TW201825119A - Method of treating cancer using anti-ccr4 antibody and anti-pd-1 antibody - Google Patents

Method of treating cancer using anti-ccr4 antibody and anti-pd-1 antibody Download PDF

Info

Publication number
TW201825119A
TW201825119A TW106142006A TW106142006A TW201825119A TW 201825119 A TW201825119 A TW 201825119A TW 106142006 A TW106142006 A TW 106142006A TW 106142006 A TW106142006 A TW 106142006A TW 201825119 A TW201825119 A TW 201825119A
Authority
TW
Taiwan
Prior art keywords
cancer
antibody
seq
set forth
sequence set
Prior art date
Application number
TW106142006A
Other languages
Chinese (zh)
Inventor
川上哲義
大山行也
玄元一人
亞蘭 J 寇曼
尼爾斯 隆伯格
新达 郑
史帝芬 D 艾爾布奇
瑪格麗特 馬歇爾
丹尼斯 海利
Original Assignee
日商協和醱酵麒麟有限公司
日商小野藥品工業股份有限公司
美商必治妥美雅史谷比公司
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 日商協和醱酵麒麟有限公司, 日商小野藥品工業股份有限公司, 美商必治妥美雅史谷比公司 filed Critical 日商協和醱酵麒麟有限公司
Publication of TW201825119A publication Critical patent/TW201825119A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Genetics & Genomics (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

A method of treating cancer comprising administering an anti-CC-chemokine receptor 4 (CCR4) antibody and an anti-programmed death-1 (PD-1) antibody to a subject is provided. Also provided is a composition or kit for use in treating a cancer comprising an anti-CCR4 antibody and an anti-PD-1 antibody.

Description

使用抗CCR4抗體及抗PD-1抗體治療癌症之方法Method for treating cancer using anti-CCR4 antibody and anti-PD-1 antibody

本發明係關於一種治療有需要之個體之癌症的方法,該方法包含將抗CCR4抗體及抗PD-1抗體投與至個體。本發明進一步提供一種用於治療有需要之個體之癌症的醫藥組合物,該醫藥組合物包含抗CCR4抗體以及抗PD-1抗體。The present invention relates to a method of treating cancer in an individual in need, the method comprising administering an anti-CCR4 antibody and an anti-PD-1 antibody to the individual. The present invention further provides a pharmaceutical composition for treating cancer in an individual in need, the pharmaceutical composition comprising an anti-CCR4 antibody and an anti-PD-1 antibody.

CC趨化因子受體4 (CCR4)被稱為兩種趨化因子之淋巴球趨化因子受體,該兩種趨化因子為胸腺及活化調節趨化因子(TARC) (亦稱為CCL17)及巨噬細胞衍生之趨化因子(MDC) (亦稱為CCL22)。此外,熟知CCR4表現於Th2細胞、CD4+ 調節性T細胞(Treg)及T淋巴瘤細胞,諸如成人T細胞白血病淋巴瘤(ATL)、皮膚T細胞淋巴瘤(CTCL)以及外周T細胞淋巴瘤(PTCL)上。近來,大部分有前景之抗CCR4人類化抗體,莫格利珠單抗(mogamulizumab)已由日本衛生勞動及福利部(Minister of Health,Labour and Welfare in Japan,MHLW)審批通過上市許可以治療CCR4陽性成人T細胞白血病淋巴瘤(ATL)(非專利文獻1)及CCR4+ 復發/頑固性CTCL及PTCL (非專利文獻2)。 此外近來免疫檢查點分子,諸如程序式細胞死亡-1 (PDCD1,PD-1)、程序式細胞死亡配位體1 (PD-L1)、細胞毒性T淋巴球相關聯4 (CTLA-4)以及殺傷抑制性受體(KIR)變得可供用於觸發一些癌症中之抗腫瘤免疫反應。 PD-1 (CD279)為屬於免疫球蛋白家族之55 kDa之I型跨膜蛋白且據報導表現於免疫細胞(特別地包括活化T細胞、調節性T細胞(Treg)、B細胞以及NK細胞)上。其配位體中之一者,PD-L1 (B7同系物-1;B7-H1;CD274)為促成免疫抑制或T細胞去活化之53 kDa之I型跨膜蛋白。PD-L2 (B7-DC或CD273)經標識為PD-1之第二配位體,其類似於PD-L1在T細胞上具有抑制活性。相比之下,CTLA-4特異性地表現於活化CD4+ T細胞上,具有與CD28類似的結構且藉由使CD80/CD86 (B7-1,B7-2)結合在抗原呈遞細胞(APC),諸如樹突狀細胞上以調節T細胞活性。 用於治療不同癌症之治療性抗體已審批通過且/或已經研發用於多個免疫檢查點分子。在美國,已審批通過阻斷PD-1活性之抗PD-1人類IgG4抗體,納武單抗(先前特指5C4、BMS-936558、MDX-1106或ONO-4538)(專利文獻1及非專利文獻3)接著伊派利單抗(ipilimumab)及若BRAF V600突變陽性,則BRAF抑制劑用於治療患有不可切除性或轉移性黑素瘤及進展性疾病之患者。在美國其亦已經審批通過用於隨著基於鉑的化學療法(platinum-based chemotherapy)進展或在其之後治療轉移性鱗狀非小細胞肺癌。OPDIVO (註冊商標)經審批通過以3 mg/kg,每兩週一次投與。在日本,OPDIVO (註冊商標)亦已審批通過用於治療患有不可切除性黑素瘤之患者且可以2 mg/kg,每三週一次投與。此外,在美國已審批通過另一抗PD-1抗體,派立珠單抗(pembrolizumab) (先前特指拉立珠單抗(lambrolizumab)或MK-3475),接著伊派利單抗及若BRAF V600突變陽性,則BRAF抑制劑用於治療患有不可切除性或轉移性黑素瘤及進展性疾病之患者。研發中之其他檢查點抑制劑抗體包括抗CTLA-4抗體(例如,YERVOY (註冊商標)之活性成份的伊派利單抗及曲美單抗)、抗PD-1抗體(例如,MEDI0608)、抗PD-L1抗體(例如,MEDI4736、MPDL3280A及MSB0010718C)及PD-L2Ig (例如,AMP-224)。[ 引用清單 ] [ 專利文獻 ] PTL1:美國專利第8,008,449號[ 非專利文獻 ] NPL1:Ishida等人,J.Clin.Oncol.,2012;30;837-842 NPL 2:Ogura等人,J.Clin.Oncol.,2014;32;1157-1163 NPL 3:Wang等人,Cancer Immunol Res.,2014;2:846-56CC chemokine receptor 4 (CCR4) is known as the lymphocyte chemokine receptor for two chemokines, the two chemokines are the thymus and activation-regulated chemokine (TARC) (also known as CCL17) And macrophage-derived chemokine (MDC) (also known as CCL22). In addition, it is well known that CCR4 is expressed in Th2 cells, CD4 + regulatory T cells (Treg) and T lymphoma cells, such as adult T cell leukemia lymphoma (ATL), cutaneous T cell lymphoma (CTCL) and peripheral T cell lymphoma ( PTCL). Recently, most promising anti-CCR4 humanized antibodies, mogulizumab (mogamulizumab), have been approved by the Ministry of Health, Labour and Welfare in Japan (MHLW) to obtain a marketing license to treat CCR4 Positive adult T cell leukemia lymphoma (ATL) (Non-Patent Document 1) and CCR4 + relapsed / refractory CTCL and PTCL (Non-Patent Document 2). In addition, recent immune checkpoint molecules, such as programmed cell death-1 (PDCD1, PD-1), programmed cell death ligand 1 (PD-L1), cytotoxic T lymphocyte associated 4 (CTLA-4), and Killer inhibitory receptors (KIR) have become available to trigger antitumor immune responses in some cancers. PD-1 (CD279) is a 55 kDa type I transmembrane protein belonging to the immunoglobulin family and is reported to be expressed in immune cells (specifically including activated T cells, regulatory T cells (Treg), B cells, and NK cells) on. One of its ligands, PD-L1 (B7 homolog-1; B7-H1; CD274) is a 53 kDa type I transmembrane protein that contributes to immunosuppression or T cell deactivation. PD-L2 (B7-DC or CD273) is identified as the second ligand of PD-1, which has inhibitory activity on T cells similar to PD-L1. In contrast, CTLA-4 is specifically expressed on activated CD4 + T cells, has a structure similar to CD28 and binds CD80 / CD86 (B7-1, B7-2) to antigen presenting cells (APC) , Such as on dendritic cells to regulate T cell activity. Therapeutic antibodies used to treat different cancers have been approved and / or have been developed for multiple immune checkpoint molecules. In the United States, an anti-PD-1 human IgG4 antibody that blocks PD-1 activity, nivolumab (previously 5C4, BMS-936558, MDX-1106, or ONO-4538) has been approved (Patent Literature 1 and Non-Patent Reference 3) Then ipilimumab (ipilimumab) and if the BRAF V600 mutation is positive, the BRAF inhibitor is used to treat patients with unresectable or metastatic melanoma and progressive disease. It has also been approved in the United States for the treatment of metastatic squamous non-small cell lung cancer with or after platinum-based chemotherapy. OPDIVO (registered trademark) is approved to be administered at 3 mg / kg once every two weeks. In Japan, OPDIVO (registered trademark) has also been approved for the treatment of patients with unresectable melanoma and can be administered at 2 mg / kg every three weeks. In addition, another anti-PD-1 antibody has been approved in the United States, pembrolizumab (previously designated lambrolizumab or MK-3475), followed by ipilimumab and rubraf If the V600 mutation is positive, the BRAF inhibitor is used to treat patients with unresectable or metastatic melanoma and progressive disease. Other checkpoint inhibitor antibodies under development include anti-CTLA-4 antibodies (e.g., ipalimumab and tremelimumab, the active ingredients of YERVOY (registered trademark)), anti-PD-1 antibodies (e.g., MEDI0608), Anti-PD-L1 antibodies (eg, MEDI4736, MPDL3280A, and MSB0010718C) and PD-L2Ig (eg, AMP-224). [ Citation List ] [ Patent Literature ] PTL1: US Patent No. 8,008,449 [ Non-Patent Literature ] NPL1: Ishida et al., J. Clin. Oncol., 2012; 30; 837-842 NPL 2: Ogura et al., J. Clin .Oncol., 2014; 32; 1157-1163 NPL 3: Wang et al., Cancer Immunol Res., 2014; 2: 846-56

本發明提供一種治療有需要之個體之癌症之方法,該方法包含將抗PD-1抗體與抗CCR4抗體組合投與至個體。 在一個實施例中,本發明包括一種治療個體體內之癌症之方法,該方法包含將特異性結合於人類CC趨化因子受體4 (CCR4)之抗體或其抗原結合部分(「抗CCR4抗體或其抗原結合部分」)及特異性結合與人類程序式死亡-1 (PD-1)之抗體或其抗原結合部分(「抗PD-1抗體或其抗原結合部分」)投與至個體。在另一實施例中,本發明包括一種將患有癌症之個體內之腫瘤大小減小至少約1%、5%、10%、15%、20%或30%之方法,該方法包含將抗CCR4抗體或其抗原結合部分及抗PD-1抗體或其抗原結合部分投與至個體。 在一些實施例中,抗CCR4抗體為嵌合抗體、人類抗體或人類化抗體。在其他實施例中,用於該方法之抗CCR4抗體或其抗原結合部分選自由以下組成之群: (i)結合於與抗體相同之抗原決定基的抗體或其抗原結合部分,其包含:包含SEQ ID NO:1中所闡述之序列之重鏈可變區(「VH」)互補決定區(CDR)1、包含SEQ ID NO:2中所闡述之序列之VH CDR2、及包含SEQ ID NO:3中所闡述之序列之VH CDR3中的一或多者,及包含SEQ ID NO:4中所闡述之序列之輕鏈可變區(「VL」) CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2以及包含SEQ ID NO:6中所闡述之序列之VL CDR3中的一或多者; (ii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:1中所闡述之序列之VH CDR1、包含SEQ ID NO:2中所闡述之序列之VH CDR2、包含SEQ ID NO:3中所闡述之序列之VH CDR3、包含SEQ ID NO:4中所闡述之序列之VL CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2,以及包含SEQ ID NO:6中所闡述之序列之VL CDR3; (iii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:7中所闡述之序列之VH及包含SEQ ID NO:8中所闡述之序列之VL; (iv)抗體,其包含:包含SEQ ID NO:9中所闡述之序列之重鏈及包含SEQ ID NO:10中所闡述之序列之輕鏈; (v)抗體或其抗原結合部分,與(ii)之該抗體交叉競爭;以及 (vi)莫格利珠單抗或其抗原結合部分。 在其他實施例中,抗PD-1抗體為嵌合抗體、人類化抗體或人類抗體。在一些其他實施例中,抗PD-1抗體或其抗原結合部分選自由以下組成之群: (i)結合於與抗體相同之抗原決定基的抗體或其抗原結合部分,其包含:包含SEQ ID NO:11中所闡述之序列之VH CDR1、包含SEQ ID NO:12中所闡述之序列之VH CDR2以及包含SEQ ID NO:13中所闡述之序列之VH CDR3中的一或多者,及包含SEQ ID NO:14中所闡述之序列之VL CDR1、包含SEQ ID NO:15中所闡述之序列之VL CDR2以及包含SEQ ID NO: 16中所闡述之序列之VL CDR3中的一或多者; (ii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:11中所闡述之序列之VH CDR1、包含SEQ ID NO:12中所闡述之序列之VH CDR2、包含SEQ ID NO:13中所闡述之序列之VH CDR3、包含SEQ ID NO:14中所闡述之序列之VL CDR1、包含SEQ ID NO:15中所闡述之序列之VL CDR2,以及包含SEQ ID NO:16中所闡述之序列之VL CDR3; (iii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:17中所闡述之序列之VH及包含SEQ ID NO:18中所闡述之序列之VL; (iv)抗體,其包含:包含SEQ ID NO:19中所闡述之序列之重鏈及包含SEQ ID NO:20中所闡述之序列之輕鏈; (v)抗體或其抗原結合部分,與(ii)之該抗體交叉競爭; (vi)納武單抗或其抗原結合部分; (vii)派立珠單抗或其抗原結合部分;以及 (viii) MEDI0608或其抗原結合部分。 在某些實施例中,可藉由本發明之方法治療之癌症選自由以下組成之群:黑素瘤癌、肝癌、肝細胞細胞癌(hepatocellular cell carcinoma)、膽管癌、腎癌、前列腺癌、乳癌、結腸癌、肺癌、骨癌、胰臟癌、皮膚癌、頭部或頸部癌、皮膚或眼內惡性黑色素瘤、子宮癌、卵巢癌、宮頸癌、子宮內膜癌、直腸癌、肛門區癌、胃癌、睾丸癌、子宮癌、輸卵管癌、子宮內膜癌、子宮頸癌、陰道癌、外陰癌、霍奇金氏病(Hodgkin's Disease)、非霍奇金氏淋巴瘤(non-Hodgkin's lymphoma)、食道癌、小腸癌、內分泌系統癌、甲狀腺癌、副甲狀腺癌、腎上腺癌、軟組織肉瘤、尿道癌、陰莖癌、包括急性骨髓白血病、慢性骨髓性白血病、急性淋巴母細胞白血病、慢性淋巴球性白血病之慢性或急性白血病、兒童固態腫瘤、淋巴球性淋巴瘤、膀胱癌、腎臟或尿管癌、腎盂癌、中樞神經系統(CNS)腫瘤、原發性CNS淋巴瘤、血管生成腫瘤(tumor angiogenesis)、膠質母細胞瘤、脊柱軸腫瘤(spinal axis tumor)、腦幹神經膠瘤、垂體腺瘤、間皮瘤、卡堡氏肉瘤(Kaposi's sarcoma)、表皮樣癌、鱗狀細胞癌、T細胞淋巴瘤、包括由石棉誘導之彼等之環境誘導癌症及其任何組合。在一些實施例中,該癌症為肺癌、胃腸癌或兩者。在其他實施例中,肺癌為非小細胞肺癌或小細胞肺癌,胃腸癌為食道癌或胃癌,以及T細胞淋巴瘤為ATL、CTCL或PTCL。在另其他實施例中,該非小細胞肺癌為非鱗狀非小細胞肺癌或鱗狀非小細胞肺癌。在一特定實施例中,該癌症為黑素瘤。在又其他實施例中,該癌症為漸進性、轉移性及/或不可切除性癌症。 在一些實施例中,抗CCR4抗體以至少約0.1、0.3、0.5、0.75、1.0、2.0或3.0 mg/kg投與。在某些實施例中,抗CCR4抗體或其抗原結合部分以至少一週一次、至少每兩週一次或至少一週一次接著每兩週一次投與。在一特定實施例中,抗CCR4抗體或其抗原結合部分在第一次給藥之後以一週一次持續四週,接著每兩週一次投與。 在某些實施例中,抗PD-1抗體或其抗原結合部分以至少約1 mg/kg、2.0 mg/kg、3.0 mg/kg或5 mg/kg、或以至少約240 毫克/身體作為平調劑量投與。在其他實施例中,抗PD-1抗體或其抗原結合部分以至少一週一次、至少每兩週一次、或至少每三週一次投與。在另其他實施例中,抗PD-1抗體或其抗原結合部分以2.0 mg/kg每兩週一次或以3.0 mg/kg每兩週一次,或以240 毫克/身體作為平調劑量每兩週一次投與。 在一些實施例中,將抗CCR4抗體或其抗原結合部分及抗PD-1抗體或其抗原結合部分並行地或依序投與至該個體。在其他實施例中,抗CCR4抗體或其抗原結合部分在抗PD-1抗體之前或之後投與。在其他實施例中,投藥導致以下效果中之一或多者: (i)減少之CCR4+ T細胞, (ii)減少之調節性T細胞, (iii)增強之腫瘤組織中之PD-L1表現, (iv)減少之PD-1陽性T細胞, (v)增加之腫瘤浸潤性淋巴球(TIL),以及 (vi)其任何組合。 在某些其他實施例中,該等方法進一步包含額外抗癌劑。在一特定實施例中,癌症中之一或多種腫瘤表現PD-L1、PD-L2、CCR4或其任何組合。 在某些實施例中,本發明亦包括一種用於治療罹患癌症之個體之套組,該套組包含: (a)介於0.1至10 mg/kg體重範圍內的劑量之特異性結合於PD-1之抗體或其抗原結合部分(「抗PD-1抗體或其抗原結合部分」); (b)介於0.1至10 mg/kg體重範圍內的劑量之特異性結合於CCR4之抗體或其抗原結合部分(「抗CCR4抗體或其抗原結合部分」);以及 (c)在該等方法中使用抗PD-1抗體及抗CCR4抗體之說明書。 在其他實施例中,本發明包括一種用於治療癌症之醫藥組合物,該醫藥組合物包含(a)特異性結合於PD-1且抑制PD-1活性之抗體或其抗原結合部分(「抗PD-1抗體或其抗原結合部分」)及特異性結合於CCR4且抑制CCR4之抗體或其抗原結合部分(「抗CCR4抗體或其抗原結合部分」)。The present invention provides a method for treating cancer in an individual in need, the method comprising administering an anti-PD-1 antibody in combination with an anti-CCR4 antibody to the individual. In one embodiment, the present invention includes a method of treating cancer in an individual, the method comprising binding an antibody or antigen-binding portion thereof that specifically binds to human CC chemokine receptor 4 (CCR4) ("anti-CCR4 antibody or The antigen-binding portion ") and an antibody or antigen-binding portion that specifically binds to human programmed death-1 (PD-1) (" anti-PD-1 antibody or antigen-binding portion ") are administered to an individual. In another embodiment, the invention includes a method of reducing the size of a tumor in an individual with cancer by at least about 1%, 5%, 10%, 15%, 20%, or 30%, the method comprising The CCR4 antibody or its antigen-binding portion and the anti-PD-1 antibody or its antigen-binding portion are administered to the individual. In some embodiments, the anti-CCR4 antibody is a chimeric antibody, a human antibody, or a humanized antibody. In other embodiments, the anti-CCR4 antibody or antigen-binding portion thereof used in the method is selected from the group consisting of: (i) an antibody or antigen-binding portion that binds to the same epitope as the antibody, which includes: comprising The heavy chain variable region ("VH") complementarity determining region (CDR) 1, the sequence set forth in SEQ ID NO: 1, the VH CDR2 containing the sequence set forth in SEQ ID NO: 2, and the SEQ ID NO: One or more of the VH CDR3 of the sequence set forth in 3, and the light chain variable region ("VL") CDR1 containing the sequence set forth in SEQ ID NO: 4, including the set forth in SEQ ID NO: 5 VL CDR2 of the sequence and one or more of the VL CDR3 containing the sequence set forth in SEQ ID NO: 6; (ii) an antibody or antigen-binding portion thereof, comprising: comprising the set forth in SEQ ID NO: 1 VH CDR1 of the sequence, VH CDR2 of the sequence set forth in SEQ ID NO: 2, VH CDR3 of the sequence set forth in SEQ ID NO: 3, VL CDR1 of the sequence set forth in SEQ ID NO: 4 VL CDR2 containing the sequence set forth in SEQ ID NO: 5, and VL CDR3 containing the sequence set forth in SEQ ID NO: 6; (iii) antibody or anti- The binding portion, comprising: VH comprising the sequence set forth in SEQ ID NO: 7 and VL comprising the sequence set forth in SEQ ID NO: 8; (iv) antibody, comprising: comprising the set forth in SEQ ID NO: 9 The heavy chain of the sequence described and the light chain comprising the sequence set forth in SEQ ID NO: 10; (v) the antibody or antigen-binding portion thereof, which cross-competes with the antibody of (ii); and (vi) Mogley beads MAb or its antigen binding portion. In other embodiments, the anti-PD-1 antibody is a chimeric antibody, a humanized antibody, or a human antibody. In some other embodiments, the anti-PD-1 antibody or antigen-binding portion thereof is selected from the group consisting of: (i) an antibody or antigen-binding portion that binds to the same epitope as the antibody, which comprises: comprising SEQ ID One or more of VH CDR1 of the sequence set forth in NO: 11, VH CDR2 containing the sequence set forth in SEQ ID NO: 12, and VH CDR3 containing the sequence set forth in SEQ ID NO: 13, and including One or more of VL CDR1 of the sequence set forth in SEQ ID NO: 14, VL CDR2 containing the sequence set forth in SEQ ID NO: 15, and VL CDR3 containing the sequence set forth in SEQ ID NO: 16; (ii) Antibody or antigen-binding portion thereof, comprising: VH CDR1 comprising the sequence set forth in SEQ ID NO: 11, VH CDR2 comprising the sequence set forth in SEQ ID NO: 12, comprising SEQ ID NO: 13 VH CDR3 of the sequence described, VL CDR1 of the sequence set forth in SEQ ID NO: 14, VL CDR2 of the sequence set forth in SEQ ID NO: 15, and the sequence of SEQ ID NO: 16 VL CDR3; (iii) antibody or antigen-binding portion thereof, comprising: comprising SEQ ID NO: 17 VH of the sequence and VL comprising the sequence set forth in SEQ ID NO: 18; (iv) Antibody comprising: a heavy chain comprising the sequence set forth in SEQ ID NO: 19 and comprising the set forth in SEQ ID NO: 20 The light chain of the sequence; (v) the antibody or its antigen-binding portion, cross-competing with the antibody of (ii); (vi) nivolumab or its antigen-binding portion; (vii) peclizumab or its antigen A binding portion; and (viii) MEDI0608 or an antigen binding portion thereof. In certain embodiments, the cancer that can be treated by the method of the present invention is selected from the group consisting of melanoma cancer, liver cancer, hepatocellular cell carcinoma (hepatocellular cell carcinoma), cholangiocarcinoma, kidney cancer, prostate cancer, breast cancer , Colon cancer, lung cancer, bone cancer, pancreatic cancer, skin cancer, head or neck cancer, skin or intraocular malignant melanoma, uterine cancer, ovarian cancer, cervical cancer, endometrial cancer, rectal cancer, anal area Cancer, stomach cancer, testicular cancer, uterine cancer, fallopian tube cancer, endometrial cancer, cervical cancer, vaginal cancer, vulvar cancer, Hodgkin's Disease, non-Hodgkin's lymphoma ), Esophageal cancer, small intestine cancer, endocrine system cancer, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, including acute myeloid leukemia, chronic myelogenous leukemia, acute lymphoblastic leukemia, chronic lymphocytic Chronic or acute leukemia of children with leukemia, solid tumors in children, lymphocytic lymphoma, bladder cancer, kidney or urinary tract cancer, renal pelvis cancer, central nervous system (CNS) swelling Neoplasms, primary CNS lymphoma, tumor angiogenesis, glioblastoma, spinal axis tumor, brain stem glioma, pituitary adenoma, mesothelioma, Kaposi's sarcoma ( Kaposi's sarcoma), epidermoid carcinoma, squamous cell carcinoma, T-cell lymphoma, including environment-induced cancers induced by asbestos, and any combination thereof. In some embodiments, the cancer is lung cancer, gastrointestinal cancer, or both. In other embodiments, the lung cancer is non-small cell lung cancer or small cell lung cancer, the gastrointestinal cancer is esophageal cancer or gastric cancer, and the T cell lymphoma is ATL, CTCL, or PTCL. In still other embodiments, the non-small cell lung cancer is non-squamous non-small cell lung cancer or squamous non-small cell lung cancer. In a specific embodiment, the cancer is melanoma. In yet other embodiments, the cancer is progressive, metastatic, and / or unresectable cancer. In some embodiments, the anti-CCR4 antibody is administered at at least about 0.1, 0.3, 0.5, 0.75, 1.0, 2.0, or 3.0 mg / kg. In certain embodiments, the anti-CCR4 antibody or antigen-binding portion thereof is administered at least once a week, at least once every two weeks, or at least once a week and then once every two weeks. In a specific embodiment, the anti-CCR4 antibody or antigen-binding portion thereof is administered once a week for four weeks after the first administration, and then administered once every two weeks. In certain embodiments, the anti-PD-1 antibody or antigen-binding portion thereof is at least about 1 mg / kg, 2.0 mg / kg, 3.0 mg / kg, or 5 mg / kg, or at least about 240 mg / body as the average Dosage adjustment. In other embodiments, the anti-PD-1 antibody or antigen-binding portion thereof is administered at least once a week, at least once every two weeks, or at least once every three weeks. In still other embodiments, the anti-PD-1 antibody or antigen-binding portion thereof is at 2.0 mg / kg every two weeks or at 3.0 mg / kg once every two weeks, or at 240 mg / body as a flat dose every two weeks Give once. In some embodiments, the anti-CCR4 antibody or antigen-binding portion thereof and the anti-PD-1 antibody or antigen-binding portion thereof are administered to the individual concurrently or sequentially. In other embodiments, the anti-CCR4 antibody or antigen-binding portion thereof is administered before or after the anti-PD-1 antibody. In other embodiments, administration results in one or more of the following effects: (i) reduced CCR4 + T cells, (ii) reduced regulatory T cells, (iii) enhanced PD-L1 expression in tumor tissue , (Iv) decreased PD-1 positive T cells, (v) increased tumor infiltrating lymphocytes (TIL), and (vi) any combination thereof. In certain other embodiments, the methods further include additional anti-cancer agents. In a particular embodiment, one or more of the tumors exhibit PD-L1, PD-L2, CCR4, or any combination thereof. In certain embodiments, the present invention also includes a kit for treating individuals suffering from cancer, the kit comprising: (a) a dose ranging from 0.1 to 10 mg / kg body weight that specifically binds to PD -1 antibody or antigen-binding portion thereof ("anti-PD-1 antibody or antigen-binding portion"); (b) an antibody or its specific binding to CCR4 at a dose in the range of 0.1 to 10 mg / kg body weight Antigen binding portion ("anti-CCR4 antibody or antigen binding portion thereof"); and (c) instructions for using anti-PD-1 antibody and anti-CCR4 antibody in these methods. In other embodiments, the invention includes a pharmaceutical composition for treating cancer, the pharmaceutical composition comprising (a) an antibody or antigen-binding portion thereof that specifically binds to PD-1 and inhibits PD-1 activity ("Anti PD-1 antibody or antigen-binding portion ") and an antibody or antigen-binding portion that specifically binds to CCR4 and inhibits CCR4 (" anti-CCR4 antibody or antigen-binding portion ").

除非另外定義,否則本文所使用之所有技術及科學術語均具有熟習本發明所屬領域者通常理解的意義。以下參考文獻為技術者提供用於本發明中之多個術語之一般定義:Singleton等人, Dictionary of Microbiology and Molecular Biology (第2版 1994); The Cambridge Dictionary of Science and Technology (Walker版, 1988); The Glossary of Genetics,第5版,R. Rieger等人(編), Springer Verlag (1991);以及 Hale & Marham, The Harper Collins Dictionary of Biology (1991)。如本文所使用,除非另外規定,否則以下術語具有下文歸屬於其之意義。 「投與」係指使用熟習此項技術者已知之多種方法及傳遞系統中之任一者以將包含治療劑之組合物物理引入至個體。抗PD-1抗體之投藥途徑包括靜脈內、肌肉內、皮下、腹膜內、脊髓或其他非經腸投藥途徑,例如藉由注射或輸注。如本文中所使用,片語「非經腸投與」意謂通常藉由注射之除腸及局部投與之外的投與模式,且包括(但不限於)靜脈內、肌肉內、動脈內、鞘內、淋巴管內、病灶內、囊內、眶內、心內、皮內、腹膜內、經氣管、皮下、表皮下、關節內、囊下、蛛膜下、脊柱內、硬膜外及胸骨內注射及輸液,以及活體內電穿孔。在一些實施例中,藥劑經由非腸胃外路徑,在某些實施例中經口投與。其他非腸胃外途徑包括局部、表皮或黏膜投藥途徑,例如鼻內、經陰道、經直腸、舌下或局部。投與亦可例如執行一次、複數次及/或歷經一或多個延長之時間段。 如本文所使用之「不良事件」 (AE)為與醫學治療之使用相關聯之任何不利且一般不希望或不合意的跡象(包括異常實驗室研究結果)、症狀或疾病。舉例而言,不良事件可回應於治療而與免疫系統之活化或免疫系統細胞(例如T細胞)之擴增相關聯。醫學治療可具有一或多個相關AE且各AE可具有相同或不同的嚴重程度。參考能夠「更改不良事件」之方法意謂降低一或多個AE之發生率及/或嚴重性之治療療法。 「經分離抗體」係指實質上不含具有不同抗原特異性之其他抗體之抗體(例如,特異性結合於PD-1之經分離抗體實質上不含特異性結合於除PD-1之外的抗原之抗體)。然而,特異性結合於PD-1之經分離抗體可與其他抗原,諸如來自不同物種之PD-1分子具有交叉反應。此外,經分離抗體可實質上不含其他細胞材料及/或化學品。 「抗-抗原」抗體係指特異性結合於抗原之抗體。舉例而言,抗PD-1抗體特異性結合於PD-1且抗CTLA-4抗體特異性結合於CTLA-4。 抗體之「抗原結合部分」(亦稱為「抗原結合片段」)係指保留特異性結合於由整個抗體所結合之抗原之能力的抗體之一或多個片段。 在本發明中,「包含(comprises/comprising)」、「含有」以及「具有」及其類似者可具有在美國專利法中歸屬其等之含義且可意謂「包括(includes/including)」及其類似者;「基本上由…組成」或「基本上組成」同樣地具有歸屬於美國專利法中之含義且術語為開放式,只要所引用之基本或新穎特徵不因大於所引用之內容的存在而改變,但排除先前技術實施例,則允許大於所引用之內容之存在。 「癌症」係指特徵為異常細胞在體內不受控生長之廣泛之各種疾病群。不受控的細胞分裂及生長分裂及生長導致形成侵入鄰近組織且亦可經由淋巴系統或血流轉移至身體之遠端部分的惡性腫瘤。癌轉移之後,遠端腫瘤可稱為「衍生自」原始,癌轉移前腫瘤。由於遠端腫瘤衍生自癌轉移前腫瘤,所以「衍生自」腫瘤亦可包含癌轉移前腫瘤。 術語「免疫療法」係指藉由包含誘導、提高、抑制或以其他方式修飾免疫反應之方法治療罹患疾病或處於感染或遭受疾病復發之風險中的個體。 個體之「治療」或「療法」係指在個體上進行之任何類型之介入或處理,或向個體投與活性劑,目標為逆轉、緩解、改善、抑制、減緩或防止症狀、併發症或病狀或與疾病相關之生物化學標誌的發作、進展、發展、嚴重程度或復發。 藥物或治療劑之「治療有效量」、「有效量」,「有效劑量」或「治療有效劑量」為當單獨的或與另一治療劑組合使用時,保護個體免於疾病發作或促進由疾病症狀之嚴重強度降低,無疾病症狀之時間段之頻率及持續時間增加證明的疾病消退,或預防由於疾病病痛而損傷或失能的藥物之任何量。治療劑促進疾病消退之能力可使用熟習此項技術者已知的多種方法評估,諸如在臨床試驗期間在人類個體中評估、在預測人類中之功效的動物模型系統中評估,或藉由在活體外分析法中分析試劑活性來評估。 在一個實施例中,本文中所描述之抗體或抗體之組合促進個體體內之癌症消退或預防其他腫瘤生長。在某些實施例中,治療有效量之藥物促進癌症消退至消除癌症之點。「促進癌症消退」意謂單獨或與抗腫瘤劑組合投與有效量之藥物,使得腫瘤生長或尺寸減小、腫瘤壞死、至少一種疾病症狀的嚴重程度降低、無疾病症狀之時間段之頻率及持續時間增加,或防止由於疾病病痛而損傷或失能。另外,關於治療之術語「有效的」及「效用」包括藥理學效用及生理學安全性兩者。藥理學效用係指藥物促進患者中之癌症消退之能力。生理學安全性係指由於投與藥物而產生之毒性程度,或對細胞、器官及/或生物體水準之其他不良生理學效果(不良效果)。 舉例而言,對於治療腫瘤,相對於未經治療之個體或在某些實施例中,相對於經標準護理療法治療之患者,治療有效量之抗體或其組合可抑制細胞生長或腫瘤生長至少約10%、至少約20%、至少約40%、至少約60%或至少約80%。在本發明之其他實施例中,腫瘤消退可觀察且持續至少約20天、至少約40天或至少約60天之時間段。不管治療效用之此等最終量測,免疫治療藥物的評估必須亦考慮到「免疫相關之」反應模式。 「免疫相關之」反應模式係指在使用免疫治療劑治療之癌症患者中通常觀測到的臨床反應模式,該等免疫治療劑藉由誘發癌症特異性免疫反應或藉由修改原生免疫過程而產生抗腫瘤效果。此反應模式之特徵為在腫瘤負荷初始增加或新病變出現之後的有益治療效果,其在傳統化學治療劑的評估中將歸類為疾病進展且將與藥物失效同義。因此,免疫治療劑之適當評估可能需要長期監測此等試劑對目標疾病之效果。 藥物之治療有效量包括「預防有效量」,其為當單獨的或與抗腫瘤劑組合投與至處於罹患癌症之風險下(例如,具有癌前病狀之個體)或遭受癌症復發之個體時,抑制癌症之發展或復發之藥物之任何量。在某些實施例中,預防有效量完全地阻止癌症之發展或復發。「抑制」癌症之發展或復發意謂降低癌症之發展或復發的可能性或完全地阻止癌症之發展或復發。 應理解,替代物(例如「或」)之使用意謂替代物中之一者、兩者或其任何組合。如本文中所使用,應理解不定冠詞「一(a/an)」係指任何引用或列舉之組分的「一或多者」。 術語「約」或「基本上包含」係指在如由一般熟習此項技術者所判定之值或組合物之可接受誤差範圍內的值或組合物,其將部分地取決於如何量測或判定值或組合物,即量測系統之侷限性。舉例而言,「約」或「基本上包含」可意謂根據此項技術中之實踐在1個或大於1個標準偏差內。可替代地,「約」或「基本上包含」可意謂至多10%之範圍(亦即,+10%至-10%)。舉例而言,約3 mg可包括介於2.7 mg與3.3 mg之間的任何數值(對於10%)。此外,尤其在生物系統或方法方面,該等術語可意指值之至多一個數量級或至多5倍。當特定值或組合物提供於本申請案及申請專利範圍中時,除非另有說明,否則「約」或「基本上包含」之含義應假設為在特定值或組合物之可接受誤差範圍內。 如本文中所使用,術語「約一週一次」意謂大致數值,且「約一週一次」可包括每六天至每八天。因此「一週一次」之給藥頻率可為每六天、每七天或每八天。 如本文所述,除非另外規定,否則任何濃度範圍、百分比範圍、比率範圍或整數範圍應理解為包括在所列舉範圍內之任何整數值及(在適當時)其分數(諸如整數之十分之一及百分之一)。 如本文中所使用之術語「CC趨化因子受體4」、「CCR4」、「CCR4肽」、「CCR4蛋白」或「CCR4多肽」可在本文中互換使用且意謂包含SEQ ID NO:22之胺基酸序列或其功能片段之多肽;包含GenBank寄存編號為NP_005499.1之胺基酸序列之多肽、包含在由SEQ ID NO:22或NP_005499.1表示之胺基酸序列中缺失、替代或添加一或多個胺基酸殘基的胺基酸序列且具有CCR4活性之多肽;包含與由SEQ ID NO:22表示之胺基酸序列具有至少約85%同源性、至少約90%同源性、至少約93%同源性以及至少約95%、約96%、約97%、約98%或約99%、更高同源性之胺基酸序列且具有CCR4活性之多肽;以及包括SNP變異體之相關多肽及其類似者。相關多肽包括SNP變異體、剪接變異體、片段、替代物、缺失以及插入,其保存CCR4活性及/或功能。 此外,CCR4多肽包括由SEQ ID NO: 21或GenBank寄存編號NM_005508.4之核苷酸序列編碼之多肽。在一些實施例中,對CCR4進行編碼之基因包括在SEQ ID NO: 21或GenBank寄存編號NM_005508.4之核苷酸序列中具有一或多個核苷酸之缺失、替代或添加的核苷酸序列。在其他實施例中,CCR4基因對具有CCR4功能之多肽進行編碼且包含與SEQ ID NO:21或GenBank寄存編號NM_005508.4之核苷酸序列具有至少約60%或更高同源性,在一些實施例中至少約80%或更高同源性,且在其他實施例中至少約95%、約96%、約97%、約98%或約99%、更高同源性之核苷酸序列。在一些實施例中,CCR4基因對具有CCR4之功能之多肽進行編碼且包含在嚴格條件下與SEQ ID NO:21或GenBank寄存編號NM_005508.4之核苷酸序列雜交之核苷酸序列。 如本文中所使用,術語「CCR4核酸分子」係指對CCR4多肽進行編碼之聚核苷酸。例示性CCR4核酸分子提供於SEQ ID NO:21或GenBank寄存編號NM_005508.4中。在對真核生物之蛋白質進行編碼之基因的核苷酸序列中,常常識別到基因多形現象。本發明中所使用之CCR4基因亦包括藉由此多形現象在核苷酸序列中產生較小修飾之基因,作為本發明中所使用之基因。 CCR4為從人類未成熟嗜鹼性細胞系KU-812克隆為K5-5的G蛋白偶聯之七個跨膜受體,且可具有由SEQ ID NO:22表示之胺基酸序列。CCR4之胞外區域在胺基酸序列中為位置1至39、位置99至111、位置176至206及位置268至284,且胞內區域在胺基酸序列(GenBank寄存編號NP_005499.1)中為位置68至77、位置134至150、位置227至242及位置309至360。 CCR4可特異性結合於不同分子,包括(但不限於)由胸腺細胞產生之TARC (胸腺及活化調節趨化因子) (J.Biol.Chem.,271,21514,1996)及與巨噬細胞分離之MDC (巨噬細胞衍生之趨化因子) (J.Exp.Med.,185,1595,1997),亦稱為STCP-1 (經刺激T細胞趨化蛋白-1) (J.Biol.Chem.,272,25229,1997)。TARC及MDC亦分別地稱為CCL17及CCL22。CCR4之一或多個功能包括其與TARC及/或MDC的結合能力。本發明中之CCR4之其他功能包括取決於CCR4配位體(諸如CCL17及/或CCL22)之表現CCR4細胞上之Ca2 + 流入、表現CCR4細胞之細胞遷移的功能。 術語「程序式細胞死亡-1」、「PD-1」、「PD-1多肽」或「CD279」在本文中互換地使用且意謂具有PD-1活性之至少一個功能(例如,藉由PD-1途徑活化T細胞及/或結合於PD-L1或PD-L2)之多肽或其片段。在一個實施例中,PD-1包含與NCBI寄存編號NP_005009至少約85%、至少約90%、至少約93%、至少約95%、約96%、約97%、約98%、約99%或約100%序列一致性之胺基酸序列或SEQ ID NO:24中所闡述之胺基酸序列,其中該PD-1蛋白具有與PD-L1及PD-L2之結合活性及/或PD-1之活性。在一些實施例中,PD-1亦包括SNP變異體、剪接變異體、片段、替代物、缺失以及插入,其保存PD-1活性及/或功能。 在其他實施例中,PD-1包含由SEQ ID NO:23或GenBank寄存編號NM_005018.2之核苷酸序列編碼之多肽。PD-1基因亦包括含有DNA之基因,該DNA包含在SEQ ID NO:23或GenBank寄存編號NM_005018.2之核苷酸序列中具有一或多個核苷酸之缺失、替代或添加的核苷酸序列。在一些實施例中,PD-1由具有與SEQ ID NO:23或GenBank寄存編號NM_005018.2之核苷酸序列至少約60%、約70%、約80%、約90%、約95%、約96%、約97%、約98%、約99%或約100%一致之核苷酸序列編碼,且具有PD-1之至少一個功能(例如,藉由PD-1途徑活化T細胞及/或結合於PD-L1或PD-L2)。在其他實施例中,PD-1由在嚴格條件下雜合有SEQ ID NO:23或GenBank寄存編號NM_005018.2之核苷酸序列之核苷酸序列編碼且具備具有PD-1之功能的多肽。 術語「PD-1核酸分子」係指對PD-1多肽進行編碼之聚核苷酸。例示性PD-1核酸分子提供於SEQ ID NO:23或GenBank寄存編號NM_005018.2中。在對真核生物之蛋白質進行編碼之基因的核苷酸序列中,常常識別到基因多形現象。本發明中所使用之PD-1基因亦包括藉由此多形現象在核苷酸序列中產生較小修飾之基因,作為本發明中所使用之基因。 PD-1經克隆為屬於免疫球蛋白家族之55 kDa之I型膜蛋白(Ishida等人,1992;11;3887-3895)。PD-1表現於活化之T淋巴球上。PD-1之胞內域含有ITSM基元(基於免疫受體酪胺酸之交換基元)及可為免疫反應之壓製域之ITIM基元(基於免疫受體酪胺酸之抑制基元)。由於缺乏PD-1之小鼠出現狼瘡類自體免疫疾病,諸如腎小球腎炎及關節炎及類似於擴張型心肌症之疾病,因此PD-1似乎係關於免疫反應之負調節劑。其他PD-1配位體,PD-L1被稱為作為PD-1之配位體及藉由結合於PD-1而作為T細胞增殖之抑制劑配位體的I型跨膜蛋白。其具有免疫球蛋白V類域、C類域及胞質尾區。已知PD-L1表現於抗原呈遞細胞及一些癌細胞上。 「抗CCR4抗體」係指選擇性地結合於人類CCR4多肽之抗體。本文提供例示性抗CCR4抗體,例如,結合於抗原決定基的由SEQ ID NO:22之胺基酸殘基2至29構成之抗CCR4抗體。在一個實施例中,抗CCR4抗體為莫格利珠單抗(POTELIGEO (註冊商標))。在另一實施例中,抗CCR4抗體減少、抑制或阻止CCR4之一或多個功能。在其他實施例中,抗CCR4抗體與莫格利珠單抗交叉競爭。 「抗PD-1抗體」係指選擇性地結合於PD-1多肽之抗體。例示性抗PD-1抗體描述於(例如)美國專利第8,008,449號中,其以引用之方式併入本文中。在一個實施例中,抗PD-1抗體為納武單抗(OPDIVO (註冊商標))。在另一實施例中,抗PD-1抗體減少、抑制或阻止PD-1之一或多個功能。在其他實施例中,抗PD1抗體與納武單抗交叉競爭。 如本揭示案中所使用,術語「抗體」係指免疫球蛋白或其片段或衍生物,且涵蓋包含抗原結合位點之任何多肽,無論其產生於活體外或活體內。該術語包括(但不限於)多株抗體、單株抗體、單特異性抗體、多特異性抗體、人類化抗體、單鏈抗體、嵌合抗體、合成抗體、重組抗體、雜交抗體、突變抗體以及移植抗體。除非藉由術語「完整」另外修改,如同「完整抗體」,否則出於本揭示案之目的,術語「抗體」亦包括保存抗原結合功能(亦即,能夠特異性地結合CCR4或PD-1)之抗體片段(或抗原結合片段),諸如Fab、F(ab')2 、Fv、單鏈Fv(scFv)、Fd、dAb以及其他抗體片段。 通常地,此類片段將包含抗原結合域(或部分)。抗體分子由被稱作重鏈(下文中被稱作H鏈)及輕鏈(下文中被稱作L鏈)之多肽形成。此外,H鏈由來自其N端之H鏈可變區(亦被稱作VH 或VH)及H鏈恆定區(亦被稱作CH)之區域構成,且L鏈由來自其N端之L鏈可變區(亦被稱作VL 或VL)及L鏈恆定區(亦被稱作CL)之區域構成。關於CH,α、δ、ε、γ及mu鏈已知用於各子類別。關於CL,已知λ及κ。IgG抗體具有兩個重鏈及兩個輕鏈,且形成由VH及VL構成之兩個抗原結合位點。因此,IgG抗體為二價抗體。 術語「抗體片段」、「抗原結合域」、「抗原結合片段」、「抗原結合部分」以及「結合片段」係指包含負責抗體與抗原之間的特異性結合之胺基酸的抗體分子之一部分。舉例而言,在抗原較大時,抗原結合域可能僅結合於抗原之一部分。負責與抗原結合域特異性相互作用之抗原分子的一部分被稱為「抗原決定基」或「抗原決定子」。抗原結合域通常包含抗體輕鏈可變區(VL 或VL)及抗體重鏈可變區(VH 或VH)。然而,不必必須包含兩者。舉例而言,Fd抗體片段僅由VH 結構域組成,但仍保留完整抗體之一定抗原結合功能。 術語「互補決定區」或「CDR」係指包含於抗體之可變區中之抗體之抗原結合功能的必需部分。在VH及VL中分別地存在三個CDR且此等在各可變區中經定義為CDR1至CDR3。 抗體之抗原結合片段可藉由重組DNA技術或藉由完整抗體之酶裂解或化學裂解而產生。抗原結合片段包括Fab、Fab'、F(ab')2 、Fv以及單鏈抗體。應理解,除「雙特異性」或「雙功能性」抗體以外的抗體之各結合位點一致。藉由酶(木瓜酶)分解抗體產生兩個相同的抗原結合片段,亦稱為「Fab」片段及「Fc」片段,該等兩個抗原結合片段無抗原結合活性但具有結晶能力。藉由酵(胃蛋白酶)分解抗體產生F(ab')2 片段,其中抗體分子之兩個臂保持鍵聯且包含兩個抗原結合位點。F(ab')2 片段能夠與抗原交聯。當在本文中使用時,「Fv」係指抗體中保留抗原識別位點及抗原結合位點兩者之最小片段。當在本文中使用時,「Fab」係指包含輕鏈之恆定域及重鏈之CH1域之抗體的片段。 術語「mAb」係指單株抗體。本發明中可使用之抗體包含(但不限於)全部天然抗體、雙特異性抗體;嵌合抗體;Fab、Fab'、單鏈V區片段(scFv)、融合多肽及非習知抗體。 結構域係指構成抗體分子之各多肽的功能性結構單元。此外,本發明中可使用之Fc係指由鉸鏈域、CH2域及CH3域形成之H鏈恆定區之部分序列及部分結構。 此外,CH由來自N端之CH1域、鉸鏈域、CH2域及CH3域形成。本發明中之CH1域、鉸鏈域、CH2域、CH3域及Fc區可根據EU指數藉由來自N端之多個胺基酸殘基識別[Kabat等人,Sequences of Proteins of Immunological Interest,US Dept.Health and Human Services(1991)]。多個胺基酸殘基由Kabat等人藉由EU指數指出且在本發明中,前多個胺基酸殘基指示多肽之原始或父代殘基及後多個胺基酸殘基指示多肽之置換或替代胺基酸殘基。 具體而言,分別地,CH1由來自EU指數之位置118至215之胺基酸序列識別,鉸鏈由來自EU指數之位置216至230之胺基酸序列識別,CH2由來自EU指數之位置231至340之胺基酸序列識別,及CH3由來自EU指數之位置341至447之胺基酸序列識別。 如本文中所使用,術語「重組抗體」係指藉由重組技術以及獲自融合瘤之單株抗體產生之抗體。重組抗體包括藉由將人類抗體恆定區與非人類抗體可變區結合而製備之嵌合抗體、藉由將非人類抗體可變區之H鏈及L鏈之CDR至人類抗體可變區之構架區(下文中簡稱為FR)中而製備之人類化抗體(或CDR移植抗體),及藉由使用產生人類抗體之動物或其類似方法製備之人類抗體。 如本文中所使用之術語「嵌合抗體」係指其中將非人類動物抗體之VH及VL之胺基酸序列移植至人類抗體之對應VH及VL中之抗體。嵌合抗體可藉由從衍生於非人類動物之產生單株抗體之融合瘤獲得對VH及VL進行編碼之cDNA,將該等cDNA插入至具有對人類抗體之CH及CL進行編碼之DNA的動物細胞之表現載體中以便構建人類嵌合抗體表現載體,且接著將該載體引入至動物細胞中以便表現抗體而產生。 術語「人類化抗體」係指其中非人類動物抗體之VH及VL之CDR之胺基酸序列經移植至人類抗體之VH及VL之對應的CDR中之抗體。除VH及VL之CDR以外,該區域稱為FR。 人類化抗體可由以下方式產生:(i)構造對由非人類抗體之VH之CDR之胺基酸序列及任何人類抗體之VH之構架區(FR)之胺基酸序列構成之VH之胺基酸序列進行編碼的cDNA及對由非人類動物抗體之VL之CDR之胺基酸序列及任何人類抗體之VL之FR之胺基酸序列構成之VL之胺基酸序列進行編碼的cDNA;(ii)將此等cDNA分別地插入至具有對人類抗體之CH及CL進行編碼之DNA的動物細胞之表現載體中以便構建人類化抗體表現載體;以及(iii)將此載體引入至動物細胞中以便表現該抗體。 如本文中所使用,術語「人類抗體」係指具有可變區之抗體,其中構架區及CDR區兩者皆衍生於人體生殖系免疫球蛋白序列。此外,若該抗體含有恆定區,則該恆定區亦衍生於人類生殖系免疫球蛋白序列。本發明之人類抗體可包括不由人類生殖系免疫球蛋白序列編碼之胺基酸殘基(例如藉由活體外隨機或位點特異性突變誘發或藉由活體內體細胞突變引入之突變)。然而,如本文中所使用,術語「人類抗體」並不意欲包括其中衍生於另一哺乳動物物種(諸如小鼠)之生殖系之CDR序列已移植於人類構架序列上的抗體。術語「人類」抗體與「完全人類」抗體同義使用。人類抗體亦包括獲自人類抗體噬菌體庫、選殖永生化人體外周血液淋巴球或根據近年來基因改造、細胞改造及研發改造中的技術改進而製備之產生人類抗體的轉殖基因動物之抗體。 人類抗體可藉由用所需抗原使具有人體免疫球蛋白基因(Tomizuka K等人,Proc Natl Acad Sci USA.97,722 -7,2000)之小鼠免疫來獲得。此外,藉由使用由人類B細胞之抗體基因擴增而形成之噬菌體展示庫來選擇具有所需結合活性之人類抗體,有可能在不進行免疫接種之情況下獲得人類抗體(Winter G等人, Annu Rev Immunol. 12: 433-55. 1994)。 此外,藉由使用艾司坦-巴爾(Epstein-Barr;EB)病毒來永生化人類B細胞以製備具有所需結合活性之產生人類抗體之細胞,有可能獲得人類抗體(Rosen A等人, Nature 267, 52-54. 1977)。 人類抗體噬菌體庫為藉由將由人類B細胞製備之抗體基因插入至噬菌體之基因中而致使抗體片段,諸如Fab及scFv表現於其表面上的噬菌體庫。藉由使用相對於固定抗原襯底之結合活性作為指數有可能自此庫恢復表現具有所需抗原結合活性之抗體片段的噬菌體。抗體片段亦可藉由基因改造技術而轉換為由兩個完整H鏈及兩個完整L鏈構成之人類抗體分子。 產生人類抗體之轉殖基因動物係指藉由將人類抗體基因整合至寄主動物之染色體中而獲得的動物。具體而言,人類抗體基因經引入至小鼠ES細胞。隨後將ES細胞移植至另一小鼠之早期胚胎。產生人類抗體之轉殖基因動物可由該胚胎產生。為自產生人類抗體之轉殖基因動物產生人類抗體,藉由正常融合瘤製備方法以獲得產生人類抗體之融合瘤。培育獲自除人類以外之哺乳動物的融合瘤後,可在該培養物中產生且表現人類抗體。 具體而言,適用於本揭示案之抗體可包括非人類動物抗體、人類化抗體以及藉由此項技術中已知之任何方法,例如融合瘤或產生抗體之細胞產生之人類抗體之VH及VL的胺基酸序列。本發明之抗體中之CL的胺基酸序列可為人類抗體之胺基酸序列或非人類動物抗體之胺基酸序列中的任一者。在某些實施例中,使用人類抗體之Cκ或Cλ之胺基酸序列。本發明之抗體中之CH可為屬於免疫球蛋白中之任一者。在某些實施例中,γ1 (IgG1)、γ2 (IgG2)及γ4 (IgG4)以及其變異體中之任一者可適用於本發明中的抗體。 本發明中所使用之術語「效應功能」係指由抗體誘導之細胞毒活性,包括藉由效應細胞,諸如自然殺手(NK)細胞之抗體依賴性細胞毒性(ADCC)、補體依賴性細胞毒性(CDC)、抗體依賴吞噬作用(ADP)或其任何組合。抗體之效應功能可藉由已知方法調節。舉例而言,此項技術中已知ADCC活性可藉由控制海藻糖(亦被稱作「核海藻糖」)之量來調節,該海藻糖藉由複合型N-糖苷鍵聯糖鏈之還原端中之α1-6鍵而結合於N-乙醯基葡糖胺(GlcNAc),該複合型N-糖苷鍵聯糖鏈結合於在抗體之Fc區中之根據EU指數(Kabat等人,Sequence of Proteins of immunological interests,第5版,1991)之位置297處的天冬醯胺(Asn) (參看WO2005/035586、WO2002/31140、WO00/61739),該等文獻中之每一者以全文引用之方式併入本文中。此外,此項技術中已知可例如藉由修改抗體之Fc區之胺基酸殘基來調節ADCC及/或CDC。 抗體之效應活性可藉由控制結合於抗體之Fc區之複合型N-糖苷鍵聯糖鏈中之核海藻糖之量而增強或降低。為減少結合於結合於該抗體之Fc之複合型N-糖苷鍵聯糖鏈之海藻糖含量,可藉由使用缺乏α1,6-海藻糖基轉移酶基因(FUT8)之CHO細胞以表現抗體而獲得並未結合海藻糖之抗體。 不含海藻糖之抗體具有較高ADCC活性。為增加結合於結合於該抗體之Fc之複合型N-糖苷鍵聯糖鏈之海藻糖含量,可藉由使用表現α1,6-海藻糖基轉移酶基因之宿主以表現抗體而獲得具有海藻糖之抗體。具有海藻糖之抗體具有比不含海藻糖之抗體更低的ADCC活性。 在一個實施例中,本發明中所使用之抗CCR4抗體為具有結合於抗體之Fc區之去海藻糖基化N-糖苷鍵聯糖鏈的抗體。在某些實施例中,本發明中所使用之抗CCR4抗體為具有結合於抗體之Fc區之去海藻糖基化N-糖苷鍵聯糖鏈的抗體,在一些實施例中,約50%或更大。在其他實施例中,抗體之去海藻糖基化範圍為約80%、約90%、約95%、約96%、約97%、約98%或約99%或更大。 此外,藉由修飾抗體之Fc區中之胺基酸殘基,可提高或降低ADCC活性或CDC活性。對Fc區中之胺基酸殘基進行修飾以提高或降低對於FcγR之結合活性,由此控制ADCC活性。對Fc區中之胺基酸殘基進行修飾以提高或降低補體之結合活性,由此控制CDC活性。 舉例而言,抗體之CDC活性可藉由使用描述於US2007/0148165之說明書中之Fc區的胺基酸序列而提高。此外,ADCC活性或CDC活性可藉由修飾如US6,737,056、US7,297,775、US7,317,091、WO2005/070963以及Oganesyan等人(Biol.Crystal.,2008;64;700-704)之說明書中所描述之胺基酸殘基而提高或降低。 在一些實施例中,抗PD-1抗體無效應功能,且僅藉由PD-L1或PD-L2結合來中和PD-1功能。在某些實施例中,適用於本發明之抗PD1抗體可為展現降低之效應功能或無效應功能之IgG2或IgG4子類別。在一個實施例中,用於本發明之抗PD-1抗體可具備具有降低效應功能或無效應功能之Fc變異體。 如本文中所使用,術語「個體」包括任何人類或非人類動物。術語「非人類動物」包括所有脊椎動物,例如哺乳動物及非哺乳動物,諸如非人類靈長類動物、綿羊、狗、貓、馬、牛、雞、兩棲動物、爬行動物等。除非指出,否則術語「患者」或「個體」在本文中互換使用。 關於本發明之組合物而使用之術語「平調劑量」或「平調量」意謂不管患者之體重或身體表面積(BSA)而向患者投與之劑量。因此,平調劑量不以mg/kg劑量形式提供,而是以試劑(例如抗-CCR4抗體及/或抗PD-1抗體)之絕對量提供。舉例而言,60 kg之人與100 kg之人將接收相同劑量之組合物(例如240 mg之抗PD1抗體及80 mg之抗CCR4抗體)。 如本文所提及,術語「基於體重之劑量」意謂投與至患者之劑量係基於患者之重量而計算的。舉例而言,當具有60 kg體重之患者需要3 mg/kg之抗PD-1抗體以及1 mg/kg之抗CCR4抗體時,吾人可攝取適當量之抗PD-1抗體(亦即,180 mg)及抗CCR4抗體(亦即,60 mg)。 在以下子部分中進一步詳細描述本發明之各種態樣。CCR4 CCR4為充當兩個趨化因子之淋巴球趨化因子受體,TARC (CCL17)及MDC (CCL22)之七個跨膜G型蛋白偶聯受體家族中之成員。CCR4表現於包括CD4+ 調節性T細胞(Treg)之CD4+ Th2細胞上。此外,包括成人T細胞白血病淋巴瘤(ATL)、皮膚T細胞淋巴瘤(CTCL)、外圍T細胞淋巴瘤(PTCL)之一些癌症及若干固態腫瘤已經展示以表現CCR4。藉由CCR4結合之配位體誘發Ca2 + 流入於CCR4+ 細胞上且誘發細胞增殖或細胞朝向配位體分泌組織遷移。CD4+ Th2細胞為體液免疫中之調節性細胞。在Th2細胞藉由B細胞促進抗體產生至額外抗原時,Th2細胞亦抑制Th1細胞功能。Th2細胞藉由產生免疫抑制細胞介素,諸如介白素(IL)-10來充當輔助T細胞,從而抑制細胞免疫。 CD4、CD25、CTLA-4以及GITR (糖皮質激素誘導腫瘤壞死因子受體家族相關之基因)為已知的Treg標記(Journal of Allergy and Clinical Immunology,110:693-701,2002)。此外,FoxP3(叉頭盒蛋白3)轉錄因子為參與調節性T細胞之分化以及功能性表現的主基因(Science,299:1057-61,2003)。此外,CCR4亦表現於Treg細胞上(美國公開案第2006/0034841號)。Treg細胞為抑制自體反應性T細胞之活化的T細胞群中之一者且負責免疫自身耐受性,防止免疫系統攻擊健康個體中之自身組織(Immunological Review,182:the whole volume,2001)。Treg細胞經由直接及/或間接路徑,諸如細胞-細胞相互作用,分泌抑制性趨化因子、介白素-10 (IL-10),及轉化生長因子(TGF)-β,耗盡T細胞刺激性趨化因子及IL-2來抑制免疫刺激。 在本文所提供之方法中靶向之例示性表現CCR4之細胞為選自CCR4+ 免疫細胞及CCR4+ 腫瘤細胞中之細胞。在某些實施例中,CCR4+ 細胞為選自以下之細胞:CCR4+ T細胞、CCR4+ Th2細胞、CD4+ CD25+ CCR4+ T細胞、CCR4+ Foxp3+ T細胞、CD4+ CD25+ CCR4+ Foxp3+ T細胞、CD4+ CD25+ CCR4+ CD127low T細胞以及CD4+ CD25+ CD45RA- CCR4+ Foxp3+ T細胞(被稱為效應Treg) (Miyara等人,Immunity,2009;30:899-911),且「Treg細胞」經定義為調節性T細胞。在其他實施例中,CCR4+ Treg由選自以下之至少一個標誌設置定義:CD4+ CD25+ CCR4+ 、CD25+ Foxp3+ 、CCR4+ Foxp3+ 、CD4+ CD25+ CCR4+ Foxp3+ 、CD4+ CD25+ CCR4+ CD127low ,且CD4+ CD25+ CD45RA- CCR4+ Foxp3+ 可為例示性。PD-1 人體中之T細胞活化及去活化係藉由多個膜蛋白同時複雜調節的。此等膜蛋白中之一者,PD-1經克隆為55 kDa之I型膜蛋白(Ishida等人,EMBO J.,1992;11;3887-3895)。PD-1表現於多個細胞類型活化T細胞、調節性T細胞(Treg)、活化B細胞及自然殺手(NK)細胞上且已知PD-1藉由結合於其配位體,PD-L1及PD-L2來調節T細胞活化。PD-1之胞內域含有ITSM基元及可為免疫反應之抑制域之ITIM基元。由PD-L1結合之PD-1誘導T細胞或其他表現PD-1之細胞中之抑制信號,由此阻斷T細胞或其他表現PD-1之細胞之活化、細胞增殖及/或發炎性細胞介素釋放或使T細胞失能。 另一方面,PD-L1 (人類PD-L1 cDNA,AF233516)及PD-L2 (人類PD-L2 NM_025239)為PD-1之配位體。PD-L1表現於多個細胞上,包括諸如活化單核球之抗原呈遞細胞及樹突狀細胞(Freeman等人,Journal of Experimental Medicine(2000),19;1027-1034)。不僅在正常細胞中且亦在若干癌症中確認PD-L1表現及PD-L2表現。 在本發明中,正常細胞上之PD-1表現及癌組織或癌細胞上之PD-L1表現可藉由已知分析,諸如流式細胞量測術、免疫組織化學(IHC)偵測到。「PD-1陽性」之含義指示至少約0.1%、至少約1%、至少約5%、至少約10%或至少約20%或更多染色之人類個體之細胞、組織、體液。本文中所使用之「PD-L1陽性」或「PD-L2陽性」可與「至少約1%之PD-L1及/或PD-L2表現」互換使用。在一個實施例中,PD-L1表現及/或PD-L2表現可藉由此項技術中已知之任何方法使用。在另一實施例中,PD-L1表現及/或PD-L2表現係藉由自動原位雜交(in situ hybridization;IHC)來量測。PD-L1及/或PD-L2陽性腫瘤可因此具有如藉由自動IHC所量測之至少約1%、至少約2%、至少約5%、至少約10%、至少約20%、至少約30%、至少約40%、或至少約50%之表現PD-L1的腫瘤細胞。 CCR4 抗體 適用於本發明之抗CCR4抗體可特異性結合於人類CCR4。在一個實施例中,抗CCR4抗體可減弱、抑制或阻止CCR4蛋白之至少一個功能。在一個實施例中,抗CCR4抗體減少或耗盡CCR4+ 免疫抑制細胞及/或CCR4+ 癌細胞。在另一實施例中,抗CCR4抗體結合於表現於免疫抑制細胞或癌細胞上之CCR4,且藉由以下抗體效應功能來減少或耗盡細胞數量:例如,由主要自然殺手(NK)細胞誘導之抗體依賴性細胞毒性(ADCC)、補體依賴性細胞毒性(CDC)或藉由諸如巨噬細胞或樹突狀細胞之吞噬細胞之抗體依賴性吞噬作用(ADP)及/或中和CCR4配位體。 在一個實施例中,用於本方法中之抗CCR4抗體結合於CCR4分子之胞外區且表現ADCC活性。在另一實施例中,抗CCR4抗體結合於CCR4蛋白之胺基酸2至29中之抗原決定基(例如,對應於SEQ ID NO:22之胺基酸2至29)且表現ADCC活性。在其他實施例中,抗CCR4抗體結合於CCR4蛋白之胺基酸12至29中之抗原決定基(例如,SEQ ID NO:22之胺基酸12至29)且表現ADCC活性。在其他實施例中,抗CCR4抗體與包含以下之抗體交叉競爭以結合於CCR4:包含SEQ ID NO:1中所闡述之序列之VH CDR1、包含SEQ ID NO:2中所闡述之序列之VH CDR2、包含SEQ ID NO:3中所闡述之序列之VH CDR3、包含SEQ ID NO:4中所闡述之序列之VL CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2以及包含SEQ ID NO:6中所闡述之序列之VL CDR3。 在一個實施例中,本發明中所使用之抗CCR4抗體或其抗原結合部分結合於由包含以下之抗體或莫格利珠單抗結合之相同抗原決定基:包含包含SEQ ID NO:1中所闡述之序列之VH CDR1、包含SEQ ID NO:2中所闡述之序列之VH CDR2、包含SEQ ID NO:3中所闡述之序列之VH CDR3、包含SEQ ID NO:4中所闡述之序列之VL CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2以及包含SEQ ID NO:6中中所闡述之序列之VL CDR3。 在另一實施例中,適用於本發明之抗CCR4抗體包含:包含SEQ ID NO:1中所闡述之序列之VH CDR1、包含SEQ ID NO:2中所闡述之序列之VH CDR2以及包含SEQ ID NO:3中所闡述之序列之VH CDR3中的一或多者,以及包含SEQ ID NO: 4中所闡述之序列之VL CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2以及包含SEQ ID NO:6中所闡述之序列之VL CDR3中的一或多者。在其他實施例中,抗CCR4抗體包含:包含SEQ ID NO: 1中所闡述之序列之VH CDR1、包含SEQ ID NO:2中所闡述之序列之VH CDR2、包含SEQ ID NO:3中所闡述之序列之VH CDR3、包含SEQ ID NO:4中所闡述之序列之VL CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2以及包含SEQ ID NO:6中所闡述之序列之VL CDR3。在一些實施例中,抗CCR4抗體包含:包含SEQ ID NO:7中所闡述之序列之VH及包含SEQ ID NO:8中所闡述之序列之VL。在某些實施例中,抗CCR4抗體包含:包含SEQ ID NO:9中所闡述之序列之重鏈及包含SEQ ID NO:10中所闡述之序列之輕鏈。在其他實施例中,抗CCR4抗體之Fc區包含未結合於Fc區之位置297處之N-乙醯基葡糖胺的核海藻糖。在一特定實施例中,抗CCR4抗體為莫格利珠單抗(POTELIGEO (註冊商標))。 在某些實施例中,用於本方法或組合物之抗CCR4抗體具有結合於抗體之Fc區的較低海藻糖基化、去海藻糖基化或無海藻糖基化之N-糖苷鍵聯糖鏈。較低海藻糖基化、去海藻糖基化或無海藻糖基化之抗體可具有比海藻糖基化抗體更高之ADCC活性。因此,在一個實施例中,用於本方法中或用於本發明之組合物中之抗CCR4抗體可由具有較低或無α1,6-海藻糖基轉移酶(FUT8)活性之細胞產生。在另一實施例中,用於本發明之方法或組合物中之抗CCR4抗體基本上無核海藻糖,該核海藻糖藉由結合於Fc區中之Asn 297之N-糖苷鍵聯糖鏈中之α1,6鍵而結合於N-乙醯基葡糖胺。 在某些實施例中,該方法進一步包含在投與抗CCR4抗體之前識別CCR4表現。在其他實施例中,可在治療前、治療期間及/或治療後監測浸潤於患者之腫瘤組織及/或諸如腹部、胸膜之組織流出物中之CCR4+ T細胞或CCR4+ Treg細胞群、CCR4+ T細胞或CCR4+ Treg之減少/消耗程度。此外,CCR4表現亦可藉由此項技術中已知之任何方法在癌細胞自身上分析。 PD-1 抗體 在一個實施例中,特異性結合於人類PD-1且抑制hPD-1活性之抗體或其抗原結合部分(「抗PD-1抗體」)用於本發明之方法。在另一實施例中,抗PD-1抗體或其抗原結合部分阻斷、抑制、阻止或中和細胞上之PD-1表現或PD-1相關之腫瘤免疫反應功能。用於本發明之方法中之抗PD-1抗體結合於具有較高特異性及親和性之PD-1,阻斷PD-L1之結合,及/或抑制PD-1傳訊路徑之免疫抑制效果。在本文所揭示之治療方法或組合物使用中之任一者中,抗PD-1抗體包括結合於PD-1且展現類似於全抗體抑制配位體結合且上調免疫系統之功能特性的抗原結合片段。 在一些實施例中,本發明之抗PD-1抗體包含:包含SEQ ID NO:11中所闡述之序列之VH CDR1、包含SEQ ID NO:12中所闡述之序列之VH CDR2以及包含SEQ ID NO:13中所闡述之序列之VH CDR3中的一或多者,及包含SEQ ID NO:14中所闡述之序列之VL CDR1、包含SEQ ID NO:15中所闡述之序列之VL CDR2以及包含SEQ ID NO:16中所闡述之序列之VL CDR3中的一或多者。在某些實施例中,抗PD-1抗體包含:包含SEQ ID NO:11中所闡述之序列之VH CDR1、包含SEQ ID NO:12中所闡述之序列之VH CDR2、及包含SEQ ID NO:13中所闡述之序列VH CDR3、包含SEQ ID NO:14中所闡述之序列之VL CDR1、包含SEQ ID NO:15中所闡述之序列之VL CDR2及包含SEQ ID NO:16中所闡述之序列之VL CDR3。在其他實施例中,抗PD-1抗體包含:包含SEQ ID NO:17中所闡述之序列之VH及包含SEQ ID NO:18中所闡述之序列之VL。在另其他實施例中,抗PD-1抗體包含:包含SEQ ID NO:19中所闡述之序列之重鏈及包含SEQ ID NO:20中所闡述之序列之輕鏈。在又其他實施例中,抗PD-1抗體為具有降低效應活性之IgG2、IgG4或其變異體。 在某些實施例中,抗PD-1抗體或其抗原結合部分與納武單抗交叉競爭以結合於人類PD-1且抗PD-1抗體或其抗原結合片段如納武單抗結合於人類PD-1之相同抗原決定基區。 在其他實施例中,抗PD-1單株抗體或其抗原結合片段為嵌合、人類化或人類單株抗體或其片段。在治療人類個體之某些實施例中,該單株抗體為人類化抗體。在治療人類個體之其他實施例中,該抗體為人類抗體。在其他實施例中,IgG1、IgG2、IgG3或IgG4同型之抗體或其抗體Fc變異體可用於本方法及組合物。 在某些實施例中,抗PD-1抗體或其抗原結合部分包含屬於人類IgG1或IgG4同型或任何可應用IgG變異體之重鏈恆定區。在其他實施例中,抗PD-1抗體或其抗原結合部分之IgG4重鏈恆定區之序列含有S228P突變,該突變利用在IgG1同型抗體中之對應位置處正常發現的脯胺酸殘基替代鉸鏈區中之絲胺酸殘基。存在於納武單抗中之此突變阻止Fab臂與內源性IgG4抗體交換,同時保持用於活化與野生型IgG4抗體相關聯之Fc受體的較低親和性(Wang等人,2014 Cancer Immunol Res.2(9):846-56)。在又其他實施例中,抗體包含作為人類κ或λ恆定區之輕鏈恆定區。 以較高親和性特異性地結合於PD-1之人類化抗體或人類抗體(HuMab)已揭示於美國專利第8,008,449號或第8,779,105號中。其他抗PD-1單株抗體已描述於例如,美國專利第6,808,710號、第7,488,802號、第8,168,757號及第8,354,509號以及PCT公開案第WO 2012/145493號中。揭示於美國專利第8,008,449號中之抗PD-1 HuMAb中之每一者已經證明呈現以下特徵中的一或多者:(a)結合於具有1×10-7 M或小於1×10- 7 M之KD 的人類PD-1,如藉由使用Biacore生物感測器系統之表面電漿子共振所測定;(b)實質上並未結合於人類CD28、CTLA-4或ICOS;(c)增大混合淋巴球反應(MLR)分析中之T細胞增殖;(d)增大MLR分析中之干擾素γ產生;(e)增大MLR分析中之IL-2分泌;(f)結合於人類PD-1及食蟹獼猴PD-1;(g)抑制PD-L1及/或PD-L2結合於PD-1;(h)刺激抗原特異性記憶體反應;(i)刺激抗體反應;以及(j)抑制活體內腫瘤細胞生長。可用於本發明中之抗PD-1抗體包括特異性地結合於人類PD-1且呈現前述特徵中之至少一個(在一些實施例中,至少五個)的單株抗體。在一些實施例中,抗PD-1抗體為納武單抗。在一個實施例中,抗PD-1抗體為派立珠單抗。 在一個實施例中,抗PD-1抗體為納武單抗。納武單抗(亦稱為「OPDIVO(註冊商標)」;先前特指5C4、BMS-936558、MDX-1106或ONO-4538)為選擇性地防止與PD-1配位體(PD-L1及PD-L2)相互作用,由此阻斷下調抗腫瘤T細胞功能之全人類IgG4 (S228P) PD-1免疫檢查點抑制抗體(美國專利第8,008,449號;Wang等人,2014 Cancer Immunol Res.2(9):846-56)。 在一些實施例中,抗PD-1抗體與相同於派立珠單抗之抗原決定基交叉競爭或結合。在一特定實施例中,抗PD-1抗體為納武單抗。派立珠單抗(亦稱為「KEYTRUDA(註冊商標)」、拉立珠單抗及MK-3475)為針對人類細胞表面受體PD-1(程序式死亡-1或程序式細胞死亡-1)之人類化單株IgG4抗體。派立珠單抗描述於例如美國專利第8,354,509號及第8,900,587號中;亦參見http://www.cancer.gov/drugdictionary?cdrid=695789 (最後訪問:2014年12月14日)。派立珠單抗已由FDA審批通過用於治療復發性或難治性黑素瘤。 在其他實施例中,抗PD-1抗體與相同於為單株抗體之MEDI0608(先前為AMP-514)之抗原決定基交叉競爭或結合。在又其他實施例中,抗PD-1抗體為MEDI0608。MEDI0608描述於例如美國專利第8,609,089B2號或http://www.cancer.gov/drugdictionary?cdrid=756047中(最後訪問:2014年12月14日)。 可用於所揭示之方法或所揭示之組合物中之抗PD-1抗體亦包括特異性地結合於人類PD-1且與納武單抗交叉競爭以結合於人類PD-1之分離抗體(參看,例如美國專利第8,008,449號及第8,779,105號;WO 2013/173223)。抗體交叉競爭以結合於抗原之能力可指示此等抗體結合於抗原之相同抗原決定基區且空間上阻礙其他交叉競爭抗體與特定抗原決定基區之結合。藉助於結合於PD-1之相同抗原決定基區,預期此等交叉競爭抗體具有與納武單抗極類似的功能性特性。可在標準PD-1結合分析,諸如Biacore分析、ELISA測定或流式細胞量測術中基於交叉競爭抗體與納武單抗交叉競爭之能力而易於識別該交叉競爭抗體(參看,例如,WO 2013/173223)。 在某些實施例中,抗PD-1抗體為納武單抗(OPDIVO (註冊商標))。在其他實施例中,抗PD-1抗體為派立珠單抗(KEYTRUDA (註冊商標))。在其他實施例中,抗PD-1抗體選自描述於美國專利第8,008,449號中之人類抗體17D8、人類抗體2D3、人類抗體4H1、人類抗體4A11、人類抗體7D3及人類抗體5F4。在另其他實施例中,抗PD-1抗體為MEDI0608 (先前為AMP-514)或AMP-224。 在某些實施例中,特異性結合於PD-1且抑制PD-1活性(例如,結合於PD-L1及/或PD-L2)之用於本方法或組合物中之抗PD-1抗體適用於治療癌症,諸如肺癌(例如非小細胞肺癌)、食道癌或胃癌。 在一個實施例中,用於本發明之方法或組合物中之抗PD-1抗體包括: (i)抗體,與該抗體交叉競爭以結合於人類PD-1,該抗體包含:包含SEQ ID NO:11中所闡述之序列之重鏈CDR1、包含SEQ ID NO:12中所闡述之序列之重鏈CDR2、包含SEQ ID NO:13中所闡述之序列之重鏈CDR3中的一或多者,及包含SEQ ID NO:14中所闡述之序列之輕鏈CDR1、包含SEQ ID NO:15中所闡述之序列之輕鏈CDR2以及包含SEQ ID NO:16中所闡述之序列之輕鏈CDR3中的一或多者; (ii)抗體,結合於與該抗體中之抗原決定基區相同之抗原決定基區,該抗體包含:包含SEQ ID NO: 11中所闡述之序列之重鏈CDR1、包含SEQ ID NO:12中所闡述之序列之重鏈CDR2、包含SEQ ID NO:13中所闡述之序列之重鏈CDR3、包含SEQ ID NO:14中所闡述之序列之輕鏈CDR1、包含SEQ ID NO:15中所闡述之序列之輕鏈CDR2以及包含SEQ ID NO:16中所闡述之序列之輕鏈CDR3; (iii)抗體,其包含:包含SEQ ID NO:11中所闡述之序列之重鏈CDR1、包含SEQ ID NO:12中所闡述之序列之重鏈CDR2、包含SEQ ID NO:13中所闡述之序列之重鏈CDR3、包含SEQ ID NO:14中所闡述之序列之輕鏈CDR1、包含SEQ ID NO:15中所闡述之序列之輕鏈CDR2,以及包含SEQ ID NO:16中所闡述之序列之輕鏈CDR3; (iv)抗體,其包含:包含SEQ ID NO:17中所闡述之序列之VH及包含SEQ ID NO:18中所闡述之序列之VL;或 (v)抗體,其包含:包含SEQ ID NO:19中所闡述之序列之重鏈及包含SEQ ID NO:20中所闡述之序列之輕鏈。 用於本發明中之抗PD-1抗體可對PD-1發揮中和或阻斷活性;即該抗PD-1抗體可藉由PD-L1結合而阻斷PD-1活性,抑制T細胞增殖、細胞介素釋放及其任何組合。在一個實施例中,抗PD-1抗體ONO-4538、BMS-936558、納武單抗/OPDIVO(註冊商標)為特異性結合於PD-1且阻斷PD-1結合於PD-L1及PD-L2之例示性抗PD-1抗體。納武單抗可在活體外緩解PD-1介導之對人類T細胞活化的抑制且經由T細胞依賴性機制抑制異種移植模型中的腫瘤生長。( i ) CCR4 抗體及 ( ii ) PD - 1 抗體之組合療法 本發明涉及一種治療癌症之方法,該方法包含投與CCR4拮抗劑及PD-1拮抗劑,其中該CCR4拮抗劑包含結合於CCR4、干擾CCR4或以其他方式阻斷CCR4結合於其配位體(例如,CCL17及/或CCL22)之任何較小或較大分子(例如,抗CCR4抗體),且該PD-1拮抗劑包含結合於PD-1、干擾PD-1或以其他方式阻斷PD-1結合於其配位體(例如,PD-L1)之任何較小或較大分子(例如,抗PD-1抗體)。在一個實施例中,本發明包括一種治療有需要之個體之至少一種癌症的方法,該方法包含投與抗CCR4抗體以及抗PD-1抗體。在另一實施例中,本方法之投藥排程包括對癌症患者之投藥排程,包含投與抗CCR4抗體以及抗PD-1抗體。在某些實施例中,抗CCR4抗體及抗PD-1抗體係以有效量投與來治療癌症。 本文亦提供一種將患有癌症之個體內之腫瘤大小減小至少約1%、5%、10%、15%、20%或30%之方法,該方法包含將(i)抗CCR4抗體或其抗原結合部分及(ii)抗PD-1抗體或其抗原結合部分投與至個體。在其他實施例中,相較於抗CCR4抗體或抗PD-1抗體之單藥療法,本發明之組合方法增強抗腫瘤療效。在某些實施例中,以有效量投與抗CCR4抗體及抗PD-1抗體來減小癌症之腫瘤大小。 本發明之此等組合方法可藉由減少或耗乏抑制抗腫瘤免疫反應之CCR4+ Treg細胞而在癌症患者之抗腫瘤免疫療法上帶來一或多種有益效果;因此抗CCR4抗體抵消CCR4+ Treg細胞之免疫抑止效應且引起以及增強抗腫瘤免疫反應。同時,抗PD-1抗體阻斷或中和T細胞、PD-1對於攻擊腫瘤微環境中之腫瘤細胞的細胞毒性T細胞之負調節劑;因此抗PD-1抗體抵消針對T細胞之負信號且引起抗腫瘤免疫反應。 用於該方法中之抗CCR4抗體之投與劑量包括每一投與約0.1 mg/kg至約10.0 mg/kg之任何劑量。在一個態樣中,用於該方法中之抗CCR4抗體之劑量包括至少約0.1 mg/kg、至少約0.2 mg/kg、至少約0.3 mg/kg、至少約0.4 mg/kg、至少約0.5 mg/kg、至少約0.6 mg/kg、至少約0.7 mg/kg、至少約0.8 mg/kg、至少約0.9 mg/kg、至少約1.0 mg/kg、至少約2.0 mg/kg及至少約3.0 mg/kg。 用於該方法中之抗PD-1抗體之投與劑量包括每一投與約1.0 mg/kg至約10.0 mg/kg之任何劑量。在一個態樣中,用於該方法中之抗PD-1抗體之劑量包括至少約1 mg/kg、至少約2 mg/kg、至少約3 mg/kg、至少約4 mg/kg、至少約5 mg/kg、至少約6 mg/kg、至少約7 mg/kg、至少約8 mg/kg、至少約9 mg/kg或至少約10 mg/kg。在其他實施例中,抗PD-1抗體之劑量為約2.0 mg/kg。在一特定實施例中,抗PD-1抗體之劑量為約3.0 mg/kg。在一特定實施例中,抗PD-1抗體之劑量為約240 毫克/身體作為平調劑量。在一些實施例中,抗PD-1抗體之劑量為至少約至少約100 毫克/身體、120 毫克/身體、140 毫克/身體、160 毫克/身體、180 毫克/身體、200 毫克/身體、220 毫克/身體、240 毫克/身體、260 毫克/身體、280 毫克/身體、300 毫克/身體、400 毫克/身體、420 毫克/身體、440 毫克/身體、460 毫克/身體、480 毫克/身體、500 毫克/身體、520 毫克/身體、540 毫克/身體、560 毫克/身體或600 毫克/身體。在某些實施例中,抗PD-1抗體之劑量為480 毫克/身體之平調劑量。 用於該方法中之抗體之投藥週期包括對於各抗體之任何適宜投藥週期。投藥週期定義為約一週一次(Q1W)、約每兩週一次(Q2W)、約每三週一次(Q3W)或約每四週一次或約每月一次(Q4W)。用於該方法中之抗CCR4抗體之投與週期包括一週一次(Q1W)持續四週至八週、一週一次(Q1W)持續四週及每兩週一次(Q2W)。在一個態樣中,抗CCR4抗體在第一次給藥之後一週一次持續四週(誘導階段)接著每兩週一次(維持階段)投與。用於該方法中之抗PD-1抗體之投與週期包括每兩週一次、每三週一次或每四週一次。 在另一實施例中,本發明之方法包括一種治療個體中之至少一種癌症的方法,該方法包含在第一次給藥之後約一週一次持續四週且接著約每兩週一次投與約0.1 mg/kg、約0.3 mg/kg、約0.5 mg/kg、約0.75 mg/kg、約1.0 mg/kg、約2.0 mg/kg、或約3.0 mg/kg之抗CCR4抗體,且與約每兩週一次、每三週一次或每四週一次投與約2.0 mg/kg、或約3.0 mg/kg、或至少約100 毫克/身體、120 毫克/身體、140 毫克/身體、160 毫克/身體、180 毫克/身體、200 毫克/身體、220 毫克/身體、240 毫克/身體、260 毫克/身體、280 毫克/身體、300 毫克/身體、400 毫克/身體、420 毫克/身體、440 毫克/身體、460 毫克/身體、480 毫克/身體、500 毫克/身體、520 毫克/身體、540 毫克/身體、560 毫克/身體、580 毫克/身體或600 毫克/身體作為平調劑量之抗PD-1抗體組合。 在其他實施例中,本發明之方法包括治療個體中之至少一種癌症,該方法包含投與;(i)在第一次給藥之後約一週一次持續四週且接著約每兩週一次之約0.1 mg/kg、約0.3 mg/kg、約0.5 mg/kg、約0.75 mg/kg、約1.0 mg/kg、約2.0 mg/kg或約3.0 mg/kg之抗CCR4抗體,且與約每兩週一次投與約2.0 mg/kg之抗PD-1抗體組合;(ii)在第一次給藥之後約一週一次持續四週且接著約每兩週一次之約0.1 mg/kg、約0.3 mg/kg、約0.75 mg/kg、約1.0 mg/kg或約3.0 mg/kg之抗CCR4抗體,且與約每兩週一次投與約3.0 mg/kg之抗PD-1抗體組合;(iii)在第一次給藥之後約一週一次持續四週且接著約每兩週一次之約0.1 mg/kg、約0.3 mg/kg、約0.5 mg/kg、約0.75 mg/kg、約1.0 mg/kg、約2.0 mg/kg或約3.0 mg/kg之抗CCR4抗體,且與約每兩週一次投與至少約100毫克/身體、120毫克/身體、140毫克/身體、160毫克/身體、180毫克/身體、200毫克/身體、220毫克/身體、240毫克/身體、260毫克/身體、280 毫克/身體、300 毫克/身體、400 毫克/身體、420 毫克/身體、440 毫克/身體、460 毫克/身體、480 毫克/身體、500 毫克/身體、520 毫克/身體、540 毫克/身體、560 毫克/身體、580 毫克/身體或600 毫克/身體作為平調劑量之抗PD-1抗體組合;(iv)在第一次給藥之後約一週一次持續四週且接著約每兩週一次之約0.1 mg/kg、約0.3 mg/kg、約0.75 mg/kg、約1.0 mg/kg、約2.0 mg/kg或約3.0 mg/kg之抗CCR4抗體,且與約每四週一次投與約2.0 mg/kg之抗PD-1抗體組合;(v)在第一次給藥之後約一週一次持續四週且接著約每兩週一次之約0.1 mg/kg、約0.3 mg/kg、約0.5 mg/kg、約0.75 mg/kg、約1.0 mg/kg或約3.0 mg/kg之抗CCR4抗體,且與約每四週一次投與約3.0 mg/kg之抗PD-1抗體組合或(vi)在第一次給藥之後約一週一次持續四週且接著約每四週一次之約0.1 mg/kg、約0.3 mg/kg、約0.75 mg/kg、約1.0 mg/kg、約2.0 mg/kg或約3.0 mg/kg之抗CCR4抗體,且與約每兩週一次投與至少約100 毫克/身體、120 毫克/身體、140 毫克/身體、160 毫克/身體、180 毫克/身體、200 毫克/身體、220 毫克/身體、240 毫克/身體、260 毫克/身體、280 毫克/身體、300 毫克/身體、400 毫克/身體、420 毫克/身體、440 毫克/身體、460 毫克/身體、480 毫克/身體、500 毫克/身體、520 毫克/身體、540 毫克/身體、560 毫克/身體、580 毫克/身體或600 毫克/身體作為平調劑量之抗PD-1抗體組合。 在一個實施例中,本發明之方法包括一種治療個體中之至少一種癌症的方法,該方法包含在第一次給藥之後約一週一次持續四週且接著約每兩週一次投與約0.1 mg/kg、約0.3 mg/kg、約0.5 mg/kg、約0.75 mg/kg、約1.0 mg/kg、約2.0 mg/kg或約3.0 mg/kg之抗CCR4抗體,且與約每兩週一次投與約2.0 mg/kg或約3.0 mg/kg、或約240 毫克/身體作為平調劑量之抗PD-1抗體,或約每四週一次約480 毫克/身體作為平調劑量之抗PD-1抗體組合,其中該抗CCR4抗體與包含以下之抗體競爭性地結合於CCR4:包含SEQ ID NO:1中所闡述之序列之VH CDR1、包含SEQ ID NO:2中所闡述之序列之VH CDR2、包含SEQ ID NO:3中所闡述之序列之VH CDR3、包含SEQ ID NO:4中所闡述之序列之VL CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2,及包含SEQ ID NO:6中所闡述之序列之VL CDR3且其中抗PD-1抗體與包含以下之抗體競爭性地結合於PD-1:包含SEQ ID NO:11中所闡述之序列之VH CDR1、包含SEQ ID NO:12中所闡述之序列之VH CDR2、包含SEQ ID NO:13中所闡述之序列之VH CDR3、包含SEQ ID NO:14中所闡述之序列之VL CDR1、包含SEQ ID NO:15中所闡述之序列之VL CDR2及包含SEQ ID NO:16中所闡述之序列之VL CDR3。 在一個實施例中,本發明之方法包括一種治療癌症患者的方法,該方法包含約一週一次持續四週且接著約每兩週一次投與約0.1 mg/kg、約0.3 mg/kg、約0.5 mg/kg、約0.75 mg/kg、約1.0 mg/kg、約2.0 mg/kg或約3.0 mg/kg之抗CCR4抗體,且與約每兩週一次投與約2.0 mg/kg或約3.0 mg/kg、或約240 毫克/身體作為平調劑量之抗PD-1抗體,或約每四週一次約480 毫克/身體作為平調劑量之抗D-1抗體組合,其中該抗CCR4抗體結合於與抗體相同之抗原決定基,該抗體包含:包含SEQ ID NO:1中所闡述之序列之VH CDR1、包含SEQ ID NO:2中所闡述之序列之VH CDR2、包含SEQ ID NO:3中所闡述之序列之VH CDR3、包含SEQ ID NO:4中所闡述之序列之VL CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2及包含SEQ ID NO:6中所闡述之序列之VL CDR3,且其中該抗PD-1抗體結合於與抗體相同之抗原決定基,該抗體包含:包含SEQ ID NO:11中所闡述之序列之VH CDR1、包含SEQ ID NO:12中所闡述之序列之VH CDR2、包含SEQ ID NO:13中所闡述之序列之VH CDR3、包含SEQ ID NO:14中所闡述之序列之VL CDR1、包含SEQ ID NO:15中所闡述之序列之VL CDR2及包含SEQ ID NO:16中所闡述之序列之VL CDR3。 在一個實施例中,本發明之方法包括一種治療癌症患者的方法,該方法包含約一週一次持續四週且接著約每兩週一次投與約0.1 mg/kg、約0.3 mg/kg、約0.5 mg/kg、約0.75 mg/kg、約1.0 mg/kg、約2.0 mg/kg或約3.0 mg/kg之抗CCR4抗體,且與約每兩週一次投與約2.0 mg/kg、或約3.0 mg/kg、或以約240 毫克/身體為平調劑量之抗PD-1抗體,或約每四週一次之以約480 毫克/身體為平調劑量的抗PD-1抗體組合,其中該抗CCR4抗體包含:包含SEQ ID NO:1中所闡述之序列之VH CDR1、包含SEQ ID NO:2中所闡述之序列之VH CDR2、包含SEQ ID NO:3中所闡述之序列之VH CDR3、包含SEQ ID NO:4中所闡述之序列之VL CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2及包含SEQ ID NO:6中所闡述之序列之VL CDR3,且其中該抗PD-1抗體包含:包含SEQ ID NO:11中所闡述之序列之VH CDR1、包含SEQ ID NO:12中所闡述之序列之VH CDR2、包含SEQ ID NO:13中所闡述之序列之VH CDR3、包含SEQ ID NO:14中所闡述之序列之VL CDR1、包含SEQ ID NO:15中所闡述之序列之VL CDR2及包含SEQ ID NO:16中所闡述之序列之VL CDR3。 在另一實施例中,本發明之方法包括一種治療癌症患者的方法,該方法包含約一週一次持續四週且接著約每兩週一次投與約0.1 mg/kg、約0.3 mg/kg、約0.5 mg/kg、約0.75 mg/kg、約1.0 mg/kg、約2.0 mg/kg或約3.0 mg/kg之抗CCR4抗體(莫格利珠單抗),且與約每兩週一次投與約2.0 mg/kg、或約3.0 mg/kg、或以約240 毫克/身體為平調劑量之抗PD-1抗體(納武單抗)或約每四週一次之以約480 毫克/身體為平調劑量之抗PD-1抗體(納武單抗)組合。 在本文中所描述之方法或組合物中,可將抗體組合並行地(或同時)或依序投與至癌症患者中。術語「並行地」或「同時」意謂至少兩種抗體溶解於醫藥學上可接受之水性液體,諸如生理鹽水或僅林格氏溶液或其混合物中,且同時藉由靜脈內線及/或輸液袋投與至患者中。術語「依序」意謂將一個抗體投與至癌症患者中,接著在投與第一抗體後容許的稍後時間將第二抗體投與至該等癌症患者中。 在某些實施例中,本揭示案包括一種治療患有HCC之患者的方法,其中無進展存活期(PFS)為至少100天、至少101天、至少102天、至少103天、至少104天、至少105天、至少106天、至少107天、至少108天、至少109天、至少110天、至少111天、至少112天、至少113天、至少114天、至少115天、至少116天、至少117天、至少118天、至少119天、至少120天、至少125天、至少130天、至少135天、至少140天、至少145天、至少150天、至少155天、至少160天、至少165天、至少170天、至少175天、至少180天、至少185天、至少190天、至少195天、至少200天、至少205天、至少210天、至少215天或至少220天。在其他實施例中,抗CCR4抗體及抗PD-1抗體之本組合療法的PFS高於抗CCR4抗體單藥療法及/或抗PD-1抗體單藥療法之PFS (高於至少一週、至少兩週、至少三週、至少四週、至少一個月、至少兩個月、至少三個月、至少四個月、至少五個月、至少六個月、至少七個月、至少八個月、至少九個月,或至少十個月)。 在其他實施例中,本揭示案提供一種治療患有HCC之患者的方法,其中總存活期為至少約200天、至少約210天、至少約220天、至少約230天、至少約240天、至少約250天、至少約260天、至少約270天、至少約280天、至少約290天、至少約300天、至少約310天、至少約320天、至少約330天、至少約340天、至少約350天、至少約360天、至少約370天、至少約380天、至少約390天、或至少約400天。在又其他實施例中,本方法之OS高於抗CCR4抗體單藥療法及/或抗PD-1抗體單藥療法之OS (高於至少約一個月、至少約兩個月、至少約三個月、至少約四個月、至少約五個月、至少約六個月、至少約七個月、至少約八個月、至少約九個月、至少約十個月、至少約11個月、至少約12個月、至少約15個月、至少約20個月、至少約兩年、至少約25個月、至少約30個月、至少約35個月、至少約三年、至少約40個月、至少約3.5年、至少約4年、至少約4.5年,或至少約5年)。用於治療癌症之包含抗 CCR4 抗體及抗 PD - 1 抗體之套組 本發明亦包含一種用於治療罹患癌症之個體之套組,該套組包含: (a)劑量介於約0.1 mg/kg至約10 mg/kg體重範圍內之特異性結合於PD-1及/或抑制PD-1活性之抗體或其抗原結合部分(「抗PD-1抗體或其抗原結合部分」); (b)劑量介於約0.1 mg/kg至約10 mg/kg體重範圍內之特異性結合於CCR4及/或抑制CCR4活性之抗體或其抗原結合部分(「抗CCR4抗體或其抗原結合部分」);以及 (c)在本文所揭示之任何方法中使用抗PD-1抗體及抗CCR4抗體之說明書。 套組通常包括指示套組之內含物之預期用途的標籤及使用說明書。術語標籤包括在套組上或與套組一起供應或以其他方式伴隨套組之任何書面或記錄材料。 在其他實施例中,抗CCR4抗體或其抗原結合部分選自由以下組成之群: (i)結合於與抗體相同之抗原決定基的抗體或其抗原結合部分,其包含:包含SEQ ID NO:1中所闡述之序列之VH CDR1、包含SEQ ID NO:2中所闡述之序列之VH CDR2及包含SEQ ID NO:3中所闡述之序列之VH CDR3中的一或多者,及包含SEQ ID NO:4中所闡述之序列之VL CDR 1、包含SEQ ID NO:5中所闡述之序列之VL CDR2及包含SEQ ID NO:6中所闡述之序列之VL CDR3中的一或多者; (ii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:1中所闡述之序列之VH CDR1、包含SEQ ID NO:2中所闡述之序列之VH CDR2、包含SEQ ID NO:3中所闡述之序列之VH CDR3、包含SEQ ID NO:4中所闡述之序列之VL CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2,以及包含SEQ ID NO:6中所闡述之序列之VL CDR3; (iii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:7中所闡述之序列之VH及包含SEQ ID NO:8中所闡述之序列之VL; (iv)抗體,其包含:包含SEQ ID NO:9中所闡述之序列之重鏈及包含SEQ ID NO:10中所闡述之序列之輕鏈; (v)抗體或其抗原結合部分,與(ii)之該抗體交叉競爭;以及 (vi)莫格利珠單抗或其抗原結合部分。 在某些實施例中,抗PD-1抗體或其抗原結合部分選自由以下組成之群: (i)結合於與抗體相同之抗原決定基的抗體或其抗原結合部分,其包含:包含SEQ ID NO:11中所闡述之序列之VH CDR1、包含SEQ ID NO:12中所闡述之序列之VH CDR2以及包含SEQ ID NO:13中所闡述之序列之VH CDR3中的一或多者,及包含SEQ ID NO:14中所闡述之序列之VL CDR1、包含SEQ ID NO:15中所闡述之序列之VL CDR2以及包含SEQ ID NO: 16中所闡述之序列之VL CDR3中的一或多者; (ii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:11中所闡述之序列之VH CDR1、包含SEQ ID NO:12中所闡述之序列之VH CDR2、包含SEQ ID NO:13中所闡述之序列之VH CDR3、包含SEQ ID NO:14中所闡述之序列之VL CDR1、包含SEQ ID NO:15中所闡述之序列之VL CDR2,以及包含SEQ ID NO:16中所闡述之序列之VL CDR3; (iii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:17中所闡述之序列之VH及包含SEQ ID NO:18中所闡述之序列之VL; (iv)抗體,其包含:包含SEQ ID NO:19中所闡述之序列之重鏈及包含SEQ ID NO:20中所闡述之序列之輕鏈; (v)抗體,與(ii)之該抗體交叉競爭; (vi)納武單抗或其抗原結合部分; (vii)派立珠單抗或其抗原結合部分;以及 (viii) MEDI0608或其抗原結合部分。用於治療癌症之包含抗 CCR4 抗體及抗 PD - 1 抗體之組合物 在一個實施例中,本發明之組合物包含用於治療癌症之抗CCR4抗體及抗PD-1抗體。 在一個實施例中,用於本發明之組合物經製備待以約每週一次持續四週且接著約每兩週一次投與約0.1 mg/kg、約0.3 mg/kg、約0.5 mg/kg、約0.75 mg/kg、約1.0 mg/kg、約2.0 mg/kg或約3.0 mg/kg劑量之抗CCR4抗體,與約每兩週一次以約2.0 mg/kg、或約3.0 mg/kg或約240 毫克/身體作為平調劑量之抗PD-1抗體或約每四週一次以約480 毫克/身體作為平調劑量之抗PD-1抗體組合。在某些實施例中,組合物經製備待以約每兩週一次之約2.0 mg/kg、或約3.0 mg/kg、或約240 毫克/身體作為平調劑量之抗PD-1抗體,或約每四週一次之約480 毫克/身體作為平調劑量之抗PD-1抗體與約每週一次持續四週且接著約每兩週一次之約0.1 mg/kg、約0.3 mg/kg、約0.5 mg/kg、約0.75 mg/kg、約1.0 mg/kg、約2.0 mg/kg或約3.0 mg/kg之劑量之抗CCR4抗體組合投與。在一些實施例中,組合物經製備待以約每週一次持續四週且接著約每兩週一次之約0.1 mg/kg、約0.3 mg/kg、約0.5 mg/kg、約0.75 mg/kg、約1.0 mg/kg、約2.0 mg/kg或約3.0 mg/kg劑量之抗CCR4抗體與約每兩週一次之約2.0 mg/kg或約3.0 mg/kg或約240 毫克/身體作為平調劑量之抗PD-1抗體或約每四週一次之約480 毫克/身體作為平調劑量之抗PD-1抗體組合投與。在其他實施例中,組合物經製備待以約每兩週一次之約2.0 mg/kg、或約3.0 mg/kg或約240 毫克/身體作為平調劑量之抗PD-1抗體與約一週一次持續四週且接著約每兩週一次之約0.1 mg/kg、約0.3 mg/kg、約1.0 mg/kg、約2.0 mg/kg或約3.0 mg/kg劑量之抗CCR4抗體組合投與。 在一個實施例中,用於本發明之組合物包含抗CCR4抗體(莫格利珠單抗),以約一週一次持續四週且接著約每兩週一次以約0.1 mg/kg、約0.3 mg/kg、約0.5 mg/kg、約0.75 mg/kg、約1.0 mg/kg、約2.0 mg/kg或約3.0 mg/kg之劑量與約每兩週一次以約2.0 mg/kg或約3.0 mg/kg之劑量或約240 毫克/身體為平調劑量或約每四週一次以約480 毫克/身體為平調劑量之抗PD-1抗體(納武單抗)組合治療個體中之癌症。在另一實施例中,該組合物包含抗PD-1抗體(納武單抗),以約每兩週一次以約2.0 mg/kg或約3.0 mg/kg劑量或以約240 毫克/身體為平調劑量或約每四週一次以約480 毫克/身體為平調劑量與以約一週一次持續四週且接著約每兩週一次約0.1 mg/kg、約0.3 mg/kg、約0.5 mg/kg、約0.75 mg/kg、約1.0 mg/kg或約3.0 mg/kg劑量之抗CCR4抗體(莫格利珠單抗)組合。 在某些實施例中,用於本發明之組合物經製備為平調劑量組合物;例如,抗CCR4抗體之平調劑量可為至少約5 mg、至少約10 mg、至少約15 mg、至少約20 mg、至少約30 mg、至少約40 mg、至少約50 mg、至少約60 mg、至少約70 mg、至少約80 mg、至少約90 mg、至少約100 mg、至少約110 mg、至少約120 mg、至少約130 mg、至少約140 mg、至少約150 mg、至少約160 mg、至少約170 mg、至少約180 mg、至少約190 mg、至少約200 mg、至少約210 mg、至少約220 mg、至少約230 mg、至少約240 mg、至少約250 mg、至少約260 mg、至少約270 mg、至少約280 mg、至少約290 mg、或至少約300 mg。在一些實施例中,抗CCR4抗體之劑量介於約1 mg至約200 mg、約2 mg至約180 mg、約3 mg至約170 mg、或約6 mg至約160 mg之範圍內。在另一實施例中,組合物包含劑量介於約6 mg至約60 mg、約10 mg至約80 mg或約20 mg至約70 mg範圍內的抗CCR4抗體。在其他實施例中,抗CCR4抗體之劑量為約20 mg、約30 mg、約40 mg、約50 mg、約60 mg、約70 mg、約80 mg、約90 mg、約100 mg、約110 mg、約120 mg、約130 mg、約140 mg、約150 mg、約160 mg、約170 mg或約180 mg。 在其他實施例中,用於本發明之組合物以平調劑量投與,其中抗PD-1抗體之劑量為至少約1 mg、至少約5 mg、至少約10 mg、至少約15 mg、至少約20 mg、至少約30 mg、至少約40 mg、至少約50 mg、至少約60 mg、至少約70 mg、至少約80 mg、至少約90 mg、至少約100 mg、至少約110 mg、至少約120 mg、至少約130 mg、至少約140 mg、至少約150 mg、至少約160 mg、至少約170 mg、至少約180 mg、至少約190 mg、至少約200 mg、至少約210 mg、至少約220 mg、至少約230 mg、至少約240 mg、至少約250 mg、至少約260 mg、至少約270 mg、至少約280 mg、至少約290 mg、至少約300 mg、至少約310 mg、至少約320 mg、至少約330 mg、至少約340 mg、至少約350 mg、至少約360 mg、至少約370 mg、至少約380 mg、至少約390 mg、至少約400 mg、至少約410 mg、至少約420 mg、至少約430 mg、至少約440 mg、至少約450 mg、至少約460 mg、至少約470 mg、至少約480 mg、至少約490 mg、至少約500 mg、至少約510 mg、至少約520 mg、至少約530 mg、至少約540 mg、至少約550 mg、至少約560 mg、至少約570 mg、至少約580 mg、至少約590 mg或至少約600 mg。在一些實施例中,抗PD-1抗體之劑量介於20 mg至300 mg、30 mg至280 mg、40 mg至270 mg、50 mg至270 mg、60 mg至260 mg、70 mg至250 mg或80 mg至240 mg之範圍內。在又其他實施例中,抗PD-1抗體之劑量為約50 mg、約60 mg、約70 mg、約80 mg、約90 mg、約100 mg、約110 mg、約120 mg、約130 mg、約140 mg、約150 mg、約160 mg、約170 mg、約180 mg、約190 mg、約200 mg、約210 mg、約220 mg、約230 mg、約240 mg、約250 mg、約260 mg、約270 mg、約280 mg、約290 mg、約300 mg、約310 mg、約320 mg、約330 mg、約340 mg、約350 mg、400 mg、約410 mg、約420 mg、約430 mg、約440 mg、約450 mg、約460 mg、約470 mg、約480 mg、約490 mg、約500 mg、約510 mg、約520 mg、約530 mg、約540 mg、約550 mg、約560 mg、約570 mg、約580 mg、約590 mg、或約600 mg。 本發明之組合物包括以供使用之任何組合物,包含具有作為活性成份之上述活性的抗CCR4抗體及抗PD-1抗體之組合,且在一個實施例中,該組合以藥物調配物之形式提供,該藥物調配物通常地藉由根據醫藥學領域中任何熟知方法來將各抗體與一或多種醫藥學上可接受之載劑混合來製備。此外,用於本發明中之抗體亦以藥物調配物之形式提供,該藥物調配物通常地藉由根據醫藥學領域中之任何熟知方法而將其與一或多種醫藥學上可接受之載劑混合來製備。 在一些實施例中,本發明之抗體係以無菌溶液形式提供,其中該抗體溶解於諸如水之水性載劑,或使用之鹽、甘胺酸、葡萄糖或人類白蛋白之水溶液中。亦可添加醫藥學上可接受之添加劑,諸如緩衝液或張力劑用於製備更加類似於生理條件及其實例之溶液,包括醋酸鈉、氯化鈉、乳酸鈉、氯化鉀、檸檬酸鈉或其類似物。其亦可藉由冷凍乾燥保藏且在實際使用中,可藉由溶解於適當溶劑中來使用。 關於用於本發明或本發明之組合物中之抗體之投藥路徑,在某些實施例中,使用對於治療最有效之路徑。其實例包括經口投與及非經腸投與,諸如口內、氣管支氣管、直腸內、皮下、肌肉內、鞘內及靜脈內投與。可使用鞘內或靜脈內投與。 膠囊、錠劑、散劑、顆粒或其類似物可使用賦形劑,諸如乳糖、葡糖、蔗糖、甘露醇、或其類似物;崩解劑,諸如澱粉、海藻酸鈉或其類似物;潤滑劑,諸如硬脂酸鎂、滑石或其類似物;黏合劑,諸如聚乙烯醇、羥丙基纖維素、明膠或其類似物;表面活性劑,諸如脂肪酸酯或其類似物;塑化劑,諸如丙三醇或其類似物作為添加劑來製備。 適用於非經腸投與之製劑之實例可包括可注射調配物、栓劑、空氣噴霧或其類似物。舉例而言,可注射調配物係使用包括鹽溶液、葡萄糖溶液或其混合物之載劑來製備的。栓劑係使用載劑,諸如可可脂、氫化脂、羧酸或其類似物來製備的。空氣噴霧係使用例如並不刺激抗體自身及待投與人之口及呼吸道黏膜,且將抗體分散為細粒而使得易於吸收之載劑來製備的。 載劑之特定實例包括乳糖、丙三醇或其類似物。取決於所使用之抗體及載劑之特性,可製備霧劑、乾粉或其類似物。此外,即使在非經腸製備中,可添加在口服製備中例示為添加劑之組分。 在一個實施例中,本發明中之癌症患者包括能夠藉由本發明之組合物或方法有效治療之任何癌症患者。舉例而言,在本發明之一個實施例中,癌症患者為患有癌症之至少一個癌症患者,該癌症選自肺癌、胃腸癌、胃癌(或胃癌)、結腸癌、結腸直腸癌、食道癌、胰腺癌、肝癌、腎癌、卵巢癌、乳癌、頭頸癌、皮膚癌、黑素瘤及包括白血病、淋巴瘤之造血癌症。在另一實施例中,本發明中之癌症包括任何階段之癌症、原發性/轉移性癌症、局部晚期/癌轉移癌症、復發/頑固性癌症、及以手術方式可切除/不可切除性癌症。 在一個實施例中,癌症患者患有選自由以下組成之群中之至少一種癌症:包含小細胞肺癌(SCLC)、非小細胞肺癌(NSCLC)之肺癌,鱗狀細胞癌,腺癌,大細胞癌,腺鱗癌瘤,具有多形性肉瘤樣或肉瘤元素之癌瘤,類癌瘤(cartinoid tumor)及經分類之唾液腺型癌瘤(carcinomas of salivary-gland type),胃癌,肝癌,肝細胞細胞癌瘤,膽管癌瘤,乳癌,腎癌,黑素瘤及其任何組合。在另一實施例中,癌症患者患有選自由以下組成之群中之胰腺癌:胰管癌、胰腺頭癌、胰腺體癌、胰腺尾癌及其任何組合。在另一實施例中,患者患有選自由以下組成之群中之頭頸癌:口腔癌、咽癌、喉癌、頸癌、唾液腺癌及其任何組合。 在本發明中,癌症患者可在投與該組合療法之前經診斷。在一個實施例中,診斷方法可藉由本背景技術中之已知方法,諸如X射線圖形、電腦斷層掃描(CT)、正子發射斷層攝影術CT(PET-CT)、磁核共振成像(MRI)及/或對患者之血漿、血清、外周血液、腫瘤組織中之一個或數個生物標記進行分析來執行。此外,癌症進展、疾病階段及/或對於治療之反應可藉由已知方法及參數與上述成像及生物標記組合來分析。 舉例而言,關於葡萄糖消耗之腫瘤代謝通常在FDG-PET上基於上調之放射性同位素標記葡糖耗費,諸如18 F氟去氧葡萄糖(FDG)來分析為癌症組織。FDG高度積聚於癌症組織中,此係由於癌細胞高度代謝葡萄糖且需要比正常組織更大量之葡萄糖。 此外,分析外周血液淋巴球(PBL)、胸膜流出物中之淋巴球(LPE)及/或腫瘤浸潤淋巴球(TIL)中的細胞群可用於評估各患者之各組織及/或整個身體中之免疫反應狀態。舉例而言,若干標記物,諸如CD4、CD8、CD25、CD45RA、CD45RO、CCR4、CD127、PD-1、PD-L1 (B7-H1、CD274)、PD-L2、CTLA-4 (CD152)、IFN-γ、IL-2、IL-4、IL-12及Foxp3可用於預測各患者之免疫反應狀態。偵測到表現CD4+ CD25+ 、CD8+ CD25+ 、PD-1+ 及/或CTLA-4+ 之活化T細胞、表現CD4+ CD25+ CCR4+ 、CD25+ Foxp3+ 、CCR4+ Foxp3high 、CD4+ CCR4+ Foxp3+ 或CD4+ CD25+ CD45RA- CCR4+ Foxp3+ 之Treg細胞,且當患者體內之活化T細胞增加及/或Treg細胞減少時,視為患者體內之免疫反應增強。 在一個分析中,可在投與該組合療法之前及/或之後分析藉由CD4、CD8、CD25、CD45RA、CD45RO、CCR4、CD127、PD-1、PD-L1 (B7-H1、CD274)、PD-L2、CTLA-4 (CD152)、IFN-γ、IL-2、IL-4、IL-12及/或Foxp3,或外周/微環境細胞介素之表現而識別之T細胞、效應Treg、耗竭T細胞、活化T細胞、效應T細胞、無變應性T細胞或骨髓衍生之抑制細胞(MSDC)。 在一些實施例中,抗CCR4抗體及抗PD-1抗體之組合療法可使得患者之外周血液、胸膜流出物及/或腫瘤組織中之Treg細胞減少及/或活化T細胞增加。因此,抗體之組合可增強對於患者體內之癌細胞的免疫反應。此外,在一個實施例中,該方法進一步包含在投與之前分析或量測腫瘤細胞上之PD-L1、PD-L2或CCR4表現水準以判定患者適用於該組合療法。在另一實施例中,患者體內之PD-L1或PD-L2表現可被視為患者適用於本發明之組合療法的標誌。 此外,患者之血漿樣本中之吲哚胺2,3-二氧酶(IDO)活性可在本發明中進行分析。此項技術中已知表現於各種器官,諸如肺、小腸、胎盤中之IDO係關於誘導Treg分化,由此將造成免疫耐受。此外,分子高度表現於多個癌細胞上,抑制免疫反應且誘導癌細胞之免疫耐受。因此,IDO活性之分析可偵測免疫反應狀態。IDO活性通常係藉由分析色胺酸、犬尿胺酸以及犬尿胺酸代謝物之含量變化來偵測。當患者體內之IDO活性增強時,色胺酸、犬尿胺酸以及犬尿胺酸代謝物亦增加,由此導致患者體內免疫反應降低。 在其他實施例中,抗CCR4抗體及抗PD-1抗體之組合療法增加患者體內之Treg細胞之數量及/或降低患者體內之IDO活性。 治療之臨床醫藥學評估將通常藉由固態腫瘤反應評估準則(RECIST)版本1.1準則以及藉由免疫相關反應準則(irRC)評定(Wolchok等人,Clin.Cancer Res.;2009,15:7412-7420)。實例 1 具有莫格利珠單抗及納武單抗之組合療法在患有晚期固態腫瘤之個體中之階段 1 研究 1.研究概述 該研究為在患有組織學或細胞學上確診之局部晚期或轉移性固態腫瘤的成人個體中之抗CCR4抗體莫格利珠單抗組合抗PD-1抗體納武單抗之兩部分、多中心、階段1、開放標記、劑量遞增的組擴增研究。 研究群體:組織學或細胞學上確診之局部晚期或轉移性固態腫瘤。 劑量遞增階段:將登記患有組織學或細胞學上確診之局部晚期或轉移性固態腫瘤的個體。 組擴增階段:登記患有晚期或轉移性固態腫瘤(包括(但不限於)非小細胞肺癌(NSCLC)、小細胞肺癌(SCLC)、胃癌(GC)、肝細胞細胞癌、胰腺癌及食道癌)之個體。 2.研究目標: 主要目標: 主要目標為表徵莫格利珠單抗及納武單抗之組合在患有局部晚期或轉移性固態腫瘤之個體中之安全性及耐受性且判定該組合之最大耐受劑量(MTD)或建議固定劑量。 次要目標: 第一次要目標為基於固態腫瘤反應評估準則(RECIST版本1.1)評估患有局部晚期或轉移性固態腫瘤之個體體內之莫格利珠單抗及納武單抗之組合之抗腫瘤活性。將針對抗腫瘤活性評定最佳整體反應率(BOR)、達至反應之時間(TTR)、反應持續時間(DOR)、無進展存活期(PFS)、總存活期(OS)。 第二次要目標表徵在莫格利珠單抗及納武單抗之組合療程中之莫格利珠單抗及納武單抗之藥代動力學(PK)特性。另一次要目標為評估莫格利珠單抗及納武單抗之免疫原性。 3.入選準則: 包括根據監管機構指導原則已自願簽署機構審查委員會(Institutional Review Board)審批通過之知情同意書且註明日期的個體。書面知情同意書必須在執行任何研究相關步驟之前獲得。入選準則包括以下: -在知情同意時,20歲或大於20歲之個體。 -根據RECIST版本1.1指南具有可評估病變之個體。 -生命預期>12週之個體。 -ECOG機能狀態為(performance status) 0至1之個體。劑量遞增階段 :個體可能已經組織學或細胞學上確診為局部晚期或轉移性固態腫瘤(不包括原發性CNS或血液科惡性疾病)。組擴增 階段 :個體可能已經組織學或細胞學上確診為局部晚期或轉移性固態腫瘤,包括(但不限於)NSCLC、SCLC、GC、肝癌、肝細胞細胞癌、胰腺癌及食道癌(不包括原發性CNS或血液科惡性疾病)。 4.排除準則 排除準則包括懷孕或哺乳期女性或預期懷孕之所有個體。 排除準則進一步包括患有不可控且明顯當前(inter-current)疾病之個體。 排除準則進一步包括患有已知CNS轉移瘤及/或軟腦膜轉移瘤之個體。已經治療之患有無症狀之腦轉移瘤的個體視為穩定的,且可包括在第1週期第1天之前至少28天未接收皮質類固醇或抗驚厥劑的個體。 排除預先已經抗PD-1、抗PD-L1、抗PD-L2、抗CD137或抗細胞毒性T淋巴球相關聯之抗原-4(CTLA-4)抗體或特異性靶向T細胞共刺激或檢查點途徑之任何其他抗體或藥物治療之個體。 排除已預先經莫格利珠單抗治療之個體。 排除具有任何先前3級或更大免疫相關不良事件(irAE)同時接受任何先前免疫療法劑或任何未定irAE之個體。 排除對藥物具有嚴重過敏反應之病史之個體。 排除在第1週期第1天之前的28天內已接收減毒活痘苗之個體。 排除具有器官移植或同種異體骨髓移植病史之個體。 排除在第1週期第1天前之28天或42天內已接收化學療法、免疫療法、生物或激素療法、另一研究性藥物、輻射或主要手術用於癌症治療(對於亞硝基脲或絲裂黴素C)之個體。 排除根據先前抗癌療法具有任何未定之毒性1級或更小(由CTCAE版本4.03定義)之個體。排除具有不可逆毒性的個體,該不可逆毒性(例如,聽覺損耗)可能在主要或副研究員酌情處理時可包括的由研究性產品不合理預期而加劇。 排除在第1週期第1天前之28天內已接收全身性類固醇療法或任何其他形式之免疫抑制藥療的個體,然而,可在主要或副研究員酌情處理時包括經局部免疫抑制藥療之個體。 排除需要全身性類固醇或免疫抑制劑之患有已知活性自體免疫疾病或症候群(例如,類風濕性關節炎、眼色素層炎、全身性紅斑狼瘡、韋格納氏肉芽腫病(Wegener's granulomatosis)或類肉瘤病)之個體。根據此研究將排除在書面知情同意之前已3年不需要全身性類固醇或免疫抑制劑的患有白斑病、禿髮或自體免疫疾病或症候群之病史之個體。 具有活性發炎性腸病(例如,發炎性結腸炎及潰瘍性結腸炎)、克隆氏症(Crohn's disease)、腸易激疾病、乳糜瀉,或與腹瀉相關聯之其他嚴重GI慢性病狀之個體。然而,可包括在書面知情同意之前已3年不需要任何療法之具有慢性GI疾病之病史之個體。 具有先天性或後天性免疫缺乏且具有肺結核或測試為陽性之人類免疫不全病毒(HIV)、人類T細胞白血病I型病毒(HTLV-1)、B型肝炎表面抗原及C型肝炎病毒抗體之先前臨床診斷已知病史的個體。 排除在第1週期第1天前之5年內具有其他創傷性惡性腫瘤之個體。然而,具有已以手術方式治癒之非侵入性惡性腫瘤,諸如子宮頸原位癌、皮膚之非黑色素瘤癌瘤或乳房之乳腺管原位癌的個體可登記。 排除已登記另一臨床研究之個體,除非該另一臨床研究為觀測性(非干預)臨床研究或干預研究之跟蹤階段。 排除具有在研究員及/或發起人之觀點中將干擾研究性產品之評估或個體安全性或研究結果之解釋之任何病況的個體。 5.研究設計劑量遞增階段 :劑量遞增階段為將鑑別組合療程之MTD或建議固定劑量之平行的3+3設計。MTD定義為經研究之莫格利珠單抗及納武單抗的一個較低劑量水準,其中經受劑量限制毒性(DLT)之6個個體組中的大於2個個體;在MTD未經判定之情況下,用於組擴增階段之建議固定劑量將根據MTD或組2中之劑量水準來判定。劑量遞增階段將登記大約3至18個個體(在至多3個組中每組3至6個個體)。劑量水準及排程描述於下文中。亦參看圖1。 [表1] 1 . 劑量遞增階段中之莫格利珠單抗及納武單抗組 組擴增 階段 :組擴增階段將進一步探索各組合在至多6種腫瘤類型中之安全性、PK、PD及抗腫瘤活性。在此階段中,將登記大約90個個體,該等個體患有局部晚期或轉移性固態腫瘤,包括(但不限於)NSCLC、SCLC、GC、肝細胞細胞癌瘤、胰腺癌及食道癌;另一腫瘤類型可被認為由劑量遞增階段之觀察結果保證以評估其他安全性。組合療法之治療 :在劑量遞增階段及組擴增階段中,個體將接收莫格利珠單抗及納武單抗之組合治療。各個體將在第1天及第15天以歷時至少1小時之靜脈內(IV)輸注形式接收3.0 mg/kg之納武單抗。各個體將亦在第一週期之第1天、第8天、第15天及第22天以及後續週期之第1天及第15天以歷時至少1小時之IV輸注形式接收0.1、0.3或1.0 mg/kg之莫格利珠單抗。將在第29天重複該組合療程。在各週期之第1天及第15天,將在完成納武單抗投與及1小時之觀察後,投與莫格利珠單抗。個體將接收組合療法直到疾病發展或死亡。安全性及療效評定 :所有個體將根據事件排程在第1週期期間之第1天、第8天、第15天及第22天監測安全性;在後續週期中,將在第1天及第15天每2週執行安全性評定。 藉由電腦斷層掃描(CT)或磁共振成像(MRI)之疾病評定將在每4週持續第一2個週期,每8週持續下一12個週期,且接著其後每12週之篩選時執行直至疾病進展或死亡。疾病進展將根據RECIST版本1.1之準則而限定。 [表2] 2 . 研究性藥品 6.療效及安全性變量主要終點安全性次要終點療效 :將利用以下療效參數: -使用RECIST版本1.1評估之最佳整體反應(BOR)。 -達至反應之時間(TTR):自第1週期第1天至確診CR/PR之第一評定日期的天數 -反應持續時間(DOR):自確診CR/PR之第一評定日期至死亡或PD (無論哪個較早)日期的天數 -無進展存活期(PFS):自第1週期第1天至死亡或PD (無論哪個較早)日期的天數 -總存活期(OS):自第1週期第1天至死亡日期的天數統計分析 :以下分析集將用於研究: -安全性分析集:包括接收研究藥物之至少一種劑量(即使部分劑量)之所有個體。全分析集(FAS):包括接收研究藥物之至少一種劑量(即使部分劑量)且具有至少一個可利用之劑量後療效數據之所有個體。 不良事件將由身體系統、嚴重性及治療比例列表。將提供關於嚴重不良事件、導致研究停藥的AE及導致死亡之AE的類似表示用於安全性分析及全分析集。 將評估個體之腫瘤反應、TTR、DOR、PFS及OS。ORR將經計算為作為反應者之個體的比例,亦即完全反應(CR)及部分反應(PR);將計算關於反應之95%的準確信賴區間。亦將計算疾病控制率(CR、PR、SD)。 7.臨床療效 如在此臨床試驗之組合療法期間之觀察結果,截至2016年9月7日,15個肝細胞細胞癌(HCC)患者中觀察到4個PR、6個SD及3個PD。另一方面,截至2016年9月7日,在15個胰腺癌(PA)患者中觀察到1個PR、5個SD及9個PD。此外,在各癌症類型中觀察到若干不良事件,然而,所有事件在癌症治療中可為可接受。 截至2017年11月1日(資料截止日期為2017年8月31日),15個HCC患者中觀察到4個PR及6個SD (客觀反應率26.7%,疾病控制率66.7%) (圖2);15個PA患者中觀察到1個PR及5個SD (客觀反應率6.7%,疾病控制率40.0%) (圖3)。對於患有HCC及PA之患者之中間無進展存活期分別地為114.5天(95% CI,28.0至223.0)及56.0天(95% CI,29.0至115.0),及中間總存活期分別地為343.0天(95% CI,123.0至不可估)及198.0天(95% CI,107.0至328.0)。 在劑量遞增階段中未觀察到劑量限制毒性。3級至4級藥物相關不良事件(AE)以25.0%發生於劑量遞增階段及組擴增部分中。未見到5級相關AE。最常相關AE為皮疹(36.5%)、皮丘疹(20.8%)及腹瀉(14.6%)。 根據此試驗之此等臨床結果,發現抗CCR4抗體及抗PD-1抗體之組合對若干類型之癌症患者(至少包括HCC及PC患者)為有效的。實例 2 莫格利珠單抗組合納武單抗在患有局部晚期或轉移性固態腫瘤之個體中之開放標記的多中心階段 1 / 2 研究 1.研究概述 該研究為抗CCR4抗體,例如莫格利珠單抗及抗PD-1抗體,例如納武單抗之組合療法在患有局部晚期或轉移性固態腫瘤之成人個體中之多中心的階段1/2開放標記的發現劑量的組擴增研究。 階段1將鑑別莫格利珠單抗及納武單抗個體之組合療程之MTD或在不存在超出MTD之情況下的最高方案限定劑量。階段1將登記至多12個個體。 階段2將探索組合療程之最高耐受劑量之安全性、療效及抗腫瘤活性。階段2將登記至多184個個體(21至36個個體/腫瘤類型),該等個體患有以下腫瘤類型之局部晚期或轉移性疾病:鱗狀細胞非小細胞肺癌(NSCLC);未表現程序式細胞死亡配位體1(PD-L1)之非鱗狀細胞NSCLC;頭部及頸部之鱗狀細胞癌(SCCHN);非微衛星不穩定性(非MSI)較高結腸直腸癌(CRC);卵巢癌;肝細胞細胞癌(HCC)及胰腺癌(PA)。 2.研究目標 主要目標 主要目標為表徵莫格利珠單抗及納武單抗之組合療程在患有局部晚期或轉移性固態腫瘤之個體中之安全性及耐受性且判定該組合療程之MTD或在不存在超出MTD之情況下的最高方案限定劑量。 次要目標 次要目標為基於RECIST版本1.1評估莫格利珠單抗及納武單抗之組合之抗腫瘤活性。抗腫瘤活性將評定為整體反應率(ORR)、TTR、DOR、PFS及OS。 探索目標 探索目標為: -當組合投與時分析莫格利珠單抗及納武單抗之血清濃度; -當組合投與時評估莫格利珠單抗及納武單抗之免疫原性; -評估莫格利珠單抗及納武單抗之組合之藥效動力學(PD)特性且判定哪一種生物標記可能與安全性及/或抗腫瘤活性相關; -基於免疫相關RECIST (irRECIST)版本1.1評估莫格利珠單抗及納武單抗之組合之ORR。 3.入選準則 個體在篩選階段期間必須符合以下入選準則中之每一者以便有資格參與研究。此外,參與階段2之個體必須符合所有入選準則且不具有對於相關腫瘤類型之排除準則中之一者。 -18歲或大於18歲之個體; -患有組織學或細胞學上確診之實體腫瘤之個體; -患有局部晚期或轉移性實體腫瘤之個體; -已發展或對任何標準治療方案已不耐受或拒絕標準治療,或不存在用於其之適當標準療法之個體; -具有可評估病灶/RECIST版本1.1之指南之個體; -具有0或1之東部腫瘤協作組(ECOG)性能狀態(PS)之個體; -若個體為可能懷孕之女性或與可能懷孕女性具有性活動之男性,則同意自簽署ICF起在參與研究期間;以及對於女性在最後劑量之研究性醫療產品(IMP)後之23週或對於男性在最後劑量之IMP後之31週使用充分避孕之個體; -具有充分血液、腎、肝及呼吸功能限定的個體; -願意在篩選階段期間經受腫瘤活檢,或若腫瘤不可存取用於活檢,則存檔腫瘤材料必須可供用於呈遞之個體;以及 -根據監管機構指導原則自願簽署機構審查委員會審批通過之知情同意書且註明日期的個體。 4.排除準則 若在篩選階段期間符合以下排除準則中之任一者,則個體將無資格參與此研究。此外,參與階段2之個體必須符合所有入選準則且不具有特定對於相關腫瘤類型之排除準則中之一者。 -懷孕或哺乳期之女性個體或在此研究期間期望懷孕或生孩子之任何個體; -具有不可控且明顯當前疾病之個體。 -具有在研究員之觀點中將限制順從研究要求之精神疾病/社會情況之個體; -具有原發性CNS腫瘤或已知CNS癌轉移及/或CNS癌轉移及/或癌性腦膜炎病史之個體;例外:若CNS癌轉移經充分治療則個體為符合條件的及在登記前之至少4週已神經性返回至基線(除關於CNS治療之殘餘病症或症狀以外)之個體。此外,個體必須在登記前停用皮質類固醇4週。 -在第1週期第1天前已接收用於癌症之先前療法或在28天或42天內用於亞硝基脲或絲裂黴素C,或14天內用於他莫昔芬(tamoxifen)之主要手術之個體; -在第1週期第1天前之14天內已接收放射線療法或放射外科手術之個體; -已預先經抗PD-1、抗PD-L1、抗PD-L2、抗CD137或抗CTLA-4抗體或特定靶向T細胞共刺激或檢查點途徑之任何其他抗體或藥物治療之個體; -已預先經莫格利珠單抗治療之個體; -具有過敏病史或對研究藥物組分有過敏之個體; -在第1週期第1天前之28天內已接收減毒活痘苗之個體; -具有器官移植或同種異體骨髓移植病史之個體; -具有來自先前抗癌療法之>1級的任何未解決毒性之個體; -在第1週期第1天前之14天內已使用免疫抑制藥療之個體。 -具有可能影響生命器官功能或需要包括全身性皮質類固醇之免疫抑制治療的已知活性自體免疫疾病或自體免疫疾病病史之個體; -具有中毒性表皮壞死溶解或史蒂芬斯強森症候群(Stevens-Johnson syndrome)病史之個體; -具有發炎性腸病、克隆氏病、潰瘍性結腸炎或韋格納氏肉芽腫病病史之個體; -具有原發性或後天性免疫缺乏或人類免疫不全病毒(HIV)測試為陽性之已知病史或已知後天免疫缺乏症候群之個體; -除具有肝細胞癌之個體外,對於指示急性或慢性感染之B型肝炎表面抗原(HBVsAg)或C型肝炎RNA測試為陽性之個體; -具有要求同時干預之另一活性惡性腫瘤之個體; -接收任何其他研究性藥劑之個體; -具有在研究員及/或發起人之觀點中將干擾IMP之評估或個體安全性或研究結果之解釋之另一病狀之個體;以及 -具有肺炎或間質性肺病病史之個體。 5.研究設計 階段1:劑量發現 階段1劑量發現研究具有針對該組合療程將鑑別MTD或在不存在超出MTD之情況下的最高方案限定劑量的3+3設計。階段1研究將登記至多12個個體(3至6個個體/組)。規劃起始劑量水準及可選劑量水準。劑量水準及排程描述於表3中。MTD定義為低於其中三分之一或更多個體經歷DLT之組的劑量水準的一種劑量水準。階段2之建議劑量方案欲為所測試之MTD或最高劑量水準。 [表3] 3 . 劑量 水準 *若>1個個體在劑量水準1下經歷DLT,則可登記此劑量水準。 用於劑量發現之準則概述於圖4中。 階段2:擴增組 為進一步表徵該組合之安全性、耐受性及抗腫瘤活性,將登記患有以下腫瘤類型中之局部晚期或轉移性疾病的至多184個個體(21至36個個體/腫瘤類型):鱗狀細胞非小細胞肺癌(NSCLC);未表現程序式細胞死亡配體1 (PD-L1)之非鱗狀細胞NSCLC;頭部及頸部之鱗狀細胞癌(SCCHN);非微衛星不穩定性(非MSI)較高結腸直腸癌(CRC);卵巢癌;肝細胞細胞癌瘤(HCC);以及胰腺癌(PA)。個體在階段1中將經視為可耐受之最高劑量之組合療程治療。將監測用於各擴增組中之給藥療程之安全性及耐受性。 個體之治療 研究性藥品(IMP) 以下IMP將用於此研究: -莫格利珠單抗:在第1週期之第1天、第8天、第15天及第22天隨後在各後續28天週期之第1天及第15天以靜脈內輸注形式歷時至少1小時投與0.3或1.0 mg/kg。 -納武單抗:在各28天週期之第1天及第15天以靜脈內輸注形式歷時至少30分鐘投與240 mg之納武單抗。 治療之持續時間 個體可自第1週期第1天接收組合療法持續至多96週。停止治療且無疾病進程及在最後劑量之IMP之12個月內體驗疾病進程之個體可額外接受額外的IMP持續至多48週,前提條件是該等個體未接受其他全身性療法用於其癌症。 安全性及療效評定 將在整個研究期間監測個體之安全性。藉由CT或MRI之腫瘤評定將在篩選階段、第10週、接著其後之至少每12週期間執行直至明確的疾病進程或死亡。將在篩選階段(除存在存檔腫瘤組織之個體以外)及第10週(除非腫瘤不可存取用於活檢)期間需要腫瘤活檢體。 6.療效及安全性變量 主要終點 安全性及耐受性將藉由評定AE及物理檢驗發現、生命徵象量測值、12導聯ECG讀取及臨床實驗室評估中之變化來評估。 次要終點 將利用以下療效參數: -使用RECIST版本1.1評估之最佳整體反應(BOR); -達至反應之時間(TTR):自第1週期第1天至使用RECIST版本1.1確診CR/PR之第一評定日期的天數 -反應持續時間(DOR):自使用RECIST版本1.1確診CR/PR之第一評定日期至死亡或PD (無論哪個為較早)之日期的天數; -無進展存活期(PFS):自第1週期第1天至死亡或PD(無論哪個為較早)之日期的天數。對於使用RECIST版本1.1其BOR為PR或CR之個體; -總存活期(OS):自第1週期第1天至死亡日期的天數 統計方法及規劃分析 用於此研究之資料之評估將主要地由資料清單及總結顯示組成。將針對安全性分析及療效分析集概括人口統計及其他基線特徵資訊。不良事件將由身體系統、嚴重性及治療比例列表。將提供關於SAE、導致IMP停止之AE及導致死亡之AE的類似表現。實驗室參數之列表將指示對於各參數之正常範圍。各值將經分類為屬於、低於或在正常範圍內。將評估個體之腫瘤反應、TTR、DOR、PFS及OS。ORR將經計算為作為反應者之個體的比例,亦即完全反應(CR)及部分反應(PR);將計算關於ORR之95%的準確信賴區間。將基於組合療法之擴增組中之療效可評估個體使用RECIST版本1.1及irRECIST版本1.1兩者來推導出ORR,該等個體包括在第1週期第1天接受組合療法之所有個體。將使用卡本-麥爾方法(Kaplan-Meier methodology)來評估反應持續時間、OS及PFS。 7.臨床療效 如在此臨床試驗之組合療法期間之觀察結果,截至2017年8月28日,10個HCC患者中1個未確診PR,21個卵巢癌患者中2個確診CR及2個確診PR;以及29個CRC患者中1個確診PR。 截至2017年10月23日,10個HCC患者中1個未確診PR及1個確診PR;21個卵巢癌患者中2個確診CR、2個確診PR及1個未確診PR;以及29個CRC患者中1個確診PR。另一方面,截至2017年10月23日,17個PA患者中未觀察到PR或CR。 根據此試驗之此等臨床結果,發現抗CCR4抗體及抗PD-1抗體之組合對至少包括HCC、卵巢癌及CRC患者之若干類型之癌症患者為有效的。實例 3 個體之外周血液中之免疫細胞亞群的流式細胞量測術分析 方法 在實例1中所描述之階段1研究中,免疫細胞亞群係藉由FCM來量測。在治療前、治療之第2週期第1天或治療結束及第3週期第1天收集外周血液且TIL在治療前及治療後(自治療之第2週期第15天至第3週期第1天或治療結束)與新製腫瘤活檢樣本分離。效應Treg之分析係根據描述於Immunity,2009,30(6)899-911中之方法藉由CD4、Foxp3及CD45RA之表現來完成。使用FlowJo版本7.6.5來分析資料。同時外周血液中之各群體之百分比係以淋巴球之數量來校正的。 結果 藉由對外周血液之初步分析,觀察到第2週期第1天(包括治療結束)及第3週期第1天之效應Treg (CD3+ CD4+ CD45RA-Foxp3high )之降低趨勢與最佳整體反應無關。相較於非反應者中之彼等,亦在反應者中觀察到第3週期第1天之活化T細胞(CD3+ CD4+ ICOS+ T細胞)增加。 藉由在治療前及治療後對11對TIL之初步分析,TIL中之效應Treg (CD3+ CD4+ CD45RA-Foxp3high )與總CD3+ T細胞之比例在治療後具有獨立於臨床反應之降低趨勢。相較於Treg,TIL中之CD3+ CD8+ T細胞與總CD3+ T細胞之比例在治療後具有整體增加趨勢。TIL中之CD3+ CD8+ T細胞/效應Treg在治療後亦具有增加趨勢。此外,藉由評估治療後之外周血液中之趨化因子(MIG及IP-10)之含量,發現增加趨勢與最佳整體反應無關。 在整個本申請案中,各種公開案以圓括號提及作者名稱及日期,或提及專利號或專利公開案號。此等公開案之揭示內容在此以全文引用之方式併入本申請案中,以更全面地描述截至本文中描述及主張之本發明之日期為止,如熟習此項技術者已知之目前先進技術。然而,本文中參考文獻之引用不應視為承認此類參考文獻為本發明之先前技術 本申請案主張2016年11月30日申請之美國臨時申請案序列號第62/428,468號及2017年9月5日申請之美國臨時申請案序列號第62/554,486號之權利,該兩個文獻之全部內容以引用之方式併入本文中。無文本序列表 SEQ ID NO.1:人工序列之描述;KW-0761_VH CDR1 SEQ ID NO.2:人工序列之描述;KW-0761_VH CDR2 SEQ ID NO.3:人工序列之描述;KW-0761_VH CDR3 SEQ ID NO.4:人工序列之描述;KW-0761_VL CDR1 SEQ ID NO.5:人工序列之描述;KW-0761_VL CDR2 SEQ ID NO.6:人工序列之描述;KW-0761_VL CDR3 SEQ ID NO.7:人工序列之描述;KW-0761_VH SEQ ID NO.8:人工序列之描述;KW-0761_VL SEQ ID NO.9:人工序列之描述;KW-0761_H SEQ ID NO.10:人工序列之描述;KW-0761_L SEQ ID NO.11:人工序列之描述;Nivolumab_VH CDR1 SEQ ID NO.12:人工序列之描述;Nivolumab_VH CDR2 SEQ ID NO.13:人工序列之描述;Nivolumab_VH CDR3 SEQ ID NO.14:人工序列之描述;Nivolumab_VL CDR1 SEQ ID NO.15:人工序列之描述;Nivolumab_VL CDR2 SEQ ID NO.16:人工序列之描述;Nivolumab_VL CDR3 SEQ ID NO.17:人工序列之描述;Nivolumab_VH SEQ ID NO.18:人工序列之描述;Nivolumab_VL SEQ ID NO.19:人工序列之描述;Nivolumab_H SEQ ID NO.20:人工序列之描述;Nivolumab_L SEQ ID NO.21:人工序列之描述;人類CCR4 cd核苷酸及胺基酸序列 SEQ ID NO.22:人工序列之描述;合成建構CCR4胺基酸序列 SEQ ID NO.23:人工序列之描述;PD-1 cd人類PD-1核苷酸及胺基酸序列 SEQ ID NO.24:人工序列之描述;合成建構人類PD-1胺基酸序列Unless otherwise defined, all technical and scientific terms used herein have the meaning commonly understood by those skilled in the art to which the present invention belongs. The following references provide the skilled person with a general definition of multiple terms used in the present invention: Singleton et al., Dictionary of Microbiology and Molecular Biology (2nd Edition 1994); The Cambridge Dictionary of Science and Technology (Walker Edition, 1988) ; The Glossary of Genetics, 5th Edition, R. Rieger et al. (Eds.), Springer Verlag (1991); and Hale & Marham, The Harper Collins Dictionary of Biology (1991). As used herein, unless otherwise specified, the following terms have the meaning attributed to them below. "Dosing" refers to using any of a variety of methods and delivery systems known to those skilled in the art to physically introduce a composition containing a therapeutic agent to an individual. The administration route of the anti-PD-1 antibody includes intravenous, intramuscular, subcutaneous, intraperitoneal, spinal cord, or other parenteral administration routes, such as by injection or infusion. As used herein, the phrase "non-intestinal administration" means the mode of administration other than enteral and local administration, usually by injection, and includes (but is not limited to) intravenous, intramuscular, intraarterial , Intrathecal, intralymphatic, intralesional, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subepidermal, intraarticular, subcapsular, subarachnoid, intraspinal, epidural And intrasternal injection and infusion, and electroporation in vivo. In some embodiments, the agent is administered orally via a non-parenteral route, in some embodiments. Other non-parenteral routes include topical, epidermal or mucosal routes of administration, such as intranasal, transvaginal, transrectal, sublingual or topical. The administration can also be performed once, plural times, and / or over one or more extended time periods, for example. As used herein, "adverse event" (AE) is any adverse and generally undesirable or undesirable sign (including abnormal laboratory findings), symptoms, or disease associated with the use of medical treatment. For example, adverse events can be associated with activation of the immune system or expansion of immune system cells (eg, T cells) in response to treatment. The medical treatment may have one or more related AEs and each AE may have the same or different severity. Reference to methods that can "modify adverse events" means treatments that reduce the incidence and / or severity of one or more AEs. "Isolated antibody" refers to an antibody that is substantially free of other antibodies with different antigen specificities (for example, an isolated antibody that specifically binds to PD-1 does not substantially contain specific binding to other than PD-1 Antigen antibody). However, isolated antibodies that specifically bind to PD-1 can cross-react with other antigens, such as PD-1 molecules from different species. In addition, the isolated antibody may be substantially free of other cellular materials and / or chemicals. "Anti-antigen" anti-system refers to an antibody that specifically binds to an antigen. For example, anti-PD-1 antibodies specifically bind to PD-1 and anti-CTLA-4 antibodies specifically bind to CTLA-4. The "antigen-binding portion" (also called "antigen-binding fragment") of an antibody refers to one or more fragments of an antibody that retain the ability to specifically bind to the antigen bound by the entire antibody. In the present invention, "comprises / comprising", "containing" and "having" and the like may have the meaning attributed to them in the US Patent Law and may mean "includes / including" and Similar to it; "consisting essentially of" or "essentially consisting of" also have the meaning attributed to the US Patent Law and the term is open-ended, as long as the basic or novel features cited are not greater than those cited Exist and change, but excluding the prior art embodiments, the existence of content greater than the cited content is allowed. "Cancer" refers to a wide variety of disease groups characterized by uncontrolled growth of abnormal cells in the body. Uncontrolled cell division and growth Division and growth result in the formation of malignant tumors that invade adjacent tissues and can also be transferred to the distal part of the body via the lymphatic system or blood flow. After the cancer has metastasized, the distant tumor can be said to be "derived from" the original, pre-metastatic tumor. Since distal tumors are derived from pre-metastatic tumors, "derived from" tumors may also include pre-metastatic tumors. The term "immunotherapy" refers to the treatment of individuals suffering from disease or at risk of infection or suffering from disease recurrence by methods that include inducing, enhancing, inhibiting or otherwise modifying the immune response. "Treatment" or "therapy" of an individual refers to any type of intervention or treatment performed on the individual, or administration of an active agent to the individual, with the goal of reversing, relieving, improving, inhibiting, alleviating or preventing symptoms, complications, or disease The onset, progression, development, severity, or recurrence of symptoms or biochemical markers associated with the disease. The "therapeutically effective amount", "effective amount", "effective dose" or "therapeutically effective dose" of a drug or therapeutic agent is when used alone or in combination with another therapeutic agent to protect the individual from the onset of the disease or promote the disease A reduction in the severity of symptoms, an increase in the frequency and duration of periods without disease symptoms, evidence of the regression of the disease, or any amount of medicine that prevents injury or disability due to the disease. The ability of a therapeutic agent to promote disease regression can be evaluated using various methods known to those skilled in the art, such as evaluation in human individuals during clinical trials, evaluation in animal model systems that predict efficacy in humans, or by In the external analysis method, the activity of the analysis reagent is evaluated. In one embodiment, the antibodies or combinations of antibodies described herein promote cancer regression or prevent other tumor growth in the individual. In certain embodiments, a therapeutically effective amount of the drug promotes cancer regression to the point of eliminating the cancer. "Promoting cancer regression" means that an effective amount of the drug is administered alone or in combination with an anti-tumor agent, resulting in tumor growth or size reduction, tumor necrosis, reduction in the severity of at least one disease symptom, frequency of time period without disease symptom and Increase the duration, or prevent injury or disability due to illness and illness. In addition, the terms "effective" and "utility" regarding treatment include both pharmacological utility and physiological safety. Pharmacological utility refers to the ability of a drug to promote cancer regression in a patient. Physiological safety refers to the degree of toxicity due to the administration of drugs, or other adverse physiological effects (adverse effects) on the level of cells, organs and / or organisms. For example, for the treatment of tumors, a therapeutically effective amount of an antibody or combination thereof can inhibit cell growth or tumor growth by at least about 100% relative to an untreated individual or in certain embodiments, relative to a patient treated with standard care therapy 10%, at least about 20%, at least about 40%, at least about 60%, or at least about 80%. In other embodiments of the invention, tumor regression is observable and lasts for a period of at least about 20 days, at least about 40 days, or at least about 60 days. Regardless of these final measures of therapeutic effectiveness, the evaluation of immunotherapy drugs must also take into account "immune-related" response patterns. "Immune-related" response patterns refer to the clinical response patterns commonly observed in cancer patients treated with immunotherapeutic agents. These immunotherapeutic agents produce resistance by inducing cancer-specific immune responses or by modifying the native immune process Tumor effect. This response pattern is characterized by beneficial therapeutic effects after the initial increase in tumor burden or the appearance of new lesions, which will be classified as disease progression in the evaluation of traditional chemotherapeutics and will be synonymous with drug failure. Therefore, proper evaluation of immunotherapeutic agents may require long-term monitoring of the effects of these agents on the target disease. A therapeutically effective amount of a drug includes a "prophylactically effective amount", which is when administered alone or in combination with an antineoplastic agent to individuals at risk of developing cancer (eg, individuals with precancerous conditions) or individuals suffering from cancer relapse , Any amount of drugs that inhibit the development or recurrence of cancer. In certain embodiments, the prophylactically effective amount completely prevents the development or recurrence of cancer. "Suppressing" the development or recurrence of cancer means reducing the possibility of the development or recurrence of cancer or completely preventing the development or recurrence of cancer. It should be understood that the use of substitutes (eg, "or") means one of the substitutes, both, or any combination thereof. As used herein, it should be understood that the indefinite article "a (an)" refers to "one or more" of any cited or enumerated component. The term "about" or "substantially encompass" refers to a value or composition that is within an acceptable error range of the value or composition as determined by those of ordinary skill in the art, which will depend in part on how to measure or The judgment value or composition is the limitation of the measurement system. For example, "about" or "substantially encompass" may mean within 1 or more than 1 standard deviation according to practice in the art. Alternatively, "about" or "substantially encompass" may mean a range of at most 10% (ie, + 10% to -10%). For example, about 3 mg may include any value between 2.7 mg and 3.3 mg (for 10%). In addition, especially in terms of biological systems or methods, these terms may mean at most an order of magnitude or at most 5 times the value. When a specific value or composition is provided in the scope of this application and patent application, unless otherwise stated, the meaning of "about" or "substantially included" shall be assumed to be within the acceptable error range of the specific value or composition . As used herein, the term "about once a week" means a rough numerical value, and "about once a week" may include every six days to every eight days. Therefore, the frequency of "once a week" administration can be every six days, every seven days, or every eight days. As described herein, unless otherwise specified, any concentration range, percentage range, ratio range, or integer range should be understood to include any integer value within the recited range and, where appropriate, its fraction (such as tenths of an integer) One and one percent). The terms "CC chemokine receptor 4", "CCR4", "CCR4 peptide", "CCR4 protein" or "CCR4 polypeptide" as used herein are used interchangeably herein and are meant to include SEQ ID NO: 22 Amino acid sequence or a functional fragment of the polypeptide; including the GenBank deposit number NP_005499.1 amino acid sequence of the polypeptide, included in the amino acid sequence represented by SEQ ID NO: 22 or NP_005499.1 deletion, substitution Or a polypeptide having an amino acid sequence of one or more amino acid residues and having CCR4 activity; comprising at least about 85% homology, at least about 90% homology with the amino acid sequence represented by SEQ ID NO: 22 Homology, at least about 93% homology, and at least about 95%, about 96%, about 97%, about 98%, or about 99%, higher homology amino acid sequences and CCR4 activity polypeptides; and including Related polypeptides of SNP variants and the like. Related polypeptides include SNP variants, splice variants, fragments, substitutes, deletions, and insertions, which preserve CCR4 activity and / or function. In addition, the CCR4 polypeptide includes a polypeptide encoded by the nucleotide sequence of SEQ ID NO: 21 or GenBank accession number NM_005508.4. In some embodiments, the gene encoding CCR4 includes nucleotides having one or more nucleotide deletions, substitutions, or additions in the nucleotide sequence of SEQ ID NO: 21 or GenBank accession number NM_005508.4 sequence. In other embodiments, the CCR4 gene encodes a polypeptide having CCR4 function and contains at least about 60% or higher homology with the nucleotide sequence of SEQ ID NO: 21 or GenBank accession number NM_005508.4, in some embodiments Nucleotide sequences that are at least about 80% or higher in homology, and in other embodiments at least about 95%, about 96%, about 97%, about 98%, or about 99%, higher homology. In some embodiments, the CCR4 gene encodes a polypeptide having the function of CCR4 and includes a nucleotide sequence that hybridizes to the nucleotide sequence of SEQ ID NO: 21 or GenBank accession number NM_005508.4 under stringent conditions. As used herein, the term "CCR4 nucleic acid molecule" refers to a polynucleotide that encodes a CCR4 polypeptide. Exemplary CCR4 nucleic acid molecules are provided in SEQ ID NO: 21 or GenBank accession number NM_005508.4. Gene polymorphism is often identified in the nucleotide sequence of genes encoding proteins in eukaryotes. The CCR4 gene used in the present invention also includes genes that generate minor modifications in the nucleotide sequence by this polymorphism, as the genes used in the present invention. CCR4 is a G protein coupled seven transmembrane receptor cloned from human immature basophil cell line KU-812 to K5-5, and may have the amino acid sequence represented by SEQ ID NO: 22. The extracellular region of CCR4 is in the amino acid sequence from position 1 to 39, position 99 to 111, position 176 to 206, and position 268 to 284, and the intracellular region is in the amino acid sequence (GenBank accession number NP_005499.1) These are positions 68 to 77, positions 134 to 150, positions 227 to 242, and positions 309 to 360. CCR4 can specifically bind to different molecules, including (but not limited to) TARC (thymus and activation regulating chemokines) produced by thymocytes (J. Biol. Chem., 271, 21514, 1996) and separated from macrophages MDC (macrophage-derived chemokine) (J. Exp. Med., 185, 1595, 1997), also known as STCP-1 (stimulated T cell chemokine-1) (J. Biol. Chem ., 272,25229,1997). TARC and MDC are also called CCL17 and CCL22, respectively. One or more functions of CCR4 include its ability to bind to TARC and / or MDC. Other functions of CCR4 in the present invention include the expression of Ca on CCR4 cells depending on the expression of CCR4 ligands (such as CCL17 and / or CCL22)2 + Inflow and express the cell migration function of CCR4 cells. The terms "programmed cell death-1", "PD-1", "PD-1 polypeptide" or "CD279" are used interchangeably herein and mean having at least one function of PD-1 activity (eg, by PD The -1 pathway activates T cells and / or binds to PD-L1 or PD-L2) polypeptides or fragments thereof. In one embodiment, PD-1 comprises at least about 85%, at least about 90%, at least about 93%, at least about 95%, about 96%, about 97%, about 98%, about 99% with NCBI deposit number NP_005009 Or an amino acid sequence of about 100% sequence identity or the amino acid sequence set forth in SEQ ID NO: 24, wherein the PD-1 protein has binding activity to PD-L1 and PD-L2 and / or PD- 1 of activity. In some embodiments, PD-1 also includes SNP variants, splice variants, fragments, substitutes, deletions, and insertions that preserve PD-1 activity and / or function. In other embodiments, PD-1 comprises a polypeptide encoded by the nucleotide sequence of SEQ ID NO: 23 or GenBank accession number NM_005018.2. The PD-1 gene also includes genes containing DNA that contains nucleosides with one or more nucleotide deletions, substitutions, or additions in the nucleotide sequence of SEQ ID NO: 23 or GenBank accession number NM_005018.2 Acid sequence. In some embodiments, PD-1 has a nucleotide sequence at least about 60%, about 70%, about 80%, about 90%, about 95%, having a nucleotide sequence with SEQ ID NO: 23 or GenBank accession number NM_005018.2, Approximately 96%, approximately 97%, approximately 98%, approximately 99%, or approximately 100% identical nucleotide sequence codes and has at least one function of PD-1 (eg, activation of T cells by the PD-1 pathway and / or Or combined with PD-L1 or PD-L2). In other embodiments, PD-1 is encoded by a nucleotide sequence hybridized with SEQ ID NO: 23 or the nucleotide sequence of GenBank accession number NM_005018.2 under stringent conditions and has the function of a PD-1 polypeptide . The term "PD-1 nucleic acid molecule" refers to a polynucleotide that encodes a PD-1 polypeptide. Exemplary PD-1 nucleic acid molecules are provided in SEQ ID NO: 23 or GenBank accession number NM_005018.2. Gene polymorphism is often identified in the nucleotide sequence of genes encoding proteins in eukaryotes. The PD-1 gene used in the present invention also includes genes that generate minor modifications in the nucleotide sequence by this polymorphism, as the genes used in the present invention. PD-1 was cloned as a 55 kDa type I membrane protein belonging to the immunoglobulin family (Ishida et al., 1992; 11; 3887-3895). PD-1 is expressed on activated T lymphocytes. The intracellular domain of PD-1 contains ITSM motifs (exchange motifs based on immunoreceptor tyrosine) and ITIM motifs (inhibitory motifs based on immune receptor tyrosine) that can be suppression domains of immune responses. Since mice lacking PD-1 develop lupus-like autoimmune diseases such as glomerulonephritis and arthritis and diseases similar to dilated cardiomyopathy, PD-1 appears to be a negative regulator of immune response. The other PD-1 ligand, PD-L1, is known as a ligand for PD-1 and a type I transmembrane protein that acts as an inhibitor ligand for T cell proliferation by binding to PD-1. It has immunoglobulin class V domain, class C domain and cytoplasmic tail region. PD-L1 is known to be expressed on antigen-presenting cells and some cancer cells. "Anti-CCR4 antibody" refers to an antibody that selectively binds to human CCR4 polypeptide. Exemplary anti-CCR4 antibodies are provided herein, for example, anti-CCR4 antibodies consisting of amino acid residues 2 to 29 of SEQ ID NO: 22 bound to an epitope. In one embodiment, the anti-CCR4 antibody is moglizumab (POTELIGEO (registered trademark)). In another embodiment, the anti-CCR4 antibody reduces, inhibits, or prevents one or more functions of CCR4. In other embodiments, anti-CCR4 antibodies cross-compete with moglizumab. "Anti-PD-1 antibody" refers to an antibody that selectively binds to PD-1 polypeptide. Exemplary anti-PD-1 antibodies are described in, for example, US Patent No. 8,008,449, which is incorporated herein by reference. In one embodiment, the anti-PD-1 antibody is nivolumab (OPDIVO (registered trademark)). In another embodiment, the anti-PD-1 antibody reduces, inhibits, or prevents one or more functions of PD-1. In other embodiments, the anti-PD1 antibody cross-competes with nivolumab. As used in this disclosure, the term "antibody" refers to immunoglobulins or fragments or derivatives thereof, and encompasses any polypeptide that includes an antigen binding site, whether produced in vitro or in vivo. The term includes, but is not limited to, polyclonal antibodies, monoclonal antibodies, monospecific antibodies, multispecific antibodies, humanized antibodies, single chain antibodies, chimeric antibodies, synthetic antibodies, recombinant antibodies, hybrid antibodies, mutant antibodies, and Transplant antibodies. Unless otherwise modified by the term "intact", like "intact antibody", for the purposes of this disclosure, the term "antibody" also includes preservation of antigen-binding function (ie, the ability to specifically bind CCR4 or PD-1) Antibody fragments (or antigen-binding fragments), such as Fab, F (ab ')2 , Fv, single chain Fv (scFv), Fd, dAb and other antibody fragments. Generally, such fragments will contain an antigen binding domain (or portion). Antibody molecules are formed from polypeptides called heavy chains (hereinafter referred to as H chains) and light chains (hereinafter referred to as L chains). In addition, the H chain is composed of the H chain variable region (also called VH Or VH) and the H chain constant region (also called CH), and the L chain is composed of the L chain variable region (also called VL Or VL) and L chain constant region (also called CL). Regarding CH, α, δ, ε, γ, and mu chains are known for each sub-category. Regarding CL, λ and κ are known. IgG antibodies have two heavy chains and two light chains, and form two antigen binding sites composed of VH and VL. Therefore, IgG antibodies are bivalent antibodies. The terms "antibody fragment", "antigen-binding domain", "antigen-binding fragment", "antigen-binding portion" and "binding fragment" refer to a portion of an antibody molecule that contains amino acids responsible for specific binding between an antibody and an antigen . For example, when the antigen is large, the antigen-binding domain may only bind to a part of the antigen. The part of the antigen molecule responsible for the specific interaction with the antigen binding domain is called the "antigenic determinant" or "antigenic determinant". The antigen binding domain usually contains the antibody light chain variable region (VL Or VL) and antibody heavy chain variable region (VH Or VH). However, it is not necessary to include both. For example, Fd antibody fragments consist of only VH The domain is composed, but still retains a certain antigen binding function of the intact antibody. The term "complementarity determining region" or "CDR" refers to the essential part of the antigen-binding function of the antibody contained in the variable region of the antibody. There are three CDRs in VH and VL, respectively, and these are defined as CDR1 to CDR3 in each variable region. Antigen-binding fragments of antibodies can be produced by recombinant DNA technology or by enzymatic or chemical cleavage of intact antibodies. Antigen-binding fragments include Fab, Fab ', F (ab')2 , Fv and single chain antibodies. It should be understood that the binding sites of antibodies other than "bispecific" or "bifunctional" antibodies are identical. Degradation of antibodies by enzymes (papain) produces two identical antigen-binding fragments, also known as "Fab" fragments and "Fc" fragments. These two antigen-binding fragments have no antigen-binding activity but have crystallization ability. F (ab ') is produced by breaking down antibodies by pepsin2 A fragment in which the two arms of an antibody molecule remain linked and contain two antigen binding sites. F (ab ')2 The fragments can be cross-linked with the antigen. As used herein, "Fv" refers to the smallest fragment of an antibody that retains both the antigen recognition site and the antigen binding site. As used herein, "Fab" refers to a fragment of an antibody that includes the constant domain of the light chain and the CH1 domain of the heavy chain. The term "mAb" refers to a monoclonal antibody. Antibodies that can be used in the present invention include, but are not limited to, all natural antibodies, bispecific antibodies; chimeric antibodies; Fab, Fab ', single chain V region fragments (scFv), fusion polypeptides, and unconventional antibodies. The domain refers to the functional structural unit of each polypeptide constituting the antibody molecule. In addition, Fc that can be used in the present invention refers to the partial sequence and partial structure of the H chain constant region formed by the hinge domain, CH2 domain, and CH3 domain. In addition, CH is formed by the CH1 domain, hinge domain, CH2 domain, and CH3 domain from the N-terminal. The CH1 domain, hinge domain, CH2 domain, CH3 domain and Fc region in the present invention can be identified by multiple amino acid residues from the N-terminal according to the EU index [Kabat et al., Sequences of Proteins of Immunological Interest, US Dept .Health and Human Services (1991)]. Multiple amino acid residues are indicated by Kabat et al. By the EU index and in the present invention, the first multiple amino acid residues indicate the original or parent residue of the polypeptide and the last multiple amino acid residues indicate the polypeptide Replace or replace amino acid residues. Specifically, CH1 is identified by the amino acid sequence from positions 118 to 215 of the EU index, the hinge is identified by the amino acid sequence from positions 216 to 230 of the EU index, and CH2 is identified by positions 231 to 231 from the EU index The amino acid sequence of 340 is identified, and CH3 is identified by the amino acid sequence from positions 341 to 447 of the EU index. As used herein, the term "recombinant antibody" refers to antibodies produced by recombinant technology and monoclonal antibodies obtained from fusion tumors. Recombinant antibodies include chimeric antibodies prepared by combining a human antibody constant region and a non-human antibody variable region, and a framework by combining CDRs of the H chain and L chain of a non-human antibody variable region into a human antibody variable region Humanized antibodies (or CDR grafted antibodies) prepared in the region (hereinafter abbreviated as FR), and human antibodies prepared by using animals producing human antibodies or the like. The term "chimeric antibody" as used herein refers to an antibody in which the amino acid sequences of VH and VL of a non-human animal antibody are grafted into the corresponding VH and VL of a human antibody. Chimeric antibodies can be obtained by obtaining cDNAs encoding VH and VL from monoclonal antibody-derived fusion tumors derived from non-human animals, and inserting these cDNAs into animals with DNA encoding human antibody CH and CL The expression vector of the cell is used to construct a human chimeric antibody expression vector, and then the vector is introduced into animal cells to generate the antibody. The term "humanized antibody" refers to an antibody in which the amino acid sequence of the CDR of the VH and VL of the non-human animal antibody has been grafted into the corresponding CDR of the VH and VL of the human antibody. Except for the CDRs of VH and VL, this region is called FR. Humanized antibodies can be produced by: (i) constructing an amino acid consisting of the amino acid sequence of the CDR of the VH of the non-human antibody and the amino acid sequence of the framework region (FR) of the VH of any human antibody CDNA encoding the sequence and cDNA encoding the amino acid sequence of the VL composed of the amino acid sequence of the CDR of the VL of the non-human animal antibody and the amino acid sequence of the FR of the VL of any human antibody; (ii) Respectively inserting these cDNAs into animal cell expression vectors with DNA encoding human antibody CH and CL to construct a humanized antibody expression vector; and (iii) introducing this vector into animal cells to express the antibody. As used herein, the term "human antibody" refers to an antibody having a variable region, where both the framework region and the CDR region are derived from human germline immunoglobulin sequences. In addition, if the antibody contains a constant region, the constant region is also derived from human germline immunoglobulin sequences. The human antibody of the present invention may include amino acid residues not encoded by human germline immunoglobulin sequences (for example, mutations induced by random or site-specific mutations in vitro or introduced by somatic mutations in vivo). However, as used herein, the term "human antibody" is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species (such as a mouse) have been grafted onto human framework sequences. The term "human" antibody is used synonymously with "fully human" antibody. Human antibodies also include antibodies obtained from human antibody phage libraries, colonization of immortalized human peripheral blood lymphocytes or human antibody-producing transgenic animals prepared according to technological improvements in recent genetic engineering, cell engineering, and R & D engineering. Human antibodies can be obtained by immunizing mice with human immunoglobulin genes (Tomizuka K et al., Proc Natl Acad Sci USA. 97,722-7,2000) with the desired antigen. In addition, by using phage display libraries formed by the amplification of antibody genes from human B cells to select human antibodies with the desired binding activity, it is possible to obtain human antibodies without immunization (Winter G et al., Annu Rev Immunol. 12: 433-55. 1994). In addition, by using the Epstein-Barr (EB) virus to immortalize human B cells to prepare human antibody-producing cells with the desired binding activity, it is possible to obtain human antibodies (Rosen A et al., Nature 267, 52-54. 1977). The human antibody phage library is a phage library that causes antibody fragments such as Fab and scFv to appear on its surface by inserting antibody genes prepared from human B cells into the genes of phage. By using the binding activity relative to the fixed antigen substrate as an index, it is possible to recover from this library phages that exhibit antibody fragments with the desired antigen binding activity. Antibody fragments can also be converted into human antibody molecules composed of two complete H chains and two complete L chains by genetic modification techniques. Transgenic animals that produce human antibodies refer to animals obtained by integrating human antibody genes into the chromosomes of host animals. Specifically, human antibody genes are introduced into mouse ES cells. The ES cells were then transferred to the early embryos of another mouse. Transgenic animals that produce human antibodies can be produced from the embryo. To produce human antibodies from transgenic animals that produce human antibodies, human antibody-producing fusion tumors are obtained by the normal fusion tumor preparation method. After culturing fusion tumors obtained from mammals other than humans, human antibodies can be produced and expressed in the culture. Specifically, antibodies suitable for the present disclosure may include non-human animal antibodies, humanized antibodies, and VH and VL of human antibodies produced by fusion of tumors or antibody-producing cells by any method known in the art Amino acid sequence. The amino acid sequence of CL in the antibody of the present invention may be any of the amino acid sequence of a human antibody or the amino acid sequence of a non-human animal antibody. In certain embodiments, the amino acid sequence of Cκ or Cλ of human antibodies is used. The CH in the antibody of the present invention may be any of immunoglobulins. In certain embodiments, any one of γ1 (IgG1), γ2 (IgG2), and γ4 (IgG4) and variants thereof may be suitable for the antibody in the present invention. The term "effector function" as used in the present invention refers to the cytotoxic activity induced by antibodies, including antibody-dependent cytotoxicity (ADCC), complement-dependent cytotoxicity (ADCC) by effector cells, such as natural killer (NK) cells ( CDC), antibody dependent phagocytosis (ADP), or any combination thereof. The effector function of antibodies can be adjusted by known methods. For example, it is known in the art that ADCC activity can be adjusted by controlling the amount of trehalose (also known as "nucleated trehalose"), which is reduced by a complex N-glycoside-linked sugar chain The α1-6 bond in the terminal binds to N-acetyl glucosamine (GlcNAc), the complex N-glycoside-linked sugar chain binds to the Fc region of the antibody according to the EU index (Kabat et al., Sequence of Proteins of immunological interests, 5th edition, 1991) asparagine (Asn) at position 297 (see WO2005 / 035586, WO2002 / 31140, WO00 / 61739), each of which is cited in full Is incorporated into this article. In addition, it is known in the art that ADCC and / or CDC can be adjusted, for example, by modifying amino acid residues in the Fc region of an antibody. The effector activity of an antibody can be enhanced or reduced by controlling the amount of nucleated trehalose in the complex N-glycoside-linked sugar chain bound to the Fc region of the antibody. To reduce the trehalose content of the complex N-glycoside-linked sugar chain bound to the Fc of the antibody, CHO cells lacking the α1,6-trehalosyltransferase gene (FUT8) can be used to express the antibody and An antibody that did not bind trehalose was obtained. Antibodies without trehalose have higher ADCC activity. To increase the trehalose content of the complex N-glycoside-linked sugar chain bound to the Fc of the antibody, trehalose can be obtained by expressing the antibody by using a host expressing the α1,6-trehalosyl transferase gene Of antibodies. Antibodies with trehalose have lower ADCC activity than antibodies without trehalose. In one embodiment, the anti-CCR4 antibody used in the present invention is an antibody having a trehalosylated N-glycoside-linked sugar chain bound to the Fc region of the antibody. In certain embodiments, the anti-CCR4 antibody used in the present invention is an antibody having a trehalosylated N-glycoside-linked sugar chain bound to the Fc region of the antibody. In some embodiments, about 50% or Bigger. In other embodiments, the antibody has a trehalosylation range of about 80%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99% or greater. In addition, by modifying amino acid residues in the Fc region of the antibody, ADCC activity or CDC activity can be increased or decreased. Amino acid residues in the Fc region are modified to increase or decrease the binding activity for FcγR, thereby controlling ADCC activity. Amino acid residues in the Fc region are modified to increase or decrease the binding activity of complement, thereby controlling CDC activity. For example, the CDC activity of an antibody can be increased by using the amino acid sequence of the Fc region described in the specification of US2007 / 0148165. In addition, ADCC activity or CDC activity can be modified as described in the specifications of US6,737,056, US7,297,775, US7,317,091, WO2005 / 070963 and Oganesyan et al. (Biol. Crystal., 2008; 64; 700-704) Increase or decrease the amino acid residue. In some embodiments, the anti-PD-1 antibody has no effector function, and only neutralizes PD-1 function by PD-L1 or PD-L2 binding. In certain embodiments, anti-PD1 antibodies suitable for use in the present invention may be IgG2 or IgG4 subclasses that exhibit reduced effector function or no effector function. In one embodiment, the anti-PD-1 antibody used in the present invention may have an Fc variant with reduced effect function or no effect function. As used herein, the term "individual" includes any human or non-human animal. The term "non-human animals" includes all vertebrates, such as mammals and non-mammals, such as non-human primates, sheep, dogs, cats, horses, cattle, chickens, amphibians, reptiles, and the like. Unless otherwise indicated, the terms "patient" or "individual" are used interchangeably herein. The term "leveling dose" or "leveling amount" used in relation to the composition of the present invention means the dose administered to the patient regardless of the patient's weight or body surface area (BSA). Therefore, the flattened dose is not provided as a mg / kg dose, but as an absolute amount of the agent (eg, anti-CCR4 antibody and / or anti-PD-1 antibody). For example, a person of 60 kg and a person of 100 kg will receive the same dose of the composition (eg 240 mg of anti-PD1 antibody and 80 mg of anti-CCR4 antibody). As mentioned herein, the term "body weight-based dose" means that the dose administered to the patient is calculated based on the weight of the patient. For example, when a patient with a weight of 60 kg requires 3 mg / kg of anti-PD-1 antibody and 1 mg / kg of anti-CCR4 antibody, we can take an appropriate amount of anti-PD-1 antibody (ie, 180 mg ) And anti-CCR4 antibody (ie, 60 mg). Various aspects of the invention are described in further detail in the following subsections.CCR4 CCR4 is a member of the seven transmembrane G-type protein-coupled receptor families of lymphocyte chemokine receptors that act as two chemokines, TARC (CCL17) and MDC (CCL22). CCR4 performance includes CD4+ CD4 of regulatory T cells (Treg)+ Th2 cells. In addition, some cancers including adult T-cell leukemia lymphoma (ATL), cutaneous T-cell lymphoma (CTCL), peripheral T-cell lymphoma (PTCL) and several solid-state tumors have been demonstrated to express CCR4. Ca induced by CCR4 bound ligand2 + Flow into CCR4+ On cells and induce cell proliferation or cell migration towards ligand secretory tissues. CD4+ Th2 cells are regulatory cells in humoral immunity. When Th2 cells promote antibody production to additional antigens by B cells, Th2 cells also inhibit Th1 cell function. Th2 cells act as helper T cells by producing immunosuppressive interleukins, such as interleukin (IL) -10, thereby suppressing cellular immunity. CD4, CD25, CTLA-4 and GITR (glucocorticoid-induced tumor necrosis factor receptor family related genes) are known Treg markers (Journal of Allergy and Clinical Immunology, 110: 693-701, 2002). In addition, FoxP3 (forkhead box protein 3) transcription factor is a major gene involved in the differentiation and functional performance of regulatory T cells (Science, 299: 1057-61, 2003). In addition, CCR4 is also expressed on Treg cells (US Publication No. 2006/0034841). Treg cells are one of a group of T cells that inhibit the activation of autoreactive T cells and are responsible for immune self-tolerance, preventing the immune system from attacking self tissues in healthy individuals (Immunological Review, 182: the whole volume, 2001) . Treg cells secrete inhibitory chemokines, interleukin-10 (IL-10), and transforming growth factor (TGF) -β via direct and / or indirect pathways, such as cell-cell interactions, to deplete T cell stimulation Sex chemokine and IL-2 to suppress immune stimulation. Exemplary cells expressing CCR4 targeted in the methods provided herein are selected from CCR4+ Immune cells and CCR4+ Cells in tumor cells. In certain embodiments, CCR4+ The cells are selected from the group consisting of: CCR4+ T cells, CCR4+ Th2 cells, CD4+ CD25+ CCR4+ T cells, CCR4+ Foxp3+ T cells, CD4+ CD25+ CCR4+ Foxp3+ T cells, CD4+ CD25+ CCR4+ CD127low T cells and CD4+ CD25+ CD45RA- CCR4+ Foxp3+ T cells (called effector Tregs) (Miyara et al., Immunity, 2009; 30: 899-911), and "Treg cells" are defined as regulatory T cells. In other embodiments, CCR4+ Treg is defined by at least one flag setting selected from: CD4+ CD25+ CCR4+ , CD25+ Foxp3+ , CCR4+ Foxp3+ , CD4+ CD25+ CCR4+ Foxp3+ , CD4+ CD25+ CCR4+ CD127low And CD4+ CD25+ CD45RA- CCR4+ Foxp3+ Can be illustrative.PD-1 T cell activation and deactivation in the human body are complexly regulated by multiple membrane proteins simultaneously. One of these membrane proteins, PD-1 was cloned as a type I membrane protein of 55 kDa (Ishida et al., EMBO J., 1992; 11; 3887-3895). PD-1 is expressed on multiple cell types activated T cells, regulatory T cells (Treg), activated B cells and natural killer (NK) cells and PD-1 is known to bind to its ligand, PD-L1 And PD-L2 to regulate T cell activation. The intracellular domain of PD-1 contains ITSM motifs and ITIM motifs that can be inhibitory domains of immune responses. PD-1 bound by PD-L1 induces inhibitory signals in T cells or other cells expressing PD-1, thereby blocking the activation, cell proliferation and / or inflammatory cells of T cells or other cells expressing PD-1 Interleukin releases or disables T cells. On the other hand, PD-L1 (human PD-L1 cDNA, AF233516) and PD-L2 (human PD-L2 NM_025239) are PD-1 ligands. PD-L1 is expressed on multiple cells, including antigen-presenting cells such as activated mononuclear cells and dendritic cells (Freeman et al., Journal of Experimental Medicine (2000), 19; 1027-1034). PD-L1 expression and PD-L2 expression were confirmed not only in normal cells but also in several cancers. In the present invention, PD-1 expression on normal cells and PD-L1 expression on cancer tissues or cancer cells can be detected by known analysis such as flow cytometry, immunohistochemistry (IHC). "PD-1 positive" means at least about 0.1%, at least about 1%, at least about 5%, at least about 10%, or at least about 20% or more stained cells, tissues, and body fluids of a human individual. As used herein, "PD-L1 positive" or "PD-L2 positive" can be used interchangeably with "at least about 1% of PD-L1 and / or PD-L2 performance." In one embodiment, the PD-L1 performance and / or PD-L2 performance can be used by any method known in the art. In another embodiment, PD-L1 performance and / or PD-L2 performance is measured by automatic in situ hybridization (IHC). PD-L1 and / or PD-L2 positive tumors may therefore have at least about 1%, at least about 2%, at least about 5%, at least about 10%, at least about 20%, at least about 1% as measured by automated IHC 30%, at least about 40%, or at least about 50% of tumor cells expressing PD-L1.anti- CCR4 antibody Anti-CCR4 antibodies suitable for the present invention can specifically bind to human CCR4. In one embodiment, the anti-CCR4 antibody can attenuate, inhibit, or prevent at least one function of the CCR4 protein. In one embodiment, the anti-CCR4 antibody reduces or depletes CCR4+ Immunosuppressive cells and / or CCR4+ cancer cell. In another embodiment, the anti-CCR4 antibody binds to CCR4 expressed on immunosuppressive cells or cancer cells, and reduces or depletes the number of cells by the following antibody effector functions: for example, induced by major natural killer (NK) cells Antibody-dependent cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC) or antibody-dependent phagocytosis (ADP) by phagocytic cells such as macrophages or dendritic cells and / or neutralizing CCR4 coordination body. In one embodiment, the anti-CCR4 antibody used in this method binds to the extracellular region of the CCR4 molecule and exhibits ADCC activity. In another embodiment, the anti-CCR4 antibody binds to the epitope of amino acids 2 to 29 of the CCR4 protein (eg, amino acids 2 to 29 corresponding to SEQ ID NO: 22) and exhibits ADCC activity. In other embodiments, the anti-CCR4 antibody binds to the epitope in amino acids 12 to 29 of the CCR4 protein (eg, amino acids 12 to 29 of SEQ ID NO: 22) and exhibits ADCC activity. In other embodiments, the anti-CCR4 antibody cross-competes with an antibody comprising the following to bind to CCR4: VH CDR1 comprising the sequence set forth in SEQ ID NO: 1, VH CDR2 comprising the sequence set forth in SEQ ID NO: 2 , A VH CDR3 comprising the sequence set forth in SEQ ID NO: 3, a VL CDR comprising the sequence set forth in SEQ ID NO: 4, a VL CDR2 containing the sequence set forth in SEQ ID NO: 5, and a VL CDR2 containing the sequence set forth in SEQ ID NO: 5. : VL CDR3 of the sequence described in 6. In one embodiment, the anti-CCR4 antibody or antigen-binding portion thereof used in the present invention binds to the same epitope bound by an antibody or moglizumab comprising the following: comprising SEQ ID NO: 1 VH CDR1 of the sequence set forth, VH CDR2 of the sequence set forth in SEQ ID NO: 2, VH CDR3 of the sequence set forth in SEQ ID NO: 3, VL of the sequence set forth in SEQ ID NO: 4 CDR1, VL CDR2 containing the sequence set forth in SEQ ID NO: 5 and VL CDR3 containing the sequence set forth in SEQ ID NO: 6. In another embodiment, an anti-CCR4 antibody suitable for use in the present invention comprises: VH CDR1 comprising the sequence set forth in SEQ ID NO: 1, VH CDR2 comprising the sequence set forth in SEQ ID NO: 2, and comprising SEQ ID One or more of the VH CDR3 of the sequence set forth in NO: 3, and the VL CDR1 containing the sequence set forth in SEQ ID NO: 4, the VL CDR2 containing the sequence set forth in SEQ ID NO: 5 and containing One or more of the VL CDR3 of the sequence set forth in SEQ ID NO: 6. In other embodiments, the anti-CCR4 antibody comprises: VH CDR1 comprising the sequence set forth in SEQ ID NO: 1, VH CDR2 comprising the sequence set forth in SEQ ID NO: 2, comprising SEQ ID NO: 3 VH CDR3 of the sequence, VL CDR1 containing the sequence set forth in SEQ ID NO: 4, VL CDR2 containing the sequence set forth in SEQ ID NO: 5, and VL CDR3 containing the sequence set forth in SEQ ID NO: 6 . In some embodiments, the anti-CCR4 antibody comprises: VH comprising the sequence set forth in SEQ ID NO: 7 and VL comprising the sequence set forth in SEQ ID NO: 8. In certain embodiments, the anti-CCR4 antibody comprises: a heavy chain comprising the sequence set forth in SEQ ID NO: 9 and a light chain comprising the sequence set forth in SEQ ID NO: 10. In other embodiments, the Fc region of the anti-CCR4 antibody comprises nucleated trehalose that does not bind to N-acetylglucosamine at position 297 of the Fc region. In a specific embodiment, the anti-CCR4 antibody is moglizumab (POTELIGEO (registered trademark)). In certain embodiments, the anti-CCR4 antibody used in the method or composition has a lower trehalosylation, detrehalosylation, or no trehalosylation of N-glycoside linkages bound to the Fc region of the antibody Sugar chain. Antibodies with lower trehalosylation, de-trehalosylation, or no trehalosylation may have higher ADCC activity than trehalosylated antibodies. Therefore, in one embodiment, the anti-CCR4 antibody used in the method or in the composition of the present invention can be produced by cells having low or no alpha 1,6-trehalosyltransferase (FUT8) activity. In another embodiment, the anti-CCR4 antibody used in the method or composition of the present invention is substantially free of nucleated trehalose, which is linked to the sugar chain by the N-glycoside linkage of Asn 297 in the Fc region The α1,6 bond is bound to N-acetylglucosamine. In certain embodiments, the method further comprises identifying CCR4 performance prior to administration of the anti-CCR4 antibody. In other embodiments, CCR4 infiltrating the tumor tissue of the patient and / or tissue effluents such as the abdomen and pleura can be monitored before, during, and / or after treatment+ T cell or CCR4+ Treg cell population, CCR4+ T cell or CCR4+ The degree of reduction / consumption of Treg. In addition, CCR4 performance can also be analyzed on cancer cells themselves by any method known in the art.anti- PD-1 antibody In one embodiment, an antibody or antigen-binding portion thereof ("anti-PD-1 antibody") that specifically binds to human PD-1 and inhibits hPD-1 activity is used in the method of the present invention. In another embodiment, the anti-PD-1 antibody or antigen-binding portion thereof blocks, inhibits, prevents or neutralizes PD-1 expression on cells or PD-1 related tumor immune response function. The anti-PD-1 antibody used in the method of the present invention binds to PD-1 with higher specificity and affinity, blocks the binding of PD-L1, and / or suppresses the immunosuppressive effect of the PD-1 signaling pathway. In any of the treatment methods or composition uses disclosed herein, anti-PD-1 antibodies include antigen binding that binds to PD-1 and exhibits ligand-like binding inhibition and upregulates the functional properties of the immune system similar to whole antibodies Fragment. In some embodiments, the anti-PD-1 antibody of the invention comprises: VH CDR1 comprising the sequence set forth in SEQ ID NO: 11, VH CDR2 comprising the sequence set forth in SEQ ID NO: 12, and SEQ ID NO : One or more of the VH CDR3 of the sequence set forth in 13 and the VL CDR1 containing the sequence set forth in SEQ ID NO: 14, the VL CDR2 containing the sequence set forth in SEQ ID NO: 15, and the VL CDR3 containing the sequence set forth in SEQ ID NO: 15 One or more of the VL CDR3 of the sequence set forth in ID NO: 16. In certain embodiments, the anti-PD-1 antibody comprises: VH CDR1 comprising the sequence set forth in SEQ ID NO: 11, VH CDR2 comprising the sequence set forth in SEQ ID NO: 12, and SEQ ID NO: The sequence VH CDR3 set forth in 13, the VL CDR 1 containing the sequence set forth in SEQ ID NO: 14, the VL CDR2 containing the sequence set forth in SEQ ID NO: 15, and the sequence set forth in SEQ ID NO: 16. VL CDR3. In other embodiments, the anti-PD-1 antibody comprises: VH comprising the sequence set forth in SEQ ID NO: 17 and VL comprising the sequence set forth in SEQ ID NO: 18. In still other embodiments, the anti-PD-1 antibody comprises: a heavy chain comprising the sequence set forth in SEQ ID NO: 19 and a light chain comprising the sequence set forth in SEQ ID NO: 20. In still other embodiments, the anti-PD-1 antibody is IgG2, IgG4, or a variant thereof that has effect-reducing activity. In certain embodiments, the anti-PD-1 antibody or antigen-binding portion thereof competes with nivolumab to bind to human PD-1 and the anti-PD-1 antibody or antigen-binding fragment such as nivolumab binds to human The same antigenic determinant region of PD-1. In other embodiments, the anti-PD-1 monoclonal antibody or antigen-binding fragment thereof is a chimeric, humanized, or human monoclonal antibody or fragment thereof. In certain embodiments of treating human subjects, the monoclonal antibody is a humanized antibody. In other embodiments for treating human subjects, the antibody is a human antibody. In other embodiments, antibodies of the IgG1, IgG2, IgG3, or IgG4 isotype or antibody Fc variants thereof can be used in the methods and compositions. In certain embodiments, the anti-PD-1 antibody or antigen-binding portion thereof comprises a heavy chain constant region belonging to a human IgG1 or IgG4 isotype or any applicable IgG variant. In other embodiments, the sequence of the IgG4 heavy chain constant region of the anti-PD-1 antibody or antigen-binding portion thereof contains the S228P mutation, which replaces the hinge with a proline residue normally found at the corresponding position in the IgG1 isotype antibody Serine residues in the region. This mutation present in nivolumab prevents the exchange of Fab arms with endogenous IgG4 antibodies, while maintaining a lower affinity for activation of Fc receptors associated with wild-type IgG4 antibodies (Wang et al., 2014 Cancer Immunol Res. 2 (9): 846-56). In still other embodiments, the antibody comprises a light chain constant region that is a human kappa or lambda constant region. Humanized antibodies or human antibodies (HuMab) that specifically bind to PD-1 with higher affinity have been disclosed in US Patent No. 8,008,449 or 8,779,105. Other anti-PD-1 monoclonal antibodies have been described in, for example, US Patent Nos. 6,808,710, 7,488,802, 8,168,757, and 8,354,509, and PCT Publication No. WO 2012/145493. Each of the anti-PD-1 HuMAbs disclosed in US Patent No. 8,008,449 has been shown to exhibit one or more of the following characteristics: (a) combined with 1 × 10-7 M or less than 1 × 10- 7 K of MD Human PD-1, as determined by surface plasmon resonance using the Biacore biosensor system; (b) is not substantially bound to human CD28, CTLA-4, or ICOS; (c) increases mixed lymph T cell proliferation in MLR analysis; (d) Increase interferon gamma production in MLR analysis; (e) Increase IL-2 secretion in MLR analysis; (f) Binding to human PD-1 and Crab-eating macaque PD-1; (g) inhibit PD-L1 and / or PD-L2 binding to PD-1; (h) stimulate antigen-specific memory response; (i) stimulate antibody response; and (j) inhibit activity Tumor cells grow in the body. Anti-PD-1 antibodies useful in the present invention include monoclonal antibodies that specifically bind to human PD-1 and exhibit at least one of the aforementioned characteristics (in some embodiments, at least five). In some embodiments, the anti-PD-1 antibody is nivolumab. In one embodiment, the anti-PD-1 antibody is peclizumab. In one embodiment, the anti-PD-1 antibody is nivolumab. Nivolumab (also known as "OPDIVO (registered trademark)"; previously specifically referred to as 5C4, BMS-936558, MDX-1106 or ONO-4538) is a selective protection against PD-1 ligands (PD-L1 and PD-L2) interaction, thereby blocking the all-human IgG4 (S228P) PD-1 immune checkpoint inhibitory antibody that down-regulates anti-tumor T cell function (US Patent No. 8,008,449; Wang et al., 2014 Cancer Immunol Res. 2 ( 9): 846-56). In some embodiments, the anti-PD-1 antibody cross-competes or binds to the same epitope as Pelicuzumab. In a specific embodiment, the anti-PD-1 antibody is nivolumab. Palivizumab (also known as "KEYTRUDA (registered trademark)", ralizumab and MK-3475) is a human cell surface receptor PD-1 (programmed death-1 or programmed cell death-1) ) Of humanized single IgG4 antibody. Palivizumab is described in, for example, US Patent Nos. 8,354,509 and 8,900,587; see also http://www.cancer.gov/drugdictionary?cdrid=695789 (last visited: December 14, 2014). Palivizumab has been approved by the FDA for the treatment of relapsed or refractory melanoma. In other embodiments, the anti-PD-1 antibody cross-competes or binds to the same epitope as MEDI0608 (formerly AMP-514) which is a monoclonal antibody. In yet other embodiments, the anti-PD-1 antibody is MEDI0608. MEDI0608 is described in, for example, US Patent No. 8,609,089B2 or http://www.cancer.gov/drugdictionary?cdrid=756047 (last visited: December 14, 2014). Anti-PD-1 antibodies that can be used in the disclosed methods or disclosed compositions also include isolated antibodies that specifically bind to human PD-1 and cross-compete with nivolumab to bind to human PD-1 (see also , For example, US Patent Nos. 8,008,449 and 8,779,105; WO 2013/173223). The ability of antibodies to cross-compete to bind to an antigen may indicate that these antibodies bind to the same epitope of the antigen and sterically hinder the binding of other cross-competing antibodies to specific epitopes. By virtue of binding to the same epitope region of PD-1, these cross-competing antibodies are expected to have very similar functional properties to nivolumab. The cross-competitive antibody can be easily identified based on its ability to cross-compete with nivolumab in standard PD-1 binding analysis, such as Biacore analysis, ELISA assay, or flow cytometry (see, for example, WO 2013 / 173223). In certain embodiments, the anti-PD-1 antibody is nivolumab (OPDIVO (registered trademark)). In other embodiments, the anti-PD-1 antibody is peclizumab (KEYTRUDA (registered trademark)). In other embodiments, the anti-PD-1 antibody is selected from human antibody 17D8, human antibody 2D3, human antibody 4H1, human antibody 4A11, human antibody 7D3, and human antibody 5F4 described in US Patent No. 8,008,449. In still other embodiments, the anti-PD-1 antibody is MEDI0608 (previously AMP-514) or AMP-224. In certain embodiments, an anti-PD-1 antibody used in the method or composition that specifically binds to PD-1 and inhibits PD-1 activity (eg, binds to PD-L1 and / or PD-L2) It is suitable for the treatment of cancer, such as lung cancer (eg non-small cell lung cancer), esophageal cancer or gastric cancer. In one embodiment, the anti-PD-1 antibody used in the method or composition of the present invention includes: (i) an antibody that cross-competes with the antibody to bind to human PD-1, the antibody includes: comprising SEQ ID NO : One or more of the heavy chain CDR1 of the sequence set forth in 11, the heavy chain CDR2 of the sequence set forth in SEQ ID NO: 12, and the heavy chain CDR3 of the sequence set forth in SEQ ID NO: 13, And the light chain CDR1 comprising the sequence set forth in SEQ ID NO: 14, the light chain CDR2 comprising the sequence set forth in SEQ ID NO: 15, and the light chain CDR3 comprising the sequence set forth in SEQ ID NO: 16. One or more; (ii) an antibody that binds to the same epitope region as the epitope region in the antibody, the antibody comprising: a heavy chain CDR1 comprising the sequence set forth in SEQ ID NO: 11, comprising SEQ The heavy chain CDR2 of the sequence set forth in ID NO: 12, the heavy chain CDR3 containing the sequence set forth in SEQ ID NO: 13, the light chain CDR1 containing the sequence set forth in SEQ ID NO: 14, including SEQ ID NO : Light chain CDR2 of the sequence set forth in 15 and light chain CDR3 containing the sequence set forth in SEQ ID NO: 16; (iii) antibody, which Including: heavy chain CDR1 containing the sequence set forth in SEQ ID NO: 11, heavy chain CDR2 containing the sequence set forth in SEQ ID NO: 12, heavy chain CDR3 containing the sequence set forth in SEQ ID NO: 13, A light chain CDR1 comprising the sequence set forth in SEQ ID NO: 14, a light chain CDR2 comprising the sequence set forth in SEQ ID NO: 15, and a light chain CDR3 containing the sequence set forth in SEQ ID NO: 16; ( iv) an antibody comprising: VH comprising the sequence set forth in SEQ ID NO: 17 and a VL comprising the sequence set forth in SEQ ID NO: 18; or (v) an antibody comprising: comprising SEQ ID NO: 19 The heavy chain of the sequence set forth in and the light chain comprising the sequence set forth in SEQ ID NO: 20. The anti-PD-1 antibody used in the present invention can exert a neutralizing or blocking activity on PD-1; that is, the anti-PD-1 antibody can block PD-1 activity through PD-L1 binding and inhibit T cell proliferation , Interleukin release and any combination thereof. In one embodiment, the anti-PD-1 antibodies ONO-4538, BMS-936558, nivolumab / OPDIVO (registered trademark) specifically bind to PD-1 and block PD-1 from binding to PD-L1 and PD -Exemplary anti-PD-1 antibody for L2. Nivolumab can alleviate PD-1 mediated inhibition of human T cell activation in vitro and inhibit tumor growth in xenograft models via T cell dependent mechanisms.( i ) anti- CCR4 Antibodies and ( ii ) anti- PD - 1 Antibody combination therapy The present invention relates to a method of treating cancer, the method comprising administering a CCR4 antagonist and a PD-1 antagonist, wherein the CCR4 antagonist comprises binding to CCR4, interfering with CCR4 or otherwise blocking the binding of CCR4 to its ligand ( For example, CCL17 and / or CCL22) any smaller or larger molecules (eg, anti-CCR4 antibodies), and the PD-1 antagonist includes binding to PD-1, interfering with PD-1, or otherwise blocking PD- 1 Any smaller or larger molecule (eg, anti-PD-1 antibody) that binds to its ligand (eg, PD-L1). In one embodiment, the invention includes a method of treating at least one cancer in an individual in need thereof, the method comprising administering an anti-CCR4 antibody and an anti-PD-1 antibody. In another embodiment, the administration schedule of the method includes the administration schedule for cancer patients, including administration of anti-CCR4 antibody and anti-PD-1 antibody. In certain embodiments, the anti-CCR4 antibody and anti-PD-1 anti-system are administered in an effective amount to treat cancer. Also provided herein is a method of reducing the size of a tumor in an individual with cancer by at least about 1%, 5%, 10%, 15%, 20%, or 30%, the method comprising (i) an anti-CCR4 antibody or The antigen-binding portion and (ii) the anti-PD-1 antibody or antigen-binding portion thereof are administered to the individual. In other embodiments, the combination method of the present invention enhances anti-tumor efficacy compared to anti-CCR4 antibody or anti-PD-1 antibody monotherapy. In certain embodiments, the anti-CCR4 antibody and the anti-PD-1 antibody are administered in an effective amount to reduce the tumor size of the cancer. The combination methods of the present invention can suppress the anti-tumor immune response by reducing or depleting CCR4+ Treg cells can bring one or more beneficial effects on anti-tumor immunotherapy of cancer patients; therefore, anti-CCR4 antibodies counteract CCR4+ The immunosuppressive effect of Treg cells causes and enhances the anti-tumor immune response. At the same time, anti-PD-1 antibodies block or neutralize T cells, and PD-1 is a negative regulator of cytotoxic T cells that attack tumor cells in the tumor microenvironment; therefore, anti-PD-1 antibodies counteract negative signals against T cells And cause anti-tumor immune response. The dose of anti-CCR4 antibody used in this method includes any dose of about 0.1 mg / kg to about 10.0 mg / kg per administration. In one aspect, the dose of anti-CCR4 antibody used in the method includes at least about 0.1 mg / kg, at least about 0.2 mg / kg, at least about 0.3 mg / kg, at least about 0.4 mg / kg, at least about 0.5 mg / kg, at least about 0.6 mg / kg, at least about 0.7 mg / kg, at least about 0.8 mg / kg, at least about 0.9 mg / kg, at least about 1.0 mg / kg, at least about 2.0 mg / kg, and at least about 3.0 mg / kg. The dose of anti-PD-1 antibody used in the method includes any dose of about 1.0 mg / kg to about 10.0 mg / kg per administration. In one aspect, the dose of anti-PD-1 antibody used in the method includes at least about 1 mg / kg, at least about 2 mg / kg, at least about 3 mg / kg, at least about 4 mg / kg, at least about 5 mg / kg, at least about 6 mg / kg, at least about 7 mg / kg, at least about 8 mg / kg, at least about 9 mg / kg, or at least about 10 mg / kg. In other embodiments, the dose of anti-PD-1 antibody is about 2.0 mg / kg. In a specific embodiment, the dose of anti-PD-1 antibody is about 3.0 mg / kg. In a specific embodiment, the dose of anti-PD-1 antibody is about 240 mg / body as a flat dose. In some embodiments, the dose of anti-PD-1 antibody is at least about at least about 100 mg / body, 120 mg / body, 140 mg / body, 160 mg / body, 180 mg / body, 200 mg / body, 220 mg / Body, 240 mg / body, 260 mg / body, 280 mg / body, 300 mg / body, 400 mg / body, 420 mg / body, 440 mg / body, 460 mg / body, 480 mg / body, 500 mg / Body, 520 mg / body, 540 mg / body, 560 mg / body or 600 mg / body. In some embodiments, the dose of anti-PD-1 antibody is 480 mg per body dose. The dosing cycle of antibodies used in this method includes any suitable dosing cycle for each antibody. The dosing cycle is defined as about once a week (Q1W), about once every two weeks (Q2W), about once every three weeks (Q3W) or about once every four weeks or about once a month (Q4W). The administration cycle of the anti-CCR4 antibody used in this method includes once a week (Q1W) for four to eight weeks, once a week (Q1W) for four weeks, and once every two weeks (Q2W). In one aspect, the anti-CCR4 antibody is administered once a week for four weeks (induction phase) and once every two weeks (maintenance phase) after the first administration. The administration cycle of the anti-PD-1 antibody used in this method includes once every two weeks, once every three weeks, or once every four weeks. In another embodiment, the method of the invention includes a method of treating at least one cancer in an individual, the method comprising administering about 0.1 mg about once a week for four weeks after the first administration and then about once every two weeks / kg, about 0.3 mg / kg, about 0.5 mg / kg, about 0.75 mg / kg, about 1.0 mg / kg, about 2.0 mg / kg, or about 3.0 mg / kg of anti-CCR4 antibody, and about every two weeks Administer about 2.0 mg / kg, or about 3.0 mg / kg, or at least about 100 mg / body, 120 mg / body, 140 mg / body, 160 mg / body, 180 mg once, once every three weeks, or once every four weeks / Body, 200 mg / body, 220 mg / body, 240 mg / body, 260 mg / body, 280 mg / body, 300 mg / body, 400 mg / body, 420 mg / body, 440 mg / body, 460 mg / Body, 480 mg / body, 500 mg / body, 520 mg / body, 540 mg / body, 560 mg / body, 580 mg / body or 600 mg / body as a flat-dose combination of anti-PD-1 antibodies. In other embodiments, the method of the invention includes treating at least one cancer in an individual, the method comprising administering; (i) about once a week for four weeks after the first administration and then about 0.1 every about once every two weeks mg / kg, about 0.3 mg / kg, about 0.5 mg / kg, about 0.75 mg / kg, about 1.0 mg / kg, about 2.0 mg / kg or about 3.0 mg / kg of anti-CCR4 antibody, and about every two weeks Administer about 2.0 mg / kg of anti-PD-1 antibody combination at one time; (ii) about 0.1 mg / kg, about 0.3 mg / kg about once a week for four weeks after the first administration and then about once every two weeks , About 0.75 mg / kg, about 1.0 mg / kg or about 3.0 mg / kg of anti-CCR4 antibody, and in combination with about 3.0 mg / kg of anti-PD-1 antibody administered about once every two weeks; (iii) About 0.1 mg / kg, about 0.3 mg / kg, about 0.5 mg / kg, about 0.75 mg / kg, about 1.0 mg / kg, about 2.0 after one dose, about once a week for four weeks and then about once every two weeks mg / kg or about 3.0 mg / kg of anti-CCR4 antibody, and at least about 100 mg / body, 120 mg / body, 140 mg / body, 160 mg / body, 180 mg / body, and about once every two weeks 200 mg / body, 220 mg / Body, 240 mg / body, 260 mg / body, 280 mg / body, 300 mg / body, 400 mg / body, 420 mg / body, 440 mg / body, 460 mg / body, 480 mg / body, 500 mg / Body, 520 mg / body, 540 mg / body, 560 mg / body, 580 mg / body or 600 mg / body as a flat-dose combination of anti-PD-1 antibodies; (iv) about one week after the first dose Anti-CCR4 antibody of about 0.1 mg / kg, about 0.3 mg / kg, about 0.75 mg / kg, about 1.0 mg / kg, about 2.0 mg / kg or about 3.0 mg / kg once for four weeks and then about every two weeks , And in combination with about 2.0 mg / kg of anti-PD-1 antibody administered about once every four weeks; (v) about 0.1 mg / kg about once a week for four weeks after the first administration and then about once every two weeks , About 0.3 mg / kg, about 0.5 mg / kg, about 0.75 mg / kg, about 1.0 mg / kg or about 3.0 mg / kg of anti-CCR4 antibody, and about 3.0 mg / kg of anti-c PD-1 antibody combination or (vi) about 0.1 mg / kg, about 0.3 mg / kg, about 0.75 mg / kg, about 1.0 after the first administration, about once a week for four weeks and then about once every four weeks mg / kg, about 2.0 mg / kg or about 3.0 mg / kg of anti-CCR4 antibody, and administered at least about 100 mg / body, 120 mg / body, 140 mg / body, 160 mg / body once every two weeks , 180 mg / body, 200 mg / body, 220 mg / body, 240 mg / body, 260 mg / body, 280 mg / body, 300 mg / body, 400 mg / body, 420 mg / body, 440 mg / body , 460 mg / body, 480 mg / body, 500 mg / body, 520 mg / body, 540 mg / body, 560 mg / body, 580 mg / body or 600 mg / body as a flat-dose anti-PD-1 antibody combination. In one embodiment, the method of the present invention includes a method of treating at least one cancer in an individual, the method comprising administering about 0.1 mg / day about once a week for four weeks after the first administration and then about once every two weeks kg, about 0.3 mg / kg, about 0.5 mg / kg, about 0.75 mg / kg, about 1.0 mg / kg, about 2.0 mg / kg or about 3.0 mg / kg of anti-CCR4 antibody, and about once every two weeks With about 2.0 mg / kg or about 3.0 mg / kg, or about 240 mg / body as an adjusted dose of anti-PD-1 antibody, or about once every four weeks about 480 mg / body as an adjusted dose of anti-PD-1 antibody Combination, wherein the anti-CCR4 antibody competitively binds to CCR4 with an antibody comprising: VH CDR1 comprising the sequence set forth in SEQ ID NO: 1, VH CDR2 comprising the sequence set forth in SEQ ID NO: 2, comprising VH CDR3 of the sequence set forth in SEQ ID NO: 3, VL CDR1 containing the sequence set forth in SEQ ID NO: 4, VL CDR2 containing the sequence set forth in SEQ ID NO: 5, and containing SEQ ID NO: VL CDR3 of the sequence set forth in 6 and wherein the anti-PD-1 antibody competitively binds to PD-1 with an antibody comprising: comprising SEQ ID NO: 11 VH CDR1 of the sequence set forth, VH CDR2 of the sequence set forth in SEQ ID NO: 12, VH CDR3 of the sequence set forth in SEQ ID NO: 13, of the sequence set forth in SEQ ID NO: 14 VL CDR1, VL CDR2 comprising the sequence set forth in SEQ ID NO: 15 and VL CDR3 comprising the sequence set forth in SEQ ID NO: 16. In one embodiment, the method of the present invention includes a method of treating a cancer patient, the method comprising about once a week for four weeks and then about once every two weeks about 0.1 mg / kg, about 0.3 mg / kg, about 0.5 mg / kg, about 0.75 mg / kg, about 1.0 mg / kg, about 2.0 mg / kg or about 3.0 mg / kg of anti-CCR4 antibody, and about 2.0 mg / kg or about 3.0 mg / about every two weeks kg, or about 240 mg / body as an adjusted dose of anti-PD-1 antibody, or about every four weeks about 480 mg / body as an adjusted dose of anti-D-1 antibody combination, wherein the anti-CCR4 antibody binds to the antibody The same epitope, the antibody contains: VH CDR1 containing the sequence set forth in SEQ ID NO: 1, VH CDR2 containing the sequence set forth in SEQ ID NO: 2, including the set forth in SEQ ID NO: 3 VH CDR3 of the sequence, VL CDR including the sequence set forth in SEQ ID NO: 4, VL CDR2 including the sequence set forth in SEQ ID NO: 5, and VL CDR3 containing the sequence set forth in SEQ ID NO: 6, And where the anti-PD-1 antibody binds to the same epitope as the antibody, the antibody includes: comprising the sequence set forth in SEQ ID NO: 11 VH CDR1, VH CDR2 containing the sequence set forth in SEQ ID NO: 12, VH CDR3 containing the sequence set forth in SEQ ID NO: 13, VL CDR1 containing the sequence set forth in SEQ ID NO: 14, including VL CDR2 of the sequence set forth in SEQ ID NO: 15 and VL CDR3 comprising the sequence set forth in SEQ ID NO: 16. In one embodiment, the method of the present invention includes a method of treating a cancer patient, the method comprising about once a week for four weeks and then about once every two weeks about 0.1 mg / kg, about 0.3 mg / kg, about 0.5 mg / kg, about 0.75 mg / kg, about 1.0 mg / kg, about 2.0 mg / kg or about 3.0 mg / kg of anti-CCR4 antibody, and about 2.0 mg / kg or about 3.0 mg administered about every two weeks / kg, or an anti-PD-1 antibody with an adjusted dose of about 240 mg / body, or an anti-PD-1 antibody combination with an adjusted dose of about 480 mg / body about once every four weeks, wherein the anti-CCR4 antibody Contains: VH CDR1 containing the sequence set forth in SEQ ID NO: 1, VH CDR2 containing the sequence set forth in SEQ ID NO: 2, VH CDR3 containing the sequence set forth in SEQ ID NO: 3, containing SEQ ID VL CDR1 of the sequence set forth in NO: 4, VL CDR2 containing the sequence set forth in SEQ ID NO: 5 and VL CDR3 containing the sequence set forth in SEQ ID NO: 6, and wherein the anti-PD-1 antibody Contains: VH CDR1 containing the sequence set forth in SEQ ID NO: 11, VH CDR2 containing the sequence set forth in SEQ ID NO: 12, including the set forth in SEQ ID NO: 13 Column of VH CDR3, comprising SEQ ID NO: VL CDR1 sequences 14 set forth, the comprising SEQ ID NO: VL sequences 15 set forth the CDR2 and comprising SEQ ID NO: VL CDR3 sequences 16 as set forth in the. In another embodiment, the method of the present invention includes a method of treating a cancer patient, the method comprising about once a week for four weeks and then about once every two weeks about 0.1 mg / kg, about 0.3 mg / kg, about 0.5 mg / kg, about 0.75 mg / kg, about 1.0 mg / kg, about 2.0 mg / kg or about 3.0 mg / kg of anti-CCR4 antibody (moglizumab), and about once every two weeks 2.0 mg / kg, or about 3.0 mg / kg, or about 240 mg / body as a flat dose of anti-PD-1 antibody (nivolumab) or about every four weeks, about 480 mg / body as a flat agent A combination of anti-PD-1 antibodies (Nivolumab). In the methods or compositions described herein, antibody combinations can be administered to cancer patients in parallel (or simultaneously) or sequentially. The term "in parallel" or "simultaneously" means that at least two antibodies are dissolved in a pharmaceutically acceptable aqueous liquid, such as physiological saline or Ringer's solution only or a mixture thereof, and both by intravenous line and / or infusion The bag is administered to the patient. The term "in order" means that one antibody is administered to cancer patients, and then the second antibody is administered to these cancer patients at a later time allowed after administration of the first antibody. In certain embodiments, the present disclosure includes a method of treating patients with HCC, wherein progression-free survival (PFS) is at least 100 days, at least 101 days, at least 102 days, at least 103 days, at least 104 days, At least 105 days, at least 106 days, at least 107 days, at least 108 days, at least 109 days, at least 110 days, at least 111 days, at least 112 days, at least 113 days, at least 114 days, at least 115 days, at least 116 days, at least 117 Days, at least 118 days, at least 119 days, at least 120 days, at least 125 days, at least 130 days, at least 135 days, at least 140 days, at least 145 days, at least 150 days, at least 155 days, at least 160 days, at least 165 days, At least 170 days, at least 175 days, at least 180 days, at least 185 days, at least 190 days, at least 195 days, at least 200 days, at least 205 days, at least 210 days, at least 215 days, or at least 220 days. In other embodiments, the PFS of the combination therapy of anti-CCR4 antibody and anti-PD-1 antibody is higher than that of anti-CCR4 antibody monotherapy and / or anti-PD-1 antibody monotherapy (higher than at least one week, at least two Week, at least three weeks, at least four weeks, at least one month, at least two months, at least three months, at least four months, at least five months, at least six months, at least seven months, at least eight months, at least nine Months, or at least ten months). In other embodiments, the present disclosure provides a method of treating patients with HCC, wherein the overall survival period is at least about 200 days, at least about 210 days, at least about 220 days, at least about 230 days, at least about 240 days, At least about 250 days, at least about 260 days, at least about 270 days, at least about 280 days, at least about 290 days, at least about 300 days, at least about 310 days, at least about 320 days, at least about 330 days, at least about 340 days, At least about 350 days, at least about 360 days, at least about 370 days, at least about 380 days, at least about 390 days, or at least about 400 days. In still other embodiments, the OS of the method is higher than the OS of anti-CCR4 antibody monotherapy and / or anti-PD-1 antibody monotherapy (higher than at least about one month, at least about two months, at least about three Month, at least about four months, at least about five months, at least about six months, at least about seven months, at least about eight months, at least about nine months, at least about ten months, at least about 11 months, At least about 12 months, at least about 15 months, at least about 20 months, at least about two years, at least about 25 months, at least about 30 months, at least about 35 months, at least about three years, at least about 40 Month, at least about 3.5 years, at least about 4 years, at least about 4.5 years, or at least about 5 years).Used to treat cancer CCR4 Antibodies and anti PD - 1 Antibody Kit The present invention also includes a kit for treating individuals suffering from cancer, the kit comprising: (a) a dose ranging from about 0.1 mg / kg to about 10 mg / kg body weight that specifically binds to PD-1 and / Or an antibody or antigen-binding portion thereof that inhibits PD-1 activity ("anti-PD-1 antibody or antigen-binding portion"); (b) a dose ranging from about 0.1 mg / kg to about 10 mg / kg body weight An antibody or antigen-binding portion thereof that specifically binds to CCR4 and / or inhibits CCR4 activity ("anti-CCR4 antibody or antigen-binding portion thereof"); and (c) use an anti-PD-1 antibody in any method disclosed herein and Instructions for anti-CCR4 antibody. The kit usually includes a label and instruction manual indicating the intended use of the contents of the kit. The term label includes any written or recorded material supplied on or with the kit or otherwise accompanying the kit. In other embodiments, the anti-CCR4 antibody or antigen-binding portion thereof is selected from the group consisting of: (i) an antibody or antigen-binding portion that binds to the same epitope as the antibody, which includes: comprising SEQ ID NO: 1 VH CDR1 of the sequence set forth in one or more of VH CDR2 including the sequence set forth in SEQ ID NO: 2 and VH CDR3 including the sequence set forth in SEQ ID NO: 3, and including SEQ ID NO : VL CDR1 of the sequence set forth in 4: one or more of VL CDR2 containing the sequence set forth in SEQ ID NO: 5 and VL CDR3 containing the sequence set forth in SEQ ID NO: 6; (ii ) Antibody or antigen-binding portion thereof, comprising: VH CDR1 comprising the sequence set forth in SEQ ID NO: 1, VH CDR2 comprising the sequence set forth in SEQ ID NO: 2, including the set forth in SEQ ID NO: 3 VH CDR3 of the sequence, VL CDR1 containing the sequence set forth in SEQ ID NO: 4, VL CDR2 containing the sequence set forth in SEQ ID NO: 5, and VL containing the sequence set forth in SEQ ID NO: 6 CDR3; (iii) antibody or antigen-binding portion thereof, comprising: VH comprising the sequence set forth in SEQ ID NO: 7 and comprising SEQ VL of the sequence set forth in ID NO: 8; (iv) an antibody comprising: a heavy chain comprising the sequence set forth in SEQ ID NO: 9 and a light chain comprising the sequence set forth in SEQ ID NO: 10; (v) an antibody or antigen-binding portion thereof that cross-competes with the antibody of (ii); and (vi) moglizumab or its antigen-binding portion. In certain embodiments, the anti-PD-1 antibody or antigen-binding portion thereof is selected from the group consisting of: (i) an antibody or antigen-binding portion that binds to the same epitope as the antibody, which comprises: comprising SEQ ID One or more of VH CDR1 of the sequence set forth in NO: 11, VH CDR2 containing the sequence set forth in SEQ ID NO: 12, and VH CDR3 containing the sequence set forth in SEQ ID NO: 13, and including One or more of VL CDR1 of the sequence set forth in SEQ ID NO: 14, VL CDR2 containing the sequence set forth in SEQ ID NO: 15, and VL CDR3 containing the sequence set forth in SEQ ID NO: 16; (ii) Antibody or antigen-binding portion thereof, comprising: VH CDR1 comprising the sequence set forth in SEQ ID NO: 11, VH CDR2 comprising the sequence set forth in SEQ ID NO: 12, comprising SEQ ID NO: 13 VH CDR3 of the sequence described, VL CDR1 of the sequence set forth in SEQ ID NO: 14, VL CDR2 of the sequence set forth in SEQ ID NO: 15, and the sequence of SEQ ID NO: 16 VL CDR3; (iii) antibody or antigen-binding portion thereof, comprising: comprising the sequence set forth in SEQ ID NO: 17 VH and VL comprising the sequence set forth in SEQ ID NO: 18; (iv) Antibodies comprising: a heavy chain comprising the sequence set forth in SEQ ID NO: 19 and comprising the sequence set forth in SEQ ID NO: 20 The light chain of the sequence; (v) an antibody that cross-competes with the antibody of (ii); (vi) nivolumab or its antigen binding portion; (vii) peclizumab or its antigen binding portion; and (viii ) MEDI0608 or its antigen binding portion.Used to treat cancer CCR4 Antibodies and anti PD - 1 Antibody composition In one embodiment, the composition of the present invention comprises an anti-CCR4 antibody and an anti-PD-1 antibody for the treatment of cancer. In one embodiment, the composition for use in the present invention is prepared to be administered about 0.1 mg / kg, about 0.3 mg / kg, about 0.5 mg / kg, about once a week for four weeks and then about once every two weeks, About 0.75 mg / kg, about 1.0 mg / kg, about 2.0 mg / kg or about 3.0 mg / kg of anti-CCR4 antibody, and about once every two weeks at about 2.0 mg / kg, or about 3.0 mg / kg or about 240 mg / body as a flat dose of anti-PD-1 antibody or about 480 mg / body as a flat dose of anti-PD-1 antibody combination every four weeks. In certain embodiments, the composition is prepared to be used as a flat dose of anti-PD-1 antibody at about 2.0 mg / kg, or about 3.0 mg / kg, or about 240 mg / body about once every two weeks, or About 480 mg / body about once every four weeks as a flat dose of anti-PD-1 antibody and about 0.1 mg / kg, about 0.3 mg / kg, about 0.5 mg about once a week for four weeks and then about once every two weeks / kg, about 0.75 mg / kg, about 1.0 mg / kg, about 2.0 mg / kg or about 3.0 mg / kg of anti-CCR4 antibody combination. In some embodiments, the composition is prepared to be about 0.1 mg / kg, about 0.3 mg / kg, about 0.5 mg / kg, about 0.75 mg / kg, about once a week for four weeks and then about once every two weeks, About 1.0 mg / kg, about 2.0 mg / kg or about 3.0 mg / kg dose of anti-CCR4 antibody and about once every two weeks about 2.0 mg / kg or about 3.0 mg / kg or about 240 mg / body as a flat dose The anti-PD-1 antibody or about 480 mg / body about once every four weeks is administered as a flat-dose combination of anti-PD-1 antibodies. In other embodiments, the composition is prepared to be administered with an anti-PD-1 antibody at a dose of about 2.0 mg / kg, or about 3.0 mg / kg, or about 240 mg / body about once every two weeks and about once a week An anti-CCR4 antibody combination is administered at a dose of about 0.1 mg / kg, about 0.3 mg / kg, about 1.0 mg / kg, about 2.0 mg / kg, or about 3.0 mg / kg that lasts for four weeks and then about once every two weeks. In one embodiment, the composition for use in the present invention comprises an anti-CCR4 antibody (moglizumab) for about four times a week and then about once every two weeks at about 0.1 mg / kg, about 0.3 mg / kg, about 0.5 mg / kg, about 0.75 mg / kg, about 1.0 mg / kg, about 2.0 mg / kg or about 3.0 mg / kg and about once every two weeks at about 2.0 mg / kg or about 3.0 mg / The dose of kg or about 240 mg / body is a flat dose or about once every four weeks with a combination of about 480 mg / body as a flat dose of anti-PD-1 antibody (nivolumab) to treat cancer in the individual. In another embodiment, the composition comprises an anti-PD-1 antibody (nivolumab) at a dose of about 2.0 mg / kg or about 3.0 mg / kg or about 240 mg / body for about once every two weeks Equalized dose or about once every four weeks with about 480 mg / body as an adjusted dose and about once a week for four weeks and then about once every two weeks about 0.1 mg / kg, about 0.3 mg / kg, about 0.5 mg / kg, Combination of anti-CCR4 antibody (moglizumab) at a dose of about 0.75 mg / kg, about 1.0 mg / kg, or about 3.0 mg / kg. In certain embodiments, the composition used in the present invention is prepared as a flat dose composition; for example, the flat dose of an anti-CCR4 antibody may be at least about 5 mg, at least about 10 mg, at least about 15 mg, at least About 20 mg, at least about 30 mg, at least about 40 mg, at least about 50 mg, at least about 60 mg, at least about 70 mg, at least about 80 mg, at least about 90 mg, at least about 100 mg, at least about 110 mg, at least About 120 mg, at least about 130 mg, at least about 140 mg, at least about 150 mg, at least about 160 mg, at least about 170 mg, at least about 180 mg, at least about 190 mg, at least about 200 mg, at least about 210 mg, at least About 220 mg, at least about 230 mg, at least about 240 mg, at least about 250 mg, at least about 260 mg, at least about 270 mg, at least about 280 mg, at least about 290 mg, or at least about 300 mg. In some embodiments, the dose of the anti-CCR4 antibody is in the range of about 1 mg to about 200 mg, about 2 mg to about 180 mg, about 3 mg to about 170 mg, or about 6 mg to about 160 mg. In another embodiment, the composition comprises an anti-CCR4 antibody in a dose ranging from about 6 mg to about 60 mg, about 10 mg to about 80 mg, or about 20 mg to about 70 mg. In other embodiments, the dose of anti-CCR4 antibody is about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, or about 180 mg. In other embodiments, the composition used in the present invention is administered in a flat dose, wherein the dose of anti-PD-1 antibody is at least about 1 mg, at least about 5 mg, at least about 10 mg, at least about 15 mg, at least About 20 mg, at least about 30 mg, at least about 40 mg, at least about 50 mg, at least about 60 mg, at least about 70 mg, at least about 80 mg, at least about 90 mg, at least about 100 mg, at least about 110 mg, at least About 120 mg, at least about 130 mg, at least about 140 mg, at least about 150 mg, at least about 160 mg, at least about 170 mg, at least about 180 mg, at least about 190 mg, at least about 200 mg, at least about 210 mg, at least About 220 mg, at least about 230 mg, at least about 240 mg, at least about 250 mg, at least about 260 mg, at least about 270 mg, at least about 280 mg, at least about 290 mg, at least about 300 mg, at least about 310 mg, at least About 320 mg, at least about 330 mg, at least about 340 mg, at least about 350 mg, at least about 360 mg, at least about 370 mg, at least about 380 mg, at least about 390 mg, at least about 400 mg, at least about 410 mg, at least About 420 mg, at least about 430 mg, at least about 440 mg, at least about 450 mg, at least about 460 mg, at least about 470 mg, at least 480 mg, at least about 490 mg, at least about 500 mg, at least about 510 mg, at least about 520 mg, at least about 530 mg, at least about 540 mg, at least about 550 mg, at least about 560 mg, at least about 570 mg, at least about 580 mg, at least about 590 mg, or at least about 600 mg. In some embodiments, the dose of the anti-PD-1 antibody is between 20 mg to 300 mg, 30 mg to 280 mg, 40 mg to 270 mg, 50 mg to 270 mg, 60 mg to 260 mg, 70 mg to 250 mg Or in the range of 80 mg to 240 mg. In yet other embodiments, the dose of anti-PD-1 antibody is about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 110 mg, about 120 mg, about 130 mg , About 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg, about 210 mg, about 220 mg, about 230 mg, about 240 mg, about 250 mg, about 260 mg, about 270 mg, about 280 mg, about 290 mg, about 300 mg, about 310 mg, about 320 mg, about 330 mg, about 340 mg, about 350 mg, 400 mg, about 410 mg, about 420 mg, About 430 mg, about 440 mg, about 450 mg, about 460 mg, about 470 mg, about 480 mg, about 490 mg, about 500 mg, about 510 mg, about 520 mg, about 530 mg, about 540 mg, about 550 mg, about 560 mg, about 570 mg, about 580 mg, about 590 mg, or about 600 mg. The composition of the present invention includes any composition for use, including a combination of an anti-CCR4 antibody and an anti-PD-1 antibody having the above activity as an active ingredient, and in one embodiment, the combination is in the form of a pharmaceutical formulation Provided that the pharmaceutical formulation is generally prepared by mixing each antibody with one or more pharmaceutically acceptable carriers according to any well-known method in the pharmaceutical field. In addition, the antibodies used in the present invention are also provided in the form of pharmaceutical formulations, which are generally combined with one or more pharmaceutically acceptable carriers by any well-known method in the medical field Mix to prepare. In some embodiments, the anti-system of the invention is provided in the form of a sterile solution, wherein the antibody is dissolved in an aqueous vehicle such as water, or an aqueous solution of the salt, glycine, glucose or human albumin used. Pharmaceutically acceptable additives such as buffers or tonicity agents can also be added to prepare solutions more similar to physiological conditions and their examples, including sodium acetate, sodium chloride, sodium lactate, potassium chloride, sodium citrate or their analog. It can also be stored by freeze-drying and in actual use, it can be used by dissolving in an appropriate solvent. Regarding the administration route of the antibody used in the present invention or the composition of the present invention, in some embodiments, the most effective route for treatment is used. Examples include oral administration and parenteral administration, such as intraoral, tracheobronchial, intrarectal, subcutaneous, intramuscular, intrathecal, and intravenous administration. It can be administered intrathecally or intravenously. Excipients such as lactose, glucose, sucrose, mannitol, or the like can be used for capsules, lozenges, powders, granules or the like; disintegrating agents such as starch, sodium alginate or the like; lubricating Agents, such as magnesium stearate, talc, or the like; binders, such as polyvinyl alcohol, hydroxypropyl cellulose, gelatin, or the like; surfactants, such as fatty acid esters or the like; plasticizers , Such as glycerin or its analogues are prepared as additives. Examples of formulations suitable for parenteral administration may include injectable formulations, suppositories, air spray or the like. For example, injectable formulations are prepared using carriers including saline solutions, glucose solutions, or mixtures thereof. Suppositories are prepared using carriers such as cocoa butter, hydrogenated fat, carboxylic acid or the like. The air spray is prepared using, for example, a carrier that does not stimulate the antibody itself and the mucosa of the mouth and respiratory tract to be administered to a person, and disperses the antibody into fine particles so that it is easily absorbed. Specific examples of carriers include lactose, glycerin, or the like. Depending on the characteristics of the antibody and carrier used, aerosols, dry powders or the like can be prepared. In addition, even in parenteral preparations, components exemplified as additives in oral preparations may be added. In one embodiment, cancer patients in the present invention include any cancer patients that can be effectively treated by the composition or method of the present invention. For example, in an embodiment of the present invention, the cancer patient is at least one cancer patient with cancer selected from lung cancer, gastrointestinal cancer, gastric cancer (or gastric cancer), colon cancer, colorectal cancer, esophageal cancer, pancreas Cancer, liver cancer, kidney cancer, ovarian cancer, breast cancer, head and neck cancer, skin cancer, melanoma, and hematopoietic cancers including leukemia and lymphoma. In another embodiment, the cancer in the present invention includes any stage of cancer, primary / metastatic cancer, locally advanced / metastatic cancer, relapsed / refractory cancer, and surgically resectable / unresectable cancer . In one embodiment, the cancer patient has at least one cancer selected from the group consisting of lung cancer including small cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), squamous cell carcinoma, adenocarcinoma, and large cell Carcinoma, adenosquamous carcinoma, carcinoma with pleomorphic sarcomatoid or sarcoma elements, cartinoid tumor and classified salivary adenocarcinoma (carcinomas of salivary-gland type), gastric cancer, liver cancer, hepatocytes Cell carcinoma, cholangiocarcinoma, breast cancer, kidney cancer, melanoma and any combination thereof. In another embodiment, the cancer patient has pancreatic cancer selected from the group consisting of pancreatic duct cancer, pancreatic head cancer, pancreatic body cancer, pancreatic tail cancer, and any combination thereof. In another embodiment, the patient has head and neck cancer selected from the group consisting of oral cancer, pharyngeal cancer, larynx cancer, neck cancer, salivary gland cancer, and any combination thereof. In the present invention, cancer patients can be diagnosed before administering the combination therapy. In one embodiment, the diagnostic method may be by methods known in the background art, such as X-ray graphics, computed tomography (CT), positron emission tomography CT (PET-CT), magnetic nuclear resonance imaging (MRI) And / or perform one or several biomarkers in the patient's plasma, serum, peripheral blood, and tumor tissue for analysis and execution. In addition, cancer progression, disease stage, and / or response to treatment can be analyzed by known methods and parameters in combination with the aforementioned imaging and biomarkers. For example, tumor metabolism with regard to glucose consumption is usually spent on FDG-PET based on the upregulated radioisotope labeling glucose, such as18 F fluorodeoxyglucose (FDG) was analyzed as cancer tissue. FDG is highly accumulated in cancer tissues because cancer cells metabolize glucose highly and require a greater amount of glucose than normal tissues. In addition, analysis of cell populations in peripheral blood lymphocytes (PBL), pleural effusion lymphocytes (LPE), and / or tumor-infiltrating lymphocytes (TIL) can be used to assess tissues in each patient and / or the entire body Immune response status. For example, several markers such as CD4, CD8, CD25, CD45RA, CD45RO, CCR4, CD127, PD-1, PD-L1 (B7-H1, CD274), PD-L2, CTLA-4 (CD152), IFN -γ, IL-2, IL-4, IL-12 and Foxp3 can be used to predict the immune response status of each patient. Performance CD4 detected+ CD25+ , CD8+ CD25+ , PD-1+ And / or CTLA-4+ Activated T cells, express CD4+ CD25+ CCR4+ , CD25+ Foxp3+ , CCR4+ Foxp3high , CD4+ CCR4+ Foxp3+ Or CD4+ CD25+ CD45RA- CCR4+ Foxp3+ Treg cells, and when the activated T cells in the patient increase and / or the Treg cells decrease, it is considered that the immune response in the patient is enhanced. In one analysis, CD4, CD8, CD25, CD45RA, CD45RO, CCR4, CD127, PD-1, PD-L1 (B7-H1, CD274), PD can be analyzed before and / or after administration of the combination therapy -T cells, effector Treg, depletion recognized by the performance of L2, CTLA-4 (CD152), IFN-γ, IL-2, IL-4, IL-12 and / or Foxp3, or peripheral / microenvironment cytokines T cells, activated T cells, effector T cells, non-allergic T cells, or bone marrow-derived suppressor cells (MSDC). In some embodiments, the combination therapy of anti-CCR4 antibody and anti-PD-1 antibody can reduce Treg cells and / or increase activated T cells in the peripheral blood, pleural effluent, and / or tumor tissue of the patient. Therefore, the combination of antibodies can enhance the immune response to cancer cells in the patient. In addition, in one embodiment, the method further includes analyzing or measuring the PD-L1, PD-L2, or CCR4 performance level on the tumor cells prior to administration to determine whether the patient is suitable for the combination therapy. In another embodiment, the PD-L1 or PD-L2 manifestation in the patient can be regarded as a sign that the patient is suitable for the combination therapy of the present invention. In addition, the indoleamine 2,3-dioxase (IDO) activity in the patient's plasma sample can be analyzed in the present invention. It is known in the art that IDO manifested in various organs, such as lung, small intestine, and placenta, is related to the induction of Treg differentiation, which will cause immune tolerance. In addition, the molecule is highly expressed on multiple cancer cells, suppresses the immune response and induces immune tolerance of the cancer cells. Therefore, the analysis of IDO activity can detect the immune response status. IDO activity is usually detected by analyzing changes in the content of tryptophan, kynurenine, and kynurenine metabolites. When the IDO activity of the patient is increased, tryptophan, kynurenine, and kynurenine metabolites also increase, thereby resulting in a reduction in the patient's immune response. In other embodiments, the combination therapy of anti-CCR4 antibody and anti-PD-1 antibody increases the number of Treg cells in the patient and / or reduces IDO activity in the patient. The clinical medical evaluation of treatment will usually be assessed by the solid tumor response assessment criteria (RECIST) version 1.1 criteria and by the immune-related response criteria (irRC) (Wolchok et al., Clin. Cancer Res .; 2009, 15: 7412-7420 ).Examples 1 Stages of combination therapy with moglizumab and nivolumab in individuals with advanced solid tumors 1 the study 1. Research Overview This study is a combination of anti-CCR4 antibody moglizumab and anti-PD-1 antibody nivolumab in adult individuals with locally advanced or metastatic solid tumors diagnosed with histology or cytology. Two-part, multicenter, stage 1, open-label, and dose-escalation group expansion study. Study group: Locally advanced or metastatic solid tumor diagnosed by histology or cytology. Dose escalation stage: Individuals with locally advanced or metastatic solid tumors diagnosed with histology or cytology will be registered. Group expansion stage: Registration of advanced or metastatic solid tumors (including but not limited to non-small cell lung cancer (NSCLC), small cell lung cancer (SCLC), gastric cancer (GC), hepatocellular carcinoma, pancreatic cancer and esophagus) Cancer). 2. Research objectives: Main objective: The main objective is to characterize the safety and tolerability of the combination of moglizumab and nivolumab in individuals with locally advanced or metastatic solid tumors and to determine the combination Maximum tolerated dose (MTD) or fixed dose is recommended. Secondary objective: The first objective is to evaluate the combined resistance of moglizumab and nivolumab in individuals with locally advanced or metastatic solid tumors based on the solid tumor response assessment criteria (RECIST version 1.1) Tumor activity. The best overall response rate (BOR), time to response (TTR), duration of response (DOR), progression-free survival (PFS), and overall survival (OS) will be evaluated for antitumor activity. The second key objective was to characterize the pharmacokinetic (PK) properties of moglizumab and nivolumab in the combined course of moglizumab and nivolumab. Another major goal was to assess the immunogenicity of moglizumab and nivolumab. 3. Selection criteria: Including individuals who have voluntarily signed the informed consent form approved by the Institutional Review Board in accordance with the guiding principles of the regulatory body and dated. Written informed consent must be obtained before performing any research-related steps. The inclusion criteria include the following:-Individuals who are 20 years of age or older with informed consent. -Individuals with assessable lesions according to RECIST version 1.1 guidelines. -Individuals with life expectancy> 12 weeks. -Individuals with ECOG performance status (performance status) 0 to 1.Dose escalation phase : The individual may have been histologically or cytologically diagnosed as locally advanced or metastatic solid tumor (excluding primary CNS or hematological malignant disease).Group expansion stage : The individual may have been histologically or cytologically diagnosed as locally advanced or metastatic solid tumors, including (but not limited to) NSCLC, SCLC, GC, liver cancer, hepatocellular carcinoma, pancreatic cancer, and esophageal cancer (excluding primary CNS or hematological malignant disease). 4. Exclusion criteria Exclusion criteria include pregnant or lactating women or all individuals who are expected to become pregnant. Exclusion criteria further include individuals with uncontrollable and apparently current (inter-current) disease. Exclusion criteria further include individuals with known CNS metastases and / or meningeal metastases. Individuals with asymptomatic brain metastases that have been treated are considered stable, and may include individuals who have not received corticosteroids or anticonvulsants for at least 28 days prior to Day 1 of Cycle 1. Exclude anti-PD-1, anti-PD-L1, anti-PD-L2, anti-CD137, or anti-cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) antibody or specifically targeted T cell co-stimulation or inspection Individuals who are treated with any other antibody or drug. Exclude individuals who have previously been treated with moglizumab. Individuals with any previous grade 3 or greater immune-related adverse events (irAE) who simultaneously received any previous immunotherapy agent or any undetermined irAE were excluded. Exclude individuals with a history of severe allergic reactions to drugs. Exclude individuals who have received live attenuated vaccinia within 28 days before the first day of the first cycle. Exclude individuals with a history of organ transplantation or allogeneic bone marrow transplantation. Excludes having received chemotherapy, immunotherapy, biological or hormonal therapy, another research drug, radiation or major surgery for cancer treatment (for nitrosourea or Individuals of mitomycin C). Individuals with any undetermined toxicity level 1 or less (as defined by CTCAE version 4.03) based on previous anti-cancer therapies are excluded. Exclude individuals with irreversible toxicity, which may be exacerbated by unreasonable expectations of research products that may be included in the discretion of the principal or associate researcher. Exclude individuals who have received systemic steroid therapy or any other form of immunosuppressive therapy within 28 days prior to the first day of the first cycle. However, treatment with local immunosuppressive drugs may be included in the discretion of the principal or associate researcher. individual. Exclude those who require systemic steroids or immunosuppressants with known active autoimmune diseases or syndromes (eg, rheumatoid arthritis, uveitis, systemic lupus erythematosus, Wegener's granulomatosis) Or sarcomatoid). Based on this study, individuals with a history of leukoplakia, baldness, or autoimmune disease or syndrome who do not require systemic steroids or immunosuppressants for 3 years prior to written informed consent will be excluded. Individuals with active inflammatory bowel disease (eg, inflammatory colitis and ulcerative colitis), Crohn's disease, irritable bowel disease, celiac disease, or other severe GI chronic conditions associated with diarrhea. However, it may include individuals with a history of chronic GI disease who have not required any therapy for 3 years prior to written informed consent. Prior to human immunodeficiency virus (HIV), human T-cell leukemia virus type 1 (HTLV-1), hepatitis B surface antigen, and hepatitis C virus antibodies with congenital or acquired immunodeficiency and having tuberculosis or testing Individuals with a clinical diagnosis of a known medical history. Exclude individuals with other traumatic malignant tumors within 5 years before the first day of the first cycle. However, individuals with non-invasive malignant tumors that have been cured surgically, such as carcinoma in situ of the cervix, non-melanoma carcinoma of the skin, or carcinoma in situ of the breast duct of the breast, can be registered. Exclude individuals who have registered another clinical study unless the other clinical study is an observational (non-intervention) clinical study or a follow-up phase of an intervention study. Exclude individuals with any condition that would interfere with the evaluation of research products or the interpretation of individual safety or research results in the opinion of the researcher and / or sponsor. 5. Research designDose escalation phase : The dose escalation phase is a parallel 3 + 3 design that will identify the MTD of the combination course or recommend a fixed dose. MTD is defined as a lower dose level of the studied moglizumab and nivolumab, of which more than 2 individuals in the 6 group of individuals undergoing dose-limiting toxicity (DLT); In this case, the recommended fixed dose for the group expansion phase will be determined according to the dose level in the MTD or group 2. During the dose escalation phase, approximately 3 to 18 individuals will be registered (3 to 6 individuals in each of up to 3 groups). Dose levels and schedules are described below. See also Figure 1. [Table 1]table 1 . Moglizumab and nivolumab in the dose-escalation phase Group expansion stage : The group expansion phase will further explore the safety, PK, PD and antitumor activity of each combination in up to 6 tumor types. At this stage, approximately 90 individuals will be registered with locally advanced or metastatic solid tumors, including (but not limited to) NSCLC, SCLC, GC, hepatocellular carcinoma, pancreatic cancer, and esophageal cancer; A tumor type can be considered to be guaranteed by observations at the dose escalation stage to assess other safety.Combination therapy : During the dose escalation phase and the group expansion phase, the individual will receive a combination of moglizumab and nivolumab. Individuals will receive nivolumab 3.0 mg / kg on day 1 and day 15 as an intravenous (IV) infusion lasting at least 1 hour. Individuals will also receive 0.1, 0.3, or 1.0 as an IV infusion over a period of at least 1 hour on Day 1, Day 8, Day 15, and Day 22 of the first cycle and Day 1 and Day 15 of subsequent cycles mg / kg of moglizumab. The combination course will be repeated on the 29th day. On days 1 and 15 of each cycle, after completion of nivolumab administration and 1 hour of observation, moglizumab will be administered. The individual will receive combination therapy until the disease progresses or dies.Safety and efficacy evaluation : All individuals will monitor safety on the 1st, 8th, 15th, and 22nd days of the 1st cycle period according to the event schedule; in subsequent cycles, on the 1st and 15th days every 2 weeks Perform safety assessment. The disease assessment by computed tomography (CT) or magnetic resonance imaging (MRI) will continue every 4 weeks for the first 2 cycles, every 8 weeks for the next 12 cycles, and then every 12 weeks after screening Perform until disease progression or death. Disease progression will be limited according to RECIST version 1.1 guidelines. [Table 2]table 2 . Research drug 6. Efficacy and safety variablesPrimary endpoint safety .Secondary endpoint efficacy : The following efficacy parameters will be used:-Best overall response (BOR) evaluated using RECIST version 1.1. -Time to response (TTR): days from the first day of the first cycle to the first assessment date of CR / PR diagnosis-Duration of response (DOR): from the first assessment date of CR / PR diagnosis to death or Days of PD (regardless of which earlier) date-progression-free survival (PFS): days from day 1 of cycle 1 to death or PD (regardless of earlier) date-total survival (OS): from 1st The number of days from the first day of the cycle to the date of deathStatistical Analysis : The following analysis set will be used for research:-Safety analysis set: includes all individuals who receive at least one dose (even a partial dose) of study drug. Full Analysis Set (FAS): Includes all individuals who receive at least one dose (even a partial dose) of study drug and have at least one available post-dose efficacy data. Adverse events will be listed by body system, severity and treatment proportion. Similar representations on serious adverse events, AEs leading to study discontinuation and AEs leading to death will be provided for safety analysis and full analysis set. The individual's tumor response, TTR, DOR, PFS and OS will be evaluated. ORR will be calculated as the proportion of individuals who are responders, that is, complete response (CR) and partial response (PR); 95% accurate confidence interval for the response will be calculated. The disease control rate (CR, PR, SD) will also be calculated. 7. Clinical efficacy As observed during the combination therapy in this clinical trial, as of September 7, 2016, 4 PRs, 6 SDs, and 3 PDs were observed in 15 patients with hepatocellular carcinoma (HCC). On the other hand, as of September 7, 2016, 1 PR, 5 SD, and 9 PD were observed in 15 patients with pancreatic cancer (PA). In addition, several adverse events were observed in each cancer type, however, all events may be acceptable in cancer treatment. As of November 1, 2017 (data cut-off date is August 31, 2017), 4 PRs and 6 SDs were observed in 15 HCC patients (objective response rate 26.7%, disease control rate 66.7%) (Figure 2 ); 1 PR and 5 SD were observed in 15 PA patients (objective response rate 6.7%, disease control rate 40.0%) (Figure 3). For patients with HCC and PA, the intermediate progression-free survival period was 114.5 days (95% CI, 28.0 to 223.0) and 56.0 days (95% CI, 29.0 to 115.0), respectively, and the intermediate overall survival period was 343.0, respectively Days (95% CI, 123.0 to inestimable) and 198.0 days (95% CI, 107.0 to 328.0). No dose limiting toxicity was observed during the dose escalation phase. Grade 3 to 4 drug-related adverse events (AE) occurred in 25.0% of the dose escalation stage and group expansion. No level 5 related AEs were seen. The most commonly associated AEs were skin rash (36.5%), skin papules (20.8%) and diarrhea (14.6%). Based on these clinical results of this trial, it was found that the combination of anti-CCR4 antibody and anti-PD-1 antibody is effective for several types of cancer patients (including at least HCC and PC patients).Examples 2 Open-labeled multicenter stage of moglizumab combined with nivolumab in individuals with locally advanced or metastatic solid tumors 1 / 2 the study 1. Research Overview This study is a multicenter anti-CCR4 antibody, such as moglizumab and anti-PD-1 antibody, such as nivolumab combination therapy in adult individuals with locally advanced or metastatic solid tumors A phase 1/2 open-labeled group expansion study of the discovered dose. Phase 1 will identify the MTD of the combination therapy of moglizumab and nivolumab individuals or the highest regimen-defined dose in the absence of excess MTD. Phase 1 will register up to 12 individuals. Phase 2 will explore the safety, efficacy and anti-tumor activity of the highest tolerated dose of the combined treatment course. Phase 2 will register up to 184 individuals (21 to 36 individuals / tumor type) with locally advanced or metastatic disease of the following tumor types: squamous cell non-small cell lung cancer (NSCLC); no program Non-squamous cell NSCLC of cell death ligand 1 (PD-L1); squamous cell carcinoma of the head and neck (SCCHN); non-microsatellite instability (non-MSI) higher colorectal cancer (CRC) ; Ovarian cancer; Hepatocellular carcinoma (HCC) and pancreatic cancer (PA). 2. Research objectives The main objective is to characterize the safety and tolerability of the combination therapy of moglizumab and nivolumab in individuals with locally advanced or metastatic solid tumors and to determine the combination therapy MTD or the highest regimen limit dose in the absence of MTD. Secondary objective The secondary objective was to evaluate the antitumor activity of the combination of moglizumab and nivolumab based on RECIST version 1.1. Antitumor activity will be assessed as overall response rate (ORR), TTR, DOR, PFS and OS. Exploring objectives Exploring objectives are:-to analyze the serum concentrations of moglizumab and nivolumab when the combination is administered;-to evaluate the immunogenicity of moglizumab and nivolumab when the combination is administered ;-Evaluate the pharmacodynamic (PD) properties of the combination of moglizumab and nivolumab and determine which biomarker may be related to safety and / or antitumor activity;-based on immune-related RECIST (irRECIST ) Version 1.1 evaluates the ORR of the combination of moglizumab and nivolumab. 3. Selection criteria Individuals must meet each of the following selection criteria during the screening phase to be eligible to participate in the study. In addition, individuals participating in stage 2 must meet all inclusion criteria and do not have one of the exclusion criteria for the relevant tumor type. Individuals -18 years old or older;-Individuals with solid tumors diagnosed histologically or cytologically;-Individuals with locally advanced or metastatic solid tumors;-Has developed or has not responded to any standard treatment regimen Individuals who tolerate or reject standard treatment, or do not have appropriate standard therapy for them;-Individuals with guidelines for assessable lesions / RECIST version 1.1;-Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1 ( PS) individual;-If the individual is a woman who may be pregnant or a man who has sexual activity with a woman who may be pregnant, agree to participate in the study period since the signing of the ICF; and for women after the final dose of the research medical product (IMP) 23 weeks or for men who used adequate contraception 31 weeks after the last dose of IMP;-individuals with adequate blood, kidney, liver, and respiratory function limitations;-willing to undergo tumor biopsy during the screening phase, or if the tumor is not Access for biopsy, the archived tumor material must be available for submission to the individual; and-according to the guidelines of the regulatory agency, the voluntary approval of the agency review committee approval Consent and indicate the date of the individual. 4. Exclusion criteria If any of the following exclusion criteria is met during the screening phase, the individual will not be eligible to participate in this study. In addition, individuals participating in stage 2 must meet all inclusion criteria and do not have one of the specific exclusion criteria for the relevant tumor type. -Female individuals who are pregnant or breastfeeding or any individuals who expect to become pregnant or have children during this study period;-Individuals with uncontrollable and obvious current disease. -Individuals with a mental illness / social situation that will limit compliance with research requirements in the researcher ’s perspective;-Individuals with a history of primary CNS tumors or known CNS cancer metastases and / or CNS cancer metastases and / or cancerous meningitis ; Exception: if the CNS cancer metastasis is adequately treated, the individual is eligible and has returned to baseline (except for residual conditions or symptoms related to CNS treatment) at least 4 weeks before registration. In addition, individuals must stop corticosteroids for 4 weeks before registration. -Received prior therapy for cancer before day 1 of cycle 1 or for nitrosourea or mitomycin C within 28 or 42 days, or tamoxifen for 14 days ) Individuals undergoing major surgery;-Individuals who have received radiation therapy or radiosurgery within 14 days before the first day of the first cycle;-Have been pre-treated with anti-PD-1, anti-PD-L1, anti-PD-L2, Individuals treated with anti-CD137 or anti-CTLA-4 antibodies or any other antibodies or drugs that specifically target T cell co-stimulation or checkpoint pathways;-Individuals who have previously been treated with moglizumab;-Patients with a history of allergies or Individuals with allergy to the study drug component;-Individuals who have received live attenuated vaccinia within 28 days before the first day of the first cycle;-Individuals with a history of organ transplantation or allogeneic bone marrow transplantation;-Individuals with previous cancer resistance Any individual with unresolved toxicity of> 1 level of therapy;-Individuals who have used immunosuppressive drugs within 14 days before the first day of the first cycle. -Individuals with known active autoimmune diseases or a history of autoimmune diseases that may affect the function of living organs or require immunosuppressive therapy including systemic corticosteroids;-have toxic epidermal necrolysis or Stevens Johnson syndrome (Stevens) -Individuals with a history of Johnson syndrome;-Individuals with a history of inflammatory bowel disease, Crohn's disease, ulcerative colitis or Wegener's granulomatosis;-Primary or acquired immunodeficiency or human immunodeficiency virus ( HIV) with a known medical history or a known acquired immunodeficiency syndrome; -Except for individuals with hepatocellular carcinoma, for hepatitis B surface antigen (HBVsAg) or hepatitis C RNA tests that indicate acute or chronic infection Individuals who are positive;-Individuals with another active malignant tumor that requires simultaneous intervention;-Individuals receiving any other investigational medicinal agents;-Have the ability to interfere with IMP's assessment or individual safety from the perspective of the researcher and / or sponsor Or an individual with another symptom that explains the results of the study; and-an individual with a history of pneumonia or interstitial lung disease. 5. Study design Phase 1: Dose discovery The Phase 1 dose discovery study has a 3 + 3 design that will identify the MTD or the highest regimen-defined dose in the absence of exceeding the MTD for this combination course. Phase 1 studies will register up to 12 individuals (3 to 6 individuals / group). Plan the initial dose level and optional dose level. The dose level and schedule are described in Table 3. MTD is defined as a dose level that is lower than the dose level of the group in which one third or more individuals experience DLT. The recommended dosage regimen for stage 2 is the MTD or highest dose level tested. [table 3]table 3 . dose level * If> 1 individual experiences DLT at dose level 1, this dose level can be registered. The guidelines for dose discovery are summarized in Figure 4. Stage 2: Expansion group To further characterize the safety, tolerability and anti-tumor activity of the combination, up to 184 individuals (21 to 36 individuals / Tumor type): Squamous cell non-small cell lung cancer (NSCLC); non-squamous cell NSCLC that does not show programmed cell death ligand 1 (PD-L1); squamous cell carcinoma of the head and neck (SCCHN); Non-microsatellite instability (non-MSI) is higher in colorectal cancer (CRC); ovarian cancer; hepatocellular carcinoma (HCC); and pancreatic cancer (PA). The individual will be treated with the highest dose of the combined course of treatment deemed to be tolerable in Phase 1. The monitoring will be used to monitor the safety and tolerability of the dosing course in each expansion group. Therapeutic Research Medicines (IMP) of the individual The following IMPs will be used in this study:-Moglizumab: on the first, eighth, fifteenth and twenty-second day of the first cycle and then on each subsequent 28 On day 1 and day 15 of the daily cycle, 0.3 or 1.0 mg / kg is administered as an intravenous infusion over a period of at least 1 hour. -Nivolumab: 240 mg of nivolumab was administered as an intravenous infusion over 1 and 15 days of each 28-day cycle for at least 30 minutes. Duration of treatment Individuals can receive combination therapy from day 1 of cycle 1 for up to 96 weeks. Individuals who discontinue treatment and have no disease progression and experience disease progression within 12 months of the last dose of IMP can receive an additional IMP for up to 48 weeks, provided that these individuals have not received other systemic therapies for their cancer. Safety and efficacy evaluation The individual's safety will be monitored throughout the study period. Tumor assessment by CT or MRI will be performed at the screening stage, at week 10, and thereafter at least every 12 weeks until a clear disease progression or death. Tumor biopsies will be required during the screening phase (except for individuals with archived tumor tissue) and week 10 (unless the tumor is not accessible for biopsy). 6. Efficacy and safety variables Main endpoints Safety and tolerability will be assessed by evaluating changes in AE and physical test findings, vital sign measurements, 12-lead ECG readings, and clinical laboratory evaluations. The secondary end point will use the following efficacy parameters:-Best overall response (BOR) evaluated using RECIST version 1.1;-Time to response (TTR): From day 1 of cycle 1 to CR / PR diagnosis using RECIST version 1.1 Days of the first assessment date-duration of response (DOR): the number of days from the first assessment date of CR / PR diagnosed using RECIST version 1.1 to the date of death or PD (whichever is earlier);-progression-free survival (PFS): The number of days from the first day of the first cycle to the date of death or PD (whichever is earlier). For individuals using RECIST version 1.1 whose BOR is PR or CR; -Overall Survival (OS): The statistical method and planning analysis of the number of days from the first day of the first cycle to the date of death Consists of data list and summary display. Demographic and other baseline characteristics information will be summarized for safety analysis and efficacy analysis sets. Adverse events will be listed by body system, severity and treatment proportion. Similar presentations will be provided regarding SAE, AE that caused IMP to stop, and AE that caused death. The list of laboratory parameters will indicate the normal range for each parameter. Each value will be classified as belonging to, below or within the normal range. The individual's tumor response, TTR, DOR, PFS and OS will be evaluated. ORR will be calculated as the proportion of individuals who are responders, that is, complete response (CR) and partial response (PR); 95% accurate confidence interval for ORR will be calculated. The ORR will be used to derive the ORR using both the RECIST version 1.1 and the irRECIST version 1.1 in the expansion group based on the combination therapy. These individuals include all individuals who received the combination therapy on Day 1 of Cycle 1. The Kaplan-Meier methodology will be used to assess reaction duration, OS, and PFS. 7. Clinical efficacy As observed during the combination therapy in this clinical trial, as of August 28, 2017, 1 out of 10 HCC patients had undiagnosed PR, 2 out of 21 ovarian cancer patients were diagnosed with CR and 2 were diagnosed PR; and PR was confirmed in 1 of 29 CRC patients. As of October 23, 2017, 1 undiagnosed PR and 1 confirmed PR among 10 HCC patients; 2 confirmed CR, 2 confirmed PR, and 1 undiagnosed PR among 21 ovarian cancer patients; and 29 CRC One of the patients was diagnosed with PR. On the other hand, as of October 23, 2017, no PR or CR was observed in 17 PA patients. Based on these clinical results of this trial, the combination of anti-CCR4 antibody and anti-PD-1 antibody was found to be effective for certain types of cancer patients including at least HCC, ovarian cancer and CRC patients.Examples 3 Flow Cytometry Analysis of Immune Cell Subgroups in the Peripheral Blood of Individuals Methods In the Phase 1 study described in Example 1, immune cell subsets were measured by FCM. Before treatment, on the first day of the second cycle of treatment or on the end of treatment and on the first day of the third cycle, collect peripheral blood and TIL before and after treatment (from the 15th day of the second cycle of the treatment to the first day of the third cycle Or end of treatment) Separated from the newly prepared tumor biopsy sample. The analysis of the effect Treg was performed by the performance of CD4, Foxp3 and CD45RA according to the method described in Immunity, 2009, 30 (6) 899-911. Use FlowJo version 7.6.5 to analyze the data. At the same time, the percentage of each group in the peripheral blood is corrected by the number of lymphocytes. Results Through preliminary analysis of peripheral blood, the effect of Treg (CD3) on day 1 of cycle 2 (including the end of treatment) and day 1 of cycle 3 was observed+ CD4+ CD45RA-Foxp3high ) Has no relation to the best overall response. Compared to those in non-responders, activated T cells (CD3) on day 1 of cycle 3 were also observed in responders+ CD4+ ICOS+ T cells) increased. By preliminary analysis of 11 pairs of TIL before and after treatment, the effect of TIL in TIL (CD3+ CD4+ CD45RA-Foxp3high ) With total CD3+ The proportion of T cells has a tendency to decrease independently of clinical response after treatment. Compared to Treg, CD3 in TIL+ CD8+ T cells and total CD3+ The proportion of T cells has an overall increasing trend after treatment. CD3 in TIL+ CD8+ T cells / effector Treg also showed an increasing trend after treatment. In addition, by evaluating the levels of chemokines (MIG and IP-10) in the peripheral blood after treatment, it was found that the increasing trend was not related to the best overall response. Throughout this application, various publications refer to the name and date of the author in parentheses, or refer to the patent number or patent publication number. The disclosures of these publications are hereby incorporated by reference in their entirety in order to more fully describe the present invention as described and claimed herein, such as the current advanced technology known to those skilled in the art . However, the citation of references herein should not be taken as an admission that such references are prior art to the present invention. This application claims the US Provisional Application Serial No. 62 / 428,468 filed on November 30, 2016 and 2017 9 The US Provisional Application Serial No. 62 / 554,486, filed on May 5, is entitled to the entire contents of these two documents and is incorporated herein by reference.No text sequence list SEQ ID NO.1: Description of artificial sequence; KW-0761_VH CDR1 SEQ ID NO.2: Description of artificial sequence; KW-0761_VH CDR2 SEQ ID NO.3: Description of artificial sequence; KW-0761_VH CDR3 SEQ ID NO.4 : Description of artificial sequence; KW-0761_VL CDR1 SEQ ID NO. 5: description of artificial sequence; KW-0761_VL CDR2 SEQ ID NO. 6: description of artificial sequence; KW-0761_VL CDR3 SEQ ID NO. 7: description of artificial sequence ; KW-0761_VH SEQ ID NO. 8: artificial sequence description; KW-0761_VL SEQ ID NO. 9: artificial sequence description; KW-0761_H SEQ ID NO. 10: artificial sequence description; KW-0761_L SEQ ID NO. 11: artificial sequence description; Nivolumab_VH CDR1 SEQ ID NO. 12: artificial sequence description; Nivolumab_VH CDR2 SEQ ID NO. 13: artificial sequence description; Nivolumab_VH CDR3 SEQ ID NO. 14: artificial sequence description; Nivolumab_VL CDR1 SEQ ID NO.15: Description of artificial sequence; Nivolumab_VL CDR2 SEQ ID NO.16: Description of artificial sequence; Nivolumab_VL CDR3 SEQ ID NO.17: Description of artificial sequence; Nivolumab_VH SEQ ID NO.18: Description of artificial sequence; Nivolumab_VL SEQ ID NO.19: Description of artificial sequence ; Nivolumab_H SEQ ID NO.20: artificial sequence description; Nivolumab_L SEQ ID NO.21: artificial sequence description; human CCR4 cd nucleotide and amino acid sequence SEQ ID NO.22: artificial sequence description; synthetic construction CCR4 Amino acid sequence SEQ ID NO.23: description of artificial sequence; PD-1 cd human PD-1 nucleotide and amino acid sequence SEQ ID NO.24: description of artificial sequence; synthesis of human PD-1 amino group Acid sequence

[圖1]圖1展示出展示抗CCR4抗體(例如,莫格利珠單抗)及抗PD-1抗體(例如,納武單抗)之投藥排程之示意圖。 [圖2]圖2展示出抗CCR4抗體(莫格利珠單抗)及抗-PD-1抗體(納武單抗)之組合療法在肝細胞細胞癌患者中之臨床療效的結果。圖2指示各癌症患者之蜘網圖(spider plot)。縱軸指示相較於組合療法前之「靶標區之腫瘤收縮率(%)」且橫軸指示治療起始後之週數。「+」標誌表示新型病變的第一次出現。 [圖3]圖3展示出抗CCR4抗體(莫格利珠單抗)及抗PD-1抗體(納武單抗)之組合療法在胰腺癌患者中之臨床療效的結果。圖3指示各癌症患者之蜘網圖。縱軸指示相較於組合療法前之「靶標區之腫瘤收縮率(%)」且橫軸指示治療起始後之週數。「+」標誌表示新型病變的第一次出現。 [圖4]圖4展示出用於劑量發現及擴增之準則。DLT為劑量限制性毒性之縮寫。[FIG. 1] FIG. 1 shows a schematic diagram showing the schedule of administration of anti-CCR4 antibody (eg, moglizumab) and anti-PD-1 antibody (eg, nivolumab). [FIG. 2] FIG. 2 shows the results of the clinical efficacy of the combination therapy of anti-CCR4 antibody (moglizumab) and anti-PD-1 antibody (nivolumab) in patients with hepatocellular carcinoma. Figure 2 indicates the spider plot of each cancer patient. The vertical axis indicates the "tumor contraction rate (%) of the target area" compared to the combination therapy and the horizontal axis indicates the number of weeks after the start of treatment. The "+" sign indicates the first appearance of a new lesion. [FIG. 3] FIG. 3 shows the results of the clinical efficacy of combination therapy of anti-CCR4 antibody (moglizumab) and anti-PD-1 antibody (nivolumab) in patients with pancreatic cancer. Figure 3 indicates the spider web of each cancer patient. The vertical axis indicates the "tumor contraction rate (%) of the target area" compared to the combination therapy and the horizontal axis indicates the number of weeks after the start of treatment. The "+" sign indicates the first appearance of a new lesion. [Figure 4] Figure 4 shows the criteria for dose discovery and amplification. DLT is an abbreviation for dose-limiting toxicity.

Claims (35)

一種治療有需要之個體之癌症之方法,該方法包含將特異性結合於人類CC趨化因子受體4 (CCR4)之抗體或其抗原結合部分(「抗CCR4抗體或其抗原結合部分」)及特異性結合於人類體程序式死亡-1 (PD-1)之抗體或其抗原結合部分(「抗PD-1抗體或其抗原結合部分」)投與至個體。A method of treating cancer in an individual in need thereof, the method comprising binding an antibody or antigen-binding portion thereof specifically to human CC chemokine receptor 4 (CCR4) ("anti-CCR4 antibody or antigen-binding portion thereof") and An antibody or antigen-binding portion thereof that specifically binds to PD-1 ("anti-PD-1 antibody or antigen-binding portion") is administered to an individual. 一種將患有癌症之個體體內的腫瘤大小減小至少約1%、5%、10%、15%、20%或30%之方法,該方法包含將抗CCR4抗體或其抗原結合部分及抗PD-1抗體或其抗原結合部分投與至個體。A method for reducing the size of a tumor in an individual with cancer by at least about 1%, 5%, 10%, 15%, 20%, or 30%, the method comprising combining an anti-CCR4 antibody or antigen-binding portion thereof and anti-PD -1 The antibody or antigen-binding portion thereof is administered to the individual. 如請求項1或2之方法,其中該抗CCR4抗體為嵌合抗體、人類化抗體或人類抗體。The method of claim 1 or 2, wherein the anti-CCR4 antibody is a chimeric antibody, a humanized antibody, or a human antibody. 如請求項1至3中任一項之方法,其中該抗CCR4抗體或其抗原結合部分選自由以下組成之群: (i)結合於與抗體相同之抗原決定基的抗體或其抗原結合部分,其包含:包含SEQ ID NO:1中所闡述之序列之重鏈可變區(「VH」)互補決定區(CDR)1、包含SEQ ID NO:2中所闡述之序列之VH CDR2,以及包含SEQ ID NO:3中所闡述之序列之VH CDR3中的一或多者,及包含SEQ ID NO:4中所闡述之序列之輕鏈可變區(「VL」)CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2以及包含SEQ ID NO:6中所闡述之序列之VL CDR3中的一或多者; (ii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:1中所闡述之序列之VH CDR1、包含SEQ ID NO:2中所闡述之序列之VH CDR2、包含SEQ ID NO:3中所闡述之序列之VH CDR3、包含SEQ ID NO:4中所闡述之序列之VL CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2,以及包含SEQ ID NO:6中所闡述之序列之VL CDR3; (iii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:7中所闡述之序列之VH及包含SEQ ID NO:8中所闡述之序列之VL; (iv)抗體,其包含:包含SEQ ID NO:9中所闡述之序列之重鏈及包含SEQ ID NO:10中所闡述之序列之輕鏈; (v)抗體或其抗原結合部分,與(ii)之該抗體交叉競爭;以及 (vi)莫格利珠單抗(mogamulizumab)或其抗原結合部分。The method according to any one of claims 1 to 3, wherein the anti-CCR4 antibody or antigen-binding portion thereof is selected from the group consisting of: (i) an antibody or antigen-binding portion thereof that binds to the same epitope as the antibody, It includes: a heavy chain variable region ("VH") complementarity determining region (CDR) comprising the sequence set forth in SEQ ID NO: 1, a VH CDR2 comprising the sequence set forth in SEQ ID NO: 2, and comprising One or more of the VH CDR3 of the sequence set forth in SEQ ID NO: 3, and the light chain variable region ("VL") CDR1 including the sequence set forth in SEQ ID NO: 4, including SEQ ID NO: One or more of the VL CDR2 of the sequence set forth in 5 and the VL CDR3 containing the sequence set forth in SEQ ID NO: 6; (ii) an antibody or antigen-binding portion thereof, comprising: comprising SEQ ID NO: 1 VH CDR1 of the sequence set forth in V1, VH CDR2 including the sequence set forth in SEQ ID NO: 2, VH CDR3 containing the sequence set forth in SEQ ID NO: 3, including the sequence set forth in SEQ ID NO: 4 VL CDR1, VL CDR2 containing the sequence set forth in SEQ ID NO: 5, and VL CDR3 containing the sequence set forth in SEQ ID NO: 6; (iii) antibody Or an antigen binding portion thereof, comprising: VH comprising the sequence set forth in SEQ ID NO: 7 and VL comprising the sequence set forth in SEQ ID NO: 8; (iv) antibody comprising: comprising SEQ ID NO: The heavy chain of the sequence set forth in 9 and the light chain containing the sequence set forth in SEQ ID NO: 10; (v) the antibody or antigen-binding portion thereof, which competes with the antibody of (ii); and (vi) Mo Glimizumab (mogamulizumab) or its antigen binding portion. 如請求項1至4中任一項之方法,其中該抗PD-1抗體為嵌合抗體、人類化抗體或人類抗體。The method according to any one of claims 1 to 4, wherein the anti-PD-1 antibody is a chimeric antibody, a humanized antibody, or a human antibody. 如請求項1至5中任一項之方法,其中該抗PD-1抗體或其抗原結合部分選自由以下組成之群: (i)結合於與抗體相同之抗原決定基的抗體或其抗原結合部分,其包含:包含SEQ ID NO:11中所闡述之序列之VH CDR1、包含SEQ ID NO:12中所闡述之序列之VH CDR2以及包含SEQ ID NO:13中所闡述之序列之VH CDR3中的一或多者,及包含SEQ ID NO:14中所闡述之序列之VL CDR1、包含SEQ ID NO:15中所闡述之序列之VL CDR2以及包含SEQ ID NO: 16中所闡述之序列之VL CDR3中的一或多者; (ii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:11中所闡述之序列之VH CDR1、包含SEQ ID NO:12中所闡述之序列之VH CDR2、包含SEQ ID NO:13中所闡述之序列之VH CDR3、包含SEQ ID NO:14中所闡述之序列之VL CDR1、包含SEQ ID NO:15中所闡述之序列之VL CDR2,以及包含SEQ ID NO:16中所闡述之序列之VL CDR3; (iii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:17中所闡述之序列之VH及包含SEQ ID NO:18中所闡述之序列之VL; (iv)抗體,其包含:包含SEQ ID NO:19中所闡述之序列之重鏈及包含SEQ ID NO:20中所闡述之序列之輕鏈; (v)抗體,與(ii)之該抗體交叉競爭; (vi)納武單抗(nivolumab)或其抗原結合部分; (vii)派立珠單抗(pembrolizumab)或其抗原結合部分;以及 (viii) MEDI0608或其抗原結合部分。The method according to any one of claims 1 to 5, wherein the anti-PD-1 antibody or antigen-binding portion thereof is selected from the group consisting of: (i) an antibody or antigen-binding which binds to the same epitope as the antibody Part, which includes: VH CDR1 containing the sequence set forth in SEQ ID NO: 11, VH CDR2 containing the sequence set forth in SEQ ID NO: 12, and VH CDR3 containing the sequence set forth in SEQ ID NO: 13. One or more of, and VL CDR1 containing the sequence set forth in SEQ ID NO: 14, VL CDR2 containing the sequence set forth in SEQ ID NO: 15, and VL containing the sequence set forth in SEQ ID NO: 16. One or more of CDR3; (ii) an antibody or antigen-binding portion thereof, comprising: VH CDR1 comprising the sequence set forth in SEQ ID NO: 11, VH CDR2 comprising the sequence set forth in SEQ ID NO: 12 , VH CDR3 containing the sequence set forth in SEQ ID NO: 13, VL CDR 1 containing the sequence set forth in SEQ ID NO: 14, VL CDR2 containing the sequence set forth in SEQ ID NO: 15, and SEQ ID NO: 15 VL CDR3 of the sequence set forth in NO: 16; (iii) antibody or antigen-binding portion thereof, comprising: comprising SEQ ID VH of the sequence set forth in NO: 17 and VL comprising the sequence set forth in SEQ ID NO: 18; (iv) Antibody, comprising: a heavy chain comprising the sequence set forth in SEQ ID NO: 19 and comprising SEQ The light chain of the sequence described in ID NO: 20; (v) antibody, which cross-competes with the antibody of (ii); (vi) nivolumab (nivolumab) or its antigen-binding portion; (vii) Pelican Mab (pembrolizumab) or its antigen binding portion; and (viii) MEDI0608 or its antigen binding portion. 如請求項1至6中任一項之方法,其中該癌症選自由以下組成之群:黑素瘤癌、肝癌、肝細胞細胞癌、膽管癌、腎癌、前列腺癌、乳癌、結腸癌、肺癌、骨癌、胰臟癌、皮膚癌、頭部或頸部之癌症、皮膚或眼內惡性黑色素瘤、子宮癌、卵巢癌、宮頸癌、子宮內膜癌、直腸癌、肛門區癌、胃癌、睪丸癌、子宮癌、輸卵管癌、子宮內膜癌、子宮頸癌、陰道癌、外陰癌、霍奇金氏疾病(Hodgkin's Disease)、非霍奇金氏淋巴瘤、食道癌、小腸癌、內分泌系統癌、甲狀腺癌、副甲狀腺癌、腎上腺癌、軟組織肉瘤、尿道癌、陰莖癌、包括急性骨髓白血病、慢性骨髓性白血病、急性淋巴母細胞白血病、慢性淋巴球性白血病之慢性或急性白血病、兒童固態腫瘤、淋巴球性淋巴瘤、膀胱癌、腎臟或尿管癌、腎盂癌、中樞神經系統(CNS)腫瘤、原發性CNS淋巴瘤、血管生成腫瘤、膠質母細胞瘤、脊柱軸腫瘤、腦幹神經膠瘤、垂體腺瘤、間皮瘤、卡堡氏肉瘤(Kaposi's sarcoma)、表皮樣癌症、鱗狀細胞癌症、T細胞淋巴瘤、包括由石棉誘導之彼等之環境誘導癌症,以及其任何組合。The method according to any one of claims 1 to 6, wherein the cancer is selected from the group consisting of melanoma cancer, liver cancer, hepatocellular carcinoma, cholangiocarcinoma, renal cancer, prostate cancer, breast cancer, colon cancer, and lung cancer , Bone cancer, pancreatic cancer, skin cancer, head or neck cancer, skin or intraocular malignant melanoma, uterine cancer, ovarian cancer, cervical cancer, endometrial cancer, rectal cancer, anal cancer, stomach cancer, Testicular cancer, uterine cancer, fallopian tube cancer, endometrial cancer, cervical cancer, vaginal cancer, vulvar cancer, Hodgkin's disease, non-Hodgkin's lymphoma, esophageal cancer, small intestine cancer, endocrine system Cancer, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, including acute myelogenous leukemia, chronic myelogenous leukemia, acute lymphoblastic leukemia, chronic or acute leukemia of chronic lymphocytic leukemia, children's solid state Tumor, lymphocytic lymphoma, bladder cancer, kidney or urinary tract cancer, renal pelvis cancer, central nervous system (CNS) tumor, primary CNS lymphoma, angiogenic tumor, gel Blastoma, spinal axis tumor, brain stem glioma, pituitary adenoma, mesothelioma, Kaposi's sarcoma, epidermoid cancer, squamous cell cancer, T cell lymphoma, including asbestos induced Their environment induces cancer, and any combination thereof. 如請求項1至6中任一項之方法,其中該癌症為肺癌、胃腸癌、肝癌、胰臟癌或其任何組合。The method of any one of claims 1 to 6, wherein the cancer is lung cancer, gastrointestinal cancer, liver cancer, pancreatic cancer, or any combination thereof. 如請求項8之方法,其中該肺癌為非小細胞肺癌或小細胞肺癌,該胃腸癌為食道癌、胃癌或結腸直腸癌、該肝癌為肝細胞細胞癌,且該胰臟癌為胰腺癌。The method of claim 8, wherein the lung cancer is non-small cell lung cancer or small cell lung cancer, the gastrointestinal cancer is esophageal cancer, gastric cancer or colorectal cancer, the liver cancer is hepatocellular carcinoma, and the pancreatic cancer is pancreatic cancer. 如請求項9之方法,其中該非小細胞肺癌為非鱗狀非小細胞肺癌或鱗狀非小細胞肺癌。The method of claim 9, wherein the non-small cell lung cancer is non-squamous non-small cell lung cancer or squamous non-small cell lung cancer. 如請求項1至6中任一項之方法,其中該癌症為黑素瘤。The method of any one of claims 1 to 6, wherein the cancer is melanoma. 如請求項1至11中任一項之方法,其中該癌症為漸進性、轉移性及/或不可切除性癌症。The method of any one of claims 1 to 11, wherein the cancer is progressive, metastatic, and / or unresectable cancer. 如請求項1至12中任一項之方法,其中該抗CCR4抗體以至少約0.1、0.3、0.5、0.75、1.0、2.0或3.0 mg/kg投與。The method of any one of claims 1 to 12, wherein the anti-CCR4 antibody is administered at least about 0.1, 0.3, 0.5, 0.75, 1.0, 2.0, or 3.0 mg / kg. 如請求項1至13中任一項之方法,其中該抗CCR4抗體或其抗原結合部分經至少一週一次、至少每兩週一次或至少一週一次接著每兩週一次投與。The method of any one of claims 1 to 13, wherein the anti-CCR4 antibody or antigen-binding portion thereof is administered at least once a week, at least once every two weeks, or at least once a week and then once every two weeks. 如請求項1至13中任一項之方法,其中該抗CCR4抗體或其抗原結合部分在第一次給藥之後一週一次持續四週,接著每兩週一次投與。The method according to any one of claims 1 to 13, wherein the anti-CCR4 antibody or antigen-binding portion thereof is administered once a week for four weeks after the first administration, and then administered once every two weeks. 如請求項1至15中任一項之方法,其中該抗PD-1抗體或其抗原結合部分以至少約1 mg/kg、2.0 mg/kg、3.0 mg/kg、或5 mg/kg或以240 毫克/身體為平調劑量投與。The method of any one of claims 1 to 15, wherein the anti-PD-1 antibody or antigen-binding portion thereof is at least about 1 mg / kg, 2.0 mg / kg, 3.0 mg / kg, or 5 mg / kg or 240 mg / body is administered as a flat dose. 如請求項16之方法,其中該抗PD-1抗體或其抗原結合部分經至少一週一次、至少每兩週一次或至少每三週一次投與。The method of claim 16, wherein the anti-PD-1 antibody or antigen-binding portion thereof is administered at least once a week, at least once every two weeks, or at least once every three weeks. 如請求項1至17中任一項之方法,其中該抗PD-1抗體或其抗原結合部分以2.0 mg/kg每兩週一次投與。The method according to any one of claims 1 to 17, wherein the anti-PD-1 antibody or antigen-binding portion thereof is administered at 2.0 mg / kg every two weeks. 如請求項1至17中任一項之方法,其中該抗PD-1抗體或其抗原結合部分以3.0 mg/kg每兩週一次,或以至少240 毫克/身體作為平調劑量每兩週一次投與。The method according to any one of claims 1 to 17, wherein the anti-PD-1 antibody or antigen-binding portion thereof is once every two weeks at 3.0 mg / kg, or once every two weeks at a flat dose of at least 240 mg / body Cast. 如請求項1至19中任一項之方法,其中將該抗CCR4抗體或其抗原結合部分及該抗PD-1抗體或其抗原結合部分並行地或依序投與至該個體。The method of any one of claims 1 to 19, wherein the anti-CCR4 antibody or antigen-binding portion thereof and the anti-PD-1 antibody or antigen-binding portion thereof are administered to the individual in parallel or sequentially. 如請求項20之方法,其中該抗CCR4抗體或其抗原結合部分在該抗PD-1抗體或其抗原結合部分之前或之後投與。The method of claim 20, wherein the anti-CCR4 antibody or antigen-binding portion thereof is administered before or after the anti-PD-1 antibody or antigen-binding portion thereof. 如請求項1至21中任一項之方法,其中該投藥導致以下效果中之一或多者: (i)減少之CCR4+ T細胞, (ii)減少之調節性T細胞, (iii)增強之腫瘤組織中之PD-L1表現, (iv)減少之PD-1陽性T細胞,及 (v)增加之腫瘤浸潤性淋巴球(TIL)。The method of any one of claims 1 to 21, wherein the administration results in one or more of the following effects: (i) reduced CCR4 + T cells, (ii) reduced regulatory T cells, (iii) enhanced PD-L1 expression in tumor tissue, (iv) decreased PD-1 positive T cells, and (v) increased tumor infiltrating lymphocytes (TIL). 如請求項1至22中任一項之方法,其進一步包含投與額外抗癌劑。The method of any one of claims 1 to 22, further comprising administering an additional anticancer agent. 如請求項23之方法,其中該癌症中之一或多個腫瘤表現PD-L1、PD-L2、CCR4或其任何組合。The method of claim 23, wherein one or more tumors in the cancer exhibit PD-L1, PD-L2, CCR4, or any combination thereof. 一種治療一或多種癌症之方法,該(該等)癌症選自由以下組成之群:非小細胞肺癌、食道癌、肝癌、胃癌及其任何組合,該方法包含在第一次給藥之後將一週一次持續四週之0.1、0.3、0.5、0.75、1.0、2.0或3.0 mg/kg之莫格利珠單抗與每兩週或三週一次之2.0或3.0 mg/kg之納武單抗,或以240 毫克/身體之納武單抗作為平調劑量組合投與至患有該(該等)癌症之個體。A method of treating one or more cancers, the cancer (s) is selected from the group consisting of non-small cell lung cancer, esophageal cancer, liver cancer, gastric cancer, and any combination thereof, the method includes one week after the first administration Mogizumab at 0.1, 0.3, 0.5, 0.75, 1.0, 2.0 or 3.0 mg / kg and nivolumab at 2.0 or 3.0 mg / kg once every two or three weeks, or 240 mg / body of nivolumab is administered as a flat-dose combination to individuals with the cancer (s). 一種治療一或多種癌症之方法,該(該等)癌症選自由以下組成之群:非小細胞肺癌、食道癌、肝癌、胃癌及其任何組合,該方法包含在第一次給藥之後將一週一次持續四週之0.1、0.3、0.5、0.75、1.0、2.0或3.0 mg/kg之莫格利珠單抗,與每兩週一次之2.0 mg/kg之納武單抗,或以240 毫克/身體之納武單抗作為平調劑量組合投與至患有該(該等)癌症之個體。A method of treating one or more cancers, the cancer (s) is selected from the group consisting of non-small cell lung cancer, esophageal cancer, liver cancer, gastric cancer, and any combination thereof, the method includes one week after the first administration Mograizumab at 0.1, 0.3, 0.5, 0.75, 1.0, 2.0 or 3.0 mg / kg for four weeks at a time, and nivolumab at 2.0 mg / kg once every two weeks, or at 240 mg / body Nivolumab is administered as an even-dose combination to individuals with the cancer (s). 一種治療一或多種癌症之方法,該(該等)癌症選自由以下組成之群:非小細胞肺癌、食道癌、肝癌、胃癌及其任何組合,該方法包含在第一次給藥之後將一週一次持續四週之0.1、0.3、0.5、0.75、1.0、2.0或3.0 mg/kg之莫格利珠單抗,與每兩週一次之3.0 mg/kg之納武單抗,或以240 毫克/身體之納武單抗作為平調劑量組合投與至患有該(該等)癌症之個體。A method of treating one or more cancers, the cancer (s) is selected from the group consisting of non-small cell lung cancer, esophageal cancer, liver cancer, gastric cancer, and any combination thereof, the method includes one week after the first administration Mograizumab at 0.1, 0.3, 0.5, 0.75, 1.0, 2.0 or 3.0 mg / kg once every four weeks, and nivolumab at 3.0 mg / kg once every two weeks, or at 240 mg / body Nivolumab is administered as an even-dose combination to individuals with the cancer (s). 一種用於治療罹患癌症之個體的套組,該套組包含: (a)介於0.1至10 mg/kg體重範圍內的劑量之特異性結合於人類PD-1之抗體或其抗原結合部分(「抗PD-1抗體或其抗原結合部分」); (b)介於0.1至10 mg/kg體重範圍內的劑量之特異性結合於人類CCR4之抗體或其抗原結合部分;(「抗CCR4抗體或其抗原結合部分」);以及 (c)在如請求項1至27中任一項之方法中使用該抗PD-1抗體及該抗CCR4抗體的說明書。A kit for treating individuals suffering from cancer, the kit comprising: (a) an antibody or antigen-binding portion thereof that specifically binds to human PD-1 at a dose in the range of 0.1 to 10 mg / kg body weight ( "Anti-PD-1 antibody or antigen-binding portion"); (b) Antibodies or antigen-binding portions that specifically bind to human CCR4 at a dose in the range of 0.1 to 10 mg / kg body weight; ("Anti-CCR4 antibody Or an antigen-binding portion thereof "); and (c) instructions for using the anti-PD-1 antibody and the anti-CCR4 antibody in the method according to any one of claims 1 to 27. 如請求項28使用之套組,其中該抗CCR4抗體或其抗原結合部分選自由以下組成之群: (i)結合於與抗體相同之抗原決定基的抗體或其抗原結合部分,其包含:包含SEQ ID NO:1中所闡述之序列之重鏈可變區(「VH」)互補決定區(CDR)1、包含SEQ ID NO:2中所闡述之序列之VH CDR2及包含SEQ ID NO:3中所闡述之序列之VH CDR3中的一或多者,及包含SEQ ID NO:4中所闡述之序列之輕鏈可變區(「VL」)CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2及包含SEQ ID NO:6中所闡述之序列之VL CDR3中的一或多者; (ii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:1中所闡述之序列之VH CDR1、包含SEQ ID NO:2中所闡述之序列之VH CDR2、包含SEQ ID NO:3中所闡述之序列之VH CDR3、包含SEQ ID NO:4中所闡述之序列之VL CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2,以及包含SEQ ID NO:6中所闡述之序列之VL CDR3; (iii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:7中所闡述之序列之VH及包含SEQ ID NO:8中所闡述之序列之VL; (iv)抗體,其包含:包含SEQ ID NO:9中所闡述之序列之重鏈及包含SEQ ID NO:10中所闡述之序列之輕鏈; (v)抗體或其抗原結合部分,與(ii)之該抗體交叉競爭;以及 (vi)莫格利珠單抗或其抗原結合部分。The kit as used in claim 28, wherein the anti-CCR4 antibody or antigen-binding portion thereof is selected from the group consisting of: (i) an antibody or antigen-binding portion that binds to the same epitope as the antibody, which includes: comprising The heavy chain variable region ("VH") complementarity determining region (CDR) of the sequence set forth in SEQ ID NO: 1, the VH CDR2 containing the sequence set forth in SEQ ID NO: 2 and the SEQ ID NO: 3 One or more of the VH CDR3 of the sequence set forth in, and the light chain variable region ("VL") CDR1 containing the sequence set forth in SEQ ID NO: 4, including the set forth in SEQ ID NO: 5 One or more of the VL CDR2 of the sequence and the VL CDR3 comprising the sequence set forth in SEQ ID NO: 6; (ii) the antibody or antigen-binding portion thereof, comprising: comprising the sequence set forth in SEQ ID NO: 1 VH CDR1, VH CDR2 containing the sequence set forth in SEQ ID NO: 2, VH CDR3 containing the sequence set forth in SEQ ID NO: 3, VL CDR1 containing the sequence set forth in SEQ ID NO: 4, including VL CDR2 of the sequence set forth in SEQ ID NO: 5, and VL CDR3 containing the sequence set forth in SEQ ID NO: 6; (iii) Antibody or antigen binding Part, which comprises: VH comprising the sequence set forth in SEQ ID NO: 7 and VL comprising the sequence set forth in SEQ ID NO: 8; (iv) Antibody, comprising: contains the set forth in SEQ ID NO: 9 The heavy chain of the sequence and the light chain comprising the sequence set forth in SEQ ID NO: 10; (v) the antibody or antigen-binding portion thereof, which cross-competes with the antibody of (ii); and (vi) mogliflozin Anti-or antigen-binding portion. 如請求項28或29使用之套組,其中該抗PD-1抗體或其抗原結合部分選自由以下組成的組: (i)結合於與抗體相同之抗原決定基的抗體或其抗原結合部分,其包含:包含SEQ ID NO:11中所闡述之序列之VH CDR1、包含SEQ ID NO:12中所闡述之序列之VH CDR2以及包含SEQ ID NO:13中所闡述之序列之VH CDR3中的一或多者,及包含SEQ ID NO:14中所闡述之序列之VL CDR1、包含SEQ ID NO:15中所闡述之序列之VL CDR2以及包含SEQ ID NO: 16中所闡述之序列之VL CDR3中的一或多者; (ii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:11中所闡述之序列之VH CDR1、包含SEQ ID NO:12中所闡述之序列之VH CDR2、包含SEQ ID NO:13中所闡述之序列之VH CDR3、包含SEQ ID NO:14中所闡述之序列之VL CDR1、包含SEQ ID NO:15中所闡述之序列之VL CDR2,以及包含SEQ ID NO:16中所闡述之序列之VL CDR3; (iii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:17中所闡述之序列之VH及包含SEQ ID NO:18中所闡述之序列之VL; (iv)抗體,其包含:包含SEQ ID NO:19中所闡述之序列之重鏈及包含SEQ ID NO:20中所闡述之序列之輕鏈; (v)抗體,與(ii)之該抗體交叉競爭; (vi)納武單抗或其抗原結合部分; (vii)派立珠單抗或其抗原結合部分;以及 (viii) MEDI0608或其抗原結合部分。A kit as used in claim 28 or 29, wherein the anti-PD-1 antibody or antigen-binding portion thereof is selected from the group consisting of: (i) an antibody or antigen-binding portion thereof that binds to the same epitope as the antibody, It contains: one of VH CDR1 containing the sequence set forth in SEQ ID NO: 11, one of VH CDR2 containing the sequence set forth in SEQ ID NO: 12, and VH CDR3 containing the sequence set forth in SEQ ID NO: 13 Or more, and in VL CDR1 containing the sequence set forth in SEQ ID NO: 14, VL CDR2 containing the sequence set forth in SEQ ID NO: 15, and VL CDR3 containing the sequence set forth in SEQ ID NO: 16. One or more of; (ii) an antibody or antigen-binding portion thereof, comprising: VH CDR1 comprising the sequence set forth in SEQ ID NO: 11, VH CDR2 comprising the sequence set forth in SEQ ID NO: 12, comprising VH CDR3 of the sequence set forth in SEQ ID NO: 13, VL CDR 1 containing the sequence set forth in SEQ ID NO: 14, VL CDR2 containing the sequence set forth in SEQ ID NO: 15, and SEQ ID NO: VL CDR3 of the sequence set forth in 16; (iii) antibody or antigen-binding portion thereof, comprising: comprising SEQ ID NO: VH of the sequence set forth in 17 and VL comprising the sequence set forth in SEQ ID NO: 18; (iv) Antibodies comprising: a heavy chain comprising the sequence set forth in SEQ ID NO: 19 and comprising SEQ ID NO : Light chain of the sequence described in 20; (v) antibody, cross-competing with the antibody of (ii); (vi) nivolumab or its antigen-binding portion; (vii) peclizumab or its antigen A binding portion; and (viii) MEDI0608 or an antigen binding portion thereof. 一種用於治療癌症之醫藥組合物,其包含(a)特異性結合於人類PD-1之抗體或其抗原結合部分(「抗PD-1抗體或其抗原結合部分」)及特異性結合於人類CCR4之抗體或其抗原結合部分(抗CCR4抗體或其抗原結合部分)。A pharmaceutical composition for treating cancer, comprising (a) an antibody or antigen-binding portion that specifically binds to human PD-1 ("anti-PD-1 antibody or antigen-binding portion thereof") and a specific binding to human CCR4 antibody or antigen binding portion thereof (anti-CCR4 antibody or antigen binding portion thereof). 如請求項31使用之醫藥組合物,其中該抗CCR4抗體或其抗原結合部分選自由以下組成之群: (i)結合於與抗體相同之抗原決定基的抗體或其抗原結合部分,其包含:包含SEQ ID NO:1中所闡述之序列之重鏈可變區(「VH」)互補決定區(CDR)1、包含SEQ ID NO:2中所闡述之序列之VH CDR2及包含SEQ ID NO:3中所闡述之序列之VH CDR3中的一或多者,及包含SEQ ID NO:4中所闡述之序列之輕鏈可變區(「VL」)CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2及包含SEQ ID NO:6中所闡述之序列之VL CDR3中的一或多者; (ii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:1中所闡述之序列之VH CDR1、包含SEQ ID NO:2中所闡述之序列之VH CDR2、包含SEQ ID NO:3中所闡述之序列之VH CDR3、包含SEQ ID NO:4中所闡述之序列之VL CDR1、包含SEQ ID NO:5中所闡述之序列之VL CDR2,以及包含SEQ ID NO:6中所闡述之序列之VL CDR3; (iii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:7中所闡述之序列之VH及包含SEQ ID NO:8中所闡述之序列之VL; (iv)抗體,其包含:包含SEQ ID NO:9中所闡述之序列之重鏈及包含SEQ ID NO:10中所闡述之序列之輕鏈; (v)抗體或其抗原結合部分,與(ii)之該抗體交叉競爭;以及 (vi)莫格利珠單抗或其抗原結合部分。The pharmaceutical composition as used in claim 31, wherein the anti-CCR4 antibody or antigen-binding portion thereof is selected from the group consisting of: (i) an antibody or antigen-binding portion thereof that binds to the same epitope as the antibody, which includes: A heavy chain variable region ("VH") complementarity determining region (CDR) comprising the sequence set forth in SEQ ID NO: 1, a VH CDR2 comprising the sequence set forth in SEQ ID NO: 2, and a SEQ ID NO: One or more of the VH CDR3 of the sequence set forth in 3, and the light chain variable region ("VL") CDR1 containing the sequence set forth in SEQ ID NO: 4, including the set forth in SEQ ID NO: 5 VL CDR2 of the sequence and one or more of the VL CDR3 comprising the sequence set forth in SEQ ID NO: 6; (ii) the antibody or antigen-binding portion thereof, comprising: comprising the set forth in SEQ ID NO: 1 VH CDR1 of the sequence, VH CDR2 of the sequence set forth in SEQ ID NO: 2, VH CDR3 of the sequence set forth in SEQ ID NO: 3, VL CDR1 of the sequence set forth in SEQ ID NO: 4 VL CDR2 containing the sequence set forth in SEQ ID NO: 5, and VL CDR3 containing the sequence set forth in SEQ ID NO: 6; (iii) antibody or anti- The original binding portion, comprising: VH comprising the sequence set forth in SEQ ID NO: 7 and VL comprising the sequence set forth in SEQ ID NO: 8; (iv) Antibody, comprising: containing SEQ ID NO: 9 The heavy chain of the sequence set forth and the light chain comprising the sequence set forth in SEQ ID NO: 10; (v) the antibody or antigen-binding portion thereof, cross-compete with the antibody of (ii); and (vi) Mogley Zumab or its antigen-binding portion. 如請求項31或32使用之醫藥組合物,其中該抗PD-1抗體或其抗原結合部分選自由以下組成之群: (i)結合於與抗體相同之抗原決定基的抗體或其抗原結合部分,其包含:包含SEQ ID NO:11中所闡述之序列之VH CDR1、包含SEQ ID NO:12中所闡述之序列之VH CDR2以及包含SEQ ID NO:13中所闡述之序列之VH CDR3中的一或多者,及包含SEQ ID NO:14中所闡述之序列之VL CDR1、包含SEQ ID NO:15中所闡述之序列之VL CDR2以及包含SEQ ID NO: 16中所闡述之序列之VL CDR3中的一或多者; (ii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:11中所闡述之序列之VH CDR1、包含SEQ ID NO:12中所闡述之序列之VH CDR2、包含SEQ ID NO:13中所闡述之序列之VH CDR3、包含SEQ ID NO:14中所闡述之序列之VL CDR1、包含SEQ ID NO:15中所闡述之序列之VL CDR2,以及包含SEQ ID NO:16中所闡述之序列之VL CDR3; (iii)抗體或其抗原結合部分,其包含:包含SEQ ID NO:17中所闡述之序列之VH及包含SEQ ID NO:18中所闡述之序列之VL; (iv)抗體,其包含:包含SEQ ID NO:19中所闡述之序列之重鏈及包含SEQ ID NO:20中所闡述之序列之輕鏈; (v)抗體或其抗原結合部分,與(ii)之該抗體交叉競爭; (vi)納武單抗或其抗原結合部分; (vii)派立珠單抗或其抗原結合部分;以及 (viii) MEDI0608或其抗原結合部分。The pharmaceutical composition for use according to claim 31 or 32, wherein the anti-PD-1 antibody or antigen-binding portion thereof is selected from the group consisting of: (i) an antibody or antigen-binding portion thereof that binds to the same epitope as the antibody Which includes: VH CDR1 containing the sequence set forth in SEQ ID NO: 11, VH CDR2 containing the sequence set forth in SEQ ID NO: 12, and VH CDR3 containing the sequence set forth in SEQ ID NO: 13 One or more, and VL CDR1 containing the sequence set forth in SEQ ID NO: 14, VL CDR2 containing the sequence set forth in SEQ ID NO: 15, and VL CDR3 containing the sequence set forth in SEQ ID NO: 16. One or more of; (ii) an antibody or antigen-binding portion thereof, comprising: VH CDR1 comprising the sequence set forth in SEQ ID NO: 11, VH CDR2 comprising the sequence set forth in SEQ ID NO: 12, VH CDR3 containing the sequence set forth in SEQ ID NO: 13, VL CDR 1 containing the sequence set forth in SEQ ID NO: 14, VL CDR2 containing the sequence set forth in SEQ ID NO: 15, and SEQ ID NO : VL CDR3 of the sequence described in 16; (iii) antibody or antigen-binding portion thereof, comprising: comprising S VH of the sequence set forth in EQ ID NO: 17 and VL containing the sequence set forth in SEQ ID NO: 18; (iv) antibodies, which include: heavy chains containing the sequence set forth in SEQ ID NO: 19 and A light chain comprising the sequence set forth in SEQ ID NO: 20; (v) an antibody or antigen-binding portion thereof that cross-competes with the antibody of (ii); (vi) nivolumab or its antigen-binding portion; (vii ) Palivizumab or its antigen binding portion; and (viii) MEDI0608 or its antigen binding portion. 如請求項31至33中任一項使用之醫藥組合物,其用於治療癌症。The pharmaceutical composition used in any one of claims 31 to 33, which is used to treat cancer. 如請求項34使用之醫藥組合物,其中該癌症選自由以下組成之群:黑素瘤癌、肝癌、肝細胞細胞癌、膽管癌、腎癌、前列腺癌、乳癌、結腸癌、肺癌、骨癌、胰臟癌、皮膚癌、頭部或頸部之癌症、皮膚或眼內惡性黑色素瘤、子宮癌、卵巢癌、宮頸癌、子宮內膜癌、直腸癌、肛門區癌、胃癌、睪丸癌、子宮癌、輸卵管癌、子宮內膜癌、子宮頸癌、陰道癌、外陰癌、霍奇金氏疾病、非霍奇金氏淋巴瘤、食道癌、小腸癌、內分泌系統癌、甲狀腺癌、副甲狀腺癌、腎上腺癌、軟組織肉瘤、尿道癌、陰莖癌、包括急性骨髓白血病、慢性骨髓性白血病、急性淋巴母細胞白血病、慢性淋巴球性白血病之慢性或急性白血病、兒童固態腫瘤、淋巴球性淋巴瘤、膀胱癌、腎臟或尿管癌、腎盂之癌瘤、中樞神經系統(CNS)腫瘤、原發性CNS淋巴瘤、血管生成腫瘤、膠質母細胞瘤、脊柱軸腫瘤、腦幹神經膠瘤、垂體腺瘤、間皮瘤、卡堡氏肉瘤、表皮樣癌症、鱗狀細胞癌症、T細胞淋巴瘤、包括由石棉誘導之彼等之環境誘導癌症,以及其任何組合。The pharmaceutical composition as claimed in claim 34, wherein the cancer is selected from the group consisting of melanoma cancer, liver cancer, hepatocellular carcinoma, cholangiocarcinoma, kidney cancer, prostate cancer, breast cancer, colon cancer, lung cancer, bone cancer , Pancreatic cancer, skin cancer, head or neck cancer, skin or intraocular malignant melanoma, uterine cancer, ovarian cancer, cervical cancer, endometrial cancer, rectal cancer, anal cancer, stomach cancer, testicular cancer, Uterine cancer, fallopian tube cancer, endometrial cancer, cervical cancer, vaginal cancer, vulvar cancer, Hodgkin's disease, non-Hodgkin's lymphoma, esophageal cancer, small intestine cancer, endocrine system cancer, thyroid cancer, parathyroid Cancer, adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, including acute myeloid leukemia, chronic myelogenous leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic or acute leukemia, solid tumors of children, lymphocytic lymphoma , Bladder cancer, kidney or urinary tract cancer, carcinoma of the renal pelvis, central nervous system (CNS) tumor, primary CNS lymphoma, angiogenic tumor, glioblastoma , Spinal axis tumors, brainstem gliomas, pituitary adenoma, mesothelioma, Kaposi's sarcoma, epidermoid cancer, squamous cell cancer, T-cell lymphoma, including asbestos-induced environment-induced cancer And any combination thereof.
TW106142006A 2016-11-30 2017-11-30 Method of treating cancer using anti-ccr4 antibody and anti-pd-1 antibody TW201825119A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662428468P 2016-11-30 2016-11-30
US62/428,468 2016-11-30
US201762554486P 2017-09-05 2017-09-05
US62/554,486 2017-09-05

Publications (1)

Publication Number Publication Date
TW201825119A true TW201825119A (en) 2018-07-16

Family

ID=60888559

Family Applications (1)

Application Number Title Priority Date Filing Date
TW106142006A TW201825119A (en) 2016-11-30 2017-11-30 Method of treating cancer using anti-ccr4 antibody and anti-pd-1 antibody

Country Status (2)

Country Link
TW (1) TW201825119A (en)
WO (1) WO2018101448A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2012345746B2 (en) 2011-12-01 2016-12-08 Chemocentryx, Inc. Substituted anilines as CCR(4) antagonists
MX2018000621A (en) 2015-07-13 2018-05-11 Cytomx Therapeutics Inc Anti-pd-1 antibodies, activatable anti-pd-1 antibodies, and methods of use thereof.
EP3843734A4 (en) * 2018-08-29 2022-05-25 ChemoCentryx, Inc. Combination therapy using c-c chemokine receptor 4 (ccr4) antagonists and one or more immune checkpoint inhibitors

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
EP2275540B1 (en) 1999-04-09 2016-03-23 Kyowa Hakko Kirin Co., Ltd. Method for controlling the activity of immunologically functional molecule
JP4896327B2 (en) 1999-08-23 2012-03-14 ダナ−ファーバー キャンサー インスティテュート,インコーポレイテッド PD-1, B7-4 receptors and uses thereof
MXPA03002974A (en) 2000-10-06 2004-05-05 Kyowa Hakko Kogyo Kk Cells producing antibody compositions.
US7317091B2 (en) 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
CA2508660C (en) 2002-12-23 2013-08-20 Wyeth Antibodies against pd-1 and uses therefor
CA2542046A1 (en) 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Fused protein composition
KR101149242B1 (en) 2004-01-12 2012-05-25 어플라이드 몰리큘라 에볼류션, 인코포레이티드 Fc region variants
US20060034841A1 (en) 2004-06-07 2006-02-16 Kyowa Hakko Kogyo Co., Ltd. Method of depleting regulatory T cell
SI2439273T1 (en) 2005-05-09 2019-05-31 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US7923538B2 (en) 2005-07-22 2011-04-12 Kyowa Hakko Kirin Co., Ltd Recombinant antibody composition
KR101586617B1 (en) 2007-06-18 2016-01-20 머크 샤프 앤 도메 비.브이. Antibodies to human programmed death receptor PD-1
EP2262837A4 (en) 2008-03-12 2011-04-06 Merck Sharp & Dohme Pd-1 binding proteins
PE20110435A1 (en) 2008-08-25 2011-07-20 Amplimmune Inc ANTAGONIST COMPOSITIONS OF PD-1
DK2699264T3 (en) 2011-04-20 2018-06-25 Medimmune Llc ANTIBODIES AND OTHER MOLECULES BINDING B7-H1 AND PD-1
JP6448533B2 (en) 2012-05-15 2019-01-09 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Cancer immunotherapy by disrupting PD-1 / PD-L1 signaling

Also Published As

Publication number Publication date
WO2018101448A1 (en) 2018-06-07

Similar Documents

Publication Publication Date Title
KR102606252B1 (en) How to Treat Cancer with Anti-PD-1 Antibodies
CN106103486B (en) anti-OX 40 antibodies and methods of use
US11866509B2 (en) Humanized antibodies against CEACAM1
JP6552621B2 (en) Anti-PD-1 antibody and method of using the same
JP5560301B2 (en) Therapeutic and diagnostic methods and compositions targeting 4IG-B7-H3 and its corresponding NK cell receptor
JP2018521019A (en) Method of treating cancer using anti-OX40 antibody
KR20170074246A (en) Methods and biomarkers for predicting efficacy and evaluation of an 0x40 agonist treatment
CN108350505A (en) Genetic marker for measuring ICOS expression
TW201622748A (en) Therapeutic combinations and methods for treating neoplasia
KR20190008962A (en) Use of anti-PD-1 antibodies in combination with anti-CD30 antibodies in the treatment of lymphoma
WO2021228178A1 (en) Compositions and methods for treating cancer
CN111094982A (en) TIM-3 antagonists for the treatment and diagnosis of cancer
Borrie et al. T Lymphocyte–Based Cancer Immunotherapeutics
US20230131598A1 (en) Combination treatment for cancer
CN109475633A (en) With receiving military MAbs blocking PD-1 in intractable Hodgkin lymphoma
TW201825119A (en) Method of treating cancer using anti-ccr4 antibody and anti-pd-1 antibody
JP2023554422A (en) Multispecific antibodies for cancer treatment
EP3765519B1 (en) Anti-cxcr4 antibody combined with activated and expanded natural killer cells for cancer immunotherapy
CN110603447A (en) Compositions and methods for treating cancer with anti-renalase antibodies and anti-PD 1 antibodies
US20240092934A1 (en) Assessment of ceacam1 expression on tumor infiltrating lymphocytes
US20230365691A1 (en) Use of anti-pd-1 antibody in treatment of nasopharyngeal carcinoma
AU2022377628A1 (en) Methods of treating cancer with a combination of an anti-pd-1 antibody and an anti-cd30 antibody-drug conjugate
EP4136105A1 (en) Combination treatment for cancer based upon an icos antibody and a pd-l1 antibody tgf-beta-receptor fusion protein
JP2023521228A (en) Cancer combination therapy