumor tissue before administration have a high ratio of phosphorylated STAT3 positive areas. , more specifically, 1% or more and 100% or less, 5% or more and 100% or less, but not limited to these.

所謂「藉由IL-6抑制劑促進體內低分子藥劑向IL-6過度表現組織中之滲透」係指於一形態中,藉由IL-6抑制劑促進分子量400以下之藥劑之滲透。又,於一形態中,藉由IL-6抑制劑促進分子量400以下之藥劑向IL-6過度表現組織中之滲透。進而,於一形態中,藉由IL-6抑制劑促進分子量400以下之藥劑向高度纖維性及/或含較密間質(結締組織增生)狀態之組織中之滲透。於「高度纖維性及/或含較密間質」之狀態下,例如觀察到經HE染色腫瘤纖維性間質組織圖像、或α-SMA陽性細胞聚集。所謂「分子量400以下之藥劑」,例如可列舉:5-氟尿嘧啶(分子量130.08)、替加氟(200.16)、吉西他濱(263.20)、順鉑(300.05)、卡培他濱(359.5)、埃羅替尼(393.436)、奧沙利鉑(397.29)等。The so-called "promoting the penetration of low-molecular-weight drugs in vivo into IL-6-overexpressing tissues by IL-6 inhibitors" refers to promoting the penetration of drugs with molecular weights below 400 by IL-6 inhibitors in one form. Furthermore, in one form, the IL-6 inhibitor promotes the penetration of an agent having a molecular weight of 400 or less into the IL-6 overexpressing tissue. Furthermore, in one form, the penetration of agents with a molecular weight of 400 or less into tissues that are highly fibrous and/or contain a denser stroma (connective tissue hyperplasia) is facilitated by an IL-6 inhibitor. In the state of "highly fibrous and/or containing dense stroma", for example, HE-stained images of tumor fibrous stroma, or aggregation of α-SMA-positive cells were observed. The so-called "drugs with a molecular weight of 400 or less" include, for example, 5-fluorouracil (molecular weight 130.08), tegafur (200.16), gemcitabine (263.20), cisplatin (300.05), capecitabine (359.5), erlotite Ni (393.436), Oxaliplatin (397.29) and so on.

所謂「促進免疫細胞之浸潤」係指相較於未投予IL-6抑制劑之情形,投予IL-6抑制劑之情形時CD45陽性細胞之比率較高。已知CD45亦稱作白血球共同抗原(LCA:leukocyte common antigen)、B220、T200,為於除紅血球及血小板以外之所有造血系統細胞之細胞膜上表現之單鏈跨膜蛋白酪胺酸磷酸酶。CD45表徵源自骨髄及其他供給源之人間質幹細胞(MSC),「CD45陽性細胞」呈陽性,有嗜中性細胞、嗜酸性細胞、嗜鹼性細胞、淋巴細胞、單核細胞。淋巴細胞有自然殺手細胞、T細胞、B細胞。The so-called "promoting the infiltration of immune cells" means that the ratio of CD45-positive cells is higher in the case of administering the IL-6 inhibitor than in the case of not administering the IL-6 inhibitor. It is known that CD45, also known as leukocyte common antigen (LCA), B220, and T200, is a single-chain transmembrane protein tyrosine phosphatase expressed on the cell membrane of all hematopoietic cells except erythrocytes and platelets. CD45 characterizes human mesenchymal stem cells (MSCs) derived from bone marrow and other sources. "CD45 positive cells" are positive, including neutrophils, eosinophils, basophils, lymphocytes, and monocytes. Lymphocytes include natural killer cells, T cells, and B cells.

IL-6 抑制劑與免疫檢查點抑制劑之併用之效果進而,本發明之另一形態係基於併用IL-6抑制劑與免疫檢查點抑制劑之情形時新發現之作用。最初發現藉由IL-6抑制劑與免疫檢查點抑制劑之組合,促進免疫細胞向靶組織之浸潤。免疫細胞之代表例可為CD45陽性細胞及B細胞。靶組織可為IL-6過度表現組織。靶組織可為腫瘤組織。腫瘤組織可為胰臟癌或肺癌之腫瘤組織,但不限定於該等。免疫檢查點抑制劑之代表例係與免疫檢查點分子結合而發揮效果者,例如,與CTLA-4結合者(即CTLA-4抑制劑)、或者與PD-1或PD-L1結合者(即PD-1/PD-L1訊號抑制劑)。免疫檢查點抑制劑例如可為抗CTLA-4抗體、抗PD-1抗體、抗PD-L1抗體或該等之任意之組合。 Effect of Combined Use of IL-6 Inhibitor and Immune Checkpoint Inhibitor Furthermore, another aspect of the present invention is based on a newly discovered effect when an IL-6 inhibitor and an immune checkpoint inhibitor are used together. It was initially discovered that the infiltration of immune cells into target tissues is facilitated by the combination of IL-6 inhibitors and immune checkpoint inhibitors. Representative examples of immune cells may be CD45 positive cells and B cells. The target tissue may be an IL-6 overexpressing tissue. The target tissue can be tumor tissue. The tumor tissue may be pancreatic cancer or lung cancer tumor tissue, but is not limited to these. Representative examples of immune checkpoint inhibitors are those that bind to immune checkpoint molecules, such as those that bind to CTLA-4 (ie, CTLA-4 inhibitors), or those that bind to PD-1 or PD-L1 (ie, those that bind to PD-1 or PD-L1). PD-1/PD-L1 signaling inhibitor). The immune checkpoint inhibitor can be, for example, an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-PD-L1 antibody, or any combination of these.

本發明之一形態為IL-6抑制劑及免疫檢查點抑制劑(例如PD-1/PD-L1訊號抑制劑或CTLA-4抑制劑)與其他抗腫瘤劑之併用,藉由IL-6訊號抑制劑改善腫瘤微環境、促進藥劑之滲透,且藉由免疫檢查點抑制劑促進免疫細胞向腫瘤組織之浸潤,從而發揮優異之抗腫瘤作用。One aspect of the present invention is the combined use of IL-6 inhibitor and immune checkpoint inhibitor (such as PD-1/PD-L1 signaling inhibitor or CTLA-4 inhibitor) and other anti-tumor agents, through IL-6 signaling Inhibitors improve the tumor microenvironment, promote the penetration of drugs, and promote the infiltration of immune cells into tumor tissues through immune checkpoint inhibitors, thereby exerting excellent anti-tumor effects.

於一形態中,本發明為用於以免疫療法為基礎之併用治療的IL-6抑制劑與免疫檢查點抑制劑之組合。其具體之一形態為抗IL-6R抗體與抗PD-L1抗體之組合。於本發明中,確認藉由併用抗IL-6R抗體與抗PD-L1抗體,顯著促進包括T細胞及嗜中性球或單核球之主要免疫細胞向胰臟癌腫瘤組織之浸潤。進而,藉由使用小鼠胰臟癌模型之試驗,實際確認到抗腫瘤效果。In one form, the present invention is a combination of an IL-6 inhibitor and an immune checkpoint inhibitor for use in immunotherapy-based concomitant therapy. One specific form is a combination of anti-IL-6R antibody and anti-PD-L1 antibody. In the present invention, it was confirmed that the combined use of anti-IL-6R antibody and anti-PD-L1 antibody significantly promoted the infiltration of major immune cells including T cells and neutrophils or monocytes into pancreatic cancer tumor tissue. Furthermore, an antitumor effect was actually confirmed by a test using a mouse pancreatic cancer model.

進而,具體之一形態為對至少一種化學治療劑併用抗IL-6R抗體與PD-L1抗體之組合。於本發明中,藉由抗IL-6R抗體+抗PD-L1抗體+吉西他濱之併用投予,確認到以樹狀細胞、T細胞、B細胞為代表之CD45陽性免疫細胞向腫瘤內之浸潤亢進,暗示誘導後天性免疫。因此認為,抗IL-6R抗體+抗PD-L1抗體+化學治療劑之併用投予藉由維持腫瘤組織中各種免疫細胞之浸潤、並誘導T細胞與B細胞活化產生後天性免疫,從而增強癌症免疫療法之效果。Furthermore, as a specific form, a combination of an anti-IL-6R antibody and a PD-L1 antibody is used in combination with at least one chemotherapeutic agent. In the present invention, by the combined administration of anti-IL-6R antibody + anti-PD-L1 antibody + gemcitabine, it was confirmed that the infiltration of CD45-positive immune cells represented by dendritic cells, T cells, and B cells into tumors was increased. , suggesting induction of acquired immunity. Therefore, it is considered that the combined administration of anti-IL-6R antibody + anti-PD-L1 antibody + chemotherapeutic agent enhances cancer by maintaining the infiltration of various immune cells in tumor tissue and inducing activation of T cells and B cells to generate acquired immunity Effects of immunotherapy.

托珠單抗及 / 或阿替利珠單抗之投予於一形態中,本發明之醫藥係用於處置個體之胰臟癌的投予托珠單抗及/或阿替利珠單抗之醫藥。 Administration of Tocilizumab and / or Atezolizumab In one form, the medicament of the present invention is the administration of tocilizumab and/or atezolizumab for the treatment of pancreatic cancer in an individual of medicine.

本發明之醫藥可為含有托珠單抗、或含有與托珠單抗併用投予之阿替利珠單抗之胰臟癌之治療劑。或者,本發明之醫藥可為含有阿替利珠單抗、或含有與阿替利珠單抗併用投予之托珠單抗之胰臟癌之治療劑。胰臟癌可為對化學治療劑具有抗藥性之胰臟癌。The medicine of the present invention may be a therapeutic agent for pancreatic cancer containing tocilizumab or atezolizumab administered in combination with tocilizumab. Alternatively, the medicine of the present invention may be a therapeutic agent for pancreatic cancer containing atezolizumab or tocilizumab administered in combination with atezolizumab. Pancreatic cancer may be pancreatic cancer that is resistant to chemotherapeutic agents.

關於本發明之特徵在於提高包含需要進行對化學治療劑具有抗藥性之胰臟癌之處置的個體之群組中之有效率之上述醫藥,於一實施形態中,包含需要進行對化學治療劑具有抗藥性之癌之處置的個體之群組亦可包含未於投予托珠單抗及/或阿替利珠單抗之前診斷檢測有無抗藥性之個體。或者關於該醫藥,於另一實施形態中,包含需要進行對化學治療劑具有抗藥性之癌之處置的個體之群組亦可包含於投予托珠單抗及/或阿替利珠單抗之前診斷檢測有無抗藥性而確定有抗藥性之個體。The present invention is characterized in that the above-mentioned medicine for improving the effectiveness in a group including individuals requiring treatment of pancreatic cancer resistant to a chemotherapeutic agent includes, in one embodiment, a need for treatment with a chemotherapeutic agent The cohort of individuals treated for drug-resistant cancer may also include individuals who have not been diagnostically tested for drug resistance prior to administration of tocilizumab and/or atezolizumab. Or with regard to the medicine, in another embodiment, a group comprising individuals in need of treatment for cancers resistant to chemotherapeutic agents may also be included in the administration of tocilizumab and/or atezolizumab Individuals identified as drug-resistant by prior diagnostic testing for drug resistance.

於一形態中,本發明之處置方法係用於處置對化學治療劑具有抗藥性之胰臟癌之處置方法。於一實施形態中,相較於未投予托珠單抗及/或阿替利珠單抗之情形,於包含需要進行對化學治療劑具有抗藥性之胰臟癌之處置之個體的群組中,該處置方法使群組中之有效率提高。In one form, the treatment method of the present invention is a treatment method for pancreatic cancer resistant to chemotherapeutic agents. In one embodiment, in a group comprising individuals in need of treatment for chemotherapeutic agent-resistant pancreatic cancer, compared to the situation where tocilizumab and/or atezolizumab were not administered , the treatment method increases the efficiency in the group.

關於本發明之特徵在於提高包含需要進行對化學治療劑具有抗藥性之胰臟癌之處置的個體之群組中之有效率之上述醫藥,於一實施形態中,包含需要進行對化學治療劑具有抗藥性之癌之處置的個體之群組亦可包含未於投予托珠單抗及/或阿替利珠單抗之前診斷檢測有無抗藥性之個體。或者關於該醫藥,於另一實施形態中,包含需要進行對化學治療劑具有抗藥性之癌之處置的個體之群組亦可包含於投予托珠單抗之前診斷檢測有無抗藥性而確定有抗藥性之個體。The present invention is characterized in that the above-mentioned medicine for improving the effectiveness in a group including individuals requiring treatment of pancreatic cancer resistant to a chemotherapeutic agent includes, in one embodiment, a need for treatment with a chemotherapeutic agent The cohort of individuals treated for drug-resistant cancer may also include individuals who have not been diagnostically tested for drug resistance prior to administration of tocilizumab and/or atezolizumab. Or with regard to the medicine, in another embodiment, a group including individuals who need to undergo treatment for cancers resistant to chemotherapeutic agents may also be included in a diagnostic test for drug resistance prior to administration of tocilizumab to determine the presence or absence of drug resistance. drug-resistant individuals.

於一形態中,本發明之用途係用於製造本發明之醫藥或套組的托珠單抗及/或阿替利珠單抗之用途。 於本發明之用途中,醫藥或套組為用以處置個體之胰臟癌之醫藥或套組。於該用途中,胰臟癌較佳為復發/治療抗性胰臟癌。 In one form, the use of the present invention is the use of tocilizumab and/or atezolizumab for the manufacture of the medicament or kit of the present invention. In the use of the present invention, a medicament or kit is a medicament or kit for treating pancreatic cancer in an individual. In this use, the pancreatic cancer is preferably recurrent/treatment resistant pancreatic cancer.

於本發明之用途中,醫藥或套組為用以處置個體之對化學治療劑具有抗藥性之胰臟癌之醫藥或套組。關於該用途,於一實施形態中,醫藥或套組包含托珠單抗及/或阿替利珠單抗。For the purposes of the present invention, a medicament or kit is a medicament or kit for treating pancreatic cancer resistant to chemotherapeutic agents in an individual. For this use, in one embodiment, the medicament or kit comprises tocilizumab and/or atezolizumab.

於本發明之用途中,醫藥或套組為用以處置對化學治療劑具有抗藥性之癌之醫藥或套組。於一實施形態中,該醫藥或套組包含托珠單抗及/或阿替利珠單抗。於一實施形態中,相較於未投予托珠單抗及/或阿替利珠單抗之情形,於包含需要進行對化學治療劑具有抗藥性之癌之處置的個體之群組中,該醫藥或套組使群組中之有效率提高。In the use of the present invention, a medicament or kit is a medicament or kit for treating cancers that are resistant to chemotherapeutic agents. In one embodiment, the medicament or kit comprises tocilizumab and/or atezolizumab. In one embodiment, in a group comprising individuals in need of treatment for chemotherapeutic agent-resistant cancers, compared to when tocilizumab and/or atezolizumab were not administered, The medicine or kit increases the effectiveness in the group.

關於本發明之特徵在於提高包含需要進行對化學治療劑具有抗藥性之癌之處置的個體之群組中之有效率之上述用途,於一實施形態中,包含需要進行對化學治療劑具有抗藥性之癌之處置的個體之群組亦可包含未於投予化學治療劑之前診斷檢測有無抗藥性之個體。或者關於該醫藥或套組,於另一實施形態中,包含需要進行對化學治療劑具有抗藥性之癌之處置的個體之群組亦可包含於投予化學治療劑之前診斷檢測有無抗藥性而確定有抗藥性之個體。又,於另一實施形態中,抗藥性可為包含化學療法之先前處置引起之抗藥性。The above-mentioned use of the present invention is characterized by increasing the effectiveness in a group comprising individuals in need of treatment of chemotherapeutic agent-resistant cancers, in one embodiment, including the need for chemotherapeutic-agent-resistant cancers. The group of individuals for the treatment of cancer may also include individuals who have not been diagnostically tested for drug resistance prior to administration of the chemotherapeutic agent. Or with regard to the medicine or kit, in another embodiment, a group comprising individuals in need of treatment of chemotherapeutic agent-resistant cancers may also be included in diagnostic testing for drug resistance prior to administration of the chemotherapeutic agent. Identify drug-resistant individuals. Also, in another embodiment, the drug resistance may be drug resistance caused by prior treatment including chemotherapy.

本發明之醫藥及套組除包含托珠單抗及/或阿替利珠單抗以外,亦可與用以處置胰臟癌之其他醫藥等、或者放射線治療或外科手術等公知之治療組合使用。In addition to tocilizumab and/or atezolizumab, the medicine and kit of the present invention can also be used in combination with other medicines for treating pancreatic cancer, or known treatments such as radiation therapy or surgery. .

本發明之醫藥及套組包含將作為有效成分之托珠單抗及/或阿替利珠單抗製劑化而成之醫藥或套組之實施形態。 本發明之處置方法係用於處置需要其之個體之胰臟癌的處置方法。包括如下步驟:按照本說明書記載之各實施形態,對個體投予本發明之醫藥、套組、以及托珠單抗及/或阿替利珠單抗。 The medicine and kit of the present invention include an embodiment of the medicine or kit prepared by formulating tocilizumab and/or atezolizumab as active ingredients. The treatment method of the present invention is a treatment method for treating pancreatic cancer in an individual in need thereof. It includes the following steps: according to each embodiment described in this specification, the medicine, the kit of the present invention, and tocilizumab and/or atezolizumab are administered to the individual.

本發明之處置方法亦可包括:利用本說明書中具體記載之本發明之醫藥、套組、以及托珠單抗及/或阿替利珠單抗以外之用以處置癌之其他醫藥、藥劑或套組等進行處置之步驟,或者實施放射線治療或外科手術等公知之治療之步驟。其他醫藥可包含一種或複數種化學治療劑。The treatment method of the present invention may also include: using the medicines, kits of the present invention, and other medicines, pharmaceuticals or drugs other than tocilizumab and/or atezolizumab for treating cancer as specifically described in this specification. Procedures for treatment such as kits, or procedures for performing known treatments such as radiation therapy or surgery. Other medicines may contain one or more chemotherapeutic agents.

本發明之用途係用於製造本發明之醫藥或套組的托珠單抗及/或阿替利珠單抗之用途。本發明之用途中除使用托珠單抗及/或阿替利珠單抗以外,亦可包括使用用以處置癌之其他藥劑、或者放射線治療或外科手術等公知之治療。其他藥劑可包含一種或複數種化學治療劑。The use of the present invention is the use of tocilizumab and/or atezolizumab for the manufacture of the medicine or kit of the present invention. In addition to the use of tocilizumab and/or atezolizumab, the use of the present invention may also include the use of other drugs for treating cancer, or known treatments such as radiation therapy or surgery. Other agents may contain one or more chemotherapeutic agents.

本發明之托珠單抗及/或阿替利珠單抗亦可與用以處置胰臟癌之其他醫藥、藥劑、套組等、或者放射線治療或外科手術等公知之治療組合使用。The tocilizumab and/or atezolizumab of the present invention can also be used in combination with other medicines, pharmaceuticals, kits, etc. for treating pancreatic cancer, or known treatments such as radiation therapy or surgery.

藉由對患有胰臟癌之個體投予托珠單抗及/或阿替利珠單抗,相較於未投予托珠單抗及/或阿替利珠單抗之情形,發揮更高效果。By administering tocilizumab and/or atezolizumab to individuals with pancreatic cancer, compared to the situation where tocilizumab and/or atezolizumab were not administered High effect.

作為本發明之一形態,選擇C反應蛋白(CRP)較高之個體群組。所謂「C反應蛋白較高」,更具體而言為血清或血漿中之濃度0.5 mg/dL以上且未達10 mg/dL、1 mg/dL以上且未達10 mg/dL、2 mg/dL以上且未達10 mg/dL、3 mg/dL以上且未達10 mg/dL、4 mg/dL以上且未達10 mg/dL、5 mg/dL以上且未達10 mg/dL、7 mg/dL以上且未達10 mg/dL之個體群組,但不限定於該等。As an aspect of the present invention, a group of individuals with higher C-reactive protein (CRP) is selected. The so-called "high C-reactive protein", more specifically, the concentration in serum or plasma is 0.5 mg/dL or more and less than 10 mg/dL, 1 mg/dL or more and less than 10 mg/dL, 2 mg/dL Above and below 10 mg/dL, above 3 mg/dL and below 10 mg/dL, above 4 mg/dL and below 10 mg/dL, above 5 mg/dL and below 10 mg/dL, 7 mg /dL or more and less than 10 mg/dL of individual groups, but not limited to these.

進而,若針對化學療法無反應性之胰臟癌,使用托珠單抗及/或阿替利珠單抗,則可確認到明顯比未使用托珠單抗及/或阿替利珠單抗之情形時高之抗癌作用。因此,本發明之醫藥等尤其於化學療法無反應性之難以治療之胰臟癌之處置中發揮其有效性。Furthermore, when tocilizumab and/or atezolizumab were used for chemotherapy-unresponsive pancreatic cancer, it was confirmed that tocilizumab and/or atezolizumab were not used. In this case, the anticancer effect is high. Therefore, the medicine and the like of the present invention are particularly effective in the treatment of difficult-to-treat pancreatic cancer that is not responsive to chemotherapy.

因此,於包含需要進行癌之處置之個體之群組中,藉由利用本發明之醫藥等處置胰臟癌,而與無此種處置之對照組相比,於群組內之各個體中獲得更高之抗癌作用。此處,對照組可為包含需要進行胰臟癌之處置之個體且未投予托珠單抗之群組。Therefore, in a group including individuals in need of cancer treatment, by using the medicine of the present invention, etc. to treat pancreatic cancer, compared with a control group without such treatment, each individual in the group obtained Higher anticancer effect. Here, the control group may be a group comprising individuals in need of pancreatic cancer treatment and not administered tocilizumab.

例如,於本發明之醫藥為含有托珠單抗及/或阿替利珠單抗之醫藥之情形時,於作為包含需要進行胰臟癌之處置之個體之群體的群組中,相較於未投予托珠單抗及/或阿替利珠單抗之情形,於群組內之各個體中獲得更高之抗癌作用。For example, in the case where the medicament of the present invention is a medicament containing tocilizumab and/or atezolizumab, in a group comprising individuals in need of pancreatic cancer treatment, compared with In the absence of administration of tocilizumab and/or atezolizumab, higher anticancer effects were obtained in each individual in the group.

如上所述,藉由利用托珠單抗及/或阿替利珠單抗進行處置,本發明之醫藥等對於化學療法無反應性之癌亦有效。因此,於上述群組為患有化學療法無反應性之癌之個體之群體之情形時,尤其發揮本發明之醫藥等之有效性。本發明之醫藥等在用於包含患有化學療法無反應性之癌之個體之群組之情形時,相較於未使用托珠單抗及/或阿替利珠單抗之情形,可增加確認到抗癌作用之個體之比率,即,提高有效率。再者,無反應性只要對化學療法無反應性或具有抗藥性,則無特別限定,可為先天抗藥性、或後天抗藥性或復發之胰臟癌中之無反應性。於一實施形態中,包含需要進行胰臟癌之處置之個體之群組包含確定有抗藥性之個體。於另一實施形態中,包含需要進行胰臟癌之處置之個體之群組包含確定有因先前處置引起之後天抗藥性之個體。於本實施形態中,例如醫藥可為含有托珠單抗及/或阿替利珠單抗之醫藥。As described above, by treating with tocilizumab and/or atezolizumab, the medicament of the present invention and the like are also effective for chemotherapy-unresponsive cancers. Therefore, in the case where the above-mentioned group is a group of individuals suffering from chemotherapy-unresponsive cancer, the medicines and the like of the present invention are particularly effective. When the medicament and the like of the present invention are used in a group including individuals with chemotherapy-unresponsive cancer, compared to the case in which tocilizumab and/or atezolizumab are not used, an increase in The ratio of individuals who confirmed the anticancer effect, that is, the effective rate was improved. In addition, the anergy is not particularly limited as long as it is not responsive to chemotherapy or has drug resistance, and may be innate drug resistance, acquired drug resistance, or anergy in recurrent pancreatic cancer. In one embodiment, the group comprising individuals in need of treatment for pancreatic cancer comprises individuals determined to be drug-resistant. In another embodiment, the group comprising individuals in need of treatment for pancreatic cancer includes individuals determined to have acquired resistance due to prior treatment. In this embodiment, for example, the medicine may be a medicine containing tocilizumab and/or atezolizumab.

於一實施形態中,包含需要進行胰臟癌之處置之個體之群組可為包含於利用托珠單抗及/或阿替利珠單抗處置前未判別對胰臟癌之化學療法是否無反應性之個體之群組。於一實施形態中,對於包含需要進行胰臟癌之處置之個體之群組中之至少一部分個體,可於利用托珠單抗及/或阿替利珠單抗處置前,判別對該胰臟癌之化學療法是否無反應性。無反應性只要對化學療法無反應性或具有抗藥性,則無特別限定,亦可為先天抗藥性。如上所述,於對化學療法無反應性之癌之情形時,尤其發揮本發明之醫藥等之有效性,因此,特別能夠提高包含需要進行胰臟癌之處置之個體之群組中之有效率。In one embodiment, the group comprising individuals in need of treatment for pancreatic cancer may be comprised of individuals who have not been identified prior to treatment with tocilizumab and/or atezolizumab for the absence of chemotherapy for pancreatic cancer. Groups of reactive individuals. In one embodiment, for at least a portion of the group including individuals in need of pancreatic cancer treatment, the pancreatic cancer can be identified before treatment with tocilizumab and/or atezolizumab. Whether the cancer is unresponsive to chemotherapy. Anergy is not particularly limited as long as it is unresponsive to chemotherapy or has drug resistance, and it may be innate drug resistance. As described above, in the case of cancer that is not responsive to chemotherapy, the medicine of the present invention is particularly effective, and therefore, the effectiveness in a group including individuals requiring pancreatic cancer treatment can be particularly improved. .

本發明之醫藥等藉由使用托珠單抗及/或阿替利珠單抗,相較於未使用托珠單抗及/或阿替利珠單抗之情形,無論對於化學療法無反應性之胰臟癌或應答性之癌,均發揮顯著更高之抗癌作用。然而,由於尤其在化學療法無反應性之難以治療之胰臟癌之處置中能夠發揮其有效性,故而對於對先前處置具有抗藥性或無反應性之個體、不適於繼續實施先前處置之胰臟癌、或對先前處置之應答性之程度未達預期之胰臟癌(例如不應答、且/或毒性之原因),即,對化學治療劑具有先天抗藥性之胰臟癌、或具有因利用化學療法等之先前處置而獲得之後天抗藥性之胰臟癌、利用化學療法等之先前處置後出現之復發性胰臟癌等難以治療之胰臟癌的處置特別有效。本發明之醫藥等尤其對於因為對化學療法無反應性而難以處置之癌特別有效。即便對於對化學療法無反應性之胰臟癌,即,對化學治療劑具有先天抗藥性之胰臟癌、或因已實施之先前處置而具有後天抗藥性之胰臟癌、或復發之胰臟癌,托珠單抗及/或阿替利珠單抗發揮與未投予托珠單抗及/或阿替利珠單抗之情形相比更高之效果,顯著提高癌之治療效果。By using tocilizumab and/or atezolizumab, the medicine of the present invention has no reactivity to chemotherapy compared to the case where tocilizumab and/or atezolizumab are not used. Pancreatic cancer or responsive cancer, both play a significantly higher anticancer effect. However, because of its effectiveness especially in the treatment of refractory pancreatic cancer that is not responsive to chemotherapy, it is not suitable to continue the previously treated pancreas for individuals who are resistant or non-responsive to the previous treatment. Cancer, or pancreatic cancer with an unexpected degree of responsiveness to prior treatment (eg, non-responsiveness, and/or cause of toxicity), i.e., pancreatic cancer with innate resistance to chemotherapeutic agents, or with It is particularly effective in the treatment of difficult-to-treat pancreatic cancer such as pancreatic cancer that has acquired resistance after previous treatment with chemotherapy or the like, or recurrent pancreatic cancer that has appeared after previous treatment with chemotherapy or the like. The medicine and the like of the present invention are particularly effective for cancers that are difficult to treat because they are unresponsive to chemotherapy. Even for pancreatic cancer that is not responsive to chemotherapy, i.e., pancreatic cancer that is innately resistant to chemotherapeutic agents, or that is acquired resistance due to previous treatments that have been performed, or that has recurred Cancer, tocilizumab and/or atezolizumab exerted a higher effect than the case where tocilizumab and/or atezolizumab were not administered, significantly improving the therapeutic effect of cancer.

又,本發明之醫藥等可用於處置例如無法藉由外科手術切除、進展性及/或復發之胰臟癌。本發明之醫藥等可較佳地用於處置無需進行治癒性切除之進展期或復發性胰臟癌、無法切除之進展期或復發性胰臟癌、或者無法手術或復發性胰臟癌。In addition, the medicament and the like of the present invention can be used to treat, for example, pancreatic cancer that cannot be surgically removed, that has advanced and/or recurred. The medicament and the like of the present invention can be preferably used for the treatment of advanced or recurrent pancreatic cancer that does not require curative resection, unresectable advanced or recurrent pancreatic cancer, or inoperable or recurrent pancreatic cancer.

於本發明之一實施形態中,本發明之醫藥等中之處置對象之胰臟癌可為於先前處置時、處置中或處置後仍在進展之胰臟癌。例如癌可為於接受先前處置後仍在進展之胰臟癌。例如可為於先前處置開始後約1、2、3、4、5、6、7、8、9、10、11或12個月以內之某一時期仍在進展之胰臟癌。於另一實施形態中,例如可為自先前處置開始起經過約3、6、9或12個月後之某一時期仍在進展之胰臟癌。於本發明之一實施形態中,胰臟癌對先前處置具有抗藥性或無反應性。於另一實施形態中,胰臟癌為復發性。即,胰臟癌最初對於利用先前處置之處置有應答性,但於先前處置中止,經過約6、7、8、9、10、11、12、24或36個月後之某一時期,癌於原發灶復發、或轉移復發。In one embodiment of the present invention, the pancreatic cancer to be treated in the medicine or the like of the present invention may be pancreatic cancer that is still progressing at the time of previous treatment, during treatment, or after treatment. For example, the cancer may be pancreatic cancer that is still progressing after previous treatment. For example, it may be pancreatic cancer that is still progressing for a period of time within about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 months after the start of the previous treatment. In another embodiment, for example, it may be pancreatic cancer that is still progressing after a period of about 3, 6, 9, or 12 months from the beginning of the previous treatment. In one embodiment of the present invention, the pancreatic cancer is drug-resistant or non-responsive to previous treatments. In another embodiment, the pancreatic cancer is recurrent. That is, pancreatic cancer is initially responsive to treatment with the previous treatment, but after the previous treatment is discontinued, after a certain period of about 6, 7, 8, 9, 10, 11, 12, 24 or 36 months, the cancer becomes Primary tumor recurrence, or metastatic recurrence.

標準療法+ IL-6 抑制劑及 / PD-1/PD-L1 訊號抑制劑本發明提供一種於使用至少一種化學治療劑之標準療法之基礎上使用IL-6抑制劑及PD-1/PD-L1訊號抑制劑之任一者或兩者之併用療法。 Standard therapy + IL-6 inhibitor and / or PD-1/PD-L1 signaling inhibitor The present invention provides an IL-6 inhibitor and PD-1/PD in addition to standard therapy using at least one chemotherapeutic agent - Therapy with either one or a combination of L1 signaling inhibitors.

本發明提供一種用於在胰臟癌之化學療法中提高化學治療劑之效果的含有抗IL-6R抗體與抗PD-L1抗體之醫藥組合物或套組。The present invention provides a pharmaceutical composition or kit containing an anti-IL-6R antibody and an anti-PD-L1 antibody for enhancing the effect of a chemotherapeutic agent in pancreatic cancer chemotherapy.

於上述醫藥組合物或套組中,胰臟癌為對化學治療劑具有先天抗藥性、或對化學治療劑產生後天抗藥性之胰臟癌,特徵在於藉由將化學治療劑與抗IL-6R抗體及抗PD-L1抗體組合使用而提高投予組之有效率。In the above pharmaceutical composition or set, pancreatic cancer is pancreatic cancer with innate resistance to chemotherapeutic agents or acquired resistance to chemotherapeutic agents, characterized by combining the chemotherapeutic agents with anti-IL-6R Antibody and anti-PD-L1 antibody were used in combination to improve the effectiveness of the administration group.

又,本發明提供一種用於處置需要進行投予化學治療劑後復發之胰臟癌之處置的個體之胰臟癌的含有抗IL-6R抗體與抗PD-L1抗體之醫藥組合物或套組。Furthermore, the present invention provides a pharmaceutical composition or kit containing an anti-IL-6R antibody and an anti-PD-L1 antibody for treating pancreatic cancer in an individual who needs treatment for pancreatic cancer that has recurred after administration of a chemotherapeutic agent .

投予之方法本發明之醫藥之投予量及投予排程根據投予對象、對象之年齡、體重、疾病之狀態、投予方法等而異,業者可依據製造廠商之說明書,結合經驗而定。例如於時間差投予計劃中,可於投予有效量之第二醫藥的1小時前、2小時前、3小時前、4小時前、5小時前、6小時前、7小時前、8小時前、9小時前、10小時前、11小時前、1天前、2天前、3天前、4天前、5天前、6天前、7天前或8天前,投予有效量之第一醫藥。進而,例如可於投予有效量之第三醫藥的1小時前、2小時前、3小時前、4小時前、5小時前、6小時前、7小時前、8小時前、9小時前、10小時前、11小時前、1天前、2天前、3天前、4天前、5天前、6天前、7天前或8天前,投予有效量之第二醫藥。進而,例如可於投予有效量之第四醫藥的1小時前、2小時前、3小時前、4小時前、5小時前、6小時前、7小時前、8小時前、9小時前、10小時前、11小時前、1天前、2天前、3天前、4天前、5天前、6天前、7天前或8天前,投予有效量之第三醫藥。 Method of Administration The dosage and schedule of administration of the medicine of the present invention vary according to the subject to be administered, the subject's age, weight, disease state, and the method of administration. Certainly. For example, in the time difference administration plan, it can be administered 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, and 8 hours before the effective dose of the second medicine is administered. , 9 hours, 10 hours, 11 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or 8 days ago, administer an effective amount of First Medicine. Furthermore, for example, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, Administer an effective amount of the second medicine 10 hours ago, 11 hours ago, 1 day ago, 2 days ago, 3 days ago, 4 days ago, 5 days ago, 6 days ago, 7 days ago, or 8 days ago. Furthermore, for example, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, Administer an effective amount of the third medicine 10 hours ago, 11 hours ago, 1 day ago, 2 days ago, 3 days ago, 4 days ago, 5 days ago, 6 days ago, 7 days ago or 8 days ago.

作為非限定之例示,就基於對診斷為胰臟癌之人進行治療之目的,IL-6抑制劑為托珠單抗、PD-1/PD-L1訊號抑制劑為阿替利珠單抗之情形時的本發明之醫藥之投予量及投予間隔進行說明。As a non-limiting example, for the purpose of treating a person diagnosed with pancreatic cancer, the IL-6 inhibitor is tocilizumab, and the PD-1/PD-L1 signaling inhibitor is atezolizumab The dosage and administration interval of the medicine of the present invention in this case will be described.

托珠單抗之通常投予量例如可根據投予路徑,於平均1天約10 ng/kg~約100 mg/kg(體重)、較佳為約1 mg/kg/天~10 mg/kg/天之範圍內變動。例如托珠單抗之投予量可為約0.1~100 mg/kg/週或顯示同等血中濃度之投予量,較佳為1~50 mg/kg/週或顯示同等血中濃度之投予量,更佳為5~10 mg/kg/週或顯示同等血中濃度之投予量。投予次數可採用一種投予形態或複數種不同之投予形態。例如可間隔1週、間隔2週、間隔3週、或間隔4週,根據投予對象患者之狀態,投予固定量或變動量。或可根據投予對象患者之狀態,縮短或延長投予間隔而投予固定量。The usual dose of tocilizumab can be, for example, according to the route of administration, about 10 ng/kg to about 100 mg/kg (body weight) per day on average, preferably about 1 mg/kg/day to 10 mg/kg Changes within the range of /day. For example, the dosage of tocilizumab can be about 0.1-100 mg/kg/week or the dosage showing the same blood concentration, preferably 1-50 mg/kg/week or the dosage showing the same blood concentration The dosage is more preferably 5-10 mg/kg/week or the dosage showing the same blood concentration. The number of injections may be one type of injection or a plurality of different types of injection. For example, a fixed amount or a variable amount may be administered at intervals of 1 week, 2 weeks, 3 weeks, or 4 weeks, depending on the state of the patient to be administered. Alternatively, a fixed amount may be administered by shortening or extending the administration interval depending on the state of the patient to be administered.

例如於靜脈內投予之情形時,可間隔1週、間隔2週、間隔3週、或間隔4週,根據投予對象患者之狀態,投予固定量或變動量。例如於靜脈滴注之情形時,作為托珠單抗(基因重組),可1次2~24 mg/kg且每隔2~4週投予一次。例如可按照1次4 mg/kg且間隔2週(Q2W)或間隔3週(Q3W)或間隔4週(Q4W)或者1次8 mg/kg且間隔2週(Q2W)或間隔3週(Q3W)或間隔4週(Q4W)之投予量,經由靜脈滴注投予托珠單抗。所謂「1次4 mg/kg且間隔2週(Q2W)之劑量」係指相對於投予對象患者之體重每1 kg,以4 mg作為1個單位劑量,每隔2週投予一次。即,將初次投予日設為第1天時,於第15天進行下一次投予。於皮下投予之情形時,例如作為托珠單抗(基因重組),可1次皮下注射20~350 mg、較佳為50~250 mg、更佳為80~200 mg,每隔1~4週注射一次。For example, in the case of intravenous administration, a fixed amount or a variable amount may be administered at intervals of 1 week, 2 weeks, 3 weeks, or 4 weeks, depending on the state of the patient to be administered. For example, in the case of intravenous drip, as tocilizumab (gene recombination), 2-24 mg/kg can be administered once every 2-4 weeks. For example, 4 mg/kg once with 2-week interval (Q2W) or 3-week interval (Q3W) or 4-week interval (Q4W) or 8 mg/kg once with 2-week interval (Q2W) or 3-week interval (Q3W) ) or at 4-week intervals (Q4W), tocilizumab was administered via intravenous infusion. The so-called "dose of 4 mg/kg once with a 2-week interval (Q2W)" refers to 4 mg as a unit dose per 1 kg of the body weight of the administered subject, administered once every 2 weeks. That is, when the first administration day is set as the first day, the next administration is performed on the 15th day. In the case of subcutaneous administration, for example, as tocilizumab (gene recombination), 20-350 mg, preferably 50-250 mg, more preferably 80-200 mg can be subcutaneously injected once, every 1-4 Inject once a week.

阿替利珠單抗之投予量例如根據投予路徑,可於平均1天約10 ng/kg~約100 mg/kg(體重)、較佳為約1 mg/kg/天~50 mg/kg/天之範圍內變動。例如阿替利珠單抗之一次投予量可為約500~2000 mg之範圍。投予次數取決於投予路徑,可採用一種投予形態或複數種不同之投予形態。The dosage of atezolizumab can be about 10 ng/kg to about 100 mg/kg (body weight), preferably about 1 mg/kg/day to 50 mg/day, on average, depending on the route of administration. Changes within the range of kg/day. For example, a single dose of atezolizumab may range from about 500 to 2000 mg. The number of injections depends on the injection route, and one type of injection or a plurality of different types of injection may be used.

例如於靜脈內投予之情形時,可間隔1週、間隔2週、間隔3週、或間隔4週,根據投予對象患者之狀態,投予固定量或變動量。例如於靜脈滴注之情形時,作為阿替利珠單抗(基因重組),可1次50~2000 mg且每隔2~4週投予一次。例如可按照1次840 mg且間隔2週(Q2W)或1次1200 mg且間隔3週(Q3W)或1次1680 mg且間隔4週(Q4W)之投予量投予阿替利珠單抗。於按照「1次840 mg且間隔2週」或「1次840 mg且間隔2週」投予之情形時,不考慮體重,1次投予840 mg,將初次投予日設為第1天時,於第15天進行下一次投予。例如於按照1次1200 mg且間隔3週(Q3W)靜脈滴注阿替利珠單抗之情形時,可設為如下投予量:1次歷時約60分鐘投予1200 mg,每隔3週投予一次。For example, in the case of intravenous administration, a fixed amount or a variable amount may be administered at intervals of 1 week, 2 weeks, 3 weeks, or 4 weeks, depending on the state of the patient to be administered. For example, in the case of intravenous drip, as atezolizumab (gene recombination), 50-2000 mg can be administered once every 2-4 weeks. For example, atezolizumab can be administered at a dose of 840 mg once with a 2-week interval (Q2W) or 1200 mg once with a 3-week interval (Q3W) or 1680 mg once with a 4-week interval (Q4W) . In the case of "840 mg once with a 2-week interval" or "840 mg once with a 2-week interval", regardless of body weight, 840 mg is administered once, and the first administration day is set as the first day , the next dose was on the 15th day. For example, in the case of intravenous infusion of atezolizumab at a 3-week interval (Q3W) at 1,200 mg once, the dose can be set as follows: 1,200 mg once every 60 minutes, every 3 weeks Give once.

作為一形態,於將含有阿替利珠單抗之醫藥與托珠單抗、吉西他濱及白蛋白結合紫杉醇加以組合用於治療人類個體之胰臟癌之情形時,可將2~4週作為一個週期,按照以下之劑量進行投予。 ・於各週期之第1天靜脈滴注托珠單抗2~10 mg/kg; ・於各週期之第1天靜脈滴注阿替利珠單抗400~2400 mg; ・於各週期中,按照依據標準療法之投予量與投予間隔,複數次投予白蛋白結合紫杉醇及鹽酸吉西他濱。關於鹽酸吉西他濱,例如成人按照以吉西他濱計1次1000 mg/m 2(體表面積)或1次750 mg/m 2(體表面積),歷時30分鐘靜脈滴注,每週投予1次,連續投予3週在第4週停藥。可將其作為一個投予過程反覆實施。再者,根據患者之狀態適當減量。又,關於白蛋白結合紫杉醇,例如成人按照以紫杉醇計1天1次125 mg/m 2(體表面積)或1天1次100 mg/m 2(體表面積),歷時30分鐘靜脈滴注,每週投予1次,連續投予3週,第4週停藥。可將其作為一個投予過程反覆實施。再者,根據患者之狀態適當減量。 As a form, 2 to 4 weeks can be used as an Period, according to the following doses administered.・Intravenous infusion of tocilizumab 2~10 mg/kg on the first day of each cycle; ・Intravenous infusion of atezolizumab 400~2400 mg on the first day of each cycle; ・In each cycle, Nab-paclitaxel and gemcitabine hydrochloride were administered in multiple doses according to the dose and interval of administration according to standard therapy. With regard to gemcitabine hydrochloride, for example, for adults, 1000 mg/m 2 (body surface area) or 750 mg/m 2 (body surface area) once, as an intravenous infusion over 30 minutes, administered once a week, continuously administered Give 3 weeks and stop at week 4. It can be implemented iteratively as an administration process. Furthermore, the dosage should be appropriately reduced according to the patient's condition. Also, with regard to albumin-bound paclitaxel, for example, for adults, 125 mg/m 2 (body surface area) once a day or 100 mg/m 2 (body surface area) once a day is administered intravenously over 30 minutes, every time It was administered once a week for 3 consecutive weeks, and the drug was discontinued in the 4th week. It can be implemented iteratively as an administration process. Furthermore, the dosage should be appropriately reduced according to the patient's condition.

作為一形態,於將含有阿替利珠單抗之醫藥與托珠單抗、吉西他濱及白蛋白結合紫杉醇加以組合用於治療人類個體之胰臟癌之情形時,可藉由包括按照如下劑量對個體進行投予之操作之方法使用: (i)托珠單抗之劑量為1次8 mg/kg且間隔4週(Q4W);及阿替利珠單抗之劑量選自由1次840 mg且間隔2週(Q2W),1次1200 mg且間隔3週(Q3W),1次1680 mg且間隔4週(Q4W)所組成之群中;或者 (ii)托珠單抗之劑量為1次4 mg/kg且間隔2週(Q2W);及阿替利珠單抗之劑量選自由1次840 mg且間隔2週(Q2W),1次1200 mg且間隔3週(Q3W),1次1680 mg且間隔4週(Q4W)所組成之群中;或者可按照如下劑量對於對象之人類個體進行投予: (iii)托珠單抗之劑量為1次8 mg/kg且間隔2週(Q2W);及阿替利珠單抗之劑量選自由1次840 mg且間隔2週(Q2W),1次1200 mg且間隔3週(Q3W),1次1680 mg且間隔4週(Q4W)所組成之群中。 As a form, in the case of combining a drug containing atezolizumab with tocilizumab, gemcitabine, and nab-paclitaxel for the treatment of pancreatic cancer in a human subject, it can be treated by including: The method of the operation of the individual to perform the administration uses: (i) The dose of tocilizumab is 8 mg/kg once with a 4-week interval (Q4W); and the dose of atezolizumab is selected from 840 mg once with a 2-week interval (Q2W), 1200 mg once mg with a 3-week interval (Q3W) and 1680 mg once with a 4-week interval (Q4W); or (ii) The dose of tocilizumab is 4 mg/kg once with 2 weeks interval (Q2W); and the dose of atezolizumab is selected from 840 mg once with 2 weeks interval (Q2W), 1200 mg once mg at 3-week intervals (Q3W), 1680 mg once at 4-week intervals (Q4W), or administered to human subjects at the following doses: (iii) The dose of tocilizumab is 8 mg/kg once with 2 weeks interval (Q2W); and the dose of atezolizumab is selected from 840 mg once with 2 weeks interval (Q2W), 1200 mg once mg with a 3-week interval (Q3W) and 1680 mg once with a 4-week interval (Q4W).

進而,白蛋白結合紫杉醇及鹽酸吉西他濱可依據標準療法組合投予。即,可於上述任一種托珠單抗之用法、劑量及阿替利珠單抗之用法、劑量上組合白蛋白結合紫杉醇及鹽酸吉西他濱之投予。例如可按照如下用法劑量對於對象之人類個體進行投予: (iv)托珠單抗之劑量為1次8 mg/kg且間隔4週(Q4W);及阿替利珠單抗之劑量為1次1680 mg且間隔4週(Q4W);及連續3週每週投予一次白蛋白結合紫杉醇與鹽酸吉西他濱,第4週停藥,白蛋白結合紫杉醇之劑量以紫杉醇計1次125 mg/m 2(體表面積)、鹽酸吉西他濱之劑量以吉西他濱計1次1000 mg/m 2(體表面積)。 Furthermore, nab-paclitaxel and gemcitabine hydrochloride can be administered in combination according to standard therapy. That is, the administration of nab-paclitaxel and gemcitabine hydrochloride can be combined with the usage and dosage of any of the above-mentioned tocilizumab and the usage and dosage of atezolizumab. For example, the following dosages can be administered to a subject human subject: (iv) tocilizumab at a dose of 8 mg/kg once at 4-week intervals (Q4W); and atezolizumab at a dose of 1 1680 mg once a week with an interval of 4 weeks (Q4W); and once a week for 3 consecutive weeks, nab-paclitaxel and gemcitabine hydrochloride, discontinued in the 4th week, and the dose of nab-paclitaxel is 125 mg/m 2 once per paclitaxel (body surface area), the dose of gemcitabine hydrochloride is calculated as 1000 mg/m 2 (body surface area) once with gemcitabine.

作為一形態,於將含有托珠單抗之醫藥與阿替利珠單抗、吉西他濱及白蛋白結合紫杉醇加以組合用於治療人類個體之癌之情形時,可按照如下劑量對於對象之人類個體進行投予: (i)托珠單抗之劑量為1次8 mg/kg且間隔4週(Q4W);及阿替利珠單抗之劑量係選自由1次840 mg且間隔2週(Q2W)、1次1200 mg且間隔3週(Q3W)、1次1680 mg且間隔4週(Q4W)所組成之群;或者 (ii)托珠單抗之劑量為1次4 mg/kg且間隔4週(Q4W);及阿替利珠單抗之劑量係選自由1次840 mg且間隔2週(Q2W)、1次1200 mg且間隔3週(Q3W)、1次1680 mg且間隔4週(Q4W)所組成之群; (iii)托珠單抗之劑量為1次8 mg/kg且間隔2週(Q2W);及阿替利珠單抗之劑量係選自由1次840 mg且間隔2週(Q2W)、1次1200 mg且間隔3週(Q3W)、1次1680 mg且間隔4週(Q4W)所組成之群。 As a form, in the case of combining a medicine containing tocilizumab with atezolizumab, gemcitabine, and nab-paclitaxel for the treatment of cancer in a human subject, the following doses may be administered to the subject human subject Throw in: (i) The dose of tocilizumab is 8 mg/kg once with a 4-week interval (Q4W); and the dose of atezolizumab is selected from 840 mg once with a 2-week interval (Q2W), once A cohort of 1200 mg at 3-week intervals (Q3W) and 1680 mg once at 4-week intervals (Q4W); or (ii) the dose of tocilizumab is 4 mg/kg once with a 4-week interval (Q4W); and the dose of atezolizumab is selected from 840 mg once with a 2-week interval (Q2W), once A group consisting of 1200 mg at 3-week intervals (Q3W) and 1680 mg once at 4-week intervals (Q4W); (iii) the dose of tocilizumab is 8 mg/kg once with a 2-week interval (Q2W); and the dose of atezolizumab is selected from 840 mg once with a 2-week interval (Q2W), once A group consisting of 1200 mg at 3-week intervals (Q3W) and 1680 mg once at 4-week intervals (Q4W).

進而,白蛋白結合紫杉醇及鹽酸吉西他濱可依據標準療法組合投予。即,可於上述任一種托珠單抗之用法、劑量及阿替利珠單抗之用法、劑量上組合白蛋白結合紫杉醇及鹽酸吉西他濱之投予。例如可按照如下用法劑量對於對象之人類個體進行投予: (iv)托珠單抗之劑量為1次8 mg/kg且間隔4週(Q4W);及阿替利珠單抗之劑量為1次1680 mg且間隔4週(Q4W);及連續3週每週投予一次白蛋白結合紫杉醇與鹽酸吉西他濱,第4週停藥,白蛋白結合紫杉醇之劑量以紫杉醇計1次125 mg/m 2(體表面積)、鹽酸吉西他濱之劑量以吉西他濱計1次1000 mg/m 2(體表面積)。 Furthermore, nab-paclitaxel and gemcitabine hydrochloride can be administered in combination according to standard therapy. That is, the administration of nab-paclitaxel and gemcitabine hydrochloride can be combined with the usage and dosage of any of the above-mentioned tocilizumab and the usage and dosage of atezolizumab. For example, the following dosages can be administered to a subject human subject: (iv) tocilizumab at a dose of 8 mg/kg once at 4-week intervals (Q4W); and atezolizumab at a dose of 1 1680 mg once a week with an interval of 4 weeks (Q4W); and once a week for 3 consecutive weeks, nab-paclitaxel and gemcitabine hydrochloride, discontinued in the 4th week, and the dose of nab-paclitaxel is 125 mg/m 2 once per paclitaxel (body surface area), the dose of gemcitabine hydrochloride is calculated as 1000 mg/m 2 (body surface area) once with gemcitabine.

作為一形態,於將含有阿替利珠單抗之醫藥與托珠單抗、吉西他濱及白蛋白結合紫杉醇加以組合用於治療人類個體之胰臟癌之情形時,可將28天(4週)作為一個週期,按照以下之劑量進行投予。 ・於各週期之第1天靜脈滴注托珠單抗4 mg/kg或8 mg/kg; ・於各週期之第1天靜脈滴注阿替利珠單抗1200 mg或1680 mg; 此處,可於距托珠單抗靜脈滴注結束經過數小時後(例如約2小時後、約3小時後、約4小時後)投予阿替利珠單抗。 ・於各週期之第1、8及15天(於各週期之第1天注入阿替利珠單抗結束後)靜脈滴注以紫杉醇計125 mg/m 2之白蛋白結合紫杉醇; ・於各週期之第1、8及15天(於各週期之第1天注入白蛋白結合紫杉醇結束後)靜脈滴注以吉西他濱計1000 mg/m 2之鹽酸吉西他濱。 As a modality, 28 days (4 weeks) of atezolizumab-containing medicine in combination with tocilizumab, gemcitabine, and nab-paclitaxel for the treatment of pancreatic cancer in human subjects As a cycle, the following doses are administered.・Intravenous tocilizumab 4 mg/kg or 8 mg/kg on day 1 of each cycle; ・Intravenous infusion of atezolizumab 1200 mg or 1680 mg on day 1 of each cycle; here , atezolizumab may be administered several hours after the end of the intravenous infusion of tocilizumab (eg, after about 2 hours, after about 3 hours, after about 4 hours).・Intravenous infusion of 125 mg/m 2 of nab-paclitaxel based on paclitaxel on days 1, 8 and 15 of each cycle (after the end of the infusion of atezolizumab on day 1 of each cycle); ・In each cycle On days 1, 8 and 15 of the cycle (after the end of the infusion of nab-paclitaxel on day 1 of each cycle), 1000 mg/m 2 of gemcitabine hydrochloride was infused intravenously.

作為一形態,於將含有阿替利珠單抗之醫藥與托珠單抗、吉西他濱及白蛋白結合紫杉醇加以組合用於治療人類個體之胰臟癌之情形時,可將21天(3週)作為一個週期,按照以下之劑量進行投予。 ・於各週期之第1天靜脈滴注托珠單抗4 mg/kg或8 mg/kg; ・於各週期之第1天靜脈滴注阿替利珠單抗840 mg或1200 mg; 此處,可於距托珠單抗靜脈滴注結束經過數小時後(例如約2小時後、約3小時後、約4小時後)投予阿替利珠單抗。 ・於各週期之第1、8及15天(於各週期之第1天注入阿替利珠單抗結束後)靜脈滴注以紫杉醇計125 mg/m 2之白蛋白結合紫杉醇; ・於各週期之第1、8及15天(於各週期之第1天注入白蛋白結合紫杉醇結束後)靜脈滴注以吉西他濱計1000 mg/m 2之鹽酸吉西他濱。 As a form, in the case of combining atezolizumab-containing medicines with tocilizumab, gemcitabine, and nab-paclitaxel for the treatment of pancreatic cancer in human subjects, 21 days (3 weeks) As a cycle, the following doses are administered.・Intravenous tocilizumab 4 mg/kg or 8 mg/kg on day 1 of each cycle; ・Intravenous infusion of atezolizumab 840 mg or 1200 mg on day 1 of each cycle; here , atezolizumab may be administered several hours after the end of the intravenous infusion of tocilizumab (eg, after about 2 hours, after about 3 hours, after about 4 hours).・Intravenous infusion of 125 mg/m 2 of nab-paclitaxel based on paclitaxel on days 1, 8 and 15 of each cycle (after the end of the infusion of atezolizumab on day 1 of each cycle); ・In each cycle On days 1, 8 and 15 of the cycle (after the end of the infusion of nab-paclitaxel on day 1 of each cycle), 1000 mg/m 2 of gemcitabine hydrochloride was infused intravenously.

作為一形態,於將含有阿替利珠單抗之醫藥與托珠單抗、吉西他濱及白蛋白結合紫杉醇加以組合用於治療人類個體之胰臟癌之情形時,可將14天(2週)作為一個週期,按照以下之劑量進行投予。 ・於各週期之第1天靜脈滴注托珠單抗4 mg/kg或8 mg/kg; ・於各週期之第1天靜脈滴注阿替利珠單抗840 mg; 此處,可於距托珠單抗靜脈滴注結束經過數小時後(例如約2小時後、約3小時後、約4小時後)投予阿替利珠單抗。 ・於各週期之第1及8天(於各週期之第1天注入阿替利珠單抗結束後)靜脈滴注以紫杉醇計125 mg/m 2之白蛋白結合紫杉醇; ・於各週期之第1及8天(於各週期之第1天注入白蛋白結合紫杉醇結束後)靜脈滴注以吉西他濱計1000 mg/m 2之鹽酸吉西他濱。 As a form, 14 days (2 weeks) can be used in the case of combining a medicine containing atezolizumab with tocilizumab, gemcitabine, and nab-paclitaxel for the treatment of pancreatic cancer in a human subject. As a cycle, the following doses are administered.・Intravenous infusion of tocilizumab 4 mg/kg or 8 mg/kg on day 1 of each cycle; ・Intravenous infusion of atezolizumab 840 mg on day 1 of each cycle; Atezolizumab is administered several hours after the end of the intravenous infusion of tocilizumab (eg, after about 2 hours, after about 3 hours, after about 4 hours).・Intravenous infusion of 125 mg/m 2 of nab-paclitaxel based on paclitaxel on Days 1 and 8 of each cycle (after the end of atezolizumab infusion on Day 1 of each cycle); On days 1 and 8 (after the end of the infusion of nab-paclitaxel on day 1 of each cycle), 1000 mg/m 2 of gemcitabine hydrochloride based on gemcitabine was administered intravenously.

作為另一例,針對用於處置化學治療劑抗藥性之癌、或用於促進化學治療劑向癌組織之滲透、或用於促進CD45陽性細胞向靶組織之浸潤的托珠單抗之用途進行說明。As another example, the use of tocilizumab for the treatment of chemotherapeutic agent-resistant cancer, or for promoting the penetration of chemotherapeutic agents into cancer tissues, or for promoting the infiltration of CD45-positive cells into target tissues, will be described. .

於一形態中,含有托珠單抗之醫藥係用於在癌症之處置中與化學治療劑組合投予,以提高該化學治療劑之效果,化學治療劑與IL-6抑制劑較佳為同時投予、分開投予、或連續投予。In one form, the drug containing tocilizumab is administered in combination with a chemotherapeutic agent in the treatment of cancer to enhance the effect of the chemotherapeutic agent, preferably the chemotherapeutic agent and the IL-6 inhibitor are simultaneously administered Dosing, separate dosing, or continuous dosing.

於一形態中,含有托珠單抗之醫藥係用於處置對化學治療劑具有抗藥性之癌,較佳為於包含需要進行對化學治療劑具有抗藥性之癌之處置的個體之群組(患者群體)中,相較於不與托珠單抗組合而投予化學治療劑之情形,提高群組中之緩解率或病情控制率。In one form, the drug containing tocilizumab is used for the treatment of chemotherapeutic-resistant cancers, preferably in a group comprising individuals in need of treatment of chemotherapeutic-resistant cancers ( patient population), the remission rate or disease control rate in the cohort was improved compared to the case where the chemotherapeutic agent was administered without tocilizumab.

於一形態中,含有托珠單抗之醫藥係與化學治療劑組合或與化學治療劑併用,而用於處置對化學治療劑具有抗藥性之癌,化學治療劑與托珠單抗同時投予、分開投予、或連續投予。In one form, the medicine containing tocilizumab is used in combination with or in combination with a chemotherapeutic agent to treat cancer resistant to the chemotherapeutic agent, and the chemotherapeutic agent and tocilizumab are administered simultaneously , separate administration, or continuous administration.

上述情形時,較佳為於包含需要處置對於化學治療劑具有抗藥性之癌症的個體之群組中,相較於不與托珠單抗組合而投予化學治療劑之情形,提高群組中之緩解率或病情控制率。In the above case, it is preferable to increase the number of patients in the group including the individual in need of treatment of cancer resistant to the chemotherapeutic agent compared to the case where the chemotherapeutic agent is administered without tocilizumab. rate of remission or disease control.

於上述情形時, (a)上述包含需要處置對於化學治療劑具有抗藥性之癌症的個體之群組包含在投予化學治療劑之前未藉由診斷來檢測有無抗藥性之個體;或 (b)上述對化學治療劑具有抗藥性之癌症可為對化學治療劑具有先天抗藥性之癌症;或 (c)上述對化學治療劑具有抗藥性之癌症可為對化學治療劑產生後天抗藥性之癌症;或 (d)上述對化學治療劑具有抗藥性之癌症可為投予化學治療劑後復發之癌症。 In the above situation, (a) the aforementioned group comprising individuals in need of treatment of chemotherapeutic-resistant cancers comprises individuals who were not tested for resistance by diagnosis prior to administration of the chemotherapeutic agent; or (b) the above-mentioned cancer that is resistant to a chemotherapeutic agent may be a cancer that is innately resistant to a chemotherapeutic agent; or (c) the above-mentioned cancers that are resistant to chemotherapeutic agents may be cancers that have acquired resistance to chemotherapeutic agents; or (d) The above-mentioned cancer resistant to a chemotherapeutic agent may be a cancer that recurs after administration of the chemotherapeutic agent.

於上述情形時,托珠單抗之通常投予量例如取決於投予路徑,並可以在每天約10 ng/kg~約100 mg/kg(體重)、較佳為約1 mg/kg/天~10 mg/kg/天之範圍內變動。例如托珠單抗之投予量可為約0.1~100 mg/kg/週或顯示同等血中濃度之投予量,較佳為1~50 mg/kg/週或顯示同等血中濃度之投予量,更佳為5~10 mg/kg/週或顯示同等血中濃度之投予量。投予次數可採用一種投予形態或複數種不同之投予形態。例如可間隔1週、間隔2週、間隔3週、或間隔4週,根據投予對象患者之狀態,投予固定量或變動量。或可根據投予對象患者之狀態,縮短或延長投予間隔而投予固定量。In the above case, the usual dose of tocilizumab depends on, for example, the route of administration, and can range from about 10 ng/kg to about 100 mg/kg (body weight) per day, preferably about 1 mg/kg/day. Varies within the range of ~10 mg/kg/day. For example, the dosage of tocilizumab can be about 0.1-100 mg/kg/week or the dosage showing the same blood concentration, preferably 1-50 mg/kg/week or the dosage showing the same blood concentration The dosage is more preferably 5-10 mg/kg/week or the dosage showing the same blood concentration. The number of injections may be one type of injection or a plurality of different types of injection. For example, a fixed amount or a variable amount may be administered at intervals of 1 week, 2 weeks, 3 weeks, or 4 weeks, depending on the state of the patient to be administered. Alternatively, a fixed amount may be administered by shortening or extending the administration interval depending on the state of the patient to be administered.

例如於靜脈滴注之情形時,作為托珠單抗(基因重組),可1次4~12 mg/kg且每隔2~4週投予一次。例如關於托珠單抗之投予量,可以按照1次4 mg/kg且間隔2週(Q2W)或間隔3週(Q3W)或間隔4週(Q4W)或者1次8 mg/kg且間隔2週(Q2W)或間隔3週(Q3W)或間隔4週(Q4W)之投予量,藉由靜脈滴注進行投予。例如於皮下投予之情形時,作為托珠單抗(基因重組),可1次162 mg且每隔1~2週皮下注射一次。For example, in the case of intravenous drip, as tocilizumab (gene recombination), 4-12 mg/kg can be administered once every 2-4 weeks. For example, the dosage of tocilizumab can be 4 mg/kg once with 2-week interval (Q2W) or 3-week interval (Q3W) or 4-week interval (Q4W) or 8 mg/kg once with 2-week interval Weekly (Q2W) or 3-weekly (Q3W) or 4-weekly (Q4W) doses are administered by intravenous infusion. For example, in the case of subcutaneous administration, as tocilizumab (gene recombination), 162 mg can be administered subcutaneously once every 1 to 2 weeks.

又,托珠單抗例如可經由靜脈滴注,對於人類對象,每次以約0.1 mg至約30,000 mg之投予量進行投予,更具體而言,每次可投予約1 mg以上且約10 mg以下、約80 mg以下、約240 mg以下、約840 mg以下、約1,000 mg以下或約1,200 mg以下,更具體而言,每次可投予約10 mg、約80 mg、約240 mg、約840 mg或約1,200 mg。或者每次可投予約1.0 mg/kg(體重)以上、約2.0 mg/kg(體重)以上、約2.5 mg/kg(體重)以上、約10 mg/kg(體重)以上、或約20 mg/kg(體重)以上且約100 mg/kg(體重)以下,更具體而言,每次可投予約2 mg/kg(體重)、約10 mg/kg(體重)、約15 mg/kg(體重)或約20 mg/kg(體重)。並且,該等投予量例如可每隔約2週或約2週以上、或者每隔約3週或約3週以上,投予一次。又,每次投予之投予時間於保證安全投予之範圍內並無限定,較佳為約30分鐘至約90分鐘。Also, tocilizumab can be administered, for example, via intravenous infusion, for a human subject, in an administration amount of about 0.1 mg to about 30,000 mg per administration, more specifically, about 1 mg or more and about 1 mg per administration. 10 mg or less, about 80 mg or less, about 240 mg or less, about 840 mg or less, about 1,000 mg or less, or about 1,200 mg or less, more specifically, about 10 mg, about 80 mg, about 240 mg, About 840 mg or about 1,200 mg. Or more than about 1.0 mg/kg (body weight), about 2.0 mg/kg (body weight) or more, about 2.5 mg/kg (body weight) or more, about 10 mg/kg (body weight) or more, or about 20 mg/kg (body weight) or more per administration. kg (body weight) or more and about 100 mg/kg (body weight) or less, more specifically, about 2 mg/kg (body weight), about 10 mg/kg (body weight), about 15 mg/kg (body weight) can be administered each time ) or about 20 mg/kg (body weight). In addition, these doses can be administered, for example, once every about 2 weeks or more, or once every about 3 weeks or more. In addition, the injection time of each injection is not limited within the range of ensuring safe injection, and it is preferably about 30 minutes to about 90 minutes.

如上所述,業者可根據該實施形態確定托珠單抗之投予量及投予排程,對於人類對象,每次投予之投予量約為1,200 mg,可每隔約1~4週投予一次,較佳為每隔約2~3週投予一次。As mentioned above, the manufacturer can determine the dosage and schedule of tocilizumab according to the embodiment. For human subjects, the dosage for each administration is about 1,200 mg, which can be administered at intervals of about 1 to 4 weeks. It is administered once, preferably every about 2 to 3 weeks.

關於托珠單抗之投予,業者可根據治療效果、患者之病情、耐受性等,適當調整投予時間、投予間隔、投予次數、投予期間。 於本發明之醫藥等中,投予托珠單抗,亦可進而組合投予一種或複數種化學治療劑。於該情形時,業者可根據治療效果、患者之病情、耐受性等,適當調整托珠單抗及一種或複數種化學治療劑之投予時間、投予間隔、投予次數、投予期間。 Regarding the administration of tocilizumab, the manufacturer can appropriately adjust the administration time, administration interval, administration frequency, and administration period according to the therapeutic effect, the patient's condition, and tolerance. In the medicine and the like of the present invention, tocilizumab can also be administered in combination with one or more chemotherapeutic agents. In this case, the practitioner can appropriately adjust the administration time, administration interval, administration frequency, and administration period of tocilizumab and one or more chemotherapeutic agents according to the therapeutic effect, the patient's condition, tolerance, etc. .

實施例以下,藉由實施例具體地說明本發明,但本發明並不限定於該等實施例。 EXAMPLES Hereinafter, although an Example demonstrates this invention concretely, this invention is not limited to these Examples.

本發明中採用之基準及定義如下所示。 病期分類基準 病期分類(Staging)基於「UICC-TNM 第8版」胰臟惡性外分泌腫瘤之病期分類。 The criteria and definitions used in the present invention are as follows. Disease classification criteria Stage classification (Staging) is based on the stage classification of malignant exocrine pancreatic tumors of the "UICC-TNM 8th Edition".

[表1] 病期分類 內容 IA 癌之長徑2 cm以下 IB 癌之長徑2 cm以上4 cm以下 IIA 癌之長徑4 cm以上 IIB 無遠處轉移性,但向所屬淋巴節之轉移個數有1〜3個 III 無遠處轉移性,但向所屬淋巴節之轉移個數有1〜3個、或者癌 浸潤至腹腔動脈、腸系膜上動脈、或總肝動脈 IV 有遠處轉移性 [Table 1] Disease classification content IA Cancer with a length of 2 cm or less IB Cancer with a long diameter of 2 cm or more and less than 4 cm IIA Cancer with a length of 4 cm or more IIB No distant metastases, but the number of metastases to the associated lymph nodes is 1 to 3 III No distant metastasis, but the number of metastases to the associated lymph node is 1 to 3, or the cancer has infiltrated into the celiac artery, superior mesenteric artery, or common hepatic artery IV have distant metastases

日常體能狀態(Performance Status)(ECOG(Eastern Cooperative Oncology Group,美國東岸癌症臨床研究合作組織)分類) ECOG之日常體能狀態(39)之日文版引用自JCOG(日本臨床腫瘤學小組,Japan Clinical Oncology Group)網頁(http://www.jcog.jp/)。 Daily Performance Status (ECOG (Eastern Cooperative Oncology Group, East Coast Cancer Clinical Research Collaborative) classification) The Japanese version of ECOG's Daily Performance Status (39) is cited from the JCOG (Japan Clinical Oncology Group) web page (http://www.jcog.jp/).

[表2] PS 內容 0 活動完全不存在問題。與發病前一樣,日常生活之進行不受影響。 1 劇烈運動受到影響,但能夠走動,能夠從事較輕鬆之工作或無需站立之工作。例如簡單之家務、辦公 2 能夠走動,生活完全自理,但無法從事工作。一天50%以上之時間於床外度過。 3 生活部分自理。一天50%以上之時間於床上或椅子上度過。 4 完全不能活動。生活完全無法自理。全天於床上或椅子上度過。 [Table 2] PS content 0 There is absolutely no problem with the activity. As before the onset of the disease, the conduct of daily life is not affected. 1 Vigorous exercise is affected, but is able to walk and perform lighter tasks or tasks that do not require standing. such as simple housework, office 2 Able to move around and take care of herself completely, but unable to work. More than 50% of the day is spent outside the bed. 3 Part of life takes care of itself. More than 50% of the day is spent in bed or chair. 4 Completely inactive. Life is completely out of control. Spend the day in bed or in a chair.

卡氏體能狀態(Karonofsky performance status)(40)之日文版引用自腦腫瘤診療指南(https://www.jsn-o.com/guideline2016/index.html)。The Japanese version of Karonofsky performance status (40) is cited from the Guidelines for the Diagnosis and Treatment of Brain Tumors (https://www.jsn-o.com/guideline2016/index.html).

[表3] 100% 正常,無臨床症狀 90% 有輕微之臨床症狀,但能夠正常活動 80% 有明顯之臨床症狀,勉強能夠正常活動 70% 能夠自我照顧,但無法正常活動、勞動 60% 有基本之自理能力,但經常需要介入幫助    50% 根據病情而可能需要看護及定期之醫療行為 40% 無法活動,需要適當之醫療及看護 30% 完全無法活動,需要住院,但不會很快死亡 20% 病情非常嚴重,必須住院且進行積極之治療 10% 臨近死亡 0% 死亡 [table 3] 100% normal, no clinical symptoms 90% Mild clinical symptoms but able to function normally 80% Have obvious clinical symptoms, barely able to perform normal activities 70% Able to take care of themselves, but unable to perform normal activities and work 60% Has basic self-care skills, but often requires intervention 50% Nursing care and periodic medical actions may be required depending on the condition 40% Inability to move, requiring appropriate medical and nursing care 30% Completely immobile, requiring hospitalization, but not dying soon 20% Very serious condition requiring hospitalization and aggressive treatment 10% near death 0% die

不良事件之事件名及分級(Grade)採用「不良事件通用術語標準v4.0日文JCOG版(Common Terminology Criteria for Adverse Events(CTCAE)v4.0-JCOG)[對應於CTCAE v4.03/MedDRA v12.0]」。The event name and grade (Grade) of adverse events adopt "Common Terminology Criteria for Adverse Events (CTCAE) v4.0-JCOG) [corresponding to CTCAE v4.03/MedDRA v12. 0]".

腫瘤縮小效果判定採用「實體癌療效評估新標準(RECIST標準)修訂版1.1-日語JCOG版-:Revised RECIST guideline(version 1.1)」。The tumor shrinkage effect was determined using the "New Response Evaluation Criteria for Solid Cancers (RECIST Criteria) Rev. 1.1-Japanese JCOG version-: Revised RECIST guideline (version 1.1)".

以下為選自「實體癌療效評估新標準(RECIST標準)修訂版1.1-日語JCOG版-:Revised RECIST guideline(version 1.1)」之摘錄。The following is an excerpt from the "New Response Evaluation Criteria in Solid Cancers (RECIST Criteria) Rev. 1.1-Japanese JCOG Edition-: Revised RECIST guideline (version 1.1)".

1.基線評估 依據檢查排程,藉由胸腹部、骨盆造影CT或腹部、骨盆造影MRI進行入組前之腫瘤性病變之特定,將各種病變分為「可測定病變」與「不可測定病變」。利用CT或MRI之橫切面圖進行腫瘤徑之測量,並非採用基於三維重建圖之矢狀切面或冠狀切面之測量。 採用入組前14天以內之最新圖像檢查進行基線評估。入組後,治療開始前再次進行圖像檢查之情形時採用再次檢查之最新圖像檢查。 1. Baseline assessment According to the examination schedule, the tumor lesions before enrollment were identified by chest-abdominal and pelvic angiography CT or abdominal and pelvic angiography MRI, and various lesions were divided into "measurable lesions" and "unmeasurable lesions". The tumor diameter was measured using the cross-section images of CT or MRI, not the sagittal or coronal slices based on the three-dimensional reconstruction images. Baseline assessments were performed using the most recent image examination within 14 days prior to enrollment. After enrollment, the latest image examination of the re-examination shall be used in the case of re-examination of images before the start of treatment.

2.可測定病變之定義 符合以下任一者之病變屬於可測定病變(measurable lesion)。 2. Definition of measurable lesions A lesion meeting any of the following is a measurable lesion.

1)滿足以下任一條件之淋巴節病變以外之病變(非淋巴節病變) ①切片厚5 mm以下之CT或MRI測得之最大徑10 mm以上 ②切片厚超過5 mm之CT或MRI測得之最大徑為切片厚之2倍以上 ③具有滿足①或②之軟組織成分之溶骨性骨轉移病變 ④無其他可測定之非囊腫性病變之情形時之滿足①或②之囊腫性轉移病變 1) Diseases other than lymph node disease (non-lymph node disease) that meet any of the following conditions ①The maximum diameter measured by CT or MRI with a slice thickness of less than 5 mm is more than 10 mm ② The maximum diameter measured by CT or MRI with slice thickness exceeding 5 mm is more than 2 times the slice thickness ③Osteolytic bone metastases with soft tissue components satisfying ① or ② ④ Cystic metastatic lesions satisfying ① or ② when there are no other measurable non-cystic lesions

2)切片厚5 mm以下之CT測得之短徑15 mm以上之淋巴節病變(短徑10 mm以上且未達15 mm之淋巴節病變屬於非目標病變,短徑未達10 mm之淋巴節不屬於病變)2) Lymph node lesions with a short diameter of more than 15 mm measured by CT with a slice thickness of less than 5 mm (lymph node lesions with a short diameter of more than 10 mm and less than 15 mm are non-target lesions, and lymph nodes with a short diameter of less than 10 mm) not a disease)

3)胸部單純X射線照片測得之最大徑20 mm以上、且周圍被肺野包圍(未觸及縱隔或胸壁) 4)可拍攝到可以量度且彩色之照片之最大徑10 mm以上之臨床病變(表淺皮膚病變等) 3) The maximum diameter measured by the pure X-ray of the chest is more than 20 mm, and the surrounding is surrounded by lung fields (without touching the mediastinum or chest wall) 4) Clinical lesions (superficial skin lesions, etc.) with a maximum diameter of 10 mm or more can be photographed in measurable and color photographs

上述以外之所有病變屬於不可測定病變(non-measurable lesion)。 請注意以下之病變與檢查法或病變之大小無關,均屬於不可測定病變。 ・骨病變(具有可測定之軟組織成分之溶骨性病變除外) ・囊腫性病變(上述1)-4)除外) ・有放射線治療等局部治療史之病變 ・軟腦膜病變 ・腹腔、胸腔、心包積液 ・皮膚或肺之淋巴管症 ・可觸知但無法藉由圖像檢查法測定之腹部腫瘤或腹部器官之腫大 All lesions other than the above were classified as non-measurable lesions. Please note that the following lesions are not measurable lesions regardless of the examination method or the size of the lesions. ・Bone lesions (excluding osteolytic lesions with measurable soft tissue components) ・Cystic lesions (except 1)-4) above) ・ Lesions with a history of local treatment such as radiation therapy ・Pial lesions ・Abdominal cavity, pleural cavity, pericardial effusion ・Lymphangiopathy of the skin or lungs ・Abdominal tumors or enlargement of abdominal organs that are palpable but not detectable by imaging methods

3.目標病變之選擇與基線記錄 入組時選擇所確認之可測定病變中之徑(非淋巴節病變:長徑、淋巴節病變:短徑)之大小排在前5位者,每個器官上最多選2處,作為目標病變(target lesion)。選擇時,儘可能將具有可測定病變之器官全部包含在內,並考慮到重複測量時之再現性即易測量性(reproducible repeated measurement)而選擇(避免選擇徑較大但不易測量之病變)。 3. Selection of target lesions and baseline recording When entering the group, select the confirmed measurable lesions (non-lymph node lesions: long diameter, lymph node lesions: short diameter) ranked in the top 5, and select at most 2 locations on each organ as target lesions (target lesion). When selecting, all organs with measurable lesions should be included as much as possible, and the reproducibility during repeated measurement, that is, reproducible repeated measurement, should be considered for selection (avoid selecting lesions with larger diameters that are difficult to measure).

針對所選擇之目標病變,按照從頭側至尾側之順序,於CRF上記錄:部位、檢查法、檢查日、非淋巴節目標病變之長徑、淋巴節目標病變之短徑、及全部目標病變的徑長之和(以下稱為徑和)。For the selected target lesions, record on the CRF in order from cephalad to caudal: site, examination method, examination day, long diameter of non-lymph node target lesions, short diameter of lymph node target lesions, and all target lesions The sum of the diameters and lengths of (hereinafter referred to as the sum of diameters).

4.非目標病變之基線記錄 對於未被選作目標病變之病變,無論能否測定,全部作為非目標病變(non-target lesion),於CRF上記錄病變之部位、檢查方法、檢查日。同一器官內之複數個非目標病變可記錄為1個病變(例如複數個腫大骨盆淋巴節、多發性肝轉移)。 4. Baseline records of non-target lesions For the lesions that are not selected as target lesions, regardless of whether they can be measured or not, they are all regarded as non-target lesions, and the location of the lesions, the inspection method, and the inspection date are recorded on the CRF. Multiple non-target lesions in the same organ can be recorded as one lesion (eg, multiple enlarged pelvic lymph nodes, multiple liver metastases).

5.腫瘤縮小效果之判定 藉由與入組時相同之檢查法,對目標病變及非目標病變進行評估,於CRF上記錄目標病變之徑長、非目標病變之消失或惡化之有無。 5. Determination of tumor shrinkage effect Target lesions and non-target lesions were evaluated by the same examination method as in the group, and the diameter of target lesions and the disappearance or deterioration of non-target lesions were recorded on CRF.

6.目標病變之效果判定標準 ・CR(Complete Response):完全緩解 非淋巴節目標病變全部消失,所有淋巴節目標病變之短徑均未達10 mm之情形。於根據基線選擇淋巴節目標病變之情形時,存在即便徑和未變成0 mm但目標病變之效果亦為CR之情況。 ・PR(Partial Response):部分緩解 目標病變之徑和較基線徑和減少30%以上 ・PD(Progressive Disease):疾病進展 由於某些原因而無法對過程中之最小徑和(於基線為過程中之最小值時,將其作為最小徑和)進行檢查之情形、或無法判定為CR、PR、PD、SD任何一者之情形 徑和之縮小比率=(治療前之徑和-評估時之徑和)/(治療前之徑和)×100% 徑和之增大比率=(評估時之徑和-最小之徑和)/(最小之徑和)×100% 6. Efficacy criteria for target lesions ・CR (Complete Response): Complete remission All non-lymph node target lesions disappeared, and the short diameter of all lymph node target lesions did not reach 10 mm. In the case where the target lesion of the lymph node is selected based on the baseline, there are cases where the effect of the target lesion is CR even if the sum of diameters does not become 0 mm. ・PR (Partial Response): Partial mitigation Target lesion diameter and baseline diameter sum reduced by more than 30% ・PD (Progressive Disease): Disease progression For some reasons, the minimum diameter sum in the process cannot be checked (if the baseline is the minimum value in the process, it is regarded as the minimum diameter sum), or it cannot be determined as any of CR, PR, PD, SD. the situation The reduction ratio of the sum of diameters = (sum of diameters before treatment - sum of diameters at the time of evaluation)/(sum of diameters before treatment) × 100% The increase ratio of the diameter sum = (the diameter sum at the time of evaluation - the smallest diameter sum)/(the smallest diameter sum) × 100%

※只要可測定(例如即便未達5 mm),目標病變之徑長則記錄實測值,於判斷目標病變之徑長「過小而無法測定(too small to measure)」之情形時,與CT之切片厚無關,於判斷腫瘤病變無殘存時記為徑0 mm,於判斷腫瘤病變有殘存時記為徑5 mm。 ※縮小比率滿足PR之條件且同時增大比率滿足PD之條件之情形時判定為PD。 ※治療過程中1個病變發生分離之情形時,將各自之徑長相加作為徑和。 ※治療過程中複數個病變融合而無法辨別邊界之情形時,將融合後之病變之徑長相加作為徑和。於病變彼此相連,但病變之邊界仍可辨別之情形時,將各病變之徑長相加作為徑和。 ※As long as it can be measured (for example, even if it is less than 5 mm), the diameter and length of the target lesion shall be recorded as the measured value. Thickness is irrelevant, and the diameter is 0 mm when it is judged that there is no residual tumor lesion, and the diameter is 5 mm when it is judged that there is a residual tumor lesion. ※When the reduction ratio satisfies the conditions of PR and the enlargement ratio satisfies the conditions of PD at the same time, it is judged as PD. ※In the case of separation of one lesion during the treatment, add up the diameters and lengths of them as the sum of the diameters. ※In the case of multiple lesions fused and the boundary cannot be distinguished during the treatment, the diameter and length of the fused lesions are added together as the diameter sum. When the lesions are connected to each other, but the boundary of the lesions can still be identified, the diameter and length of each lesion are added as the diameter sum.

7.非目標病變之效果判定標準 ・CR(Complete Response):完全緩解 非淋巴節非目標病變全部消失,所有淋巴節非目標病變之短徑未達10 mm之情形。 ・Non-CR/non-PD:非CR/非PD 有1個以上之非目標病變殘存(亦包括淋巴節非目標病變之短徑10 mm以上之殘存)之情形。 ・PD(Progressive Disease):疾病進展 現有之非目標病變「明顯惡化」(包括復發)。 7. Criteria for determining the effect of non-target lesions ・CR (Complete Response): Complete remission All non-lymph node non-target lesions disappeared, and the short diameter of all lymph node non-target lesions did not reach 10 mm. ・Non-CR/non-PD: non-CR/non-PD There is one or more non-target lesions remaining (including the remaining non-target lesions of lymph nodes with a short diameter of more than 10 mm). ・PD (Progressive Disease): Disease progression "Significant deterioration" (including recurrence) of existing non-target lesions.

具有可測定病變之情形:即便目標病變之效果為SD或PR,基於非目標病變之變化而判定「明顯惡化」時,必須觀察到腫瘤量之整體增加值達到足以中止治療之程度的非目標病變之明顯惡化。於目標病變之效果為SD或PR之情形時,將使腫瘤量之減少大幅回升之程度的非目標病變之腫瘤量之增加判定為「明顯惡化」,反之,判定為Non-CR/non-PD。When there is a measurable lesion: Even if the effect of the target lesion is SD or PR, when "significant deterioration" is judged based on the change in the non-target lesion, the non-target lesion must be observed to have an overall increase in tumor volume sufficient to discontinue treatment markedly worsened. When the effect of the target lesion is SD or PR, the increase in the tumor volume of the non-target lesion to the extent that the reduction of the tumor volume can be greatly recovered is determined as "significant deterioration", otherwise, it is determined as Non-CR/non-PD. .

僅具有不可測定病變之情形:將如判斷明顯超過作為標準之相當於徑長增大20%、腫瘤體積增大73%之腫瘤量的非目標病變之增大判定為「明顯惡化」。Only in the case of unmeasurable lesions: If the increase of the non-target lesion significantly exceeds the tumor volume equivalent to a 20% increase in diameter and length and a 73% increase in tumor volume as the standard, it is judged as "significant deterioration".

・NE(Not Evaluable):無法評估 由於某些原因而無法進行檢查之情形、或無法判定為CR、Non-CR/non-PD、PD任何一者之情形。 ・NE (Not Evaluable): Unevaluable Cases that cannot be examined due to some reasons, or cases that cannot be judged as CR, Non-CR/non-PD, PD.

8.有無出現新病變 治療開始後確認到於基線檢查時不存在之病變之情形時,判定有「新病變」出現。 8. Whether new lesions appear When a lesion that did not exist at the baseline examination was confirmed after the start of treatment, it was determined that a "new lesion" appeared.

其中,判定為「新病變」必須並非因與基線評估時之檢查於攝像方法上不同或變更影像診斷工具(imaging modality)所致之圖像上之變化、或者因腫瘤以外之病情所致之圖像上之變化。例如因肝轉移灶之壞死導致病灶內產生之囊腫性病變不屬於新病變。基線(入組前評估)時未設為必須之部位經檢查新確認到之病變屬於新病變。Among them, the determination of "new lesions" must not be due to changes in imaging methods or changes in imaging modality from the examination at the baseline assessment, or changes in images due to conditions other than tumors. Like the change above. For example, cystic lesions in the lesions caused by necrosis of liver metastases are not new lesions. At baseline (pre-enrollment assessment), the newly confirmed lesions in the sites that were not set as required by examination are new lesions.

某病變消失後再次出現之情形時,繼續測定。其中,病變再次出現之時間點上之效果根據其他病變之狀態而異。於CR後再次出現病變之情形時,再次出現之時間點上判定為PD。另一方面,於PR或SD之情形時,暫時消失之病變再次出現時,將該病變之徑長與殘存病變之徑和相加而計算效果。即,於多個病變殘存之狀態下,1個病變看似「消失」後再次出現,僅憑此不能判定為PD,於全部病變之徑和滿足PD之標準之情形時判定為PD。其原因在於有如下認知:大部分病變並非真地「消失」,只是受使用之影像診斷工具之分辨率所限未被顯現。When a lesion disappears and reappears, continue to measure. Among them, the effect at the time point when the lesions reappeared varies according to the status of other lesions. When lesions reappeared after CR, PD was determined at the time point of reappearance. On the other hand, in the case of PR or SD, when the temporarily disappeared lesion reappears, the effect is calculated by adding the diameter of the lesion to the sum of the diameter of the residual lesion. That is, in a state where multiple lesions remain, one lesion appears to "disappear" and then reappears, which alone cannot be judged as PD. PD is determined when the diameter of all the lesions and the criteria for PD are met. The reason for this is the perception that most lesions do not really "disappear", but are not visualized due to the limitation of the resolution of the diagnostic imaging tools used.

於存在新病變之可能性但並不能確定之情形時,不判定為新病變,臨床上適當隔一段時間再次進行圖像檢查。根據再次進行之圖像檢查確定為新病變之情形時,以確定新病變之時間點之圖像檢查日,判定出現新病變。When there is a possibility of a new lesion but it cannot be determined, it will not be determined as a new lesion, and the image examination will be performed again at appropriate intervals in clinical practice. If it is determined to be a new lesion according to the image inspection performed again, the image inspection day at the time point of the new lesion is determined, and it is determined that a new lesion occurs.

於根據基線之FDG-PET顯示陰性之部位出現FDG-PET陽性(於衰減校正圖像中觀察到FDG之攝取超周圍組織2倍之FDG聚集)病變之情形時判定出現新病變。未實施基線之FDG-PET,而於治療開始後藉由所進行之FDG-PET發現FDG-PET陽性病變之情形時,於FDG-PET陽性之部位藉由CT或MRI確認到於基線檢查時未觀察到之病變時,判定出現新病變。New lesions were judged when FDG-PET positive lesions (FDG aggregation with an uptake of FDG that was 2 times greater than the surrounding tissue was observed in attenuation-corrected images) were found at sites that were negative for FDG-PET according to baseline. Baseline FDG-PET was not performed, and when FDG-PET-positive lesions were found by FDG-PET after the start of treatment, the FDG-PET-positive site was confirmed by CT or MRI to be absent at baseline. When lesions were observed, new lesions were determined.

9.總療效(Overall Response) 總療效(Overall response)係基於目標病變之效果、非目標病變之效果、新病變出現與否之組合,依據以下之表a進行判定。於基線檢查時不存在非目標病變之情形時之總療效藉由根據目標病變之效果及新病變出現來判定,於基線檢查時不存在目標病變之情形時之總療效藉由非目標病變之效果及新病變出現,依據表b來判定。 9. Overall Response The overall response (Overall response) is based on the combination of the effect of target lesions, the effect of non-target lesions, and the presence or absence of new lesions, and is determined according to Table a below. The overall efficacy in the absence of non-target lesions at baseline was determined by the effect of target lesions and the appearance of new lesions, and the overall efficacy in the absence of target lesions at baseline was determined by the effect of non-target lesions And new lesions appear, according to Table b to determine.

[表4a] 表a      各時間點上之總療效:存在目標病變之情形 目標病變 非目標病變 新病變 總療效 CR CR CR CR Non-CR /non-PD PR CR 無評估 PR PR Non-PD或評估不全 PR SD Non-PD或評估不全 SD 評估不全 Non-PD NE PD 不問 有或無 PD 不問 PD 有或無 PD 不問 不問 PD [Table 4a] Table a Overall efficacy at each time point: in the presence of target lesions target lesion non-target lesions new lesions total efficacy CR CR none CR CR Non-CR / non-PD none PR CR No assessment none PR PR Non-PD or incomplete assessment none PR SD Non-PD or incomplete assessment none SD Incomplete assessment Non-PD none NE PD Do not ask with or without PD Do not ask PD with or without PD Do not ask Do not ask Have PD

[表4b] 表b      各時間點上之總療效:僅存在非目標病變之情形 非目標病變 新病變 總療效 CR PR Non -CR /non-PD Non -CR /non-PD 評估不全 NE 明顯惡化 有或無 PD 不問 PD [Table 4b] Table b Overall efficacy at each time point: only in the presence of non-target lesions non-target lesions new lesions total efficacy CR none PR Non - CR / non-PD none Non - CR / non-PD Incomplete assessment none NE Significant deterioration with or without PD Do not ask Have PD

10.最佳總療效(Best Overall Response) 總療效(overall response)按CR>PR>SD>PD>NE之順序評定「良好度」,基於整個臨床試驗期間之總療效,依據以下之標準,判定最佳總療效(Best Overall Response)(表c)。於符合複數個區間之定義之情形時,按CR>PR>SD>PD>NE之順序歸類至更優之區間。 10. Best Overall Response The overall response (overall response) was evaluated in the order of CR > PR > SD > PD > NE. Based on the overall response during the entire clinical trial period, the Best Overall Response was determined according to the following criteria (Table 1). c). When the definitions of multiple intervals are met, they are classified into better intervals in the order of CR>PR>SD>PD>NE.

・CR(Complete Response):完全緩解 獲得間隔超過4週(28天)之連續2次以上之總療效CR之情形。第2次確認到總療效CR而確定最佳總療效CR之日為「CR確定日」。 ・PR(Partial Response):部分緩解 獲得間隔超過4週(28天)之連續2次以上之PR以上之總療效(CR或PR)之情形。第2次確認到PR以上之總療效而確定最佳總療效PR之日為「PR確定日」。 ・SD(Stable Disease):穩定 未獲得CR或PR之最佳總療效,但於治療開始後6週以內判定之總療效未出現PD、且總療效有1次以上為SD以上之情形。 ・PD(Progressive Disease):進展 最佳總療效不符合CR、PR、SD之任一者,總療效為PD之情形。 ・NE(Not Evaluable):無法評估 總療效均為NE之情形。 ・CR (Complete Response): Complete remission The case of obtaining 2 or more consecutive total curative effect CRs with an interval of more than 4 weeks (28 days). The day on which the CR was confirmed for the second time and the best overall CR was determined was the "CR Confirmation Day". ・PR (Partial Response): Partial mitigation Obtain the total efficacy (CR or PR) of more than 2 consecutive PRs with an interval of more than 4 weeks (28 days). The day on which the total effect of PR or above is confirmed for the second time and the best total effect PR is determined is the "PR confirmation day". ・SD (Stable Disease): Stable The best overall curative effect of CR or PR was not obtained, but no PD occurred in the overall curative effect determined within 6 weeks after the start of treatment, and the overall curative effect was more than SD for more than 1 time. ・PD (Progressive Disease): Progress The best overall curative effect does not meet any one of CR, PR, SD, and the overall curative effect is PD. ・NE (Not Evaluable): Unevaluable The overall efficacy is the case of NE.

[表4c] 表c  最佳總療效 最初之總療效 下一次之總療效 再下一次之總療效 最佳總療效 PR、CR之任一者 SD PD SD PR、CR之任一者 SD NE SD PR、CR之任一者 PD - PD PR、CR之任一者 NE NE NE PR、CR之任一者 NE SD SD SD PD - PD SD SD PD SD SD NE PD PD NE NE PD PD NE NE NE NE NE NE PD PD [實施例1] [Table 4c] Table c Best overall efficacy initial overall efficacy The total effect of the next time The total effect of the next time best overall efficacy Either PR or CR SD PD SD Either PR or CR SD NE SD Either PR or CR PD - PD Either PR or CR NE NE NE Either PR or CR NE SD SD SD PD - PD SD SD PD SD SD NE PD PD NE NE PD PD NE NE NE NE NE NE PD PD [Example 1]

實施例 1吉西他濱(G)+白蛋白結合紫杉醇(N)無反應例中之藉由托珠單抗(TCZ)+吉西他濱+白蛋白結合紫杉醇(N)併用所產生之臨床治療效果 Example 1 The clinical therapeutic effect of tocilizumab (TCZ) + gemcitabine + nab-paclitaxel (N) in the unresponsive case of gemcitabine (G) + nab-paclitaxel (N)

臨床試驗之概要為了研究基於藉由阻斷胰臟癌之腫瘤微環境之介白素6(IL-6)訊號而抑制結締組織增生(desmoplasia)之原理獲得之治療效果,以GN無反應之無法切除、復發之胰臟癌患者作為對象,對10名患者實施併用作為IL-6訊號抑制劑之抗IL-6R抗體托珠單抗(TCZ)之單次投予與GN療法之「針對吉西他濱與白蛋白結合紫杉醇無反應之轉移性胰臟癌的托珠單抗與吉西他濱-白蛋白結合紫杉醇併用療法之I期臨床試驗(EPOC1804)=“A Phase I study of tocilizumab plus gemcitabine/nab-paclitaxel in patient with gemcitabine/nab-paclitaxel-refractory metastatic pancreatic cancer(EPOC1804)”」。由於迄今為止無對人類併用實施TCZ投予與GN療法之案例,故目的在於研究TCZ+GN之安全性及耐受性。併用TCZ之目的在於解除GEM抗藥性,因此,對象設為對無法切除、復發之胰臟癌進行GN療法而獲得基於RECIST標準ver.1.1之部分緩解(partial response:PR)以上或與PR相當之抗腫瘤效果後變得無反應的有肝轉移之胰臟癌患者。 Summary of the clinical trial In order to investigate the therapeutic effect based on the principle of inhibiting desmoplasia by blocking interleukin 6 (IL-6) signaling in the tumor microenvironment of pancreatic cancer, the inability to respond to GN For patients with resected and recurrent pancreatic cancer, 10 patients were administered a single dose of the anti-IL-6R antibody tocilizumab (TCZ), which is an IL-6 signaling inhibitor, and the GN therapy "targeting gemcitabine and Phase I clinical trial of tocilizumab plus gemcitabine-nab-paclitaxel combination therapy for metastatic pancreatic cancer unresponsive to nab-paclitaxel (EPOC1804)="A Phase I study of tocilizumab plus gemcitabine/nab-paclitaxel in patient with gemcitabine/nab-paclitaxel-refractory metastatic pancreatic cancer (EPOC1804)”. Since there is no case of co-administration of TCZ and GN therapy in humans so far, the purpose is to study the safety and tolerability of TCZ+GN. The purpose of concomitant use of TCZ is to relieve GEM resistance. Therefore, the subject is set to obtain a partial response (PR) or higher or equivalent to PR based on RECIST criteria ver. 1.1 by GN therapy for unresectable and recurrent pancreatic cancer Pancreatic cancer patients with liver metastases who became unresponsive after antitumor effects.

用法劑量之設定TCZ: GEM+TCZ試驗(Mitsunaga S. et al., J Med Diagn Meth 6(1): 234, 2017)之結果表明,TCZ投予之耐受性為8 mg/kg。於本臨床試驗中,併用GN療法與TCZ,GN療法會高頻度地引發比GEM單劑時更嚴重之嗜中性球減少,另外預想到TCZ引起之嗜中性球減少,因此,考慮到安全性,將TCZ之劑量水平減半,亦設定為4 mg/kg。先前實施之針對Castleman氏病之II期臨床試驗中,每隔2週投予TCZ 2mg/kg、4 mg/kg、8 mg/kg(7例),於2 mg/kg投予時,1例由於抗IL-6R抗體表現之原因而中止。又,於投予低劑量之TCZ之情形時,由於在血中存在大量未與IL-6受體結合之IL-6之狀態下,TCZ容易自血中消失,故而有IL-6訊號傳遞系統急遽作動而引發大量炎症反應之虞。因此暗示,低劑量之TCZ、尤其2 mg/kg時,由於抗TCZ抗體之表現、或早期之TCZ消失,有可能無法獲得充分之IL-6抑制作用。綜上考慮,於本臨床試驗中,將TCZ起始劑量設為與GEM+TCZ試驗相同之8 mg/kg,亦將劑量水平減半之4 mg/kg作為可評估設定,2 mg/kg不予評估。以28天作為1個週期,第1週期之Day 1為投予TCZ之日,追加週期之Day 1為各週期中最初投予TCZ+GEM或TCZ+GN之日。 Dose and Administration TCZ: The results of the GEM+TCZ trial (Mitsunaga S. et al., J Med Diagn Meth 6(1): 234, 2017) showed that TCZ administration was tolerated at 8 mg/kg. In this clinical trial, GN therapy and TCZ were used together. GN therapy frequently caused more severe neutropenia than GEM single-dose, and TCZ-induced neutropenia was expected. Therefore, considering the safety The dose level of TCZ was halved, also set at 4 mg/kg. In a previously conducted Phase II clinical trial for Castleman's disease, TCZ was administered at 2 mg/kg, 4 mg/kg, and 8 mg/kg every 2 weeks (7 cases), and 1 case was administered at 2 mg/kg. Discontinued due to anti-IL-6R antibody performance. In addition, when a low dose of TCZ is administered, TCZ easily disappears from the blood when there is a large amount of IL-6 in the blood that is not bound to the IL-6 receptor, so there is an IL-6 signaling system. There is a risk of triggering a large amount of inflammatory response with abrupt movements. Therefore, it is suggested that at low doses of TCZ, especially 2 mg/kg, sufficient IL-6 inhibitory effect may not be obtained due to the expression of anti-TCZ antibodies or the disappearance of TCZ in the early stage. To sum up, in this clinical trial, the initial dose of TCZ was set to 8 mg/kg, which was the same as that of the GEM+TCZ trial, and the dose level was halved to 4 mg/kg as an evaluable setting, and 2 mg/kg was not evaluated. . Taking 28 days as a cycle, Day 1 of the first cycle is the day of TCZ injection, and Day 1 of the additional cycle is the day of the first injection of TCZ+GEM or TCZ+GN in each cycle.

併用藥(Gem+Nab-PTX): 於以遠處轉移性胰臟癌作為對象之大規模III期比較試驗即MPACT試驗中,以28天為1個週期,於Day 1、Day 8、Day 15投予GEM 1000 mg/m 2+nab-PTX 125 mg/m 2,結果,GEM之相對劑量強度(relative dose intensity)為75%,nab-PTX為80%(Von Hoff DD. et al., N Engl J Med. 2013 Oct 31; 369(18): 1691-703)。於將抑制IL-6之細胞內訊號之魯索替尼與GN併用之I期試驗中,按相同用法投予之GN之推薦劑量為GEM:750 mg/m 2、nab-PTX:100 mg/m 2(Bauer TM. et al., Onco Targets Ther. 2018 Apr 30; 11: 2399-2407)。綜上考慮,於對TCZ於用GN之情形時,以28天為1個週期,於Day 2、Day 9、Day 16(追加週期中為Day 1、Day 8、Day 15)投予較標準劑量減量之GEM 750 mg/m 2+nab-PTX 100 mg/m 2Concomitant medication (Gem+Nab-PTX): In the MPACT trial, a large-scale phase III comparative trial targeting distant metastatic pancreatic cancer, GEM was administered on Day 1, Day 8, and Day 15 with a cycle of 28 days 1000 mg/m 2 + nab-PTX 125 mg/m 2 . As a result, the relative dose intensity of GEM was 75% and that of nab-PTX was 80% (Von Hoff DD. et al., N Engl J Med. 2013 Oct 31; 369(18): 1691-703). In the phase I trial of ruxolitinib, which inhibits the intracellular signaling of IL-6, and GN, the recommended doses of GN administered in the same way are GEM: 750 mg/m 2 , nab-PTX: 100 mg/ m 2 (Bauer TM. et al., Onco Targets Ther. 2018 Apr 30; 11: 2399-2407). In summary, when TCZ is used in GN, a cycle of 28 days is used to administer a more standard dose on Day 2, Day 9, and Day 16 (Day 1, Day 8, and Day 15 in the additional cycle). Reduced GEM 750 mg/m 2 + nab-PTX 100 mg/m 2 .

投予期於GEM+TCZ試驗(Mitsunaga S. et al., J Med Diagn Meth 6(1): 234, 2017)中,CRP 2-10 mg/dL群體之PFS為3.6個月(95%信賴區間:1.0-4.4),故認為TCZ+GN預計持續投予1個月以上。但考慮到二次治療以後之胰臟癌患者之脆弱性及TCZ引起之CRP抑制或散熱受阻會導致感染症嚴重化之風險,本臨床試驗之投予期設定為可實施1個週期之TCZ+GN之28天。作為追加週期實施基準,僅針對不符合方案治療中止基準及臨床試驗中止基準、且於第1週期結束之時間點滿足以下全部基準之患者,進行追加週期之TCZ(8 mg/kg)+GN投予。於追加週期中無法投予GEM或GN之情形時,不單獨投予TCZ。 In the GEM+ TCZ trial (Mitsunaga S. et al., J Med Diagn Meth 6(1): 234, 2017), the PFS in the CRP 2-10 mg/dL population was 3.6 months (95% confidence interval: 1.0- 4.4), so it is considered that TCZ+GN is expected to be continuously invested for more than 1 month. However, considering the fragility of pancreatic cancer patients after the second treatment and the risk of severe infection caused by CRP inhibition or heat dissipation caused by TCZ, the administration period of this clinical trial was set to be 28% of TCZ+GN for one cycle. sky. As the implementation standard of the additional cycle, only patients who do not meet the treatment discontinuation standard and clinical trial discontinuation standard and meet all the following standards at the end of the first cycle, the additional cycle of TCZ (8 mg/kg) + GN administration will be administered . In cases where GEM or GN cannot be administered during the additional cycle, TCZ will not be administered separately.

獲得基於RECIST標準ver.1.1之PR以上或與PR相當(腫瘤縮小20%以上或腫瘤標記物減少50%以上)之抗腫瘤效果、或者病情得到控制之情形時,研究者判斷繼續實施本治療方案具有臨床意義。When the anti-tumor effect based on the RECIST criteria ver.1.1 is above or equivalent to PR (the tumor shrinks by more than 20% or the tumor marker is reduced by more than 50%), or the disease is controlled, the investigator judges that the treatment plan should continue. have clinical significance.

臨床試驗設計為了確保受驗者之安全,採用抗癌劑之I期試驗中通常所用之3+3設計。圖1表示臨床試驗設計之概要。 Clinical trial design In order to ensure the safety of the subjects, the 3+3 design commonly used in phase I trials of anticancer agents is adopted. Figure 1 shows an outline of the clinical trial design.

<劑量發現隊列(Dose finding cohort)> 為了確定TCZ之推薦劑量,進行1週期之TCZ 8 mg/kg+GN併用療法,根據DLT發生頻度來評估耐受性。於無法耐受TCZ 8 mg/kg之情形時,進行1週期之TCZ 4 mg/kg+GN併用療法,根據DLT發生頻度來評估耐受性。DLT評估對象者最多12名。 <Dose finding cohort> To determine the recommended dose of TCZ, 1 cycle of TCZ 8 mg/kg + GN concomitant therapy was performed, and tolerance was assessed according to the frequency of DLT. When TCZ 8 mg/kg could not be tolerated, 1 cycle of TCZ 4 mg/kg + GN combined therapy was performed, and the tolerance was evaluated according to the frequency of DLT. Up to 12 subjects for DLT assessment.

<擴展隊列(Expansion cohort)> 於藉由劑量發現隊列確定了TCZ之推薦劑量、且劑量發現隊列之入組者為10名以內之情形時,探索性地評估藉由劑量發現隊列確定之TCZ之推薦劑量與GN併用時之有效性及安全性。 <Expansion cohort> When the recommended dose of TCZ determined by the dose-discovery cohort and the enrollment of the dose-discovery cohort is less than 10, exploratory assessment of the efficacy of the recommended dose of TCZ determined by the dose-discovery cohort when used in combination with GN sex and safety.

評估項目對於作為主評估項目之不良事件之發生率、及劑量限制性毒性(DLT)之發生率、作為主要之次評估項目之由研究者判定之緩解率(Response rate:RR)及病情控制率(Disease control rate:DCR),進行評估。投予開始後28天為劑量限制性毒性(DLT)評估期。 The incidence of adverse events and the incidence of dose-limiting toxicity (DLT) as the main evaluation items, the remission rate (Response rate: RR) and disease control rate determined by the investigator as the main secondary evaluation items. (Disease control rate: DCR) for evaluation. The dose-limiting toxicity (DLT) evaluation period was 28 days after the start of administration.

評估、監控於整個臨床試驗期間,針對全體患者就不良事件進行周密監控,依據「不良事件通用術語標準v4.0日文JCOG版(Common Terminology Criteria for Adverse Events(CTCAE)v4.0-JCOG)[對應於CTCAE v4.03/MedDRA v12.0]」劃分不良事件之事件名及等級。患者於入組時(投予前)及第1週期之第28天接受腫瘤評估。其後實施追加週期之患者於該週期之第22天接受腫瘤評估。採用腫瘤縮小效果判定「實體癌療效判定新標準(RECIST標準)修訂版1.1-日文JCOG版-:Revised RECIST guideline (version 1.1)」,由研究者對應答進行評估。 Evaluation and monitoring During the entire clinical trial period, all patients were carefully monitored for adverse events, according to the "Common Terminology Criteria for Adverse Events (CTCAE) v4.0-JCOG) [corresponding to the "Common Terminology Criteria for Adverse Events (CTCAE) v4.0-JCOG) In CTCAE v4.03/MedDRA v12.0]", the event name and grade of adverse events were classified. Patients underwent tumor assessment at enrollment (before administration) and on Day 28 of Cycle 1. Patients who subsequently underwent a booster cycle underwent tumor assessment on day 22 of that cycle. Responses were assessed by investigators using the "New Response Evaluation Criteria in Solid Cancers (RECIST Criteria) Revised 1.1-Japanese JCOG version-: Revised RECIST guideline (version 1.1)" for tumor shrinkage effect.

關於劑量限制性毒性(DLT),評估期設為以TCZ投予開始日作為起算日計28天。再者。第28天後繼續發生之不良事件為與DLT判定相關之事件之情形時,延長DLT評估期。由研究者判定是否屬於DLT。其中,將能夠否定與TCZ+GN併用療法之因果關係之情況判定為不屬於DLT。又,於發生DLT但不符合方案治療中止基準之情形時,繼續治療方案。選擇於Day1投予TCZ、Day2投予GN,獲得DLT評估所需之足夠資料之受驗者作為DLT之評估對象。其中,將未投予GN時亦發生DLT之情形設為評估對象。Regarding dose-limiting toxicity (DLT), the evaluation period was set to be 28 days from the start date of TCZ administration. Again. If the adverse events that continue to occur after the 28th day are events related to the DLT determination, the DLT evaluation period will be extended. It is up to the investigator to determine whether it is a DLT. Among them, the case where the causal relationship with TCZ+GN combined therapy can be denied is determined as not belonging to DLT. Also, in the event of DLT occurring but not meeting the protocol treatment discontinuation criteria, the treatment regimen was continued. Select the subjects who were administered TCZ on Day 1 and GN on Day 2, and who obtained sufficient data for DLT assessment as the subjects of DLT assessment. Among them, the case where DLT occurred even when GN was not administered was set as the evaluation object.

<DLT基準> (1)持續超7天之Grade 4(<500/mm 3)之嗜中性球數減少 (2)持續超3天之伴有38.5度以上之發熱之Grade 3(<1,000/mm 3)以上之嗜中性球數減少 (3)持續超7天之Grade 4(<25,000/mm 3)之血小板數減少 (4)Grade 3以上之血小板數減少導致需進行輸血之情形 (5)Grade 4之AST/ALT上升或持續超7天之Grade 3之AST/ALT上升 (6)Grade 3以上之非血液毒性(其中,能夠藉由支持療法進行控制之Grade 3之消化系統症狀、從臨床上判斷無問題之Grade 3以上之電解質異常及檢查值異常、可控制之Grade 3之高血圧壓、以及Grade 3之輸液反應(infusion reaction)除外) <DLT standard> (1) Decreased neutrophils in Grade 4 (<500/mm 3 ) for more than 7 days (2) Grade 3 (<1,000/ mm 3 ) or more neutropenia (3) Grade 4 (<25,000/mm 3 ) thrombocytopenia that persisted for more than 7 days (4) Blood transfusion due to Grade 3 or more thrombocytopenia (5) ) AST/ALT elevation of Grade 4 or AST/ALT elevation of Grade 3 that persists for more than 7 days (6) Non-hematological toxicity of Grade 3 or higher (among which, digestive system symptoms of Grade 3 that can be controlled by supportive therapy, Electrolyte abnormalities above Grade 3 and abnormal test values, controllable Grade 3 high blood pressure, and Grade 3 infusion reactions are excluded)

基準作為本臨床試驗之對象患者群體,設定暗示更能承受自胰臟癌組織中之腫瘤細胞、間質細胞、免疫活性細胞產生之IL-6之參與的血中C反應蛋白(CRP)為0.5 mg/dL以上且10.0 mg/dL以下之GN無反應之進展期胰臟癌患者為選擇入組基準。 As the benchmark for the patient population of this clinical trial, the C-reactive protein (CRP) in blood, which suggests that it is more resistant to the participation of IL-6 produced by tumor cells, stromal cells, and immunocompetent cells in pancreatic cancer tissue, is set at 0.5 Patients with advanced pancreatic cancer with unresponsive GN above mg/dL and below 10.0 mg/dL were selected as the inclusion criteria.

滿足以下全部選擇基準、且不符合任何排除基準之受驗者合乎入組資格。 <選擇基準> (1)徵得同意時之年齡為20歲以上。 (2)病理診斷顯示為浸潤性胰臟導管癌、腺癌、腺鱗狀上皮癌、或診斷為癌症且臨床診斷與轉移性胰臟癌不矛盾。 Subjects who meet all of the following selection criteria and do not meet any exclusion criteria are eligible. <Criteria for selection> (1) The age at the time of obtaining consent is 20 years or older. (2) The pathological diagnosis shows invasive pancreatic ductal carcinoma, adenocarcinoma, adenosquamous epithelial carcinoma, or cancer and the clinical diagnosis is not inconsistent with metastatic pancreatic cancer.

(3)對無法切除、復發之胰臟癌進行GN療法而獲得基於實體腫瘤療效判定基準(RECIST)ver.1.1之部分緩解(partial response:PR)以上或與PR相當(基於RECIST ver1.1之可測定病變中之腫瘤長徑和縮小20%以上,或腫瘤標記物CA19-9血中濃度降低50%以上)之抗腫瘤效果後變得無反應。(3) GN therapy for unresectable and recurrent pancreatic cancer to obtain partial response (PR) or higher based on Response Evaluation Criteria for Solid Tumors (RECIST) ver.1.1 or equivalent to PR (based on RECIST ver1.1 The tumor length and diameter in the lesions can be measured and the anti-tumor effect of tumor marker CA19-9 reduced by more than 20%, or the blood concentration of tumor marker CA19-9 is reduced by more than 50%) and becomes unresponsive.

(4)判斷能夠對無法切除、復發之胰臟癌安全地實施GEM 750 mg/m 2+nab-PTX 100 mg/m 2之投予。 (5)存在基於RECIST標準ver 1.1之目標病變之肝轉移、且能夠進行肝活檢。又,同意提供所採集之腫瘤組織及血液檢體。 (4) It is judged that GEM 750 mg/m 2 + nab-PTX 100 mg/m 2 can be administered safely to unresectable and recurrent pancreatic cancer. (5) There is liver metastases based on the target lesions of RECIST criteria ver 1.1, and liver biopsy can be performed. Also, agree to provide the collected tumor tissue and blood samples.

(6)能夠實施造影CT檢查或造影MRI檢查。 (7)ECOG日常體能狀態(PS)為0-1。 (8)入組前7天以內保持滿足以下條件之充足器官功能。再者,存在複數個檢查值之情形時,採用最接近入組日之檢查值。 ・嗜中性球數 1,500/mm 3以上 ・血小板數 100,000/mm 3以上 ・血紅蛋白 8.5 g/dL以上 ・血清肌酐 未達1.2 mg/dL ・血清總膽紅素 2.0 mg/dL以下 ・AST、ALT 120 U/L以下 (6) Contrast CT examination or contrast MRI examination can be performed. (7) ECOG daily performance status (PS) is 0-1. (8) Maintain adequate organ function meeting the following conditions within 7 days before enrollment. Furthermore, when there are multiple check values, the check value closest to the date of enrollment is used.・Neutrophil count 1,500/ mm3 or more ・Platelet count 100,000/ mm3 or more ・Hemoglobin 8.5 g/dL or more ・Serum creatinine less than 1.2 mg/dL ・Serum total bilirubin less than 2.0 mg/dL ・AST, ALT Below 120 U/L

(9)CRP為0.5 mg/dL以上且10.0 mg/dL以下。 (10)周圍感覺神經病變為Grade 2以下。 (11)至少有望存活2個月以上。 (12)被充分告知本臨床試驗內容,經患者本人書面同意。 (9) CRP is 0.5 mg/dL or more and 10.0 mg/dL or less. (10) Peripheral sensory neuropathy is below Grade 2. (11) At least expected to survive for more than 2 months. (12) Be fully informed of the content of this clinical trial, with the written consent of the patient.

<排除基準> (1)有明顯之體腔積液、浮腫。 (2)存在伴有症狀之腦轉移。 (3)患有活動性重複癌(允許有3年以上之無病期。又,活動性重複癌並不包括在上皮內癌及黏膜內癌之病變藉由局部治療而被判斷為治癒之情形)。 <Exclusion criteria> (1) There is obvious body cavity effusion and edema. (2) Brain metastases with symptoms are present. (3) Patients with active repeat cancer (a disease-free period of more than 3 years is allowed. In addition, active repeat cancer does not include cases where intraepithelial cancer and intramucosal cancer are judged to be cured by local treatment) .

(4)既往有膽管空腸吻合術史。 (5)入組前4週以內接受過輸血。 (6)截至入組時間點,距最後一次實施手術(經皮經肝膽道引流或內窺鏡下膽道引流除外)之日未達4週。 (4) There was a history of cholangiojejunostomy in the past. (5) Received blood transfusion within 4 weeks before entering the group. (6) As of the enrollment time point, the last surgery (except percutaneous transhepatic biliary drainage or endoscopic biliary drainage) has not reached 4 weeks.

(7)於臨床試驗期間預定進行手術。 (8)既往有IL-6訊號抑制劑(JAK抑制劑等)投予史。 (9)既往有免疫療法(癌症疫苗、免疫檢查點抑制劑等)投予史。 (7) Surgery is scheduled to be performed during the clinical trial. (8) There is a history of administration of IL-6 signaling inhibitors (JAK inhibitors, etc.) in the past. (9) There is a history of administration of immunotherapy (cancer vaccine, immune checkpoint inhibitor, etc.) in the past.

(10)於入組前4週以內投予過生物學製劑(TCZ、曲妥珠單抗、英利昔單抗、依那西普、阿達木單抗等)。 (11)於入組前4週以內投予過其他臨床試驗藥。 (12)併發需全身投予抗生素、抗病毒藥、抗真菌藥等之感染疾病。 (10) Administered biological agents (TCZ, trastuzumab, infliximab, etanercept, adalimumab, etc.) within 4 weeks before enrollment. (11) Administered other clinical trial drugs within 4 weeks before enrollment. (12) Concurrent infectious diseases requiring systemic administration of antibiotics, antiviral drugs, and antifungal drugs.

(13)為HCV抗體陽性、HBs抗原陽性抗體陽性、HBs抗體陽性或HBc抗體陽性或者在兩者均為陽性之情形時HBV-DNA定量檢查呈陽性、或HIV抗體陽性。 (14)患有需全身投予腎上腺皮質激素劑之非腫瘤性疾病(過敏性疾病、哮喘等)。 (15)併發特發性肺纖維化症、間質性肺炎、塵肺症、藥物性肺炎等肺損傷及結核、或有相關既往病史。 (13) Positive for HCV antibody, positive for HBs antigen antibody, positive for HBs antibody or positive for HBc antibody, or positive for HBV-DNA quantitative test when both are positive, or positive for HIV antibody. (14) Suffering from non-neoplastic diseases (allergic diseases, asthma, etc.) requiring systemic administration of adrenocortical hormones. (15) Complicated with idiopathic pulmonary fibrosis, interstitial pneumonia, pneumoconiosis, drug-induced pneumonia and other lung injury and tuberculosis, or have related past medical history.

(16)患有臨床上成為問題之心臟疾病(不可控制高血壓、不穩定心絞痛、鬱血性心臟衰竭、需進行藥物治療之重度心律不整、入組前12個月以內發生過心肌梗塞等)。 (17)患有被研究者或助理研究者判斷為對同意或臨床試驗實施的理解會造成妨礙之精神疾病。 (16) Patients with clinically problematic heart disease (uncontrollable hypertension, unstable angina pectoris, stagnant heart failure, severe arrhythmia requiring drug treatment, myocardial infarction within 12 months before enrollment, etc.). (17) Suffering from a mental illness that is judged by the researcher or assistant researcher to hinder the understanding of consent or clinical trial implementation.

(18)患有控制不佳之糖尿病(標準為HbA1c值7.5%,以臨床判斷優先)。 (19)既往有對左旋氧氟沙星等藥物之嚴重過敏史。 (18) Suffering from poorly controlled diabetes mellitus (standard HbA1c value is 7.5%, clinical judgment is preferred). (19) There is a history of severe allergy to levofloxacin and other drugs in the past.

(20)併發有以下之疾病,研究者或助理研究者基於醫學根據而判斷為不適合參加臨床試驗之患者。 循環器官疾病、血液/造血器官疾病、呼吸系統疾病、神經肌肉疾病、內分泌疾病、泌尿系統疾病、消化系統疾病、黃斑浮腫、骨髓纖維化症、真性紅細胞增多症、類風濕性關節炎等全身性炎症性疾病。 (20) Patients with the following diseases, which the researcher or assistant researcher judges not suitable for participating in clinical trials based on medical basis. Circulatory organ diseases, blood/hematopoietic organ diseases, respiratory system diseases, neuromuscular diseases, endocrine diseases, urinary system diseases, digestive system diseases, macular edema, myelofibrosis, polycythemia vera, rheumatoid arthritis and other systemic diseases Inflammatory disease.

(21)對於可能妊娠之女性,入組7天以內之妊娠檢查結果(尿檢結果有疑問時實施血清檢查)並非陰性。 (22)不能同意自徵得同意後至最後一次投予後至少1個月內之適當避孕。又,對於女性而言,自徵得同意後至最後一次投予後至少1個月內不哺乳是不能同意的。 (23)其他研究者或助理研究者判斷為不適合參加本臨床試驗之情況。 (21) For women who may be pregnant, the pregnancy test results within 7 days of enrollment (serum test when the results of urine test are in doubt) are not negative. (22) Inability to agree to adequate contraception for at least 1 month after the last dose from the time of consent. Also, for women, it is not acceptable to abstain from breastfeeding for at least 1 month from the time of consent to the last dose. (23) Other investigators or assistant investigators judge that they are not suitable to participate in this clinical trial.

結果將該臨床試驗之結果示於表1。主評估項目為劑量限制性毒性之出現率,未確認到出現該事件。作為次評估項目之緩解率,評估結果為PR(partial response:部分緩解)1名、SD(stable disease:穩定)6名、PD(progressive disease:進展)3名。根據先行研究之報告,GN療法對於化學治療劑無反應之胰臟癌患者之緩解率(RR)為0%(西岡等人, 癌治療學會2019)、3.2%(Yang et al. J Formos Med Assoc. 2020 Jan; 119 (1 Pt 1): 97-105),相對於此,將GN療法與抗IL-6R抗體併用之療法之RR為10%。上述SD(穩定)之6名中3名患者觀察到腫瘤標記物CA19-9之減少最高達30%以上、且長期保持SD,再加上部分緩解1名,從而顯示與部分緩解(PR)相當之效果之患者共4名(40%)。先行研究中GN療法對於化學治療劑無反應之胰臟癌患者之病情控制率(DCR)為33%(西岡等人, 癌治療學會2019)、19.4%(Yang et al. J Formos Med Assoc. 2020 Jan; 119 (1 Pt 1): 97-105),相對於此,抗IL-6R抗體+GN併用療法之DCR為70%。關於針對標準治療無效之胰臟癌群體之治療效果,確認到部分緩解率及病情控制率均可謂良好之抗腫瘤效果。 Results The results of this clinical trial are shown in Table 1. The primary assessment was the incidence of dose-limiting toxicity, which was not confirmed. As the remission rate of the sub-assessment item, the evaluation results were 1 PR (partial response: partial remission), 6 SD (stable disease: stable), and 3 PD (progressive disease: progressive). According to a pilot study, the response rate (RR) of GN therapy for pancreatic cancer patients who did not respond to chemotherapeutic agents was 0% (Nishioka et al., Cancer Therapy Society 2019), 3.2% (Yang et al. J Formos Med Assoc 2020 Jan; 119 (1 Pt 1): 97-105), the RR of GN therapy combined with anti-IL-6R antibody was 10%. Among the above-mentioned 6 patients with SD (stable), 3 patients observed a reduction of the tumor marker CA19-9 by more than 30%, and maintained SD for a long time, and 1 patient had a partial response, which showed that it was equivalent to a partial response (PR). A total of 4 patients (40%) had the effect. In a pilot study, the disease control rate (DCR) of GN therapy for pancreatic cancer patients who did not respond to chemotherapeutic agents was 33% (Nishioka et al., Cancer Therapy Society 2019), 19.4% (Yang et al. J Formos Med Assoc. 2020 Jan; 119 (1 Pt 1): 97-105), the DCR of the anti-IL-6R antibody + GN combination therapy was 70%. Regarding the therapeutic effect on the pancreatic cancer group ineffective in standard treatment, it was confirmed that both the partial remission rate and the disease control rate can be regarded as a good anti-tumor effect.

可以確認,該等意想不到之優異之併用效果得益於後述實施例2及3中發現之包含腫瘤間質之微環境之改善效果所帶來之藥劑滲透之改善,進而,GN+TCZ併用療法對於GN無反應患者之優異之抗腫瘤效果與腫瘤標記物CA19-9之抑制密切相關(圖2)。It can be confirmed that these unexpected and excellent combined effects are due to the improvement of drug penetration due to the improvement effect of the microenvironment including tumor stroma found in Examples 2 and 3 described later. The excellent antitumor effect of non-responders was closely related to the inhibition of the tumor marker CA19-9 (Fig. 2).

[表5] 表1      臨床試驗之結果 年齡 性別 CRP 基線CA19-9 TCZ+GN Cycle(s) CA19-9_ 相對於基線之最佳變化 RECIST_ 最佳總療效 GAP001 66 F 4.1 50077 6 -80% SD GAP002 66 F 0.3 1344 1 無減少傾向 PD GAP003 62 M 20.1 729 1 無減少傾向 PD GAP004 76 M 17.8 37005 3 -55% SD GAP005 50 M 2.8 15065 1 無減少傾向 SD GAP006 49 F 2.9 241 1 -23% SD GAP007 74 F 2.3 18547 1 -5% PD GAP008 68 M 2.4 2188 2 -24% PR-in GAP009 48 M 3.7 220 4 -48% SD GAP010 75 M 9.2 25.1 1 -15% SD [實施例2] [Table 5] Table 1 Results of clinical trials age gender CRP Baseline CA19-9 TCZ+GN Cycle(s) CA19-9_ Best change from baseline RECIST_ Best Overall Response GAP001 66 F 4.1 50077 6 -80% SD GAP002 66 F 0.3 1344 1 No tendency to decrease PD GAP003 62 M 20.1 729 1 No tendency to decrease PD GAP004 76 M 17.8 37005 3 -55% SD GAP005 50 M 2.8 15065 1 No tendency to decrease SD GAP006 49 F 2.9 241 1 -twenty three% SD GAP007 74 F 2.3 18547 1 -5% PD GAP008 68 M 2.4 2188 2 -twenty four% PR-in GAP009 48 M 3.7 220 4 -48% SD GAP010 75 M 9.2 25.1 1 -15% SD [Example 2]

實施例 2臨床上抗IL-6R抗體對結締組織增生之抑制 富纖維化之腫瘤間質稱作結締組織增生,其為胰臟癌之特徵,被認為是化學療法無反應的原因之一。認為胰臟癌結締組織增生之原因在於如下,因自胰臟癌細胞產生之生長因子或細胞激素等而使得表現α-平滑肌肌動蛋白(α-SMA)之活化纖維母細胞旺盛生長,釋放出膠原蛋白或纖維黏連蛋白等細胞外基質。於EPOC1804中,採集GN+TCZ投予前後之胰臟癌肝轉移灶之腫瘤組織檢體,研究IL-6抑制誘導之結締組織增生形成抑制與抗腫瘤效果之關係。 Example 2 Clinically, anti-IL-6R antibody inhibits connective tissue hyperplasia The fibrotic-rich tumor stroma is called connective tissue hyperplasia, which is a feature of pancreatic cancer and is considered to be one of the reasons for non-response to chemotherapy. The reason for the proliferation of connective tissue in pancreatic cancer is considered to be as follows. Activated fibroblasts expressing α-smooth muscle actin (α-SMA) are vigorously grown due to growth factors, cytokines, etc. produced from pancreatic cancer cells. Extracellular matrix such as collagen or fibronectin. In EPOC1804, tumor tissue samples of pancreatic cancer liver metastases before and after GN+TCZ administration were collected, and the relationship between the inhibition of IL-6 inhibition-induced connective tissue hyperplasia formation and the antitumor effect was studied.

於徵得參加實施例1記載之將抗IL-6R抗體托珠單抗(TCZ)之單次投予與GN療法併用之I期臨床試驗(EPOC1804)的患者同意後,使用18號針採集GN+TCZ投予前(Day 0)及投予後(Day 28)之患者之胰臟癌肝轉移灶之腫瘤組織,立即使用10%中性福馬林固定。對於固定之腫瘤檢體,於48小時以內開始脫水操作,進行石蠟包埋而製作腫瘤檢體塊(FFPE塊)。使用切片機自該FFPE塊切下厚4 μm之腫瘤組織薄切片,貼附於載玻片上而製作腫瘤切片檢體。對該腫瘤切片實施蘇木精-伊紅(HE)染色、以抗α-SMA抗體及抗磷酸化STAT3抗體作為初級抗體之免疫組織化學染色(Immunohistochemistry;IHC)。After obtaining the consent of the patients who participated in the phase I clinical trial (EPOC1804) of single administration of the anti-IL-6R antibody tocilizumab (TCZ) in combination with GN therapy described in Example 1, GN+TCZ was collected using an 18-gauge needle. Before administration (Day 0) and after administration (Day 28), tumor tissues of pancreatic cancer liver metastases of patients were immediately fixed with 10% neutral formalin. The fixed tumor specimens were dehydrated within 48 hours and embedded in paraffin to prepare tumor specimen blocks (FFPE blocks). A thin section of tumor tissue with a thickness of 4 μm was cut from the FFPE block using a microtome, and attached to a glass slide to prepare a tumor section specimen. The tumor sections were subjected to hematoxylin-eosin (HE) staining and immunohistochemical staining (Immunohistochemistry; IHC) with anti-α-SMA antibody and anti-phospho-STAT3 antibody as primary antibodies.

將該結果示於圖3。根據源於試驗治療成效良好之受驗者(GAP01)之試樣解析,相較於GN+TCZ投予前,GN+TCZ投予後之HE染色組織圖像顯示腫瘤之纖維性間質組織被明顯破壞,又,藉由IHC法可確認,纖維化標記物α-SMA於腫瘤組織內之陽性面積比率於投予前(pre)為19%,相對於此,投予後(post)變為7%,較投予前減少63%,出現意想不到之顯著減少,從而確認了GN+TCZ投予後之腫瘤間質之結締組織增生抑制作用。關於該結締組織增生抑制作用, GN+TCZ投予後的IL-6訊號下游因子STAT3之磷酸化(phosho-STAT3)(0.9%)較投予前(5%)減少83%,由於藉由對GN無反應患者投予TCZ+GN而使得IL-6訊號下游因子STAT3之磷酸化(phosho-STAT3)得到顯著抑制,故確認TCZ有效。綜上所述,確認到TCZ+GN療法於胰臟癌中誘導優異之結締組織增生抑制,而發揮出實施例1中出現之抗腫瘤效果。 [實施例3] The results are shown in FIG. 3 . According to the analysis of the samples from the subjects with good therapeutic effect (GAP01), compared with before the administration of GN+TCZ, the HE-stained tissue images after the administration of GN+TCZ showed that the fibrous interstitial tissue of the tumor was significantly destroyed. It was confirmed by IHC method that the positive area ratio of fibrosis marker α-SMA in tumor tissue was 19% before administration (pre), but it became 7% after administration (post), which was higher than that after administration. An unexpectedly significant reduction was observed before, which was 63%, confirming the inhibitory effect of tumor stroma on connective tissue proliferation after GN+TCZ administration. Regarding this connective tissue proliferation inhibitory effect, the phosphorylation of IL-6 signaling downstream factor STAT3 (phosho-STAT3) (0.9%) after GN+TCZ administration was reduced by 83% compared to before administration (5%), due to the lack of response to GN. The phosphorylation of STAT3 (phosho-STAT3), a downstream factor of IL-6 signaling, was significantly inhibited by administration of TCZ+GN to the patient, so it was confirmed that TCZ was effective. From the above, it was confirmed that TCZ+GN therapy induces excellent inhibition of connective tissue proliferation in pancreatic cancer, and exerts the antitumor effect shown in Example 1. [Example 3]

實施例 3臨床上之TCZ作用下之低分子浸潤(TCZ作用下之左旋氧氟沙星浸潤)評估 採用TCZ+GN投予前後之患者之胰臟癌肝轉移灶之腫瘤組織檢體,藉由使用基質輔助雷射脫附電離法(matrix assisted laser desorption/ionization;MALDI)之質譜分析成像(MALDI-MSI)法評估IL-6R抑制介導之低分子藥劑向腫瘤內部之滲透亢進,研究與抗腫瘤效果之關係。 Example 3 Clinical evaluation of low molecular weight infiltration under the action of TCZ (levofloxacin infiltration under the action of TCZ) Evaluation of tumor tissue samples of pancreatic cancer liver metastases from patients before and after administration of TCZ+GN, by using stroma Mass spectrometry imaging (MALDI-MSI) of matrix assisted laser desorption/ionization (MALDI) to evaluate the hyperpenetration of low molecular weight drugs into tumors mediated by IL-6R inhibition, research and anti-tumor effects relationship.

於實施例1記載之「針對吉西他濱與白蛋白結合紫杉醇無反應之轉移性胰臟癌的托珠單抗與吉西他濱-白蛋白結合紫杉醇併用療法之I期臨床試驗(EPOC1804)」中,徵得患者之知情同意後,於第0天之GN+TCZ投予前(投予前)及第28天之藥劑投予後(投予後),使用18號針,自患者之肝轉移部位採集胰臟癌組織之粗針(core needle)活檢試樣。於活檢前,預先對患者投予低分子抗菌藥左旋氧氟沙星。將收集之活檢試樣立即於-20℃左右冷凍。試樣於使用之前-80℃保存。使用低溫恆溫器,自各試樣獲得3片連續切片,進而固定於塗有氧化銦錫之玻片上。使用TM Sprayer(HTX Imaging),於組織切片上噴塗基質(α-氰基-4-羥基肉桂酸;αCHCA)及作為內部標準物質之同位素標記左旋氧氟沙星。使用Solarix(MALDI-FTICR)(Bruker Daltonics)獲得質譜及位置資訊。MALD-MSI分析後,去除基質,利用蘇木精及伊紅將試樣染色,評估組織結構及左旋氧氟沙星分佈。最後,使用Multimaging軟體(ImaBiotech)解析MALDI-MSI資料,利用熱圖描繪左旋氧氟沙星分佈。基於內部標準之強度將圖像標準化。In the "Phase I clinical trial of tocilizumab and gemcitabine-nab-paclitaxel combination therapy for metastatic pancreatic cancer unresponsive to gemcitabine and nab-paclitaxel" described in Example 1 (EPOC1804), a patient After informed consent was obtained, before the administration of GN+TCZ on day 0 (before administration) and after administration of the drug on day 28 (after administration), an 18-gauge needle was used to collect the thickness of pancreatic cancer tissue from the patient's liver metastases. Core needle biopsy samples. Before biopsy, the low molecular weight antibiotic levofloxacin was pre-administered to the patient. The collected biopsy samples were immediately frozen at about -20°C. Samples were stored at -80°C before use. Using a cryostat, 3 serial sections were obtained from each sample and mounted on indium tin oxide coated glass slides. The tissue sections were sprayed with matrix (α-cyano-4-hydroxycinnamic acid; αCHCA) and isotope-labeled levofloxacin as an internal standard using a TM Sprayer (HTX Imaging). Mass spectra and positional information were obtained using Solarix (MALDI-FTICR) (Bruker Daltonics). After MALD-MSI analysis, the matrix was removed, and the samples were stained with hematoxylin and eosin to evaluate the tissue structure and distribution of levofloxacin. Finally, the MALDI-MSI data were parsed using Multimaging software (ImaBiotech), and the distribution of levofloxacin was depicted using a heat map. Images were normalized based on the intensity of the internal standard.

將該結果示於圖4。自實施例1中發現之TCZ+GN投予有效例(患者ID GAP001)之患者之胰臟癌肝轉移灶採集之腫瘤檢體中,投予前之腫瘤組織中之左旋氧氟沙星濃度為10.4 μg/g,相對於此,投予後之左旋氧氟沙星濃度為17.1 μg/g,GN+TCZ投予後之腫瘤中之左旋氧氟沙星濃度之上升率較投予前為64.4%。相較於投予前左旋氧氟沙星蓄積於試樣之腫瘤區,自同一患者之同一胰臟癌肝轉移目標病變區採集之投予後試樣顯示,左旋氧氟沙星向腫瘤中聚集。綜上,藉由MALDI-MSI法可確認,如實施例2中發現之包含腫瘤間質之微環境之改善效果使低分子化學治療劑左旋氧氟沙星向腫瘤內部之滲透亢進。認為其原因在於,相較於GN療法,TCZ+GN療法藉由重建(remodel)腫瘤微環境,結果誘導吉西他濱或白蛋白結合紫杉醇之類的低分子藥劑、以及高分子量之抗體藥劑或如實施例4所示之免疫細胞向腫瘤內部之浸潤亢進。該等結果佐證了實施例1中發現之抗腫瘤效果。 [實施例4] The results are shown in FIG. 4 . The concentration of levofloxacin in the tumor tissue before the administration was 10.4 μg in the tumor specimen collected from the pancreatic cancer liver metastases of the patient of the TCZ+GN administration effective case (patient ID GAP001) found in Example 1 On the other hand, the levofloxacin concentration after administration was 17.1 μg/g, and the increase rate of levofloxacin concentration in tumors after GN+TCZ administration was 64.4% compared with that before administration. Compared with the accumulation of levofloxacin in the tumor area of the sample before administration, the post-administration sample collected from the same target lesion area of pancreatic cancer liver metastases in the same patient showed that levofloxacin accumulated in the tumor. In conclusion, it was confirmed by the MALDI-MSI method that the improvement effect of the microenvironment including the tumor stroma as found in Example 2 increased the penetration of the low molecular weight chemotherapeutic agent levofloxacin into the tumor. The reason is believed to be that, compared with GN therapy, TCZ+GN therapy induces low molecular weight drugs such as gemcitabine or albumin-bound paclitaxel, and high molecular weight antibody drugs or as shown in Example 4 by remodeling the tumor microenvironment. It shows the infiltration of immune cells into the tumor. These results corroborate the antitumor effect found in Example 1. [Example 4]

實施例 4基於GN+TCZ之模擬之TCZ用法劑量 為了選擇針對胰臟癌之TCZ+GN+阿替利珠單抗(ATZ、抗PD-L1抗體製劑)併用方案中TCZ之最佳劑量,使用半機理藥動學(PK)-藥效學(PD)模型,研究G、N及TCZ對嗜中性球數之影響。嗜中性球之生命週期模型應用Friberg等人(J Clin Oncol. 2002 Dec15; 20(24): 4713-21)所報告者。G及N係作為抑制嗜中性球生長過程者組入模型。另一方面,據報告TCZ會使循環血中之嗜中性球減少(Chen et al. J Clin Pharmacol. 2014 Oct; 54(10): 1097-107., Laurence et al. Eur J Clin Invest. 2017 Oct; 47(10): 736-745.),其係作為增加嗜中性球自循環血中之清除率者組入模型。G、N及TCZ於各過程中之模型參數採用基於文獻者(Daniel et al. N Engl J Med. 2013; 369: 1691-703, Chen et al. J Clin Pharmacol. 2014 Oct; 54(10):1097-107., Gibiansky et al. J Pharmacokinet Pharmacidyn. 2012 Feb; 39(1): 5-16.)。將該半機理PK-PD模型之結構示於圖5。 Example 4 Dosage of TCZ based on the simulation of GN+TCZ In order to select the optimal dose of TCZ in the combination regimen of TCZ+GN+atezolizumab (ATZ, anti-PD-L1 antibody preparation) for pancreatic cancer, semi-mechanical pharmacokinetics were used (PK)-Pharmacodynamic (PD) model to study the effect of G, N and TCZ on neutrophil number. The neutrophil life cycle model was reported by Friberg et al. (J Clin Oncol. 2002 Dec15; 20(24): 4713-21). Lines G and N were used as models to inhibit the growth of neutrophils. On the other hand, TCZ has been reported to decrease circulating neutrophils (Chen et al. J Clin Pharmacol. 2014 Oct; 54(10): 1097-107., Laurence et al. Eur J Clin Invest. 2017 Oct; 47(10): 736-745.), which was modeled as one that increases neutrophil clearance from circulating blood. The model parameters of G, N and TCZ in each process are based on literature (Daniel et al. N Engl J Med. 2013; 369: 1691-703, Chen et al. J Clin Pharmacol. 2014 Oct; 54(10): 1097-107., Gibiansky et al. J Pharmacokinet Pharmacidyn. 2012 Feb; 39(1): 5-16.). The structure of this semi-mechanical PK-PD model is shown in FIG. 5 .

為了確認該模型是否準確記述嗜中性球數之變化,將實施例1記載之「針對吉西他濱與白蛋白結合紫杉醇無反應之轉移性胰臟癌的托珠單抗與吉西他濱-白蛋白結合紫杉醇併用療法之I期臨床試驗(EPOC1804)」中實際觀察到之嗜中性球數與預測結果示於圖6。觀察到之嗜中性球數存在較大差異,但預測結果較佳地展現了整體觀察結果之傾向。In order to confirm whether this model accurately describes the change in neutrophil number, "Tocilizumab for metastatic pancreatic cancer unresponsive to gemcitabine and nab-paclitaxel" described in Example 1 was used in combination with gemcitabine-nab-paclitaxel The actual observed and predicted neutrophil counts in the Phase I clinical trial of the therapy (EPOC1804) are shown in Figure 6. There were large differences in observed neutrophil numbers, but the predictions showed a good trend towards the overall observations.

其次,為了確定胰臟癌中之TCZ之劑量,關於4種不同劑量:1次4 mg/kg兩週一次(q2w)、1次8 mg/kg兩週一次(q2w)、1次4 mg/kg四週一次(q4w)、1次8 mg/kg四週一次(q4w),預測TCZ+GN併用情況下嗜中性球數之演變。藉由使用上述模型,模擬研究嗜中性球數之減少是否達到針對胰臟癌之Abraxane之隨附說明書中記載之GN之減量基準。其結果,投予開始後7天後,TCZ之劑量4 mg/kg q2w、8 mg/kg q2w、4 mg/kg q4w及8 mg/kg q4w下,分別有26%、27%、23%及27%之患者為劑量減少基準(未達1000 cells/mm 3)以下(圖7及表2)。同樣地,第14天,嗜中性球數未達劑量減少基準之患者分別為18%、21%、18%及22%。又,投予第28天,未恢復至投予延期之判斷基準(未達1500 cells/mm 3)之患者分別為9%、8%、10%、7%。 Second, to determine the dose of TCZ in pancreatic cancer, for 4 different doses: once 4 mg/kg biweekly (q2w), once 8 mg/kg biweekly (q2w), once 4 mg/kg kg once every four weeks (q4w) and 8 mg/kg once every four weeks (q4w), to predict the evolution of neutrophil numbers in the case of combined use of TCZ+GN. By using the model described above, it was simulated whether the reduction in neutrophil number meets the GN reduction benchmark described in the accompanying instructions for Abraxane for pancreatic cancer. As a result, 7 days after the start of administration, the TCZ doses of 4 mg/kg q2w, 8 mg/kg q2w, 4 mg/kg q4w and 8 mg/kg q4w were 26%, 27%, 23% and 23%, respectively. Twenty-seven percent of patients were below the dose reduction baseline (less than 1000 cells/mm 3 ) (Figure 7 and Table 2). Likewise, on day 14, 18%, 21%, 18% and 22% of patients did not meet the dose-reduction baseline for neutrophil counts, respectively. In addition, on the 28th day of administration, 9%, 8%, 10%, and 7% of patients did not recover to the criteria for delaying administration (less than 1500 cells/mm 3 ).

根據模擬之結果,需要劑量變更/投予延期之患者之比率於所研究之全部時間點(第7、14及28天),所研究之各種TCZ劑量(4 mg/kg q2w、8 mg/kg q2w、4 mg/kg q4w及8 mg/kg q4w)之間未顯示臨床上有意義差。Based on the results of the simulation, the ratio of patients requiring a dose change/dosing delay was at all time points studied (Days 7, 14 and 28), at the various TCZ doses studied (4 mg/kg q2w, 8 mg/kg No clinically meaningful differences were shown between q2w, 4 mg/kg q4w, and 8 mg/kg q4w).

若考慮到TCZ之劑量為1次8 mg/kg Q4W時對胰臟癌(參照實施例1)顯示有效性,則可料想1次8 mg/kg Q2W(核准劑量)亦有效果。於針對胰臟癌之TCZ+GN+ATZ併用方案中,我們就安全性之觀點出發,基於經模擬推測之嗜中性球數之見解,得出可按所研究之各種TCZ劑量(1次4 mg/kg Q2W、1次8 mg/kg Q2W、1次4 mg/kg Q4W、及1次8 mg/kg Q4W)進行併用之結論。 [表6] 表2      需要劑量變更/投予延期之患者之比率(%)    Day7 Day14 Day28 嗜中性球數<1500 (cells/mm 3)    嗜中性球數<1000 (cells/mm 3) 嗜中性球數<500 (cells/mm 3) 嗜中性球數 <1000 (cells/mm 3) 嗜中性球數<500 (cells/mm 3) Gem+nabPac 16 6 27 9 11 TCZ q2w_4 mg 26 8 18 6 9 TCZ q2w_8 mg 27 8 21 7 8 TCZ q4w_4 mg 23 7 18 6 10 TCZ q4w_8 mg 27 8 22 8 7 參照預包裝內說明書對Gem+Nab-PTX進行必要調整 減小劑量 -Nab-PTX按 25 mg/m 2-Gem按 200 mg/m 2 暫停 Day8之投予 減小劑量 (個性化) 暫停 Day15之投予 下一投予週期 延期 [實施例5] Considering that the TCZ dose of 8 mg/kg Q4W once showed efficacy on pancreatic cancer (refer to Example 1), it is expected that 8 mg/kg Q2W once (approved dose) will also be effective. In the TCZ+GN+ATZ combination regimen for pancreatic cancer, from the point of view of safety, based on the insights of the neutrophil numbers predicted by simulation, we concluded that the doses of TCZ under study (4 mg/kg Q2W once a time) can be used. , 8 mg/kg Q2W once, 4 mg/kg Q4W once, and 8 mg/kg Q4W once) were used together. [Table 6] Table 2 Percentage of patients requiring dose change/dosing delay (%) Day7 Day14 Day28 Neutrophil number <1500 (cells/mm 3 ) Neutrophil number <1000 (cells/mm 3 ) Neutrophil number <500 (cells/mm 3 ) Neutrophil number <1000 (cells/mm 3 ) Neutrophil number <500 (cells/mm 3 ) Gem+nabPac 16 6 27 9 11 TCZ q2w_4 mg 26 8 18 6 9 TCZ q2w_8 mg 27 8 twenty one 7 8 TCZ q4w_4 mg twenty three 7 18 6 10 TCZ q4w_8 mg 27 8 twenty two 8 7 Refer to the instructions in the prepackage to make necessary adjustments to Gem+Nab-PTX Reduced dose - Nab-PTX at 25 mg/m 2 - Gem at 200 mg/m 2 Suspension of Day8 investment Dose reduction (personalized) Suspend day15 investment Next dosing cycle postponed [Example 5]

實施例 5非臨床自發性胰臟癌轉基因小鼠模型(KPΔC小鼠)中之抗IL-6R抗體+抗PD-L1抗體+吉西他濱(G)併用療法作用下CD45陽性細胞(嗜中性球、淋巴細胞、單核球)、CD3陽性細胞(T細胞)、CD8陽性細胞(細胞毒殺性T細胞、一部分NK細胞)及樹狀細胞等免疫細胞向腫瘤內浸潤之確認。 Example 5 CD45-positive cells (neutrophils, neutrophils, Confirmation of infiltration of immune cells such as lymphocytes, monocytes), CD3 positive cells (T cells), CD8 positive cells (cytotoxic T cells, some NK cells) and dendritic cells into tumors.

包圍腫瘤組織之腫瘤微環境內能夠檢測出多種多樣之免疫細胞之浸潤。近年來逐漸發現,該等免疫細胞並非無害之旁觀者,而是引發胰臟癌之致癌相關因子。因此,藉由對KPΔC小鼠併用投予抗IL-6R抗體、抗PD-L1抗體及吉西他濱,分析腫瘤浸潤淋巴細胞(TIL)之基因表現譜(profiling),探明IL-6於免疫抑制性腫瘤微環境中之詳細作用。為此,使胰臟癌自然發病之轉基因KPΔC小鼠與3種帶有以下所示之遺傳背景之小鼠系統(LSL-Kras G12D(Kras tm1Tyj)(Hingorani等人, 2003; Cancer Cell 4, 437-450)、Ptf1a-cre ex1(Ptf1a tm1(cre)Hnak)(Nakhai等人, 2007; Development 134, 1151-1160)、Trp53 flox/flox(Trp53 tm1Brn)(Marino等人, 2000; Genes Dev 14, 994-1004))之小鼠交配,製作LSL-Kras G12D、Ptf1a-cre ex1、Trp53 flox/flox,進行實驗。 The infiltration of a variety of immune cells can be detected in the tumor microenvironment surrounding the tumor tissue. In recent years, it has been gradually discovered that these immune cells are not harmless bystanders, but are carcinogenic-related factors that cause pancreatic cancer. Therefore, by administering anti-IL-6R antibody, anti-PD-L1 antibody and gemcitabine to KPΔC mice, the gene profiling of tumor-infiltrating lymphocytes (TIL) was analyzed, and the role of IL-6 in immunosuppression was determined. Detailed roles in the tumor microenvironment. For this purpose, transgenic KPΔC mice with natural onset of pancreatic cancer and 3 mouse systems with the genetic background shown below (LSL-Kras G12D (Kras tm1Tyj ) (Hingorani et al., 2003; Cancer Cell 4, 437 -450), Ptf1a-cre ex1 ( Ptf1a tm1(cre)Hnak ) (Nakhai et al., 2007; Development 134, 1151-1160), Trp53 flox / flox (Trp53 tm1Brn ) (Marino et al., 2000; Genes Dev 14, 994-1004)) mice were mated, and LSL-Kras G12D , Ptf1a-cre ex1 , Trp53 flox / flox were produced, and experiments were carried out.

全部試驗均於特定之無病原體條件下飼育小鼠。KPΔC小鼠達到4~5週齡後,將小鼠分成5組,單獨或併用小鼠IgG1同型對照抗體、抗小鼠PD-L1(B7-H1)抗體(購自BioXCell公司)、抗小鼠IL-6R抗體(MR16-1)(中外製藥提供)、及吉西他濱(Klinikum Rechts der Isar(慕尼黑工業大學附屬醫院)之醫院藥局提供))對小鼠進行投予。Mice were bred under specific pathogen-free conditions for all experiments. After KPΔC mice reached 4 to 5 weeks of age, the mice were divided into 5 groups, and the mice were treated with mouse IgG1 isotype control antibody, anti-mouse PD-L1 (B7-H1) antibody (purchased from BioXCell), anti-mouse antibody alone or in combination. IL-6R antibody (MR16-1) (provided by Zhongwai Pharmaceutical) and gemcitabine (provided by the Hospital Pharmacy of Klinikum Rechts der Isar (Affiliated Hospital of Technical University of Munich)) were administered to the mice.

投予之組合如下所示:組1(對照投予組)投予同型對照IgG抗體、組2(IL-6R抗體投予組)投予抗IL-6R抗體、組3(PD-L1抗體投予組)投予抗PD-L1抗體、組4(IL-6R抗體+PD-L1抗體併用投予組)併用投予抗IL-6R抗體與抗PD-L1抗體、以及組5(IL-6R抗體+PD-L1抗體+吉西他濱併用投予組)併用投予抗IL-6R抗體與抗PD-L1抗體及吉西他濱三劑。The combination of administration was as follows: group 1 (control administration group) was administered with isotype control IgG antibody, group 2 (IL-6R antibody administration group) was administered with anti-IL-6R antibody, and group 3 (PD-L1 antibody administration group) was administered with anti-IL-6R antibody. group) anti-PD-L1 antibody, group 4 (IL-6R antibody + PD-L1 antibody combined administration group) combined with anti-IL-6R antibody and anti-PD-L1 antibody, and group 5 (IL-6R antibody +PD-L1 antibody + gemcitabine concomitant administration group) and administered three doses of anti-IL-6R antibody, anti-PD-L1 antibody and gemcitabine in combination.

各種藥劑均經由腹腔內注射投予。對小鼠投予時,將InVivoPlus小鼠IgG1同型對照、InVivoMAb抗小鼠PD-L1抗體(B7-H1)、及抗小鼠IL-6R抗體(MR16-1)利用InVivoPure pH 6.5稀釋緩衝液進行稀釋。按30 μg/kg之劑量對4~5週齡之KPΔC小鼠4次投予(第0、1、7及14天)同型對照抗體或抗IL-6R抗體(MR16-1)。5次投予(第0、3、7、10及14天)抗PD-L1抗體(25 μg/kg)或吉西他濱(100 mg/kg)。投予結束後,使動物安樂死,自胰臟癌組織及脾臟採集細胞,藉由以下記載之方法實施進一步分析。The various agents were administered via intraperitoneal injection. When administered to mice, InVivoPlus mouse IgG1 isotype control, InVivoMAb anti-mouse PD-L1 antibody (B7-H1), and anti-mouse IL-6R antibody (MR16-1) were prepared in InVivoPure pH 6.5 dilution buffer. dilution. KPΔC mice aged 4 to 5 weeks were administered four times (days 0, 1, 7 and 14) an isotype control antibody or an anti-IL-6R antibody (MR16-1) at a dose of 30 μg/kg. Anti-PD-L1 antibody (25 μg/kg) or gemcitabine (100 mg/kg) was administered 5 times (days 0, 3, 7, 10 and 14). After the administration, the animals were euthanized, and cells were collected from pancreatic cancer tissue and spleen, and further analysis was performed by the method described below.

自胰臟癌組織及脾臟分離細胞,製備單細胞化之細胞懸浮液(單細胞懸浮液),藉由螢光激活細胞分選(FACS)評估各自所含之免疫細胞之表型分析。自投予結束後之KPΔC小鼠採集新鮮之胰臟及脾臟,製備各組織之單細胞懸浮液。Cells were isolated from pancreatic cancer tissue and spleen to prepare a single-celled cell suspension (single cell suspension), and the phenotypic analysis of the immune cells contained in each was assessed by fluorescence-activated cell sorting (FACS). Fresh pancreas and spleen were collected from KPΔC mice after administration, and single-cell suspensions of each tissue were prepared.

以下揭示其方法。將採集之胰腺立即切碎,與含有1.2 mg/ml膠原酶IV型(Sigma Aldrich)與0.1 mg/ml大豆胰蛋白酶抑制劑(Sigma Aldrich)之PBS一起於37℃下培養10分鐘,繼而,使之通過70 μm細胞濾網,而製備細胞懸浮液。將採集之脾臟立即粉碎,使之通過70 μm細胞濾網,而製備細胞懸浮液。將各細胞懸浮液分別利用PBS/1%FCS清洗1次,利用紅血球溶解緩衝液(Sigma Aldrich)處理90秒去除紅血球。再次清洗細胞懸浮液後,利用由Fc受體經過含1%FCS之PBS稀釋而成之Fc-block(CD16/32 Fc-block eBiosciences),於20分鐘、4℃之條件下培養,藉此進行阻斷單株抗體之FC區之非特異性結合之處理。其後,使用離心分離機清洗細胞懸浮液,使用經過含1%FCS之PBS稀釋之以下抗體,於30分鐘、避光、4℃之條件下對細胞表面標記物進行染色:CD19(選殖eBio1D3,eBioscience)、F4/80(選殖BM8,eBioscience)、CD11b(選殖M1/70,eBioscience)、CD45(選殖30-F11,Bioscience)、CD3e(選殖145-2C11,eBioscience)、CD4(選殖GK1.5,BioLegend)、CD8(選殖53-6.7,BioLegend)、CD11c(選殖N418,BioLegend)、CD206(選殖C068C2,BioLegend)、Gr-1(選殖RB6-8C5,BioLegend)。採用使用各抗體之單染色試樣實施螢光補償,並使用無染色對照排除假陽性訊號。於流式細胞分析之前,藉由碘化丙啶(PI,Sigma Aldrich)染色評估存活率後,進行門控排除死細胞後,使用Gallios流式細胞儀(Beckman Coulter)實施流式細胞分析。使用FlowJo軟體套件進行資料解析。The method is disclosed below. The harvested pancreas was immediately minced and incubated with PBS containing 1.2 mg/ml collagenase type IV (Sigma Aldrich) and 0.1 mg/ml soybean trypsin inhibitor (Sigma Aldrich) at 37°C for 10 minutes, followed by incubation. The cell suspension was prepared by passing it through a 70 μm cell strainer. The harvested spleen was immediately crushed and passed through a 70 μm cell strainer to prepare a cell suspension. Each cell suspension was washed once with PBS/1% FCS and treated with erythrocyte lysis buffer (Sigma Aldrich) for 90 seconds to remove erythrocytes. After washing the cell suspension again, using Fc-block (CD16/32 Fc-block eBiosciences) prepared by diluting Fc receptors in PBS containing 1% FCS, and incubating at 4°C for 20 minutes. Treatment to block non-specific binding of the FC region of monoclonal antibodies. Thereafter, the cell suspension was washed with a centrifuge, and the cell surface markers were stained with the following antibodies diluted in PBS containing 1% FCS for 30 minutes, protected from light, at 4°C: CD19 (colonized eBio1D3). , eBioscience), F4/80 (selection BM8, eBioscience), CD11b (selection M1/70, eBioscience), CD45 (selection 30-F11, Bioscience), CD3e (selection 145-2C11, eBioscience), CD4 ( GK1.5, BioLegend), CD8 (53-6.7, BioLegend), CD11c (N418, BioLegend), CD206 (C068C2, BioLegend), Gr-1 (RB6-8C5, BioLegend) . Fluorescence compensation was performed using single-stained samples with each antibody, and no-stained controls were used to exclude false positive signals. Flow cytometry was performed using a Gallios flow cytometer (Beckman Coulter) after assessing viability by propidium iodide (PI, Sigma Aldrich) staining prior to flow cytometry and gating to exclude dead cells. Data analysis was performed using the FlowJo software suite.

各圖表中記載之細胞百分比係指細胞表面標記陽性細胞相對於自特定小鼠檢體獲得之活細胞總數之比率。資料係以平均±標準偏差之形式表示。為了進行組間比較,實施雙側學生t檢驗。將統計顯著性設定為*P<0.05、**P<0.01、***P<0.001。使用GraphPad PRISM(註冊商標)8.0軟體進行統計分析。The percentage of cells reported in each graph refers to the ratio of cells positive for cell surface markers relative to the total number of viable cells obtained from a particular mouse specimen. Data are presented as mean ± standard deviation. For comparison between groups, a two-sided Student's t-test was performed. Statistical significance was set as *P<0.05, **P<0.01, ***P<0.001. Statistical analysis was performed using GraphPad PRISM (registered trademark) 8.0 software.

將對流式細胞分析中使用之各種免疫細胞進行鑑定之標記物之組合歸納列於以下之表3。如圖8所示,藉由IL-6R抗體投予組(73.3%)或IL-6R抗體+PD-L1抗體併用投予組(87.1%)之投予,相較於對照投予組(40.8%),浸潤至胰臟癌腫瘤組織中之CD45陽性淋巴細胞(包括嗜中性球、單核球)呈統計學上有意義之浸潤亢進狀態。又,根據胰臟癌腫瘤組織微環境中之免疫應答之詳細解析,表明相較於對照投予組,IL-6R抗體+PD-L1抗體併用投予組中之CD3陽性T細胞、CD8陽性細胞毒殺性T細胞(CTL)、樹狀細胞(DC)及B細胞等免疫細胞之浸潤呈統計學上有意義之亢進狀態(表4)。IL-6R抗體+PD-L1抗體併用投予組較對照投予組之增加比率分別為CD3陽性T細胞:161.4%、CD8陽性CTL:529.2%、DC:152.9%、B細胞:68.9%。進而,藉由併用化學治療劑吉西他濱與抗IL-6R抗體及抗PD-L1抗體,浸潤B細胞較對照組呈統計學上有意義之增加,其比率為487.2%。可知藉由IL-6R抗體+PD-L1抗體+吉西他濱併用投予,相較於對照投予組,不僅是B細胞,所評估之其他免疫細胞向腫瘤內之浸潤亦增加,其比率分別為CD45陽性淋巴細胞:33.4%、CD3陽性及CD45陽性T細胞:69.5%、CD8陽性及CD45陽性CTL:406.5%、以及DC:250.5%。藉由IL-6R抗體+PD-L1抗體+吉西他濱併用投予所產生之B細胞之顯著之浸潤亢進之確認強烈表明後天性免疫之誘導。The combinations of markers used to identify various immune cells used in flow cytometry are summarized in Table 3 below. As shown in Fig. 8, by the administration of the IL-6R antibody administration group (73.3%) or the IL-6R antibody + PD-L1 antibody combined administration group (87.1%), compared with the control administration group (40.8%) ), the CD45-positive lymphocytes (including neutrophils and monocytes) infiltrating into pancreatic cancer tumor tissue showed a statistically significant hyperinfiltration state. In addition, according to the detailed analysis of the immune response in the pancreatic cancer tumor tissue microenvironment, it was shown that the IL-6R antibody + PD-L1 antibody in the combined administration group was killed by CD3 positive T cells and CD8 positive cells compared with the control administration group. The infiltration of immune cells such as sex T cells (CTL), dendritic cells (DC) and B cells was statistically significant (Table 4). The ratios of increase in the combined IL-6R antibody + PD-L1 antibody administration group compared with the control administration group were CD3 positive T cells: 161.4%, CD8 positive CTL: 529.2%, DC: 152.9%, B cells: 68.9%. Furthermore, by using the chemotherapeutic agent gemcitabine in combination with anti-IL-6R antibody and anti-PD-L1 antibody, there was a statistically significant increase in infiltrating B cells compared to the control group, with a ratio of 487.2%. It can be seen that by co-administration of IL-6R antibody + PD-L1 antibody + gemcitabine, compared with the control group, not only B cells, but also other assessed immune cells infiltrated into the tumor, and the ratios were CD45 positive. Lymphocytes: 33.4%, CD3-positive and CD45-positive T cells: 69.5%, CD8-positive and CD45-positive CTLs: 406.5%, and DCs: 250.5%. The confirmation of marked hyperinfiltration of B cells generated by administration of IL-6R antibody + PD-L1 antibody + gemcitabine strongly suggests the induction of acquired immunity.

後天性免疫係樹狀細胞等抗原提呈細胞藉由識別特定之抗原活化而激活T細胞或B細胞之生物體所具有之免疫應答。於本研究中,首次確認藉由對胰臟癌腫瘤組織併用投予IL-6R抗體+PD-L1抗體+吉西他濱,以DC、T細胞、B細胞為代表之CD45陽性免疫細胞向腫瘤內之浸潤亢進,這是證明樹狀細胞藉由識別特定之癌抗原活化而激活T細胞或B細胞,其結果發揮實施例6-2所示之抗腫瘤效果之後天性免疫誘導的重要之有劃時代意義之發現。Antigen-presenting cells such as dendritic cells of the acquired immune system activate T cells or B cells by recognizing specific antigens and activate the immune response of living organisms. In this study, it was confirmed for the first time that by administering IL-6R antibody + PD-L1 antibody + gemcitabine to pancreatic cancer tumor tissue, the infiltration of CD45-positive immune cells represented by DC, T cells, and B cells into the tumor was increased. , this is an important and epoch-making discovery that proves that dendritic cells activate T cells or B cells by recognizing specific cancer antigens, and as a result, the anti-tumor effect shown in Example 6-2 is induced after the induction of innate immunity.

根據該等結果,確認藉由抗IL-6R抗體+抗PD-L1抗體之併用,包括T細胞、以及嗜中性球或單核球在內之主要之免疫細胞向胰臟癌腫瘤組織之浸潤得到顯著促進。於併用抗IL-6R抗體+抗PD-L1抗體之基礎上追加標準治療之吉西他濱,結果不僅維持胰臟癌腫瘤組織中之各種免疫細胞之浸潤,且確認證明藉由T細胞與B細胞之活化而誘導後天性免疫之證據,成功獲得有劃時代意義之見解。Based on these results, it was confirmed that the infiltration of major immune cells including T cells, neutrophils or monocytes into pancreatic cancer tumor tissue by the combined use of anti-IL-6R antibody + anti-PD-L1 antibody been significantly promoted. The addition of standard treatment gemcitabine on the basis of concomitant use of anti-IL-6R antibody + anti-PD-L1 antibody not only maintained the infiltration of various immune cells in pancreatic cancer tumor tissue, but also confirmed that the activation of T cells and B cells And the evidence of inducing acquired immunity has successfully obtained epoch-making insights.

[表7] 表3  對流式細胞分析中使用之各種免疫細胞進行鑑定之標記物一覽 標記物 TIL CD45 淋巴細胞 CD3 T細胞 CD8 細胞毒殺性T細胞 CD11c 樹狀細胞 CD19 B細胞 [Table 7] Table 3 List of markers used to identify various immune cells used in flow cytometry Mark TIL CD45 lymphocytes CD3 T cells CD8 cytotoxic T cells CD11c dendritic cells CD19 B cells

[表8] 表4      浸潤至胰臟癌組織中之各種免疫細胞之陽性細胞率之平均值    組1 (對照抗體投予組) 組2 (IL-6R抗體投予組) 組3 (PD-L1抗體投予組) 組4 (IL-6R抗體+PD-L1抗體併用投予組) 組5 (IL-6R抗體+PD-L1抗體+吉西他濱併用投予組) 陽性細胞平均 (%) CD45+ 40.82 73.29 49.32 87.09 54.47 CD3+ CD45+ 17.80 30.57 19.02 46.53 30.17 CD8+ CD45+ 2.09 2.27 13.59 13.17 10.60 CD11c+ CD45+ 1.62 2.36 0.49 4.09 5.67 CD19+ CD45+ 3.81 9.72 3.91 6.44 22.38 藉由流式細胞分析所獲得之浸潤至KPΔC小鼠之胰臟癌中之CD45陽性細胞、CD3陽性・CD45陽性細胞、CD8陽性・CD45陽性細胞、CD11c陽性・CD45陽性細胞、及CD19陽性・CD45陽性細胞之陽性細胞比率之平均值 [實施例6] [Table 8] Table 4 Average values of positive cell rates of various immune cells infiltrating pancreatic cancer tissue Group 1 (control antibody administration group) Group 2 (IL-6R antibody administration group) Group 3 (PD-L1 antibody administration group) Group 4 (IL-6R antibody + PD-L1 antibody combined administration group) Group 5 (IL-6R antibody + PD-L1 antibody + gemcitabine combined administration group) Average positive cells (%) CD45+ 40.82 73.29 49.32 87.09 54.47 CD3+ CD45+ 17.80 30.57 19.02 46.53 30.17 CD8+ CD45+ 2.09 2.27 13.59 13.17 10.60 CD11c+ CD45+ 1.62 2.36 0.49 4.09 5.67 CD19+ CD45+ 3.81 9.72 3.91 6.44 22.38 CD45-positive cells, CD3-positive, CD45-positive cells, CD8-positive, CD45-positive cells, CD11c-positive, CD45-positive cells, and CD19-positive, CD45-positive cells infiltrating pancreatic cancer in KPΔC mice obtained by flow cytometry Average value of positive cell ratio of positive cells [Example 6]

實施例 6-1抗IL-6R抗體+抗PD-L1抗體+吉西他濱(G)併用療法: 胰臟癌自然發病轉基因小鼠模型(KPΔC小鼠)中之抗腫瘤效果之確認(生長標記物:Ki67及pErk) Example 6-1 Combination therapy of anti-IL-6R antibody + anti-PD-L1 antibody + gemcitabine (G): Confirmation of antitumor effect in a transgenic mouse model of natural onset of pancreatic cancer (KPΔC mice) (growth marker: Ki67 and pErk)

藉由實施作為癌症生長標記物的磷酸化ERK1/2(p-ERK)與Ki67之免疫組織化學染色(IHC)評估,評估抗IL-6R抗體+抗PD-L1抗體+吉西他濱(G)併用對抗腫瘤效果產生之效果。Anti-IL-6R antibody + anti-PD-L1 antibody + gemcitabine (G) combined with The effect of the tumor effect.

如實施例5之記載,對胰臟癌自然發病轉基因小鼠模型(KPΔC小鼠),於小鼠之腹腔內單獨地或組合地投予同型IgG抗體、抗IL-6R抗體、抗PD-L1抗體、吉西他濱。投予之組合示於以下:組1(對照投予組)投予同型對照IgG抗體、組2(IL-6R抗體投予組)投予抗IL-6R抗體、組3(PD-L1抗體投予組)投予抗PD-L1抗體、組4(IL-6R抗體+PD-L1抗體併用投予組)併用投予抗IL-6R抗體與抗PD-L1抗體、以及組5(IL-6R抗體+PD-L1抗體+吉西他濱併用投予組)併用投予抗IL-6R抗體與抗PD-L1抗體與吉西他濱三劑。As described in Example 5, to the transgenic mouse model of natural onset of pancreatic cancer (KPΔC mice), the mice were intraperitoneally administered with isotype IgG antibody, anti-IL-6R antibody, anti-PD-L1 antibody alone or in combination Antibodies, gemcitabine. The combination of administration is shown below: group 1 (control administration group) administration of isotype control IgG antibody, group 2 (IL-6R antibody administration group) administration of anti-IL-6R antibody, group 3 (PD-L1 antibody administration group) administration. group) anti-PD-L1 antibody, group 4 (IL-6R antibody + PD-L1 antibody combined administration group) combined with anti-IL-6R antibody and anti-PD-L1 antibody, and group 5 (IL-6R antibody +PD-L1 antibody + gemcitabine concomitant administration group) and three doses of anti-IL-6R antibody, anti-PD-L1 antibody and gemcitabine were administered in combination.

取出含有胰臟癌之胰臟,於室溫下、於4%(wt/vol)緩衝多聚甲醛中固定一晩,包埋至石蠟中。自FFPE塊切下厚3 μm之胰臟癌組織切片,製成載有胰臟癌組織切片之載玻片,實施蘇木精/伊紅(H/E)染色、天狼星紅染色、或Ki67及p-ERK之免疫組織化學染色(IHC)。IHC中使用以下之抗體:p-ERK1/2(Cell Signaling Technology;1:100)、Ki67(Abcam;1:1000)。使用3,3'-二胺基聯苯胺受質(DAB)套組(Vector Laboratories),藉由抗生物素蛋白-生物素過氧化酶法使染色訊號顯色。藉由省略初級抗體之對照實驗,對各抗體確認染色之特異性。利用裝有AxioCam HRc(Zeiss)相機之Zeiss Axio1攝像裝置取得亮視野圖像。使用Aperio ImageScope Software,進行胰臟癌組織中之IHC染色之定量圖像解析。用各生長標記物陽性細胞核數除以細胞核總數所算出之比率為Ki-67及p-ERK陽性率,以平均值±標準偏差之形式表示。藉由雙側學生t檢驗進行群間比較。統計顯著性設定為*P<0.05、**P<0.01。The pancreas containing pancreatic cancer was removed, fixed in 4% (wt/vol) buffered paraformaldehyde overnight at room temperature, and embedded in paraffin. Pancreatic cancer tissue sections with a thickness of 3 μm were cut from FFPE blocks to prepare slides containing pancreatic cancer tissue sections, and were subjected to hematoxylin/eosin (H/E) staining, Sirius red staining, or Ki67 and Immunohistochemical staining (IHC) for p-ERK. The following antibodies were used in IHC: p-ERK1/2 (Cell Signaling Technology; 1:100), Ki67 (Abeam; 1:1000). The staining signal was visualized by the avidin-biotin peroxidase method using a 3,3'-diaminobenzidine substrate (DAB) kit (Vector Laboratories). The specificity of staining was confirmed for each antibody by omitting control experiments with primary antibodies. Bright field images were acquired using a Zeiss Axio1 camera unit equipped with an AxioCam HRc (Zeiss) camera. Quantitative image interpretation of IHC staining in pancreatic cancer tissue was performed using Aperio ImageScope Software. The ratio calculated by dividing the number of positive nuclei for each growth marker by the total number of nuclei was the positive rate of Ki-67 and p-ERK, and was expressed in the form of mean ± standard deviation. Intergroup comparisons were made by two-sided Student's t-test. Statistical significance was set as *P<0.05, **P<0.01.

如圖9所示,IL-6R抗體+PD-L1抗體+吉西他濱併用投予組之腫瘤組織中之生長標記物Ki-67為7.1%,與對照投予組(19%)相比觀察到統計學上有意義之抑制。同樣地,IL-6R抗體+PD-L1抗體+吉西他濱併用投予組之腫瘤組織中之生長標記物p-ERK為14.4%,與對照投予組(25.2%)相比觀察到統計學上有意義之抑制(表5)。As shown in Fig. 9, the growth marker Ki-67 in the tumor tissue of the IL-6R antibody + PD-L1 antibody + gemcitabine combined administration group was 7.1%, which was statistically significant compared with the control administration group (19%). meaningful inhibition. Similarly, the growth marker p-ERK in the tumor tissue of the IL-6R antibody + PD-L1 antibody + gemcitabine combined administration group was 14.4%, which was statistically significant compared with the control administration group (25.2%). inhibition (Table 5).

綜上所述,藉由IL-6R抗體+PD-L1抗體+吉西他濱併用治療,腫瘤細胞生長標記物Ki-67及p-ERK陽性比率均顯著降低。發現抗IL-6R抗體與抗PD-L1抗體與化學治療劑吉西他濱之併用發揮任何單獨或併用之藥劑均無法獲得之協同效應,表現出顯著之抗腫瘤效果。得出該結果有助於實施例6-2所示之卓越之抗腫瘤效果的結論。In conclusion, the positive rates of tumor cell growth markers Ki-67 and p-ERK were significantly reduced by the combined treatment of IL-6R antibody + PD-L1 antibody + gemcitabine. It was found that the combined use of anti-IL-6R antibody and anti-PD-L1 antibody with the chemotherapeutic agent gemcitabine exerted a synergistic effect that could not be obtained by any single or combined drug, showing a significant anti-tumor effect. It is concluded that this result contributes to the excellent antitumor effect shown in Example 6-2.

[表9] 表5  藉由IHC染色解析獲得之KPΔC小鼠之胰臟癌組織中之生長標記物Ki67及pErk陽性細胞核比率之平均值    組1 (對照抗體投予組) 組2 (IL-6R抗體投予組) 組3 (PD-L1抗體投予組) 組4 (IL-6R抗體+PD-L1抗體併用投予組) 組5 (IL-6R抗體+PD-L1抗體+吉西他濱併用投予組) 平均陽性核(%) Ki-67 18.96 14.41 9.65 8.78 7.08 p-ERK1/2 25.21 17.12 22.35 16.32 14.39 [Table 9] Table 5 The average value of the ratio of growth marker Ki67 and pErk-positive nuclei in pancreatic cancer tissue of KPΔC mice obtained by IHC staining analysis Group 1 (control antibody administration group) Group 2 (IL-6R antibody administration group) Group 3 (PD-L1 antibody administration group) Group 4 (IL-6R antibody + PD-L1 antibody combined administration group) Group 5 (IL-6R antibody + PD-L1 antibody + gemcitabine combined administration group) Average positive nuclei (%) Ki-67 18.96 14.41 9.65 8.78 7.08 p-ERK1/2 25.21 17.12 22.35 16.32 14.39

實施例 6-2抗IL-6R抗體+抗PD-L1抗體+吉西他濱(G)併用療法: 胰臟癌自然發病轉基因小鼠模型(KPΔC小鼠)中之抗腫瘤效果評估之確認(藉由MRI評估之腫瘤體積之縮小) Example 6-2 Anti-IL-6R Antibody + Anti-PD-L1 Antibody + Gemcitabine (G) Combination Therapy: Confirmation of Anti-tumor Effect Evaluation in Pancreatic Cancer Natural Onset Transgenic Mouse Model (KPΔC Mice) (by MRI Assessed tumor volume reduction)

如實施例5之記載,對KPΔC小鼠單獨地或併用地投予同型對照抗體、抗IL-6R抗體、抗PD-L1抗體、吉西他濱,繼而藉由磁共振成像法(MRI)進行腫瘤體積評估。投予組與實施例6-1中記載者相同。KPΔC小鼠於28~35日齡時開始MRI實驗,每週重複進行(Ruess等人, Nat Med. 2018 Jul; 24(7): 954-960)。具體而言,使用獸醫用麻醉裝置(Pentland Medical),藉由持續吸入含2%異氟醚之O 2(Abbott)來實施鎮靜。為了維持麻醉下小鼠之體溫,而適當進行監控,並使用眼用軟膏進行護眼。使用3.0T醫療設備(PHILIPS)內之顯微鏡表面線圈及軸方向多層T2加權快速自旋回波序列(分辨力:0.3×0.3×0.7 mm 3、30層、回波時間=90毫秒、重複時間>3秒)進行圖像收集。藉由使用OsiriX Lite DICOM Viewer,將相鄰切片上腫瘤區域之間之截切稜錐體積相加而算出實體腫瘤體積。 As described in Example 5, KPΔC mice were administered with isotype control antibody, anti-IL-6R antibody, anti-PD-L1 antibody, gemcitabine alone or in combination, followed by tumor volume assessment by magnetic resonance imaging (MRI) . The administration group was the same as that described in Example 6-1. KPΔC mice started MRI experiments at 28-35 days of age and were repeated weekly (Ruess et al., Nat Med. 2018 Jul; 24(7): 954-960). Specifically, sedation was performed by continuous inhalation of 2% isoflurane in O2 (Abbott) using a veterinary anesthesia device (Pentland Medical). In order to maintain the body temperature of the mice under anesthesia, it was properly monitored, and ophthalmic ointment was used for eye protection. Using a microscope surface coil in a 3.0T medical device (PHILIPS) and a multi-layer T2-weighted fast spin echo sequence in the axial direction (resolution: 0.3×0.3×0.7 mm 3 , 30 slices, echo time = 90 ms, repetition time > 3 s) for image collection. Solid tumor volume was calculated by adding the volume of truncated pyramids between tumor areas on adjacent sections using the OsiriX Lite DICOM Viewer.

以投予開始時(基線)之含有胰臟癌之胰臟體積作為基準,以%算出上述含有胰臟癌之胰臟體積增加率之比,逐個小鼠進行繪圖,並進行投予組間比較,結果發現如圖10所示,相較於其他任何治療組,抗IL-6R抗體+抗PD-L1抗體+化學療法吉西他濱併用治療幾乎對全部小鼠產生顯著抑制含有胰臟癌之胰臟體積之增加比率之效果。IL-6R抗體+PD-L1抗體+吉西他濱投予組自基線之平均胰臟體積變化率為37.15%,相較於IL-6R抗體+PD-L1抗體併用投予組(167.33%)及對照投予組(183.07%),明顯確認到腫瘤生長抑制效果(表6)。Based on the volume of the pancreas containing pancreatic cancer at the start of administration (baseline), the ratio of the above-mentioned increase rate of the volume of the pancreas containing pancreatic cancer was calculated in %, plotted for each mouse, and compared between administration groups , it was found that as shown in Figure 10, the combined treatment of anti-IL-6R antibody + anti-PD-L1 antibody + chemotherapy gemcitabine significantly inhibited the volume of pancreas containing pancreatic cancer in almost all mice compared to any other treatment group The effect of increasing the ratio. The mean pancreatic volume change rate from baseline in the IL-6R antibody + PD-L1 antibody + gemcitabine administration group was 37.15%, compared with the IL-6R antibody + PD-L1 antibody combined administration group (167.33%) and the control administration group (183.07%), the tumor growth inhibitory effect was clearly confirmed (Table 6).

進而,經過本研究新發現了相較於抗IL-6R抗體+抗PD-L1抗體之2劑併用投予,將抗IL-6R抗體+抗PD-L1抗體與標準化學療法吉西他濱組合之三劑併用投予更能發揮抗腫瘤效果,顯著抑制含有胰臟癌之胰臟體積之增加,其效果超過先前技術(非專利文獻1:Mace TA. et al., Gut. 2018 Feb; 67(2): 320-332)中報告之抗IL-6R抗體+抗PD-L1抗體之併用效果。Furthermore, as a result of this study, it was newly discovered that three doses of anti-IL-6R antibody + anti-PD-L1 antibody combined with standard chemotherapy gemcitabine were administered compared to two doses of anti-IL-6R antibody + anti-PD-L1 antibody in combination. Concomitant administration can exert more anti-tumor effect, significantly suppress the increase in the volume of pancreas including pancreatic cancer, and its effect exceeds that of the prior art (Non-Patent Document 1: Mace TA. et al., Gut. 2018 Feb; 67(2) : 320-332) reported the combined effect of anti-IL-6R antibody + anti-PD-L1 antibody.

根據以前之文獻可知於對該KPΔC小鼠單獨投予吉西他濱時無抗腫瘤效果(Wörmann等人, Gastroenterology. 2016 Jul; 151(1): 180-193),因此確認,藉由對抗IL-6R抗體與抗PD-L1抗體併用吉西他濱,發揮單獨投予或其他組合併用無法達成之協同效應,表現出顯著之抗腫瘤效果。According to previous literature, there was no antitumor effect when gemcitabine was administered alone to the KPΔC mice (Wörmann et al., Gastroenterology. 2016 Jul; 151(1): 180-193), so it was confirmed that by anti-IL-6R antibody The use of gemcitabine in combination with anti-PD-L1 antibody exerts a synergistic effect that cannot be achieved by single administration or other combined use, showing a significant anti-tumor effect.

綜上所述,證明了抗IL-6R抗體+抗PD-L1抗體與胰臟癌標準化學療法吉西他濱之併用治療法發揮先前技術無法達成之優異之協同效應而表現出顯著之抗腫瘤效果這一劃時代發現。To sum up, it is proved that the combination therapy of anti-IL-6R antibody + anti-PD-L1 antibody and the standard chemotherapy of pancreatic cancer gemcitabine exerts an excellent synergistic effect that cannot be achieved by the prior art, and shows a significant anti-tumor effect. An epoch-making discovery.

[表10] 表6  藉由MRI獲得之KPΔC小鼠之含有胰臟癌之胰臟體積自基線之變化率之平均值    組1 (對照抗體投予組) 組2 (IL-6R抗體投予組) 組3 (PD-L1抗體投予組) 組4 (IL-6R抗體+PD-L1抗體併用投予組) 組5 (IL-6R抗體+PD-L1抗體+吉西他濱併用投予組) 平均胰腺體積變化率(%) 183.07 130.36 169.14 167.33 37.15 [Table 10] Table 6 Mean value of the rate of change from baseline in the volume of pancreas containing pancreatic cancer in KPΔC mice obtained by MRI Group 1 (control antibody administration group) Group 2 (IL-6R antibody administration group) Group 3 (PD-L1 antibody administration group) Group 4 (IL-6R antibody + PD-L1 antibody combined administration group) Group 5 (IL-6R antibody + PD-L1 antibody + gemcitabine combined administration group) Average pancreatic volume change rate (%) 183.07 130.36 169.14 167.33 37.15

實施例 6-3臨床上抗IL-6R抗體作用下PD-L1陽性癌細胞之增加 於實施例6-2中,確認了抗IL-6R抗體+抗PD-L1抗體+吉西他濱(G)併用療法對胰臟癌自然發病轉基因小鼠模型(KPΔC小鼠)之抗腫瘤效果。驗證藉由抗IL-6R抗體+抗PD-L1抗體+吉西他濱(G)+白蛋白結合紫杉醇(N)併用療法能夠對人類胰臟癌發揮同樣之抗腫瘤效果。 Example 6-3 Clinical increase of PD-L1 positive cancer cells by anti-IL-6R antibody In Example 6-2, the combination therapy of anti-IL-6R antibody + anti-PD-L1 antibody + gemcitabine (G) was confirmed Antitumor effect on natural onset transgenic mouse model of pancreatic cancer (KPΔC mice). It was verified that the anti-IL-6R antibody + anti-PD-L1 antibody + gemcitabine (G) + nab-paclitaxel (N) combination therapy can exert the same anti-tumor effect on human pancreatic cancer.

於實施例1記載之「針對吉西他濱與白蛋白結合紫杉醇無反應之轉移性胰臟癌的托珠單抗與吉西他濱-白蛋白結合紫杉醇併用療法之I期臨床試驗(EPOC1804)」中,徵得患者之知情同意後,於第0天之GN+TCZ投予前(pre)及第28天之藥劑投予後(post),使用18號針,自患者之肝轉移部位採集胰臟癌組織,立即利用10%中性福馬林固定。於48小時以內對福馬林固定之針活檢腫瘤檢體開始脫水操作,進行石蠟包埋而製成腫瘤檢體塊(FFPE塊)。對於判斷不適合進行GN+TCZ投予前後之腫瘤微環境變化解析之症例及無法獲得GN+TCZ投予後之檢體之症例之外的7個症例,藉由以下之方法解析胰臟癌肝轉移灶之腫瘤微環境中之PD-L1表現之變化。In the "Phase I clinical trial of tocilizumab and gemcitabine-nab-paclitaxel combination therapy for metastatic pancreatic cancer unresponsive to gemcitabine and nab-paclitaxel" described in Example 1 (EPOC1804), a patient After informed consent, the pancreatic cancer tissue was collected from the patient's liver metastases using an 18-gauge needle before the administration of GN+TCZ on day 0 (pre) and after the administration of the drug on day 28 (post), and immediately used 10% Neutral formalin fixation. Within 48 hours, the formalin-fixed needle biopsy tumor specimens were dehydrated and embedded in paraffin to prepare tumor specimen blocks (FFPE blocks). For the 7 cases other than the cases in which it was judged to be unsuitable for the analysis of the changes in the tumor microenvironment before and after GN+TCZ administration and the cases in which the specimen after GN+TCZ administration could not be obtained, the tumor microenvironment of pancreatic cancer liver metastases was analyzed by the following method Changes in PD-L1 expression.

使用切片機自製作之腫瘤檢體FFPE塊切下厚4 μm之腫瘤組織薄切片,貼附於載玻片上而製作腫瘤切片檢體。使用15種識別各種免疫細胞之蛋白質之初級抗體,對該腫瘤切片實施免疫組織化學螢光多重染色(Gerdes. M. J. et al. PNAS 110, 11982-11987 (2013))。免疫組織化學螢光多重染色分析存在如下優點:具有與標準之免疫組織化學染色相同之染色特性,且於同一細胞上直接同時比較評估複數個生物標記,藉此可鑑定特定之細胞集群。將分別與2種氰色素標記(Cy3、Cy5)直接結合之初級抗體設為一對,按照各染色循環(round)進行免疫染色,取得每個循環之染色之螢光免疫染色圖像。各循環後使色素標記失活,藉此對1枚腫瘤切片檢體反覆進行複數個染色循環,最終藉由重合各染色資料之圖像解析而取得多重染色資料。具體而言,使用如表AA所示之15種初級抗體,藉由如表BB所示之標記物之組合來鑑定特定之細胞集群。免疫組織化學螢光多重染色中使用以下之抗體作為初級抗體:CD15(選殖Carb-3,Dako)、HEPATOCYTE ANTIGEN(選殖OCH1E5,Abcam)、CK-19(選殖A53-B/A2,Abcam)、CD3(選殖F7.2.38,Dako)、CD4(選殖EPR6855,Abcam)、CD8(選殖C8/144B,Dako)、CD14(選殖EPR3652,Abcam)、α-SMA(選殖1A4,Sigma-Aldrich)、PD-L1(選殖SP263,Ventana)、Collagen IV(選殖EPR20966,Abcam)、CD31(選殖89C2,Cell Signaling Technologies)、FOXP3(選殖206D,BioLegend)、PD-1(選殖EPR4877,Abcam)、Ki67(選殖SP6,Abcam)、CD68(選殖KP1,BioLegend)。 [表11] 表AA免疫組織化學螢光多重染色之染色循環一覽 Round Cy3 Cy5 1 CD15 PD-L1 2 HEPATOCYTE ANTIGEN COLLAGEN IV 3 CK-19 CD31 4 CD3 FOXP3 5 CD4 PD-1 6 CD8 Ki67 7 CD14 CD68 8 α-SMA    [表12] 表BB藉由各種染色標記物之組合而鑑定之特定之細胞集群一覽 共表達表型 CD3+CD4+ 輔助性T細胞 CD3+CD4+FOXP3+ 調節性T細胞 CD3+PD-1+ 表達PD-1之T細胞 CD3+CD8+ 細胞毒性T細胞 CD3+CD8+PD-1+ 下調/失能之細胞毒性T細胞 CD3+CD8+Ki67+ 生長狀態之細胞毒性T細胞 CD68+PD-L1+ 表達PD-L1之巨噬細胞 CD3+CD4+Ki67+ 生長狀態之輔助性T細胞 PD-L1+CK-19+ 表達PD-L1之腫瘤細胞 CK-19+Ki67+ 生長狀態之腫瘤細胞 α-SMA+Ki67+ 生長狀態之SMA陽性細胞 α-SMA+PDL1+ 表達PD-L1之SMA陽性細胞 CD31+Ki67+ 生長狀態之CD31陽性細胞 CD68+Ki67+ 生長狀態之巨噬細胞 HFPATOCYTE ANTIGEN HERATOCYTE COLLAGEN IV 膠原蛋白(細胞間基質) Using a microtome, a thin section of tumor tissue with a thickness of 4 μm was cut from the FFPE block of the tumor specimen, and attached to a glass slide to prepare a tumor section specimen. The tumor sections were subjected to immunohistochemical fluorescence multiplex staining using 15 primary antibodies that recognize proteins of various immune cells (Gerdes. MJ et al. PNAS 110, 11982-11987 (2013)). Immunohistochemical fluorescence multiplexing assays have the advantage of having the same staining properties as standard immunohistochemical staining and the direct simultaneous comparative assessment of multiple biomarkers on the same cell, thereby allowing the identification of specific cell clusters. A pair of primary antibodies directly conjugated to two cyanochrome labels (Cy3, Cy5) was used to perform immunostaining according to each staining round, and a fluorescent immunostaining image of the staining in each round was obtained. After each cycle, the pigment label is inactivated, whereby a plurality of staining cycles are repeatedly performed on one tumor section specimen, and finally multiple staining data are obtained by superimposing the image analysis of each staining data. Specifically, using the 15 primary antibodies shown in Table AA, specific cell populations were identified by the combination of markers shown in Table BB. The following antibodies were used as primary antibodies in immunohistochemical fluorescence multiplex staining: CD15 (clone Carb-3, Dako), HEPATOCYTE ANTIGEN (clone OCH1E5, Abcam), CK-19 (clone A53-B/A2, Abcam) ), CD3 (clone F7.2.38, Dako), CD4 (clone EPR6855, Abcam), CD8 (clone C8/144B, Dako), CD14 (clone EPR3652, Abcam), α-SMA (clone 1A4, Sigma-Aldrich), PD-L1 (Collection SP263, Ventana), Collagen IV (Collection EPR20966, Abcam), CD31 (Collection 89C2, Cell Signaling Technologies), FOXP3 (Collection 206D, BioLegend), PD-1 (Collection 89C2, Cell Signaling Technologies) EPR4877, Abcam), Ki67 (SP6, Abcam), CD68 (KP1, BioLegend) were cloned. [Table 11] Table AA List of staining cycles for immunohistochemical fluorescence multiplex staining Round Cy3 Cy5 1 CD15 PD-L1 2 HEPATOCYTE ANTIGEN COLLAGEN IV 3 CK-19 CD31 4 CD3 FOXP3 5 CD4 PD-1 6 CD8 Ki67 7 CD14 CD68 8 α-SMA [Table 12] Table BB Summary of specific cell clusters identified by various combinations of staining markers co-expression phenotype CD3+CD4+ helper T cells CD3+CD4+FOXP3+ regulatory T cells CD3+PD-1+ T cells expressing PD-1 CD3+CD8+ cytotoxic T cells CD3+CD8+PD-1+ Down-regulated/disabled cytotoxic T cells CD3+CD8+Ki67+ Cytotoxic T cells in growing state CD68+PD-L1+ Macrophages expressing PD-L1 CD3+CD4+Ki67+ helper T cells in growth state PD-L1+CK-19+ Tumor cells expressing PD-L1 CK-19+Ki67+ growing tumor cells α-SMA+Ki67+ SMA-positive cells in growth state α-SMA+PDL1+ SMA-positive cells expressing PD-L1 CD31+Ki67+ CD31 positive cells in growth state CD68+Ki67+ growing macrophage HFPATOCYTE ANTIGEN HERATOCYTE COLLAGEN IV Collagen (Intercellular Matrix)

鑑定各腫瘤切片檢體之作為CK-19陽性區域之腫瘤區域,算出複數個評估區域(Region of Interest;ROI)所含之每個特定之細胞之ROI內平均每mm 2之細胞數後,算出各個腫瘤區域之特定細胞密度之平均值,以平均值±標準誤差之形式表示。藉由威爾卡遜(Wilcoxon)檢驗進行群間比較。使用NeoVUE或ImageJ軟體進行螢光染色圖像之解析。並實施相鄰腫瘤切片之蘇木精-伊紅(H/E)染色,得到光學顯微鏡水平之腫瘤區域及免疫細胞之組織結構之整體圖像。將該等結果示於圖13。 The tumor region as the CK-19 positive region in each tumor section was identified, and the average number of cells per mm2 in the ROI for each specific cell included in a plurality of evaluation regions (Region of Interest; ROI) was calculated. Mean values of specific cell densities for each tumor area are presented as mean ± standard error. Intergroup comparisons were made by Wilcoxon test. Fluorescence staining images were analyzed using NeoVUE or ImageJ software. Hematoxylin-eosin (H/E) staining of adjacent tumor sections was performed to obtain the overall image of the tumor area and the tissue structure of immune cells at the light microscope level. These results are shown in FIG. 13 .

可知無論實施例1所示之治療成效如何,腫瘤組織內之PD-L1陽性腫瘤細胞密度(每mm 2之個數)之平均值於投予前(pre)為69.04,相對於此,GN+TCZ投予後(post)為239.27,較投予前增加246.57%。威爾卡遜檢驗中之P=0.0845,可確認出現明顯增加。據此得出如下結論事項:於GN+TCZ投予後,由於癌細胞之PD-L1表現亢進所引起之免疫檢查點之亢進作用而抑制對腫瘤之免疫作用。 It can be seen that regardless of the therapeutic effect shown in Example 1, the average value of the PD-L1-positive tumor cell density (number per mm 2 ) in the tumor tissue before the administration (pre) was 69.04. After the injection (post) was 239.27, an increase of 246.57% compared with that before the injection. P=0.0845 in the Wilcarson test, confirming a significant increase. Based on this, the following conclusions are drawn: After GN+TCZ administration, the immune effect on the tumor is suppressed due to the hyperactivity of the immune checkpoint caused by the hyperactivity of PD-L1 expression in cancer cells.

無論實施例1所示之治療成效如何,腫瘤組織內之IL-6訊號下游之因子STAT3之磷酸化(phosho-STAT3)陽性面積比率於GN+TCZ投予後(0.018%)較投予前(0.031%)減少41.94%(p=0.0832),眾所周知,STAT3磷酸化抑制為TCZ之生物標記,因此,於所獲得之腫瘤檢體中明確地確認到GN+TCZ投予之效果(圖14)。Regardless of the therapeutic effect shown in Example 1, the ratio of phosphorylation of STAT3, a factor downstream of IL-6 signaling in tumor tissue (phosho-STAT3), was significantly higher after GN+TCZ administration (0.018%) than before administration (0.031%). It was reduced by 41.94% (p=0.0832). It is well known that inhibition of STAT3 phosphorylation is a biomarker of TCZ. Therefore, the effect of GN+TCZ administration was clearly confirmed in the obtained tumor specimens ( FIG. 14 ).

綜合該等結果,基本上看不到於GN無反應患者之腫瘤細胞上表現PD-L1之現象,GN+TCZ投予後PD-L1表現明顯增加,由此可確認TCZ併用投予產生了意想不到之效果。圖15表示源於實施例2及實施例3所示之受驗者(GAP01)之試樣之免疫組織化學螢光多重染色之解析結果。圖中箭頭表示PD-L1陽性癌細胞。Based on these results, it is basically impossible to see the phenomenon of PD-L1 expression on tumor cells of GN non-responsive patients, and the expression of PD-L1 is significantly increased after GN + TCZ administration, which confirms that TCZ combined administration produces unexpected effects. . 15 shows the analysis results of immunohistochemical fluorescence multiplex staining of samples derived from the subjects (GAPO1) shown in Example 2 and Example 3. FIG. Arrows in the figure indicate PD-L1 positive cancer cells.

綜上所述,確認於胰臟癌中GN+TCZ療法引起PD-L1表現亢進而誘導腫瘤之免疫檢查點,揭示了佐證於人類臨床上藉由將以抗PD-L1抗體阿替利珠單抗為代表之免疫檢查點抑制劑與GN+TCZ療法併用而發揮GN+TCZ併用療法或單獨療法無法達成之抗腫瘤效果的根據。In conclusion, it is confirmed that GN+TCZ therapy induces PD-L1 hyperactivity in pancreatic cancer and induces tumor immune checkpoints, revealing evidence in human clinical practice by using the anti-PD-L1 antibody atezolizumab as an immune checkpoint. The representative immune checkpoint inhibitor is used in combination with GN+TCZ therapy to exert an antitumor effect that cannot be achieved by GN+TCZ combination therapy or single therapy.

總結已有資料暗示腫瘤組織內之PD-L1表現上升有助於免疫檢查點抑制劑阿替利珠單抗之效果增強(背景技術)。確認於自投予GN+TCZ後之患者之胰臟癌肝轉移灶採集之腫瘤組織檢體中PD-L1陽性癌細胞增加,該等組織中之IL-6訊號下游之因子STAT3之磷酸化得到顯著抑制,從而確認TCZ之效果。該結果暗示GN+TCZ療法誘導腫瘤之免疫檢查點,揭示了佐證藉由對人類胰臟癌之GN+TCZ療法追加併用免疫檢查點抑制劑阿替利珠單抗而實現單獨投予或其他組合無法達成之抗腫瘤效果的根據。 [實施例7] Summary of existing data suggests that increased PD-L1 expression in tumor tissue contributes to the enhanced efficacy of the immune checkpoint inhibitor atezolizumab (Background Art). It was confirmed that PD-L1-positive cancer cells increased in tumor tissue samples collected from pancreatic cancer liver metastases of patients after administration of GN+TCZ, and the phosphorylation of STAT3, a factor downstream of IL-6 signaling in these tissues, was significantly inhibited , thus confirming the effect of TCZ. These results suggest that GN+TCZ therapy induces immune checkpoints in tumors, revealing evidence that by supplementing GN+TCZ therapy for human pancreatic cancer with the immune checkpoint inhibitor atezolizumab, resistance that cannot be achieved by administration alone or in other combinations can be achieved Basis for tumor effect. [Example 7]

實施例 7研究將無全身療法治療史之轉移性胰腺導管腺癌患者作為對象之以免疫療法為基礎之複數種併用療法之有效性與安全性的多中心共同、非盲檢、隨機化、Ib/II期傘式試驗(MORPHEUS-PANCREATIC CANCER)。 Example 7 To study the efficacy and safety of multiple immunotherapy-based concomitant therapies in patients with metastatic pancreatic ductal adenocarcinoma without a history of systemic therapy. Multicenter, unblinded, randomized, Ib / Phase II umbrella trial (MORPHEUS-PANCREATIC CANCER).

設計該Ib/II期非盲檢多中心隨機化試驗之目的在於評估以免疫療法為基礎之併用療法於轉移性胰臟導管腺癌(PDAC)參加患者中之有效性、安全性、及藥物動態。以下簡單說明試驗之具體目的及相應之評估項目。This phase Ib/II open-label, multicenter, randomized trial was designed to evaluate the efficacy, safety, and pharmacodynamics of immunotherapy-based concomitant therapy in enrolled patients with metastatic pancreatic ductal adenocarcinoma (PDAC). . The specific purpose of the test and the corresponding evaluation items are briefly described below.

[表13-1] 有效性之主要目的 對應之評估項目 l   評估臨床試驗治療中之併用免疫療法之有效性 l   臨床試驗治療期間,間隔4週以上之2次連續之時間點上確認到的研究(助理研究)者基於RECIST v1.1所判定之客觀緩解(定義:完全緩解或部分緩解) 有效性之次要目的 對應之評估項目 l   評估臨床試驗治療中之併用免疫療法之有效性 l   研究(助理研究)者基於RECIST v1.1所判定之臨床試驗治療中之隨機化後之PFS(定義:自隨機化至第一次病情進展或任何原因死亡之任一先發事件之間的時間) l   隨機化後之OS(定義:自隨機化至任何原因死亡之間的時間) l   特定之時間點(例如6個月)中之OS(助理研究者) l   相對於研究(助理研究)者基於RECIST v1.1所判定之臨床試驗治療中之DOR(定義為:自第一次確認客觀緩解至病情進展或任何原因死亡之任一先發事件之間的時間)的,定義為自第一次書面記錄產生客觀腫瘤縮小效果開始之時間的研究(助理研究)者基於RECISTv1.1所判定之病情控制(定義:12週以上之穩定、或者完全緩解或部分緩解) [Table 13-1] main purpose of effectiveness Corresponding assessment items l Evaluate the effectiveness of concomitant immunotherapy in clinical trial treatments l During the treatment period of the clinical trial, the objective remission (definition: complete remission or partial remission) determined by RECIST v1. secondary purpose of effectiveness Corresponding assessment items l Evaluate the effectiveness of concomitant immunotherapy in clinical trial treatments l PFS (definition: PFS between randomization and first disease progression or any preceding event of death from any cause) based on RECIST v1.1 Time) l OS after randomization (definition: time from randomization to death from any cause) l OS at a specified time point (eg 6 months) (Assistant Investigator) l Relative to study (Associate Study) Those based on the DOR (defined as the time between the first confirmed objective response to disease progression or any preceding event of death from any cause) in clinical trial treatment as determined by RECIST v1. A study (assistant study) with a written record of the time to onset of an objective tumor shrinkage effect based on disease control as determined by RECIST v1.1 (definition: stable for more than 12 weeks, or complete remission or partial remission)

[表13-2] 有效性之探索目的 對應之評估項目 l   評估臨床試驗治療中之併用免疫療法之有效性 l   研究(助理研究)者基於iRECIST所判定之客觀緩解 l   研究(助理研究)者基於iRECIST所判定之隨機化後之PFS l   研究(助理研究)者基於iRECIST所判定之DOR l   研究(助理研究)者基於iRECIST所判定之病情控制 l   CA19-9之自基線時起至其後之時間點為止的變化量 安全性之目的 對應之評估項目 l   評估臨床試驗治療中之併用免疫療法之安全性 l   不良事件及臨床檢查值異常之出現率、性質及重症度。重症度依據美國國立癌研究所之不良事件通用術語標準4.0版進行確定。 l   生命徵象與心電圖(ECG)參數相對於基線之變化量 l   目標臨床檢查值相對於基線之變化量 藥動學之探索目的 對應之評估項目 l   探明作為臨床試驗治療中併用免疫療法之一環所投予之藥劑之藥動學圖譜(PK profile)之特徵 l   特定之時間點上之各藥劑(適宜)之血漿中濃度或血清中濃度 l   評估臨床試驗治療中之藥劑之暴露與併用免疫療法之有效性及安全性的潛在關係 l   各藥劑之血漿中濃度或血清中濃度或PK參數(基於所獲得資料而適宜)與有效性評估項目之關係 l   各藥劑之血漿中濃度或血清中濃度或PK參數(基於所獲得資料而適宜)與安全性評估項目之關係 ADA=抗藥物抗體;DOR=緩解期;iRECIST=適用於免疫療法之改良之RECIST v1.1;OS=總生存期;PFS=無進展生存期;PK=藥動學;RECIST=實體癌療效判定基準 註:單一時間點上之總療效由研究(助理研究)者採用RECIST v1.1進行評估。基於iRECIST之總療效不記錄於eCRF上,贊助者基於經過研究(助理研究)者評估並記錄於eCRF上之各個病變資料,根據程式計算。 [Table 13-2] Exploring Purpose of Effectiveness Corresponding assessment items l Evaluate the effectiveness of concomitant immunotherapy in clinical trial treatments l Study (assistant study) based on iRECIST-judged objective response l Study (assistant study) based on iRECIST-judged post-randomized PFS l Study (assistant study) based on iRECIST-judged DOR l Study (assistant study) The amount of change from baseline to subsequent time points in CA19-9 based on disease control determined by iRECIST security purpose Corresponding assessment items l Evaluate the safety of concurrent immunotherapy in clinical trial treatments l The occurrence rate, nature and severity of adverse events and abnormal clinical examination values. Severity was determined according to the National Cancer Institute's General Terminology Criteria for Adverse Events, Version 4.0. l Changes from baseline in vital signs and electrocardiogram (ECG) parameters l Changes from baseline in target clinical examination values Exploratory Purpose of Pharmacokinetics Corresponding assessment items l Identify the characteristics of the pharmacokinetic profile (PK profile) of the drug administered as part of the concurrent immunotherapy in the clinical trial treatment l Plasma or serum concentration of each agent (appropriate) at a specific time point l Assess the potential relationship between exposure to an agent in a clinical trial treatment and the efficacy and safety of concomitant immunotherapy l The relationship between the plasma concentration or serum concentration or PK parameter of each drug (appropriate based on the obtained data) and the efficacy evaluation items l The plasma concentration or serum concentration or PK parameter of each drug (appropriate based on the obtained data) ) and the relationship between safety assessment items ADA=anti-drug antibody; DOR=period of remission; iRECIST=modified RECIST v1.1 for immunotherapy; OS=overall survival; PFS=progression-free survival; PK=pharmacokinetics; Benchmark Note: Overall efficacy at a single time point was assessed by the investigator (assistant investigator) using RECIST v1.1. The total efficacy based on iRECIST was not recorded on the eCRF, and was calculated by the sponsor based on the individual lesion data assessed by the researcher (assistant researcher) and recorded on the eCRF.

試驗設計本試驗由無針對轉移性PDAC之全身療法治療史之患者構成,將有參加資格之患者隨機分配至對照藥組[化學治療劑:吉西他濱+nab-紫杉醇(Gem/Nab-pac)]或試驗藥組[對阿替利珠單抗(Atezolizumab、Atezo)與化學治療劑併用托珠單抗(Tocilizumab、TCZ)之(Atezo+Chemo+TCZ)]之任一組(概要見圖11)。 Trial Design The trial consisted of patients with no history of systemic therapy for metastatic PDAC, and eligible patients were randomly assigned to either the control group [chemotherapeutic agent: gemcitabine + nab-paclitaxel (Gem/Nab-pac)] or the trial Any of the drug groups [Atezo + Chemo + TCZ for atezolizumab (Atezolizumab, Atezo) combined with a chemotherapeutic agent (Atezo + Chemo + TCZ)] (see Figure 11 for an overview).

Atezo+Chemo+TCZ試驗藥組之入組分兩期(預備期與其次之擴大期)進行。為了確保足夠數量之C-反應性蛋白高值患者,促進該部分群體中之效益風險評估,於預備期入組約30名患者。於預備期在Atezo+Chemo+TCZ組中觀察到臨床效果之情形時,於擴大期進而對該投予組追加入組約25例患者。為了保持投予組間關於潛在之預測性生物標記等之人口統計學特性及基線特性之均衡,實現進一步之部分群體解析,可允許追加患者入組。隨機化時,考慮到Atezo+Chemo+TCZ組之特異性排除基準,分配至對照組之隨機化比率不得超過35%。Atezo+Chemo+TCZ test drug group was enrolled in two phases (preparatory phase and second expansion phase). In order to ensure a sufficient number of patients with high C-reactive protein values and to facilitate benefit-risk assessment in this part of the population, about 30 patients were enrolled in the preliminary phase. When clinical effects were observed in the Atezo+Chemo+TCZ group in the preliminary phase, about 25 patients were added to the administration group in the expansion phase. In order to maintain the balance of demographic characteristics and baseline characteristics with respect to potential predictive biomarkers, etc. between the administration groups, and to achieve further partial population analysis, additional patient enrollment may be allowed. During randomization, taking into account the specific exclusion criteria of the Atezo+Chemo+TCZ group, the randomization rate assigned to the control group should not exceed 35%.

對於對照藥組之患者,持續投予直至依據不可接受之毒性或實體癌療效判定基準第1.1版(RECIST v1.1)確認到病情進展。對於試驗藥組之患者,綜合評估X射線圖像檢查及生物化學檢查之資料、局部活檢之結果(能夠獲取的話)、以及臨床狀態(例如疾病引起之疼痛等徵候之惡化),持續實施如表A中簡要記載之投予,直至確認到經研究(助理研究)者判斷為不可接受之毒性或失去臨床效益。阿替利珠單抗及其他癌症免疫療法(CIT)所誘發之T細胞應答引起免疫細胞浸潤而有初期腫瘤量增大(稱為假性惡化)之可能性,因此,基於RECIST v1.1之放射線學上所見之進展有時並不能代表真實之病情進展。如未確認到不可接受之毒性,於實施CIT併用療法過程中滿足依據RECIST v1.1之病情進展基準之患者在符合以下基準之情況下,允許繼續治療。 ●     研究(助理研究)者評審所獲得之全部資料後判定存在臨床效益之證據 ●     未觀察到明確顯示病情進展之症狀及徵候(包括高鈣血症之新發現或惡化等臨床檢查值) ●     未觀察到一般認為由病情進展引起之ECOG之日常體能狀態之惡化 ●     未觀察到無法藉由臨床試驗實施計劃書中許可之醫學介入進行管理的重要之解剖學部位上之腫瘤進展(例如軟腦膜疾病) For patients in the comparator arm, dosing was continued until disease progression was confirmed based on unacceptable toxicity or Response Criteria in Solid Cancer Version 1.1 (RECIST v1.1). For patients in the experimental drug group, comprehensively evaluate the data of X-ray image examination and biochemical examination, the results of local biopsy (if available), and clinical status (such as the deterioration of symptoms such as pain caused by the disease), and continue to implement the following table. Administration as briefly described in A until confirmation of unacceptable toxicity or loss of clinical benefit judged by the study (assistant study). T-cell responses induced by atezolizumab and other cancer immunotherapies (CITs) cause immune cell infiltration with the potential for initial tumor growth (called pseudomalignancy). Therefore, based on RECIST v1.1 Progress seen on radiology sometimes does not represent true disease progression. If no unacceptable toxicity is identified, patients who meet the criteria for disease progression according to RECIST v1.1 during the course of CIT concomitant therapy are allowed to continue treatment if they meet the following criteria. ● The researcher (assistant researcher) judges that there is evidence of clinical benefit after reviewing all the data obtained ● Symptoms and signs that clearly indicate disease progression (including clinical examination values such as new findings or worsening of hypercalcemia) have not been observed ● No deterioration in daily performance status of ECOG, generally believed to be caused by disease progression, was observed ● No observed tumor progression (eg, leptomeningeal disease) in important anatomical sites that cannot be managed by medical intervention approved in the clinical trial implementation plan

[表14] 表A Atezo+Chemo+TCZ組之治療方案 週期長度 投予量、投予路徑、及方案 (藥劑按投予順序顯示) 28天 •    托珠單抗於各週期之Day 1靜脈內投予8 mg/kg •    阿替利珠單抗於各週期之Day 1靜脈內投予1680 mg  a •    nab-紫杉醇於各週期之Day 1、8及15靜脈內投予125 mg/m 2  b •    吉西他濱於各週期之Day 1、8及15靜脈內投予1000 mg/m 2 Atezo+Chemo+TCZ=阿替利珠單抗+化學療法(nab-紫杉醇及吉西他濱)+托珠單抗;nab-紫杉醇=奈米粒子白蛋白結合紫杉醇 a:於最初之兩個週期之Day 1,靜脈注射托珠單抗結束後2小時後投予阿替利珠單抗。於其後之週期之Day 1,靜脈注射托珠單抗結束後,可按照臨床試驗實施計劃書中記載之指示投予阿替利珠單抗。 b:於靜脈注射阿替利珠單抗結束後投予nab-紫杉醇。 [Table 14] Table A Treatment regimen of Atezo+Chemo+TCZ group cycle length Dosage, route of administration, and regimen (drugs are displayed in the order of administration) 28 days • Tocilizumab 8 mg/kg iv on Day 1 of each cycle • Atezolizumab 1680 mg a iv on Day 1 of each cycle • nab-paclitaxel on Day 1, 8 and 15 iv at 125 mg/m 2 b • Gemcitabine 1000 mg/m 2 iv on Days 1, 8 and 15 of each cycle Atezo+Chemo+TCZ=atezolizumab+chemotherapy (nab-paclitaxel and gemcitabine)+tocilizumab; nab-paclitaxel=nanoparticle nab-paclitaxel a: On Day 1 of the first two cycles, intravenous tocilizumab Atezolizumab was administered 2 hours after the end of benzumab. On Day 1 of the subsequent cycle, after the intravenous injection of tocilizumab is completed, atezolizumab can be administered according to the instructions recorded in the clinical trial implementation plan. b: nab-paclitaxel was administered after the end of intravenous atezolizumab.

安全性評估期為了確認Atezo+Chemo+TCZ組中之毒性重疊之可能性,於約6例患者入組後,暫停入組,以便進行安全性評估。於至少一次之完整週期中投予治療用藥1次(即,併用之各藥劑1次)以上,基於自完成安全性追蹤評估之6例以上之患者獲得之安全性資料來進行安全性評估。於判定該併用療法充分安全之情形時,再次開放入組。 Safety evaluation period In order to confirm the possibility of overlapping toxicity in the Atezo+Chemo+TCZ group, after about 6 patients were enrolled, enrollment was suspended for safety evaluation. The therapeutic agents were administered more than once in at least one complete cycle (ie, each agent was administered in combination), and safety assessments were performed based on safety data obtained from more than 6 patients who completed safety follow-up evaluations. When it is judged that the combined therapy is sufficiently safe, the group will be opened again.

評估及監控於整個臨床試驗期間,密切注意觀察全部患者之不良事件,依據美國國立癌研究所之不良事件通用術語標準4.0版(NCI CTCAE v4.0),對不良事件進行分類。 於最初之48週(自第1週期之第1天起)內每隔6週進行腫瘤評估,之後每隔6週或12週進行腫瘤評估。研究(助理研究)者採用RECIST v1.1對效果進行評估。基於研究(助理研究)者所評估之各個病變資料,贊助者藉由程式判定依據適用於免疫治療劑之改良RECIST v1.1(iRECIST)之緩解。 Assessment and Monitoring Throughout the clinical trial, all patients were closely monitored for adverse events, classified according to the National Cancer Institute's Common Terminology Criteria for Adverse Events Version 4.0 (NCI CTCAE v4.0). Tumor assessments were performed at 6-week intervals for the first 48 weeks (starting on Day 1 of Cycle 1), followed by 6- or 12-week intervals thereafter. The researcher (assistant researcher) used RECIST v1.1 to evaluate the effect. Based on the individual lesion data as assessed by the investigator (assistant investigator), the sponsor determined the response based on the modified RECIST v1.1 (iRECIST) for immunotherapeutics by program adjudication.

選擇基準入組基準 滿足以下全部基準之患者有資格參加臨床試驗治療。 Selected Criteria Enrollment Criteria Patients who meet all of the following criteria are eligible for clinical trial treatment.

[表15] 截至同意書上署名之時間點18歲以上之患者 ECOG日常體能狀態為0或1之患者 於組織學或細胞學上診斷為轉移性PDAC之患者 藉由評估組織病理學檢查資料,並結合臨床資料及X射線圖像資料,確診轉移性PDAC。 胰臟內分泌癌或胰臟腺泡細胞癌(endocrine or acinar pancreatic carcinoma)之患者不適合參加本臨床試驗。 無PDAC之全身治療史 研究(助理研究)者判斷存活期3個月以上之患者 可獲得適於國家檢測機構進行PD-L1及/或追加生物標記判定之代表性腫瘤檢體之患者 自全部患者採集基線腫瘤組織檢體。較理想為藉由臨床試驗入組時實施之活檢進行該檢體採集。於研究(助理研究)者不能判斷無法實施活檢之情形時、於保存之腫瘤組織為入組前3個月內實施之活檢中採集之組織、且患者於活檢後未接受抗癌療法之情形時,可於獲得醫學監查員(Medical Monitor)許可後,提交保存之腫瘤組織。 須提交福馬林固定石蠟包埋腫瘤塊狀標本(推薦)、或16片以上之載有未染色之新切之連續切片之載玻片,以及相關之病理檢查報告書。所獲得之載玻片僅有10~15片時,於獲得醫學監查員許可後,該患者仍有可能參加本試驗。 研究(助理研究)者判斷有能力遵守臨床試驗實施計劃書之患者 存在基於RECIST v1.1之可測定病變(至少1個目標病變)之患者 有放射線照射史之病變僅在照射後於該部位確認到明確之進展之情況下,允許視為可測定病變。具有以下之臨床檢查結果所規定之充分之血液學功能及終端器官功能之患者。規定臨床檢查結果為於開始投予臨床試驗藥前14天以內獲得者。 − ANC1.5×10 9/L(1500/μL)以上。規定於篩查期臨床檢查前14天以內不得接受粒細胞菌落刺激因子之投予。 − 白血球數2.5×10 9/L(2500/μL)以上 − 淋巴細胞數0.5×10 9/L(500/μL)以上 − 血小板數≥100×10 9/L(100,000/μL)以上。規定於篩查期臨床檢查前7天以內不得接受輸血。 − 血紅蛋白90 g/L(9.0 g/dL)以上 與醫學監查員協商後,亦可對患者進行輸血以滿足該基準。 − AST、ALT及ALP為2.5×基準值上限(upper limit of normal;ULN)以下。但以下之情況例外。 確認發生肝轉移之患者:AST及ALT為5×ULN以下 確認發生肝轉移或骨轉移之患者:ALP為5×ULN以下 − 血清膽紅素1.5×ULN以下。但以下之情況例外。 判明患有既知之吉爾伯特(Gilbert)病之患者:血清膽紅素值為3×ULN以下 − 肌酐清除率50 mL/min以上(採用Cockcroft-Gault式計算) − 血清白蛋白25 g/L(2.5 g/dL)以上 − 未在實施抗凝治療之患者:國際標準化比值(international normalized ratio;INR)或活化部分凝血激素時間(aPTT)1.5×ULN以下 正在實施抗凝治療之患者:抗凝藥之投予方案穩定 有能夠進行活檢之腫瘤之患者 對於能夠妊娠之女性:如各投予組中所示,同意保持禁欲(避免與異性性交)或採取避孕措施,並同意不捐獻卵子;對於患者男性:如各投予組中所示,同意保持禁欲(避免與異性性交)或採取避孕措施,並同意不捐獻精子 [Table 15] Patients over 18 years of age as of the time point signed on the consent form Patients with ECOG daily performance status of 0 or 1 Patients with histologically or cytologically diagnosed metastatic PDAC By evaluating histopathological examination data, Combined with clinical data and X-ray image data, metastatic PDAC was diagnosed. Patients with endocrine or acinar pancreatic carcinoma of the pancreas are not eligible to participate in this clinical trial. Patients with a history of systemic treatment without PDAC (assistant research) judged that patients with a survival period of more than 3 months can obtain representative tumor samples suitable for PD-L1 and/or additional biomarker judgment by national testing institutions. Collect baseline tumor tissue samples. Ideally, the specimen collection is performed by biopsy performed at the time of clinical trial enrollment. When the researcher (assistant researcher) cannot judge that the biopsy cannot be performed, the preserved tumor tissue is the tissue collected in the biopsy performed within 3 months before the enrollment, and the patient has not received anticancer therapy after the biopsy , after obtaining the permission of the Medical Monitor (Medical Monitor), the preserved tumor tissue can be submitted. Formalin-fixed paraffin-embedded tumor mass specimens (recommended), or more than 16 slides containing unstained freshly cut serial sections, and related pathological examination reports must be submitted. When only 10 to 15 slides are obtained, the patient may still participate in this trial after obtaining the permission of the medical monitor. Patients judged by the researcher (assistant researcher) to be able to comply with the clinical trial implementation plan Patients with measurable lesions (at least 1 target lesion) based on RECIST v1.1 Lesions with a history of radiation exposure are confirmed only at the site after exposure Deemed measurable lesions are permitted until definite progression is reached. Patients with sufficient hematological function and end-organ function as specified by the following clinical examination results. It is stipulated that the clinical examination results are obtained within 14 days before the start of administration of the clinical trial drug. − ANC 1.5×10 9 /L (1500/μL) or more. It is stipulated that the administration of granulocyte colony-stimulating factor is not allowed within 14 days before the clinical examination during the screening period. − The number of white blood cells is more than 2.5×10 9 /L (2500/μL) − The number of lymphocytes is more than 0.5×10 9 /L (500/μL) − The number of platelets is more than 100×10 9 /L (100,000/μL). It is stipulated that no blood transfusion shall be accepted within 7 days before the clinical examination during the screening period. − Blood transfusions may also be given to patients to meet this benchmark after consultation with a medical monitor for hemoglobin levels above 90 g/L (9.0 g/dL). − AST, ALT and ALP are below 2.5 × upper limit of normal (ULN). The following exceptions apply. Patients with confirmed liver metastases: AST and ALT below 5×ULN Patients with confirmed liver metastases or bone metastases: ALP below 5×ULN − Serum bilirubin below 1.5×ULN. The following exceptions apply. Patients with known Gilbert disease: serum bilirubin value below 3×ULN − creatinine clearance rate above 50 mL/min (calculated by Cockcroft-Gault formula) − serum albumin 25 g/L (2.5 g/dL) - Patients not on anticoagulation therapy: International normalized ratio (INR) or activated partial thromboplastin time (aPTT) less than 1.5×ULN patients on anticoagulation therapy: Anticoagulation Dosing regimen of the drug Stable patients with biopsy-capable tumors For women capable of pregnancy: As indicated in each dosing group, agree to remain abstinent (avoid heterosexual intercourse) or use contraception, and agree not to donate eggs; for Patient males: As indicated in each administration group, agree to remain abstinent (avoid intercourse with the opposite sex) or use contraception, and agree not to donate sperm

排除基準 滿足以下任一基準之患者從臨床試驗中排除。 Exclude benchmarks Patients meeting any of the following criteria were excluded from clinical trials.

[表16-1] 有T細胞共刺激劑或免疫檢查點抑制劑(抗CTLA-4抗體藥、抗PD-1抗體藥及抗PD-L1抗體藥等)投予史之患者;於開始投予臨床試驗藥前28天以內接受過臨床試驗治療之患者 前期抗癌治療之不良事件未改善至Grade 1以下之患者。但不問等級之禿頭症及Grade 2以下之周圍神經病變除外。僅適合參加對照藥組之患者 具有需實施反覆(即每月複數次)引流之控制不佳之胸腔積液、心包積液或腹腔積液之患者的控制不佳之腫瘤相關疼痛 ・需要麻藥性鎮痛藥之患者於試驗入組時投予方案必須穩定。 ・可實施姑息性放射線療法之症狀性病變(例如骨轉移或引起神經卡壓之轉移)應於開始投予臨床試驗藥前進行治療。患者需從放射線之影響中恢復。所需之最小限度之停藥期無規定。 ・對於若進一步生長則有很大可能引起功能障礙或難治性疼痛之無症狀性之轉移性病變(例如當前為未伴有脊髓壓迫之硬腦膜外轉移),於合適情況下,應於開始投予臨床試驗藥前研究局部療法。 具有症狀性、未治療或活動性之進展中樞神經系統轉移之患者 有中樞神經系統病變治療史之患者於滿足以下全部基準之情況下有資格參加。 − 能夠基於RECIST v1.1進行測定之病變為中樞神經系統以外之病變。 − 無顱內出血史或脊髓出血史。 − 轉移限於小腦或幕上區(即,無向中腦、腦橋、髓質或脊髓之轉移)。 − 在中樞神經系統治療完成後直至篩查時腦掃描為止之期間未觀察到進展。 − 患者於開始投予臨床試驗藥前7天以內未接受定向放射線療法、或於開始投予臨床試驗藥前14天以內未接受全腦放射線療法。 − 無需持續使用腎上腺皮質類固醇以治療中樞神經系統病變。允許以穩定之劑量投予抗痙攣藥。 篩查時確認到新發中樞神經系統轉移之無症狀患者於實施放射線療法或手術後有資格參加本試驗,無需重複進行篩查用腦掃描。 控制不佳之高鈣血症(離子化鈣>1.5 mmol/L、鈣>12 mg/dL、或修正血清鈣>ULN)、或需使用雙磷酸鹽持續治療之症狀性高鈣血症患者 ・正因其他原因(例如骨轉移或骨質疏鬆症)而投予雙磷酸鹽,但無重大之高鈣血症臨床病史之患者有資格參加本試驗。 有軟腦膜病史之患者 自體免疫性疾病或免疫缺乏症(包含但不限於嚴重肌無力症、肌炎、自體免疫性肝炎、全身性紅斑狼瘡、類風濕性關節炎、炎症性腸疾病、抗磷脂質抗體症候群、韋格納肉芽腫病、修格蘭氏症候群、格-巴二氏症候群或多發性硬化症)、或有其等相關病史之患者。再者,以下為例外。 ・有自體免疫相關甲狀腺功能減退症病史且正在實施甲狀腺激素補充療法之患者有資格參加本試驗。 ・患有受到控制之1型糖尿病且正在實施穩定之胰島素療法之患者有資格參加本試驗。 ・患有僅呈皮膚症狀之濕疹、牛皮癬、慢性單純性苔癬或尋常性白斑之患者(例如牛皮癬性關節炎患者除外)於滿足以下全部條件之情況下有資格參加本試驗。 •  發疹面積未達體表面積之10% •  基線時之病情控制良好,僅需使用低效之腎上腺皮質類固醇外用藥 過去12個月以內未觀察到需使用補骨脂素長波紫外線療法、甲胺喋呤、視黃醇、生物學製劑、經口鈣調磷酸酶抑制劑、或者高效或經口之腎上腺皮質類固醇之原有疾病存在急性惡化 [Table 16-1] Patients with a history of administration of T-cell co-stimulators or immune checkpoint inhibitors (anti-CTLA-4 antibody drugs, anti-PD-1 antibody drugs, and anti-PD-L1 antibody drugs, etc.); before starting the administration of clinical trial drugs28 Patients who have received clinical trial treatment within days Patients whose adverse events did not improve to below Grade 1 on prior anticancer therapy. However, alopecia regardless of grade and peripheral neuropathy below Grade 2 are excluded. Only suitable for patients in the control group Uncontrolled tumor-related pain in patients with poorly controlled pleural, pericardial, or ascites requiring repeated (ie, multiple monthly) drainage ・Patients who need narcotic analgesics must have a stable dosing regimen at the time of trial enrollment. ・Symptomatic lesions that can be treated with palliative radiation therapy (such as bone metastases or metastases causing nerve entrapment) should be treated before starting the administration of the clinical trial drug. Patients need to recover from the effects of radiation. The minimum required withdrawal period is not specified. ・For asymptomatic metastatic lesions that are likely to cause functional disability or intractable pain if further growth (such as current epidural metastases without spinal cord compression), when appropriate, treatment should be started at Investigate topical therapy before administering a clinical trial drug. Patients with symptomatic, untreated or active progressive CNS metastases Patients with a history of treatment for central nervous system disorders are eligible to participate if they meet all of the following criteria. − Lesions that can be measured based on RECIST v1.1 are outside the central nervous system. − No history of intracranial hemorrhage or spinal cord hemorrhage. − Metastases limited to the cerebellum or supratentorial regions (ie, no metastases to the midbrain, pons, medulla, or spinal cord). − No progression was observed after completion of CNS therapy until the time of screening brain scan. − The patient did not receive targeted radiation therapy within 7 days before the start of the clinical trial drug administration, or did not receive whole-brain radiation therapy within 14 days before the start of the clinical trial drug administration. − Continued use of corticosteroids for the treatment of CNS lesions is not required. Anticonvulsants are allowed to be administered in stable doses. Asymptomatic patients with new-onset CNS metastases identified at screening were eligible for the trial after radiation therapy or surgery without repeat screening brain scans. Poorly controlled hypercalcemia (ionized calcium >1.5 mmol/L, calcium >12 mg/dL, or corrected serum calcium >ULN), or patients with symptomatic hypercalcemia requiring continuous treatment with bisphosphonates ・Patients who are being administered bisphosphonates for other reasons (eg, bone metastases or osteoporosis) without a significant clinical history of hypercalcemia are eligible for this trial. Patients with a history of leptomeningeal disease Autoimmune disease or immunodeficiency (including but not limited to myasthenia gravis, myositis, autoimmune hepatitis, systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, antiphospholipid antibody syndrome, Patients with Wegener's granulomatosis, Sjogren's syndrome, Guillain-Barré syndrome, or multiple sclerosis), or those with a history of related diseases. Furthermore, the following are exceptions. ・Patients with a history of autoimmune-related hypothyroidism who are undergoing thyroid hormone replacement therapy are eligible to participate in this trial. ・Patients with controlled type 1 diabetes who are on stable insulin therapy are eligible to participate in this trial. ・Patients with eczema, psoriasis, lichen simplex chronicus, or leukoplakia vulgaris (eg, patients with psoriatic arthritis except those with psoriatic arthritis) are eligible to participate in this trial if they meet all of the following conditions. • The rash area is less than 10% of the body surface area • Good disease control at baseline with only low-efficiency topical corticosteroids No need to use psoralen long-wave ultraviolet therapy, methotrexate, retinol, biological agents, oral calcineurin inhibitors, or high-potency or oral corticosteroids in the past 12 months. Acute exacerbation of pre-existing disease

[表16-2] 有特發性肺纖維化症、機化肺炎(例如閉塞性細支氣管炎)、藥物誘發性肺炎或特發性肺炎病史之患者、或者篩查時藉由胸部電腦斷層攝影觀察到活動性肺炎之患者 ・有照射野內放射性肺炎(纖維化症)病史患者可以參加。 篩查時或篩查前之任一時間點HIV檢查呈陽性之患者 ・除非各國法規不允許,否則過去在HIV檢查中未曾獲得過陽性結果之患者將在篩查時進行HIV檢查。 活動性B型肝炎病毒(HBV)感染(慢性或急性)患者,該活動性B型肝炎病毒(HBV)感染(慢性或急性)被定義為在篩查時B型肝炎表面抗原(HBsAg)檢查呈陽性 ・有HBV感染病史或治療史之患者有資格參加本試驗,該HBV感染病史或治療史被定義為在篩查時HBsAg檢查呈陰性、B型肝炎核心抗體檢查呈陽性之情形時,在HBV DNA定量中未達500 IU/mL。 活動性C型肝炎病毒(HCV)感染患者,該活動性C型肝炎病毒(HCV)感染被定義為在篩查時HCV抗體檢查呈陽性且其後HCV RNA檢查呈陽性 ・HCV RNA檢查僅針對HCV抗體檢查呈陽性之患者進行。 患有臨床上已知之重大肝病(酒精性肝炎、肝硬化、脂肪性肝病及遺傳性肝病等)之患者 患有活動性結核之患者 於開始投予臨床試驗藥前4週以內有因感染症之併發症入院、菌血症或重度肺炎等(但不限於此)重度感染症之患者 於開始投予臨床試驗藥前2週以內接受過抗菌藥之經口投予或靜脈內投予之患者 ・接受了抗菌藥之預防性投予(例如預防尿路感染或慢性阻塞性肺疾病惡化)之患者有資格參加本試驗。 於開始投予臨床試驗藥前12個月以內出現過重大心血管疾病(依照紐約心臟病學會(New York Heart Association)心功能分級評定為II級以上之心臟疾病、心肌梗塞或腦血管發作等)、或於開始投予臨床試驗藥前3個月以內出現過不穩定之心律不整或不穩定心絞痛之患者 於開始投予臨床試驗藥前28天以內出現過Grade 3以上之出血或出血性事件之患者 有同種幹細胞移植或實體器官移植史之患者 於開始投予臨床試驗藥前4週以內接受過診斷目的以外之大手術之患者,或預見需於臨床試驗中進行大手術之患者 中心靜脈通路導管(例如輸液港(port)等)之置放並不被視為大手術,故得到允許。 於篩查前2年以內有除胰臟癌以外之惡性病史之患者。其中,經過適當治療之宮頸上皮內癌、非黑色素瘤皮膚癌、局部性前列腺癌、非浸潤性乳管癌或I期子宮癌等轉移或致死風險達到可忽視程度(例如5年總生存率超90%)者除外 [Table 16-2] Patients with a history of idiopathic pulmonary fibrosis, organizing pneumonia (eg, bronchiolitis obliterans), drug-induced pneumonitis, or idiopathic pneumonitis, or those with active pneumonitis observed by chest CT at screening patient ・Patients with a history of radiation pneumonitis (fibrosis) in the irradiation field can participate. HIV-positive patients at screening or at any point prior to screening ・Unless not permitted by the laws of each country, patients who have not obtained a positive result in an HIV test in the past will be tested for HIV at the time of screening. Patients with active hepatitis B virus (HBV) infection (chronic or acute) defined as a positive hepatitis B surface antigen (HBsAg) test at screening positive ・Patients with a history of HBV infection or treatment history, defined as a negative HBsAg test at screening and a positive hepatitis B core antibody test, are eligible for this trial. Quantitation did not reach 500 IU/mL. Patients with active hepatitis C virus (HCV) infection defined as a positive HCV antibody test at screening and a positive HCV RNA test thereafter ・HCV RNA test is performed only for patients with positive HCV antibody test. Patients with clinically known major liver diseases (alcoholic hepatitis, liver cirrhosis, fatty liver disease and hereditary liver disease, etc.) patients with active tuberculosis Patients who have been admitted to hospital due to complications of infectious diseases, bacteremia or severe pneumonia (but not limited to) within 4 weeks before the start of clinical trial drug administration Patients who have received oral or intravenous antibacterial drugs within 2 weeks before the start of clinical trial drug administration ・Patients who have received prophylactic administration of antibacterial drugs (eg, to prevent urinary tract infection or exacerbation of chronic obstructive pulmonary disease) are eligible to participate in this trial. Major cardiovascular disease (heart disease, myocardial infarction or cerebrovascular attack, etc., assessed as grade II or above according to the New York Heart Association's cardiac function classification) within 12 months before the start of the clinical trial drug administration. , or patients with unstable arrhythmia or unstable angina pectoris within 3 months before the start of administration of the clinical trial drug Patients who have experienced bleeding or hemorrhagic events of Grade 3 or higher within 28 days before starting the administration of the clinical trial drug Patients with a history of allogeneic stem cell transplantation or solid organ transplantation Patients who have undergone major surgery for purposes other than diagnostic purposes within 4 weeks before the start of clinical trial drug administration, or patients who are expected to undergo major surgery in clinical trials Placement of central venous access catheters (eg, ports, etc.) is not considered major surgery and is allowed. Patients with a history of malignancy other than pancreatic cancer within 2 years before screening. Among them, appropriately treated cervical intraepithelial cancer, non-melanoma skin cancer, localized prostate cancer, non-invasive breast duct cancer, or stage I uterine cancer have a negligible risk of metastasis or death (e.g., 5-year overall survival rate exceeding 50%). 90%) except

[表16-3] 於禁忌使用臨床試驗藥之其他疾病、代謝功能障礙、身體診察所見或臨床檢查值被觀察到之情形時,有可能影響結果之解釋、或增加患者出現治療併發症之風險。 妊娠期及哺乳期女性、或臨床試驗過程中計劃妊娠之患者 可能妊娠之女性必須獲得開始投予臨床試驗藥前14天以內之血清妊娠檢查陰性結果。 於開始投予臨床試驗藥前4週以內接受過減毒活疫苗接種之患者,或預計於阿替利珠單抗投予期內或阿替利珠單抗最後一次投予後5個月以內需要減毒活疫苗之患者 曾對嵌合抗體或者人源化抗體或融合蛋白有過重度過敏反應之患者 對中國倉鼠卵巢細胞之產生物或重組人抗體有過敏症病史之患者 對任一臨床試驗藥或其添加劑有已知過敏或過敏症之患者 於開始投予臨床試驗藥前4週或藥劑之5個半衰期(以較長者為準)以內投予過全身性免疫刺激藥(包含但不限於干擾素及介白素2)之患者 於開始投予臨床試驗藥前2週以內投予過全身性免疫抑制藥(包含但不限於腎上腺皮質類固醇、環磷醯胺、硫唑嘌呤、甲胺喋呤、沙利竇邁及抗腫瘤壞死因子α製劑)之患者、或預計需於臨床試驗中投予全身性免疫抑制藥之患者。其中,以下之情況例外。 接受過低劑量之全身性免疫抑制藥之急性投予、或全身性免疫抑制藥之單次脈衝投予(例如針對造影劑過敏之48小時之腎上腺皮質類固醇)之患者於獲得醫學監查員之許可後有資格參加本試驗。 投予有礦物皮質素(例如氟氫可的松)、針對慢性阻塞性肺病或哮喘之腎上腺皮質類固醇、或者針對起立性低血壓或腎上腺功能不全之低劑量腎上腺皮質類固醇之患者有資格參加本試驗。 患有已知之中樞神經系統脫髓鞘疾病或癲癇發作之患者 既往有憩室炎、慢性潰瘍性下消化道疾病(例如克隆氏病、潰瘍性大腸炎)、或可能導致消化道穿孔之其他症狀性下消化道疾病之病史之患者 研究(助理研究)者判斷為患有與作為原有疾病之癌症之診斷無關的肝病之患者 當前有活動性感染、或者復發性之細菌、病毒、真菌(甲床真菌感染除外)、分枝桿菌或其他感染症(包含但不限於非典型分枝桿菌症、HBV、HCV及帶狀疱疹)病史之患者 於感染症發生全球大流行或流行之情形時,應按照各國或醫療機關之指南、或專門團體(例如美國臨床腫瘤學會或歐州臨床腫瘤學會)之相應指南,研究篩查活動性感染症。 開始投予臨床試驗藥前3個月以內,純化蛋白衍生物(PPD)皮膚檢查或結核血液檢查結果顯示活動性結核且胸部X射線檢查呈陽性而被確診之患者 PPD皮膚檢查或結核血液檢查呈陽性,但其後胸部X射線檢查呈陰性之患者可能有資格參加本臨床試驗。 未治療之潛伏結核感染 活動性之原發性或續發性免疫缺乏症、或有既往病史之患者 ALT或AST超1.5×ULN、或者有肝轉移之患者之情況下超5×ULN C-反應性蛋白超10 mg/dL之患者 [Table 16-3] Other diseases that contraindicate the use of clinical trial drugs, metabolic dysfunction, physical examination findings or clinical examination values observed may affect the interpretation of results or increase the risk of treatment complications for patients. Pregnant and lactating women, or patients planning pregnancy during clinical trials Potentially pregnant women must obtain a negative serum pregnancy test result within 14 days prior to starting the administration of the clinical trial drug. Patients who have received live attenuated vaccines within 4 weeks prior to the start of administration of the clinical trial drug, or who are expected to require reductions during the atezolizumab administration period or within 5 months after the last administration of atezolizumab. patients with live vaccine Patients with a history of severe allergic reactions to chimeric antibodies or humanized antibodies or fusion proteins Patients with a history of hypersensitivity to Chinese hamster ovary cell-producing products or recombinant human antibodies Patients with known allergies or hypersensitivity to any clinical trial drug or its additives Patients who have been administered systemic immunostimulatory drugs (including but not limited to interferon and interleukin 2) within 4 weeks or within 5 half-lives of the drug (whichever is longer) before the start of administration of the clinical trial drug Administered systemic immunosuppressive drugs (including but not limited to adrenal corticosteroids, cyclophosphamide, azathioprine, methotrexate, thalidomide and anti-tumor necrosis) within 2 weeks before the start of clinical trial drug administration factor alpha preparations), or patients who are expected to be administered systemic immunosuppressive drugs in clinical trials. Among them, the following exceptions. Patients who have received acute administration of low-dose systemic immunosuppressive drugs, or single-pulse administration of systemic immunosuppressive drugs (eg, 48-hour adrenal corticosteroids for Eligible to participate in this trial after permission. Patients who were administered mineralcorticoids (eg, fludrocortisone), corticosteroids for chronic obstructive pulmonary disease or asthma, or low-dose corticosteroids for orthostatic hypotension or adrenal insufficiency were eligible to participate in this trial . Patients with known central nervous system demyelinating disease or seizures Patients with a history of diverticulitis, chronic ulcerative lower gastrointestinal disease (eg, Crohn's disease, ulcerative colitis), or other symptomatic lower gastrointestinal disease that may lead to gastrointestinal perforation Patients judged by the researcher (assistant researcher) to have liver disease unrelated to the diagnosis of cancer as a pre-existing disease Current active infection, or recurrent bacterial, viral, fungal (except nail bed fungal infection), mycobacterial or other infectious disease (including but not limited to atypical mycobacterial disease, HBV, HCV and herpes zoster) patient with medical history In the event of a global pandemic or epidemic of infectious diseases, research and screening of active infectious diseases should be conducted in accordance with the guidelines of various countries or medical institutions, or the corresponding guidelines of specialized groups (such as the American Society of Clinical Oncology or the European Society of Clinical Oncology). Patients who have been diagnosed with active tuberculosis and positive chest X-rays as a result of purified protein derivative (PPD) skin test or tuberculosis blood test results within 3 months before the start of clinical trial drug administration Patients with a positive PPD skin test or blood test for tuberculosis but a subsequent negative chest X-ray may be eligible for this clinical trial. Untreated latent tuberculosis infection Active primary or secondary immunodeficiency, or patients with previous medical history ALT or AST over 1.5×ULN, or over 5×ULN in patients with liver metastases Patients with C-reactive protein exceeding 10 mg/dL

統計方法主要解析 如表A中之規定,有效性之主評估項目為試驗中之客觀緩解。緩解判定缺失或未實施之患者屬於非緩解例。 針對各群組,計算獲得了完全緩解或部分緩解之患者之比率即客觀緩解率(Objective response rate;ORR)及其90%信賴區間(Clopper-Pearson法)。還計算試驗藥組與對照藥組之ORR之差以及其90%信賴區間。根據病例數,利用漸近正態性來推測信賴區間。 The main analysis of statistical methods is as specified in Table A, and the main evaluation item of effectiveness is the objective mitigation in the test. Patients with missing or unimplemented response determinations are non-responders. For each cohort, the ratio of patients who achieved complete or partial remission, the objective response rate (ORR), and its 90% confidence interval (Clopper-Pearson method) were calculated. The difference between the ORRs of the test drug group and the control drug group and its 90% confidence interval were also calculated. Confidence intervals were inferred using asymptotic normality based on the number of cases.

結果相較於標準治療G+N,托珠單抗+阿替利珠單抗+G+N之4劑併用療法於臨床上顯示較高療效。更具體而言,與nab-紫杉醇及吉西他濱之2劑併用療法相比,於臨床上顯示較高療效。又,作為顯示臨床效益之組,可列舉C-反應性蛋白高值患者組。進而,作為其他之顯示臨床效益之組,可列舉磷酸化STAT3陽性患者組。 Results Compared with the standard treatment G+N, the 4-dose combination therapy of tocilizumab+atezolizumab+G+N showed higher clinical efficacy. More specifically, compared with the 2-dose combination therapy of nab-paclitaxel and gemcitabine, it shows a higher clinical efficacy. Moreover, as a group which shows clinical benefit, the C-reactive protein high value patient group can be mentioned. Furthermore, as another group showing clinical benefit, the phosphorylated STAT3 positive patient group can be mentioned.

圖1係表示臨床試驗設計之概要。 圖2係表示腫瘤標記物CA19-9相對於基線之變化率。 圖3係表示投予GN+TCZ之前及之後的腫瘤檢體中之α-SMA及磷酸化STAT3之產生量。 圖4係表示投予GN+TCZ之前及之後的腫瘤檢體中之低分子藥劑(使用左旋氧氟沙星作為指標劑)之濃度。 圖5係表示有關嗜中性球(neutrophil)數量變化的組入有G、N、TCZ之半機械PK-PD模型之結構。 嗜中性球數之基線(Base)、循環嗜中性球數(CircNeut)、G或N之藥劑效果(Drug_Effect)、反饋參數(FP)、嗜中性球之消失速度常數(Kcirc)、米氏(Michaelis-Menten)常數(Km)、嗜中性球之生長速度(kprol)、速度常數(ktr)、平均分化時間(MTT)、分化隔室(n)、TCZ之藥劑效果(TCZ_Effect)、最大反應速度(Vmax)。 圖6係表示實際觀察到之嗜中性球數與預測之結果。關於嗜中性球數之預測結果,以線來顯示第5百分位數、第50百分位數(實線)及第95百分位數。使用實際之給藥量,對10名受驗者實施100次模擬。以點顯示「將吉西他濱/白蛋白結合紫杉醇抗性之轉移性者作為對象之托珠單抗+吉西他濱/白蛋白結合紫杉醇之I期試驗(EPOC1804)」中所實際觀察到之嗜中性球數。 圖7係表示TCZ+GN併用之情形時嗜中性球數演變之模擬結果。按照以下所示方案實施1000次模擬。 投予方案:GN及GN+TCZ之4種不同劑量:1次4 mg/kg且間隔2週(Q2W),1次8 mg/kg且間隔2週(Q2W),1次4 mg/kg且間隔4週(Q4W)、1次8 mg/kg且間隔4週(Q4W)。關於嗜中性球數(細胞/mm 3)之預測結果,將第5~95百分位數之預測區間填充顏色以顯示,以中央之實線顯示第50百分位數。1000 細胞/mm 3之水平線為減量基準。 圖8-1係圖8表示藉由IL-6抗體+PD-L1抗體+吉西他濱所產生之各種免疫細胞向胰臟癌腫瘤組織中之浸潤促進效果。藉由流式細胞分析獲得之KPΔ(δ)C小鼠之胰臟癌中浸潤之以下之細胞表面標記陽性細胞之定量:CD45陽性細胞(A);CD3陽性及CD45陽性細胞(B);CD8陽性及CD45陽性細胞(B);CD11c陽性及CD45陽性細胞(C);及CD19陽性及CD45陽性細胞(D)。藉由雙側學生T檢驗進行群間比較。統計顯著性設定為*P<0.05、**P<0.01、***P<0.001。 A:CD45陽性免疫細胞之浸潤促進;B:T細胞(CD3陽性及CD45陽性細胞;CD8陽性及CD45陽性細胞)之浸潤促進。 圖8-2上接圖8。 C:樹狀細胞之浸潤促進;D:B細胞之浸潤促進。 圖9係表示藉由抗IL-6R抗體+抗PD-L1抗體+吉西他濱來抑制胰臟癌組織生長的效果。藉由IHC染色解析來定量KPΔC小鼠胰臟癌組織中之生長標記物Ki67及pErk訊號。結果係以平均值±標準偏差之形式表示。統計顯著性基於雙側學生T檢驗(*P<0.05、**P<0.01)。 圖10係利用抗IL-6R抗體、抗PD‐L1抗體、化學治療劑(吉西他濱)及併用其等時之腫瘤體積(胰臟導管腺癌(PDAC))的評估。以MRI來評估各個KPΔC小鼠中之含胰臟癌之胰臟體積,將投予開始時之體積作為基準,對其變化率進行繪圖。 圖11係表示臨床試驗設計之概要。 圖12係表示針對胰臟癌之標準化學療法「胰臟癌化學療法之演算法」之圖(轉自胰臟診療指南2019年度版)。 圖13係GN+TCZ投予前後之PD-L1陽性癌細胞密度之比較。 圖14係GN+TCZ投予前後之磷酸化STAT3於腫瘤組織內之陽性面積比率之比較。 圖15係基於源自GN+TCZ投予前後之受驗者(GAP01)之試樣之免疫組織化學螢光多重染色的PD-L1陽性癌細胞之解析。 Figure 1 shows an outline of the clinical trial design. Figure 2 shows the rate of change from baseline in the tumor marker CA19-9. Fig. 3 shows the production levels of α-SMA and phosphorylated STAT3 in tumor specimens before and after administration of GN+TCZ. FIG. 4 shows the concentration of the low-molecular-weight drug (levofloxacin was used as an indicator agent) in the tumor specimen before and after administration of GN+TCZ. Figure 5 shows the structure of a hemi-mechanical PK-PD model incorporating G, N, and TCZ regarding changes in neutrophil number. Baseline of neutrophil count (Base), circulating neutrophil count (CircNeut), drug effect of G or N (Drug_Effect), feedback parameter (FP), neutrophil disappearance rate constant (Kcirc), meter Michaelis-Menten constant (Km), neutrophil growth rate (kprol), rate constant (ktr), mean differentiation time (MTT), differentiation compartment (n), TCZ effect (TCZ_Effect), Maximum response speed (Vmax). Figure 6 shows the actual observed and predicted neutrophil counts. The 5th percentile, 50th percentile (solid line), and 95th percentile are shown as lines for the predicted results of neutrophil count. Using actual doses, 100 simulations were performed on 10 subjects. The number of neutrophils actually observed in the "Phase I trial of tocilizumab + gemcitabine/nab-paclitaxel (EPOC1804) in patients with metastatic gemcitabine/nab-paclitaxel resistance" is shown as a dot . Fig. 7 shows the simulation results of the evolution of the number of neutrophils when TCZ+GN is used together. 1000 simulations were performed according to the protocol shown below. Dosing regimen: 4 different doses of GN and GN+TCZ: 4 mg/kg once with 2-week intervals (Q2W), 8 mg/kg once with 2-week intervals (Q2W), 4 mg/kg once with 4-week intervals Weekly (Q4W), once at 8 mg/kg and 4 weeks apart (Q4W). Regarding the prediction result of the number of neutrophils (cells/mm 3 ), the prediction interval of the 5th to 95th percentiles was filled with color and displayed, and the 50th percentile was shown by the solid line in the center. The horizontal line of 1000 cells/mm 3 is the decrement reference. Fig. 8-1 shows the effect of promoting the infiltration of various immune cells into pancreatic cancer tumor tissue by IL-6 antibody + PD-L1 antibody + gemcitabine. Quantification of infiltrating cells positive for the following cell surface markers in pancreatic cancer of KPΔ(δ)C mice obtained by flow cytometric analysis: CD45 positive cells (A); CD3 positive and CD45 positive cells (B); CD8 Positive and CD45 positive cells (B); CD11c positive and CD45 positive cells (C); and CD19 positive and CD45 positive cells (D). Intergroup comparisons were made by two-sided Student's t-test. Statistical significance was set as *P<0.05, **P<0.01, ***P<0.001. A: Infiltration promotion of CD45-positive immune cells; B: Infiltration promotion of T cells (CD3-positive and CD45-positive cells; CD8-positive and CD45-positive cells). Figure 8-2 continues from Figure 8. C: Infiltration promotion of dendritic cells; D: Infiltration promotion of B cells. FIG. 9 shows the effect of suppressing the growth of pancreatic cancer tissue by anti-IL-6R antibody + anti-PD-L1 antibody + gemcitabine. Growth markers Ki67 and pErk signaling in KPΔC mouse pancreatic cancer tissues were quantified by IHC staining analysis. Results are presented as mean ± standard deviation. Statistical significance is based on two-sided Student's t-test (*P<0.05, **P<0.01). Figure 10 is an evaluation of tumor volume (pancreatic ductal adenocarcinoma (PDAC)) using anti-IL-6R antibody, anti-PD-L1 antibody, chemotherapeutic agent (gemcitabine), and their isochronous use. Pancreatic volume containing pancreatic cancer in each KPΔC mouse was assessed by MRI, and the rate of change was plotted using the volume at the start of administration as a baseline. Figure 11 shows an outline of the clinical trial design. Figure 12 is a diagram showing the standard chemotherapy for pancreatic cancer "The Algorithm of Pancreatic Cancer Chemotherapy" (translated from the Pancreatic Diagnosis and Treatment Guidelines 2019 Edition). Figure 13 is a comparison of the density of PD-L1 positive cancer cells before and after GN+TCZ administration. Figure 14 is a comparison of the positive area ratio of phosphorylated STAT3 in tumor tissue before and after GN+TCZ administration. 15 is an analysis of PD-L1-positive cancer cells based on immunohistochemical fluorescence multiple staining of samples derived from subjects (GAPO1) before and after GN+TCZ administration.

Claims (15)

一種醫藥組合物,其係含有免疫治療劑作為有效成分者,且與IL-6抑制劑、及至少一種化學治療劑組合使用以治療胰臟癌。A pharmaceutical composition, which contains an immunotherapy agent as an active ingredient, is used in combination with an IL-6 inhibitor and at least one chemotherapeutic agent to treat pancreatic cancer. 如請求項1之醫藥組合物,其中上述化學治療劑為選自氟尿嘧啶、替加氟、吉西他濱及卡培他濱中之嘧啶系藥劑、及/或選自由紫杉醇、白蛋白結合紫杉醇及歐洲紫杉醇所組成之群中之紫杉烷系藥劑、及/或選自順鉑及奧沙利鉑中之鉑系藥劑。The pharmaceutical composition of claim 1, wherein the chemotherapeutic agent is a pyrimidine-based agent selected from fluorouracil, tegafur, gemcitabine and capecitabine, and/or selected from paclitaxel, nab-paclitaxel and paclitaxel A taxane-based agent in the group, and/or a platinum-based agent selected from cisplatin and oxaliplatin. 如請求項1或2之醫藥組合物,其中上述IL-6抑制劑為抗IL-6R抗體。The pharmaceutical composition according to claim 1 or 2, wherein the above-mentioned IL-6 inhibitor is an anti-IL-6R antibody. 如請求項1至3中任一項之醫藥組合物,其中上述免疫治療劑為PD-1/PD-L1訊號抑制劑。The pharmaceutical composition according to any one of claims 1 to 3, wherein the immunotherapeutic agent is a PD-1/PD-L1 signaling inhibitor. 如請求項1至4中任一項之醫藥組合物,其中化學治療劑、IL-6抑制劑及PD-1/PD-L1訊號抑制劑係同時投予、分開投予、或連續投予。The pharmaceutical composition of any one of claims 1 to 4, wherein the chemotherapeutic agent, the IL-6 inhibitor, and the PD-1/PD-L1 signaling inhibitor are administered simultaneously, separately, or sequentially. 如請求項1至5中任一項之醫藥組合物,其中上述PD-1/PD-L1訊號抑制劑為抗PD-L1抗體。The pharmaceutical composition according to any one of claims 1 to 5, wherein the PD-1/PD-L1 signaling inhibitor is an anti-PD-L1 antibody. 一種含有阿替利珠單抗之醫藥,其係藉由下述方法來使用, 該方法包括為了將含有阿替利珠單抗之醫藥與托珠單抗、吉西他濱及白蛋白結合紫杉醇組合來治療人類個體之胰臟癌,而按照如下用法劑量對個體進行投予: (i)托珠單抗之劑量為1次8 mg/kg且間隔4週(Q4W);及阿替利珠單抗之劑量係選自由1次840 mg且間隔2週(Q2W),1次1200 mg且間隔3週(Q3W)、1次1680 mg且間隔4週(Q4W)所組成之群; (ii)托珠單抗之劑量為1次4 mg/kg且間隔2週(Q2W);及阿替利珠單抗之劑量係選自由1次840 mg且間隔2週(Q2W)、1次1200 mg且間隔3週(Q3W)、1次1680 mg且間隔4週(Q4W)所組成之群; (iii)托珠單抗之劑量為1次8 mg/kg且間隔2週(Q2W);及阿替利珠單抗之劑量係選自由1次840 mg且間隔2週(Q2W)、1次1200 mg且間隔3週(Q3W)、1次1680 mg且間隔4週(Q4W)所組成之群; (iv)托珠單抗之劑量為1次8 mg/kg且間隔4週(Q4W);及阿替利珠單抗之劑量為1次1680 mg且間隔4週(Q4W);及連續3週每週投予一次白蛋白結合紫杉醇與鹽酸吉西他濱並在第4週停藥,白蛋白結合紫杉醇之劑量以紫杉醇計為1次125 mg/m 2(體表面積),鹽酸吉西他濱之劑量以吉西他濱計為1次1000 mg/m 2(體表面積)。 A medicine containing atezolizumab, which is used by the following method, the method comprising treating the medicine containing atezolizumab in combination with tocilizumab, gemcitabine and nab-paclitaxel Pancreatic cancer in a human subject administered the following dosages: (i) tocilizumab at a dose of 8 mg/kg once at 4-week intervals (Q4W); and atezolizumab Doses were selected from the group consisting of 840 mg once with a 2-week interval (Q2W), 1200 mg once with a 3-week interval (Q3W), and 1680 mg once with a 4-week interval (Q4W); (ii) Tocilizumab The dose of antibacterial was 4 mg/kg once with 2-week interval (Q2W); and the dose of atezolizumab was selected from 840 mg once with 2-week interval (Q2W), 1200 mg once with 3-week interval (Q3W), a cohort of 1680 mg once with a 4-week interval (Q4W); (iii) tocilizumab at a dose of 8 mg/kg once with a 2-week interval (Q2W); and atezolizumab The dose of the antifungal drug was selected from the group consisting of 840 mg once with 2-week interval (Q2W), 1200 mg once with 3-week interval (Q3W), and 1680 mg once with 4-week interval (Q4W); The dose of atezolizumab is 8 mg/kg once with a 4-week interval (Q4W); and the dose of atezolizumab is 1680 mg once with a 4-week interval (Q4W); and administered weekly for 3 consecutive weeks A single dose of nab-paclitaxel and gemcitabine hydrochloride was discontinued in the 4th week. The dose of nab-paclitaxel was 125 mg/m 2 (body surface area) calculated as paclitaxel, and the dose of gemcitabine hydrochloride was calculated as 1000 mg once. /m 2 (body surface area). 一種含有抗PD-L1抗體之醫藥,其用於在癌症之處置中與IL-6抑制劑及化學治療劑組合投予,以使該化學治療劑之效果較先前之治療提高。A medicament containing an anti-PD-L1 antibody for administration in combination with an IL-6 inhibitor and a chemotherapeutic agent in the treatment of cancer to enhance the effect of the chemotherapeutic agent over previous treatments. 一種含有抗IL-6抗體與抗PD-L1抗體之醫藥,其用於在胰臟癌之處置中與化學治療劑組合投予,以提高該化學治療劑之效果。A medicine containing an anti-IL-6 antibody and an anti-PD-L1 antibody for administration in combination with a chemotherapeutic agent in the treatment of pancreatic cancer to enhance the effect of the chemotherapeutic agent. 如請求項8或9之醫藥,其中提高化學治療劑之效果包括:在包含需要處置有化學治療劑抗藥性之癌症之個體的群組中使有效率、緩解率及/或病情控制率提高。The medicine of claim 8 or 9, wherein increasing the effect of a chemotherapeutic agent comprises: increasing the response rate, remission rate and/or disease control rate in a group comprising individuals in need of treatment of chemotherapeutic agent-resistant cancer. 一種含有IL-6抑制劑與PD-1/PD-L1訊號抑制劑之組合物,其用於在使用癌症治療劑來處置個體之癌症時促進上述治療劑向癌組織滲透。A composition comprising an IL-6 inhibitor and a PD-1/PD-L1 signaling inhibitor for promoting the penetration of a cancer therapeutic agent into cancer tissue when the cancer therapeutic agent is used to treat cancer in an individual. 一種含有PD-1/PD-L1訊號抑制劑之組合物,其與IL-6抑制劑併用投予、或含有IL-6抑制劑且用於促進免疫細胞向靶組織浸潤。A composition containing a PD-1/PD-L1 signaling inhibitor, which is administered in combination with an IL-6 inhibitor, or contains an IL-6 inhibitor and is used to promote immune cell infiltration into target tissues. 如請求項12之組合物,其中上述免疫細胞為CD45陽性細胞。The composition of claim 12, wherein said immune cells are CD45 positive cells. 如請求項12或13之組合物,其中上述靶組織為IL-6過度表現組織。The composition of claim 12 or 13, wherein said target tissue is an IL-6 overexpressing tissue. 如請求項12至14中任一項之組合物,其中上述靶組織為腫瘤組織。The composition of any one of claims 12 to 14, wherein the target tissue is a tumor tissue.
TW110133456A 2020-10-22 2021-09-08 Pancreatic cancer quadruplet therapeutic agent combining atezolizumab, tocilizumab, gemcitabine, and nab-paclitaxel TW202228772A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2020-177655 2020-10-22
JP2020177655 2020-10-22

Publications (1)

Publication Number Publication Date
TW202228772A true TW202228772A (en) 2022-08-01

Family

ID=81290440

Family Applications (1)

Application Number Title Priority Date Filing Date
TW110133456A TW202228772A (en) 2020-10-22 2021-09-08 Pancreatic cancer quadruplet therapeutic agent combining atezolizumab, tocilizumab, gemcitabine, and nab-paclitaxel

Country Status (3)

Country Link
JP (1) JPWO2022085322A1 (en)
TW (1) TW202228772A (en)
WO (1) WO2022085322A1 (en)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130149302A1 (en) * 2010-05-28 2013-06-13 Chugai Seiyaku Kabushiki Kaisha Therapeutic agents for pancreatic cancer

Also Published As

Publication number Publication date
JPWO2022085322A1 (en) 2022-04-28
WO2022085322A1 (en) 2022-04-28

Similar Documents

Publication Publication Date Title
US20230115328A1 (en) Anti-B7-H1 and Anti-CTLA-4 Antibodies for Treating Non-Small Cell Lung Cancer
US11827706B2 (en) Anti-B7-H1 antibodies for treating tumors
JP2022536854A (en) Patient selection for enhanced anti-tumor immunity in cancer patients
US20210388090A1 (en) Method and medicament for treating cancer unresponsive to pd-1/pd-l1 signaling inhibitor
Zhou et al. A global phase 3 study of serplulimab plus chemotherapy as first-line treatment for advanced squamous non-small-cell lung cancer (ASTRUM-004)
TW202228772A (en) Pancreatic cancer quadruplet therapeutic agent combining atezolizumab, tocilizumab, gemcitabine, and nab-paclitaxel
CN112915202A (en) Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody
US20220411499A1 (en) LAG-3 Antagonist Therapy for Melanoma