TW201823471A - Methods of treating patients with a retinoic acid receptor-[alpha] agonist and an anti-CD38 antibody - Google Patents

Methods of treating patients with a retinoic acid receptor-[alpha] agonist and an anti-CD38 antibody Download PDF

Info

Publication number
TW201823471A
TW201823471A TW106134613A TW106134613A TW201823471A TW 201823471 A TW201823471 A TW 201823471A TW 106134613 A TW106134613 A TW 106134613A TW 106134613 A TW106134613 A TW 106134613A TW 201823471 A TW201823471 A TW 201823471A
Authority
TW
Taiwan
Prior art keywords
rara
tamibarotene
irf8
level
individual
Prior art date
Application number
TW106134613A
Other languages
Chinese (zh)
Inventor
邁可 R 麥基翁
辛蒂 柯林斯
馬修 盧卡斯 伊頓
馬修 G 關瑟
萳 柯
杰洛米 羅佩茲
玫葳 陳
大衛 A 奧蘭多
凱薩琳 奧斯珍
克里斯多佛 菲奥里
艾蜜莉 裴頓 李
克里斯堤安 C 費理茲
Original Assignee
美商錫羅斯製藥公司
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 美商錫羅斯製藥公司 filed Critical 美商錫羅斯製藥公司
Publication of TW201823471A publication Critical patent/TW201823471A/en

Links

Classifications

    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Genetics & Genomics (AREA)
  • Analytical Chemistry (AREA)
  • Microbiology (AREA)
  • Pathology (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Biophysics (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • Hospice & Palliative Care (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The invention provides methods that define cellular populations that are sensitive to RARA agonists and identify patient subgroups that will benefit from treatment with RARA agonists in combination with another therapy, more specifically an anti-CD38 therapy. The invention also provides packaged pharmaceutical compositions that comprise a RARA agonist and instructions for determining if such combination therapy is suitable for use in treatment.

Description

以視黃酸受體-α促效劑及抗CD38抗體治療病患之方法Method for treating patients with retinoic acid receptor-α agonist and anti-CD38 antibody

類視黃素為一類結構上與維生素A相關之化合物,包含天然及合成化合物。已發現若干系列類視黃素臨床上適用於皮膚及致癌疾病之治療。視黃酸及其其他天然存在類視黃素類似物(9-順視黃酸、全反3,4-二去氫視黃酸、4-側氧基視黃酸及視黃醇)為多效性調節化合物,其調節廣泛多種炎症細胞、免疫及結構細胞之結構及功能。其為肺中之上皮細胞增殖、分化及形態發生之重要調節劑。類視黃素經由一系列激素核受體發揮其生物學效應,該等激素核受體為屬於類固醇/甲狀腺受體超家族之配位體誘導性轉錄因子。 類視黃素受體分為兩個家族,視黃酸受體(RAR)及類視黃素X受體(RXR),各自由三個不同亞型(α、β及γ)組成。RAR基因家族之各亞型編碼可變數目之同功異型物,其由兩個初級RNA轉錄物之差異剪接產生。全反視黃酸為視黃酸受體之生理激素,且以大致相等的親和力與全部三個RAR亞型結合,但不與以9-順視黃酸為天然配位體之RXR受體結合。類視黃素具有消炎作用,改變上皮細胞分化之進程,及抑制基質細胞基質產生。此等特性已使得針對諸如牛皮癬、痤瘡及肥厚性皮膚疤痕之皮膚病症的局部及全身類視黃素治療之進展。其他應用包括急性前髓細胞性白血病、腺癌及鱗狀細胞癌及肝纖維化之控制。 類視黃素之治療用途中之侷限性源於用天然存在類視黃素、全反視黃酸及9-順視黃酸觀測到的相對毒性。此等天然配位體就RAR亞型而言為非選擇性的,且因此在整個身體中具有通常有毒的多效性作用。 已描述選擇性地或專門與RAR或RXR受體或與一個類別內的特異性亞型(α、β、γ)相互作用之各種類視黃素。RARA特異性促效劑對癌症治療具有廣闊的前景,且許多已進入人體臨床試驗。然而,僅一種RARA特異性促效劑他米巴羅汀(tamibarotene)曾經經批准用於治療癌症。此外,他米巴羅汀僅在日本經批准且僅經批准用於治療急性前髓細胞性白血病,儘管在美國及歐洲進行試驗。RARA促效劑在癌症中之理論療效與此類藥劑之監管批准的缺乏之間的脫節引發此類促效劑為何在人體內無效且不安全的問題。因此,需要更好地瞭解為何RARA促效劑仍未滿足其治療潛力。 分化簇38 (CD38)為在表面上、主要在白血球上表現之蛋白質,且視為指示分化開始之細胞表面標記。其充當在細胞信號傳導中起作用之環ADP-核糖羥化酶(Mehta, K. & Cheema, S.,Leuk. Lymphoma , 32, 441-449 (1999))。通常發現其在B細胞及漿細胞譜系之細胞中高度表現。在多發性骨髓瘤中,病患子組具有較高CD38表現,其已導致諸如達土木單抗(daratumumab)之抗CD38治療性抗體的進展(Lokhorst, H. M.等人,N . Engl . J . Med ., 373, 1207-1219 (2015); de Weers, M.等人,J . Immunol ., 186, 1840-1848 (2011))。因此,可選擇性地靶向表現CD38之癌細胞以用於藉由免疫系統使用此等治療性抗體來消除。在多發性骨髓瘤中,達土木單抗在較高CD38表現(CD38hi )的腫瘤細胞中最有效,而CD38表現之較低水準使得治療性抗體作用較小至沒有作用(Nijhof, I. S.等人,Blood , 128, 959-970 (2016))。此係為何正常白血球或初始CD38表現較低(或隨抗藥性機制變低)的多發性骨髓瘤細胞並未藉由抗CD38治療得到有效清除。 AML中之CD38表現視為一般較低(CD38lo ),但病患子組中之表現視為中等(CD38dim )。天然存在CD38hi AML細胞為不典型的,且AML之陽性百分比一般較低。因此,認為AML不可能對抗CD38治療起反應,已用達土木單抗單藥療法證明之研究結果(Dos Santos, C.等人,Blood , 124, 2312-2312 (2014))。 基因組技術之最新進展及對基因調節迴路之理解已導致超強化子之發現。鑒於指定組織中之許多基因或癌症類型可藉由接近基因編碼區之強化子的存在來調節,少數此等基因表示高度不對稱及轉錄標記及機構相對於所有其他活性基因之不成比例地大量負載。最新發現表明,此類強化子結合至與含有其之細胞之功能及存活特定相關的基因。因此,超強化子與基因之相關性指示該基因對該細胞之存活之相對重要性。Retinoids are a class of structural compounds related to vitamin A, including natural and synthetic compounds. Several series of retinoids have been found to be clinically suitable for the treatment of skin and carcinogenic diseases. Retinoic acid and its other naturally occurring retinoid analogs (9-cis retinoic acid, all-trans 3,4-didehydroretinoic acid, 4-oxo-retinoic acid and retinol) are mostly A potency-modulating compound that regulates the structure and function of a wide variety of inflammatory cells, immune and structural cells. It is an important regulator of epithelial cell proliferation, differentiation and morphogenesis in the lung. Retinoids exert their biological effects through a series of hormone nuclear receptors, which are ligand-inducible transcription factors belonging to the steroid / thyroid receptor superfamily. Retinoid receptors are divided into two families, retinoic acid receptors (RAR) and retinoid X receptors (RXR), each consisting of three different subtypes (α, β, and γ). Each subtype of the RAR gene family encodes a variable number of isoforms, which results from the differential splicing of two primary RNA transcripts. All-trans retinoic acid is a physiological hormone of the retinoic acid receptor and binds to all three RAR subtypes with approximately equal affinity, but does not bind to the RXR receptor with 9-cis retinoic acid as the natural ligand . Retinoids have anti-inflammatory effects, change the process of epithelial cell differentiation, and inhibit stromal cell matrix production. These properties have led to the progress of local and systemic retinoid treatments for skin disorders such as psoriasis, acne and hypertrophic skin scars. Other applications include the control of acute promyelocytic leukemia, adenocarcinoma and squamous cell carcinoma, and liver fibrosis. Limitations in the therapeutic use of retinoids stem from the relative toxicity observed with naturally occurring retinoids, all-trans retinoic acid, and 9-cis retinoic acid. These natural ligands are non-selective with respect to RAR subtypes, and therefore have a generally toxic pleiotropic effect throughout the body. Various retinoids have been described that interact selectively or exclusively with RAR or RXR receptors or with specific subtypes (α, β, γ) within a class. RARA-specific agonists have broad prospects for cancer treatment, and many have entered human clinical trials. However, only one RARA-specific agonist, tamibarotene, has been approved for the treatment of cancer. In addition, Tamibarotene is only approved in Japan and is only approved for the treatment of acute promyelocytic leukemia, despite trials in the United States and Europe. The disconnect between the theoretical efficacy of RARA agonists in cancer and the lack of regulatory approval for such agents raises the question of why such agonists are ineffective and unsafe in the human body. Therefore, there is a need to better understand why RARA agonists have not yet met their therapeutic potential. Differentiation cluster 38 (CD38) is a protein that appears on the surface, mainly on white blood cells, and is regarded as a cell surface marker that indicates the beginning of differentiation. It acts as a cyclic ADP-ribose hydroxylase that plays a role in cell signaling (Mehta, K. & Cheema, S., Leuk. Lymphoma , 32, 441-449 (1999)). It is usually found to be highly expressed in cells of the B cell and plasma cell lineages. In multiple myeloma, patients with high CD38 expression subset, which has led to the progress of civil engineering, such as monoclonal antibody (daratumumab) of anti-CD38 therapeutic antibody (Lokhorst, HM et al., N. Engl. J. Med ., 373, 1207-1219 (2015) ;. de Weers, M. et al., J Immunol, 186, 1840-1848 ( 2011)).. Therefore, cancer cells expressing CD38 can be selectively targeted for elimination by the immune system using these therapeutic antibodies. In multiple myeloma, datulimumab is most effective in tumor cells with higher CD38 expression (CD38 hi ), and the lower level of CD38 expression makes the therapeutic antibody less effective to no effect (Nijhof, IS et al. , Blood , 128, 959-970 (2016)). This is why multiple myeloma cells with normal white blood cells or low initial CD38 performance (or decreased with resistance mechanism) have not been effectively cleared by anti-CD38 treatment. The performance of CD38 in AML was considered generally low (CD38 lo ), but the performance in the patient subgroup was considered moderate (CD38 dim ). Naturally occurring CD38 hi AML cells are atypical, and the percentage of positive AML is generally low. Therefore, it is believed that AML is unlikely to respond to anti-CD38 treatment, which has been proved by the study of dalmatumab monotherapy (Dos Santos, C. et al., Blood , 124, 2312-2312 (2014)). Recent advances in genomic technology and understanding of gene regulatory circuits have led to the discovery of superenhancers. Given that many genes or cancer types in a given tissue can be regulated by the presence of enhancers close to the gene coding region, a few of these genes represent a high degree of asymmetry and a disproportionately large load of transcriptional markers and mechanisms relative to all other active genes . Recent findings indicate that such enhancers bind to genes specifically related to the function and survival of cells containing them. Therefore, the correlation between the superenhancer and the gene indicates the relative importance of the gene for the survival of the cell.

本發明提供用於藉由向個體投與類視黃素(例如,視黃酸受體α特異性促效劑,例如他米巴羅汀)與抗CD38抗體(例如,達土木單抗)之組合來治療患有血液癌症(例如,多發性骨髓瘤(MM)、急性骨髓性白血病(AML)、骨髓發育不良症候群(MDS)、多形性大細胞淋巴瘤(ALCL)、B細胞急性淋巴母細胞白血病(BALL)、B細胞非霍奇金淋巴瘤(B-cell non-Hodgkin lymphoma) (BNHL)、伯基特氏淋巴瘤(Burkitt's lymphoma)、慢性骨髓性淋巴瘤(CML)、彌漫性大型B細胞淋巴瘤(DLBCL)、霍奇金淋巴瘤(Hodgkin lymphoma)或T細胞急性淋巴母細胞白血病(TALL))之個體的方法,其中血液癌症:a)對作為單藥療法之CD38抑制劑無反應;且/或b)表徵為CD38- 、CD38lo 或CD38dim 。 本發明亦提供包含例如當RARA生物標記或IRF8生物標記呈現高於預定臨限值位準時偵測RARA生物標記或IRF8生物標記並投與與CD38特異性抗體組合的RARA促效劑以治療不屬於急性前髓細胞性白血病類型之急性骨髓性白血病(「非APL AML」)、骨髓發育不良症候群(「MDS」)或多發性骨髓瘤(「MM」)之方法。偵測RARA生物標記或IRF8生物標記包括測定一或多種RARAIRF8 基因組分或產物之存在、位準、形式及/或活性,包括例如RARAIRF8 超強化子強度、序數分級或盛行分級及RARA或IRF8 mRNA位準或盛行分級。本發明表明含有與RARAIRF8 基因相關聯的超強化子中之一或多者之細胞(例如,癌細胞或來自患有非APL AML、MDS或MM之個體的細胞)更易受RARA促效劑(諸如他米巴羅汀)與CD38特異性抗體組合之作用影響。 本發明之各種實施例、態樣及替代方案解決定義何種細胞群體對視黃酸受體α (「RARA」)之促效劑與CD38特異性抗體的組合敏感、鑑別將受益於RARA促效劑及CD38特異性抗體之治療之病患子群(例如,分級病患以治療;將反應者與未反應者分離)以及提供針對此類病患子群之治療療法的問題。解決方案至少部分地基於吾人之發現:RARA生物標記或IRF8生物標記在非APL AML、MDS或MM中之較高表現指示此細胞將對用RARA促效劑及CD38特異性抗體之治療起反應。 在一第一實施例中,本發明係關於一種診斷及治療患有非APL AML、MDS或MM之個體之方法,該方法包含:a)基於以下診斷個體是否患有RARA促效劑敏感(例如,他米巴羅汀敏感)型疾病:i)預先測定為等於或大於預定臨限值之來自個體的患病細胞之樣本中之視黃酸受體α mRNA位準;及/或ii)預先測定為等於或大於預定臨限值之來自個體的患病細胞之樣本中之IRF8 mRNA位準;b)向個體投與可有效治療疾病的一定量之RARA促效劑(例如,他米巴羅汀);以及c)向個體共投與CD38特異性抗體。 在第一實施例之一些態樣中,RARA促效劑為他米巴羅汀。在第一實施例之一些態樣中,CD38特異性抗體為達土木單抗。在第一實施例之一些態樣中,在投與CD38特異性抗體之前,向個體投與他米巴羅汀一段時間;且在開始投與他米巴羅汀之後,僅在個體中之CD38位準已測定為CD38hi 時共投與CD38特異性抗體。在第一實施例之一些態樣中,在開始投與他米巴羅汀之後的6至72小時之間測定個體中之CD38位準。 在一第二實施例中,本發明提供一種治療患有非APL AML、MDS或MM之人類個體之方法,其中個體的患病細胞中之視黃酸受體α mRNA之位準及/或IRF8 mRNA之位準已經測定具有等於或大於預定臨限值之視黃酸受體α mRNA位準,或等於或大於預定臨限值之IRF8 mRNA位準,該方法包含以下步驟:向個體投與有效治療疾病的一定量之RARA促效劑(例如,他米巴羅汀);以及向個體共投與CD38特異性抗體。 在第二實施例之一些態樣中,RARA促效劑為他米巴羅汀。在第二實施例之一些態樣中,CD38特異性抗體為達土木單抗。在第二實施例之一些態樣中,在投與CD38特異性抗體之前,向個體投與他米巴羅汀一段時間;且在開始投與他米巴羅汀之後,僅在個體中之CD38位準已測定為CD38hi 時共投與CD38特異性抗體。在第二實施例之一些態樣中,在開始投與他米巴羅汀之後的6至72小時之間測定個體中之CD38位準。 在一第三實施例中,本發明提供一種診斷及治療患有non APL AML、MDS或MM之人類個體之方法,該方法包含:a)基於視黃酸受體α mRNA之位準及/或IRF8 mRNA之位準診斷個體是否患有RARA促效劑敏感(例如,他米巴羅汀敏感)型疾病,該等位準中之任一者或兩者預先測定為存在於來自個體之患病細胞的樣本中;以及b)向個體投與療法,其中i)若位準指示疾病對他米巴羅汀敏感,則療法包含投與有效治療疾病的一定量之RARA促效劑(例如,他米巴羅汀)及共投與CD38特異性抗體;且ii)若位準指示疾病對RARA促效劑不敏感(例如,對他米巴羅汀不敏感),則療法包含投與除RARA促效劑外(例如,除他米巴羅汀外)之藥劑。 在第三實施例之一些態樣中,視黃酸受體α mRNA及/或IRF8 mRNA位準指示,若其大於預定臨限值,則疾病對RARA促效劑敏感(例如,對他米巴羅汀敏感),並且若其小於預定臨限值,則疾病對RARA促效劑不敏感(例如,對他米巴羅汀不敏感)。在第三實施例之一些態樣中,RARA促效劑為他米巴羅汀。在第三實施例之一些態樣中,CD38特異性抗體為達土木單抗。在第三實施例之一些態樣中,若視黃酸受體α mRNA或IRF8 mRNA之位準指示疾病對RARA促效劑敏感(例如,對他米巴羅汀敏感),則在投與CD38特異性抗體之前,向個體投與RARA促效劑(例如,他米巴羅汀)一段時間;且在開始投與RARA促效劑(例如,他米巴羅汀)之後,僅在個體中之CD38位準經測定為CD38hi 時共投與CD38特異性抗體。在第三實施例之一些態樣中,在開始投與RARA促效劑(例如,他米巴羅汀)之後的6至72小時之間測定個體中之CD38位準。 在一第四實施例中,本發明提供一種用於治療患有血液癌症之個體的方法,該個體在無類視黃素存在下對用抗CD38抗體之治療無反應,其中該方法包含向個體共投與類視黃素及抗CD38抗體。在第四實施例之一些態樣中,個體患有選自以下之血液癌症:多發性骨髓瘤(MM)、急性骨髓性白血病(AML)、骨髓發育不良症候群(MDS)、多形性大細胞淋巴瘤(ALCL)、B細胞急性淋巴母細胞白血病(BALL)、B細胞非霍奇金淋巴瘤(BNHL)、伯基特氏淋巴瘤、慢性骨髓性淋巴瘤(CML)、彌漫性大型B細胞淋巴瘤(DLBCL)、霍奇金淋巴瘤或T細胞急性淋巴母細胞白血病(TALL)。在第四實施例之一些更特定態樣中,個體患有選自以下之血液癌症:多形性大細胞淋巴瘤(ALCL)、B細胞急性淋巴母細胞白血病(BALL)、B細胞非霍奇金淋巴瘤(BNHL)、伯基特氏淋巴瘤、慢性骨髓性淋巴瘤(CML)、彌漫性大型B細胞淋巴瘤(DLBCL)、霍奇金淋巴瘤或T細胞急性淋巴母細胞白血病(TALL)。 在第四實施例之一些態樣中,類視黃素為RARA特異性促效劑。在第四實施例之更特定態樣中,類視黃素為他米巴羅汀。在第四實施例之其他態樣中,抗CD38抗體為達土木單抗。 在第四實施例之一些態樣中,在投與抗CD38抗體(例如,達土木單抗)之前,向個體投與類視黃素(例如,RARA促效劑,例如他米巴羅汀)一段時間;且在開始投與類視黃素(例如,他米巴羅汀)之後,僅在個體中(例如,在個體的血液癌細胞中)之CD38位準經測定為CD38hi 時共投與抗CD38抗體。在第四實施例之一些態樣中,在開始投與類視黃素(例如,他米巴羅汀)之後的6至120小時之間測定個體中之CD38位準。 在第四實施例之一些態樣中,僅向具有以下之個體投與類視黃素(例如,他米巴羅汀)及抗CD38抗體(例如,達土木單抗):i)預先測定為等於或大於預定臨限值之來自個體的患病細胞之樣本中之視黃酸受體α mRNA位準;及/或ii)預先測定為等於或大於預定臨限值之來自個體的患病細胞之樣本中之IRF8 mRNA位準。 在第四實施例之一些態樣中,在進行任何治療之前對個體的患病細胞之樣本進行CD38誘導測試。通常,此將涉及獲得個體的患病細胞之樣本;離體生長此類細胞;視情況量測該等細胞中之CD38誘導之基線位準;用類視黃素(例如,他米巴羅汀)治療離體細胞;以及在此類治療之後測定CD38誘導之位準。若在用類視黃素治療後,個體的患病細胞之此樣本中之CD38位準經測定為CD38HI ,則該個體經測定為類視黃素(例如,他米巴羅汀)/抗CD38抗體(例如,達土木單抗)組合治療之候選者。 在第四實施例之一些態樣中,在投與類視黃素(例如,RARA促效劑,例如他米巴羅汀)之前或之後投與抗CD38抗體(例如,達土木單抗)。在第四實施例之一些態樣中,當使用上文所描述之離體技術測定個體的患病細胞中之CD38位準為可由類視黃素誘導時,治療方案不要求投與類視黃素及抗CD38抗體之任何特定次序。因此,在類視黃素之前投與抗CD38抗體可為有利的,以便限制或消除抗體之任何副作用及/或使各藥劑之藥物動力學最佳化。在第四實施例之一些態樣中,在投與類視黃素(例如,RARA促效劑,例如他米巴羅汀)之前的6至120小時之間投與抗CD38抗體(例如,達土木單抗)。 在一第五實施例中,本發明提供一種用於治療患有表徵為CD38- 、CD38lo 或CD38dim 之血液癌症之個體的方法,其中該方法包含向個體共投與類視黃素及抗CD38抗體。在第五實施例之一些態樣中,個體患有選自以下之血液癌症:急性骨髓性白血病(AML)、骨髓發育不良症候群(MDS)、多形性大細胞淋巴瘤(ALCL)、B細胞急性淋巴母細胞白血病(BALL)、B細胞非霍奇金淋巴瘤(BNHL)、伯基特氏淋巴瘤、慢性骨髓性淋巴瘤(CML)、彌漫性大型B細胞淋巴瘤(DLBCL)、霍奇金淋巴瘤或T細胞急性淋巴母細胞白血病(TALL)。在第五實施例之一些更特定態樣中,個體患有選自以下之血液癌症:多形性大細胞淋巴瘤(ALCL)、B細胞急性淋巴母細胞白血病(BALL)、B細胞非霍奇金淋巴瘤(BNHL)、伯基特氏淋巴瘤、慢性骨髓性淋巴瘤(CML)、彌漫性大型B細胞淋巴瘤(DLBCL)、霍奇金淋巴瘤或T細胞急性淋巴母細胞白血病(TALL)。 在第五實施例之一些態樣中,類視黃素為RARA特定促效劑。在第五實施例之更特定態樣中,類視黃素為他米巴羅汀、ATRA或其衍生物(例如,非瑞替尼(fenretinide))。在第五實施例之其他態樣中,抗CD38抗體為達土木單抗。 在第五實施例之另外其他態樣中,在投與療法之前測定個體中(例如在個體的血液癌細胞中)之CD38位準。 在第五實施例之一些態樣中,在進行如針對第四實施例所描述之任何治療之前對個體的患病細胞之樣本進行CD38誘導測試。若在用類視黃素治療後,個體的患病細胞之此樣本中之CD38位準經測定為CD38HI ,則該個體經測定為類視黃素(例如,他米巴羅汀)/抗CD38抗體(例如,達土木單抗)組合治療之候選者。 在第五實施例之一些態樣中,僅向具有以下之個體投與類視黃素(例如,他米巴羅汀)及抗CD38抗體(例如,達土木單抗):i)預先測定為等於或大於預定臨限值之來自個體的患病細胞之樣本中之視黃酸受體α mRNA位準;及/或ii)預先測定為等於或大於預定臨限值之來自個體的患病細胞之樣本中之IRF8 mRNA位準。 在第五實施例之又其他態樣中,在投與抗CD38抗體(例如,達土木單抗)之前,向個體投與類視黃素(例如,RARA促效劑,例如他米巴羅汀)一段時間;且在開始投與類視黃素(例如,他米巴羅汀)之後,僅在個體中(例如,在個體的血液癌細胞中)之CD38位準經測定為CD38hi 時共投與抗CD38抗體。在第五實施例之一些態樣中,在開始投與類視黃素(例如,他米巴羅汀)之後的6至120小時之間測定個體中之CD38位準。 在第五實施例之一些態樣中,在投與類視黃素(例如,RARA促效劑,例如他米巴羅汀)之前或之後投與抗CD38抗體(例如,達土木單抗)。在第五實施例之一些態樣中,當使用上文所描述之離體技術測定個體的患病細胞中之CD38位準為可由類視黃素誘導時,治療方案不要求投與類視黃素及抗CD38抗體之任何特定次序。因此,在類視黃素之前投與抗CD38抗體可為有利的,以便限制或消除抗體之任何副作用及/或使各藥劑之藥物動力學最佳化。在第五實施例之一些態樣中,在投與類視黃素(例如,RARA促效劑,例如他米巴羅汀)之前的6至120小時之間投與抗CD38抗體(例如,達土木單抗)。 在任何及所有實施例中,在一些態樣中,本發明提供一種用於治療患有癌症(例如,非APL AML、MDS或MM)之個體的方法,其中組合物包含與CD38特異性抗體(例如,達土木單抗)共投與之RARA促效劑(例如,他米巴羅汀)。在一些態樣中,他米巴羅汀經口投與。在一些態樣中,以6 mg/m2 /天至12 mg/m2 /天之間的劑量向個體投與他米巴羅汀,其中該劑量分為兩個劑量。在一些態樣中,一週不多於一次以10至20 mg/kg個體體重之間的劑量投與抗CD38抗體。 本文中闡述本發明之一或多個實施例之細節。本發明的其他特徵、目標及優點自實施方式、圖式、實例及實施例將顯而易見。The present invention provides for the administration of retinoids (eg, retinoic acid receptor alpha specific agonists, such as Tamibarotene) and anti-CD38 antibodies (eg, dalteumab) to individuals Combination to treat blood cancers (eg, multiple myeloma (MM), acute myelogenous leukemia (AML), myelodysplastic syndrome (MDS), pleomorphic large cell lymphoma (ALCL), B-cell acute lymphoblastoma Cell leukemia (BALL), B-cell non-Hodgkin lymphoma (BNHL), Burkitt's lymphoma, chronic myeloid lymphoma (CML), diffuse large Methods for individuals with B-cell lymphoma (DLBCL), Hodgkin lymphoma or T-cell acute lymphoblastic leukemia (TALL), where blood cancer: a) is not available for CD38 inhibitors as monotherapy reaction; and / or b) is characterized by CD38 -, CD38 lo or CD38 dim. The present invention also provides RARA agonists including, for example, detecting RARA biomarkers or IRF8 biomarkers and administering RARA agonists in combination with CD38-specific antibodies when the RARA biomarkers or IRF8 biomarkers are above a predetermined threshold level to treat A method of acute myeloid leukemia ("non-APL AML"), myelodysplastic syndrome ("MDS"), or multiple myeloma ("MM") of the acute promyelocytic leukemia type. Detecting RARA biomarkers or IRF8 biomarkers includes determining the presence, level, form, and / or activity of one or more RARA or IRF8 gene components or products, including, for example, RARA or IRF8 superenhancer intensity, ordinal or prevalent grading, and RARA Or IRF8 mRNA level or prevalent classification. The present invention shows that cells containing one or more of the super- enhancers associated with the RARA or IRF8 genes (eg, cancer cells or cells from individuals with non-APL AML, MDS, or MM) are more susceptible to RARA agonists (Such as Tamibarotene) The effect of combining with CD38 specific antibody. Various embodiments, aspects and alternatives of the present invention address the definition of which cell populations are sensitive to the combination of retinoic acid receptor alpha ("RARA") agonists and CD38 specific antibodies, and identification will benefit from RARA agonists Treatment and the subgroup of patients treated with CD38-specific antibodies (eg, grading patients for treatment; separating responders from non-responders) and the problem of providing therapeutic treatment for such subgroups of patients. The solution is based at least in part on our findings: the higher performance of the RARA biomarker or IRF8 biomarker in non-APL AML, MDS, or MM indicates that this cell will respond to treatment with RARA agonist and CD38 specific antibody. In a first embodiment, the present invention relates to a method of diagnosing and treating individuals with non-APL AML, MDS, or MM, the method comprising: a) based on the diagnosis of whether the individual has RARA agonist sensitivity (eg , Tamibarotene sensitive) type disease: i) pre-determined retinoic acid receptor alpha mRNA level in a sample of diseased cells from an individual equal to or greater than a predetermined threshold; and / or ii) in advance IRF8 mRNA level in a sample of diseased cells from an individual determined to be equal to or greater than a predetermined threshold; b) administer a certain amount of RARA agonist (eg, Tamibalo) to the individual to effectively treat the disease Ting); and c) Co-administer CD38-specific antibodies to individuals. In some aspects of the first embodiment, the RARA agonist is Tamibarotene. In some aspects of the first embodiment, the CD38 specific antibody is daclizumab. In some aspects of the first embodiment, before the administration of the CD38-specific antibody, the individual is administered Tamibarotene for a period of time; and after starting the administration of Tamibarotene, only CD38 in the individual When the level has been determined to be CD38 hi , CD38 specific antibody was co-administered. In some aspects of the first embodiment, the CD38 level in the individual is determined between 6 and 72 hours after the start of administration of Tamibarotene. In a second embodiment, the present invention provides a method for treating a human subject suffering from non-APL AML, MDS or MM, wherein the level of retinoic acid receptor α mRNA and / or IRF8 in the diseased cells of the individual The mRNA level has been determined to have a retinoic acid receptor alpha mRNA level equal to or greater than a predetermined threshold, or an IRF8 mRNA level equal to or greater than a predetermined threshold. The method includes the following steps: administration to the individual is effective A certain amount of RARA agonist (eg, Tamibarotene) to treat the disease; and co-administer CD38 specific antibody to the individual. In some aspects of the second embodiment, the RARA agonist is Tamibarotene. In some aspects of the second embodiment, the CD38 specific antibody is daclizumab. In some aspects of the second embodiment, before administering the CD38-specific antibody, the individual is administered Tamibarotene for a period of time; and after starting the administration of Tamibarotene, only CD38 in the individual When the level has been determined to be CD38 hi , CD38 specific antibody was co-administered. In some aspects of the second embodiment, the CD38 level in the individual is determined between 6 and 72 hours after the start of administration of Tamibarotene. In a third embodiment, the present invention provides a method for diagnosing and treating human subjects with non APL AML, MDS or MM, the method comprising: a) level based on retinoic acid receptor alpha mRNA and / or The level of IRF8 mRNA diagnoses whether an individual has a RARA agonist sensitive (eg, Tamibarotene sensitive) type disease, either or both of these levels are pre-determined to be present in the disease from the individual In a sample of cells; and b) administration of the therapy to the individual, where i) if the level indicates that the disease is sensitive to Tamibarotene, then the therapy includes the administration of a certain amount of RARA agonist (eg, he Mibarotene) and co-administration of CD38-specific antibodies; and ii) If the level indicates that the disease is not sensitive to RARA agonists (eg, not sensitive to Tamibarotene), then the therapy includes administration of RARA Agents other than effective agents (eg, other than Tamibarotene). In some aspects of the third embodiment, the level of retinoic acid receptor alpha mRNA and / or IRF8 mRNA indicates that if it is greater than a predetermined threshold, the disease is sensitive to RARA agonists (eg, to Tamiba Rotin is sensitive), and if it is less than a predetermined threshold, the disease is not sensitive to RARA agonists (eg, not sensitive to Tamibarotene). In some aspects of the third embodiment, the RARA agonist is Tamibarotene. In some aspects of the third embodiment, the CD38 specific antibody is daclizumab. In some aspects of the third embodiment, if the level of retinoic acid receptor alpha mRNA or IRF8 mRNA indicates that the disease is sensitive to RARA agonists (eg, sensitive to Tamibarotene), then CD38 is administered Before specific antibodies, the RARA agonist (eg, Tamibarotene) is administered to the individual for a period of time; and after starting the RARA agonist (eg, Tamibarotene), only in the individual When the CD38 level was determined to be CD38 hi , CD38 specific antibody was co-administered. In some aspects of the third embodiment, the CD38 level in the individual is determined between 6 and 72 hours after the start of administration of the RARA agonist (eg, Tamibarotene). In a fourth embodiment, the present invention provides a method for treating an individual with a blood cancer who does not respond to treatment with an anti-CD38 antibody in the absence of retinoids, wherein the method includes Administration of retinoids and anti-CD38 antibodies. In some aspects of the fourth embodiment, the individual has a blood cancer selected from the group consisting of multiple myeloma (MM), acute myelogenous leukemia (AML), myelodysplastic syndrome (MDS), and pleomorphic large cells Lymphoma (ALCL), B-cell acute lymphoblastic leukemia (BALL), B-cell non-Hodgkin's lymphoma (BNHL), Burkitt's lymphoma, chronic myeloid lymphoma (CML), diffuse large B cells Lymphoma (DLBCL), Hodgkin lymphoma, or T-cell acute lymphoblastic leukemia (TALL). In some more specific aspects of the fourth embodiment, the individual has a blood cancer selected from the group consisting of pleomorphic large cell lymphoma (ALCL), B-cell acute lymphoblastic leukemia (BALL), and B-cell non-Hodgkin Gold lymphoma (BNHL), Burkitt's lymphoma, chronic myeloid lymphoma (CML), diffuse large B-cell lymphoma (DLBCL), Hodgkin's lymphoma, or T-cell acute lymphoblastic leukemia (TALL) . In some aspects of the fourth embodiment, the retinoid is a RARA-specific agonist. In a more specific aspect of the fourth embodiment, the retinoid is Tamibarotene. In other aspects of the fourth embodiment, the anti-CD38 antibody is daclizumab. In some aspects of the fourth embodiment, prior to the administration of the anti-CD38 antibody (eg, daclizumab), the individual is administered a retinoid (eg, RARA agonist, such as Tamibarotene) For a period of time; and after starting retinoids (eg, Tamibarotene), co-administer only when the CD38 level in the individual (eg, in the individual's blood cancer cells) is determined to be CD38 hi With anti-CD38 antibody. In some aspects of the fourth embodiment, the CD38 level in the individual is determined between 6 and 120 hours after the start of administration of retinoid (eg, Tamibarotene). In some aspects of the fourth embodiment, only retinoids (eg, Tamibarotene) and anti-CD38 antibodies (eg, darumumab) are administered to individuals with the following: The retinoic acid receptor α mRNA level in a sample of diseased cells from an individual equal to or greater than a predetermined threshold; and / or ii) diseased cells from an individual previously determined to be equal to or greater than a predetermined threshold IRF8 mRNA level in the sample. In some aspects of the fourth embodiment, a sample of diseased cells of an individual is subjected to a CD38 induction test before any treatment is performed. Typically, this will involve obtaining a sample of the individual ’s diseased cells; growing such cells in vitro; measuring the baseline level of CD38 induction in these cells as appropriate; using retinoids (eg, Tamibarotene ) Treatment of ex vivo cells; and determination of the level of CD38 induction after such treatment. If, after treatment with retinoids, the CD38 level in this sample of an individual's diseased cells is determined to be CD38 HI , then the individual is determined to be retinoid (eg, Tamibarotene) / anti- Candidate for combination therapy with CD38 antibodies (eg, datulimumab). In some aspects of the fourth embodiment, the anti-CD38 antibody (eg, darumumab) is administered before or after the administration of retinoid (eg, RARA agonist, such as Tamibarotene). In some aspects of the fourth embodiment, when using the ex vivo technique described above to determine the CD38 level in an individual's diseased cells as retinoid-inducible, the treatment regimen does not require administration of retinoid In any specific order. Therefore, it may be advantageous to administer anti-CD38 antibodies before retinoids in order to limit or eliminate any side effects of the antibodies and / or to optimize the pharmacokinetics of each agent. In some aspects of the fourth embodiment, the anti-CD38 antibody (eg, up to 6 hours) is administered between 6 and 120 hours before the administration of retinoid (eg, RARA agonist, eg, Tamibarotene) Civil engineering monoclonal antibody). In a fifth embodiment, the present invention provides a method for treating a subject characterized by CD38 -, CD38 lo method of individual blood or CD38 dim of cancers, wherein the method comprises co-administering to a subject and retinoid anti CD38 antibody. In some aspects of the fifth embodiment, the individual has a blood cancer selected from the group consisting of acute myelogenous leukemia (AML), myelodysplastic syndrome (MDS), pleomorphic large cell lymphoma (ALCL), and B cells Acute lymphoblastic leukemia (BALL), B-cell non-Hodgkin's lymphoma (BNHL), Burkitt's lymphoma, chronic myeloid lymphoma (CML), diffuse large B-cell lymphoma (DLBCL), Hodge Gold lymphoma or T-cell acute lymphoblastic leukemia (TALL). In some more specific aspects of the fifth embodiment, the individual has a blood cancer selected from the group consisting of pleomorphic large cell lymphoma (ALCL), B-cell acute lymphoblastic leukemia (BALL), and B-cell non-Hodgkin Gold lymphoma (BNHL), Burkitt's lymphoma, chronic myeloid lymphoma (CML), diffuse large B-cell lymphoma (DLBCL), Hodgkin's lymphoma, or T-cell acute lymphoblastic leukemia (TALL) . In some aspects of the fifth embodiment, retinoid is a RARA specific agonist. In a more specific aspect of the fifth embodiment, the retinoid is Tamibarotene, ATRA, or a derivative thereof (eg, fenretinide). In other aspects of the fifth embodiment, the anti-CD38 antibody is daclizumab. In still other aspects of the fifth embodiment, the CD38 level in the individual (eg, in the individual's blood cancer cells) is determined before administration of the therapy. In some aspects of the fifth embodiment, a sample of diseased cells of an individual is subjected to a CD38 induction test prior to any treatment as described for the fourth embodiment. If, after treatment with retinoids, the CD38 level in this sample of an individual's diseased cells is determined to be CD38 HI , then the individual is determined to be retinoid (eg, Tamibarotene) / anti- Candidate for combination therapy with CD38 antibodies (eg, datulimumab). In some aspects of the fifth embodiment, only individuals with the following are administered retinoids (eg, Tamibarotene) and anti-CD38 antibodies (eg, darumumab): i) pre-determined as The retinoic acid receptor α mRNA level in a sample of diseased cells from an individual equal to or greater than a predetermined threshold; and / or ii) diseased cells from an individual previously determined to be equal to or greater than a predetermined threshold IRF8 mRNA level in the sample. In yet other aspects of the fifth embodiment, prior to the administration of the anti-CD38 antibody (eg, daclizumab), the individual is administered a retinoid (eg, RARA agonist, such as Tamibarotene) ) For a period of time; and after starting retinoids (eg, Tamibarotene), only the CD38 level in the individual (eg, in the individual ’s blood cancer cells) has been determined to be CD38 hi Anti-CD38 antibody is administered. In some aspects of the fifth embodiment, the CD38 level in the individual is determined between 6 and 120 hours after the start of administration of retinoid (eg, Tamibarotene). In some aspects of the fifth embodiment, the anti-CD38 antibody (eg, darumumab) is administered before or after administration of retinoid (eg, RARA agonist, such as Tamibarotene). In some aspects of the fifth embodiment, when using the ex vivo technique described above to determine the CD38 level in an individual's diseased cells as retinoid-inducible, the treatment regimen does not require administration of retinoid In any specific order. Therefore, it may be advantageous to administer anti-CD38 antibodies before retinoids in order to limit or eliminate any side effects of the antibodies and / or to optimize the pharmacokinetics of each agent. In some aspects of the fifth embodiment, the anti-CD38 antibody (eg, up to 6 hours) is administered between 6 and 120 hours before the administration of retinoid (eg, RARA agonist, eg, Tamibarotene) Civil engineering monoclonal antibody). In any and all embodiments, in some aspects, the invention provides a method for treating an individual suffering from cancer (eg, non-APL AML, MDS, or MM), wherein the composition comprises an antibody specific for CD38 ( For example, darcimumab) is co-administered with a RARA agonist (eg, Tamibarotene). In some forms, he was given orally by Mibarotene. In some aspects, the Tamibarotene is administered to the individual at a dose between 6 mg / m 2 / day and 12 mg / m 2 / day, where the dose is divided into two doses. In some aspects, the anti-CD38 antibody is administered at a dose between 10 and 20 mg / kg of the individual's body weight no more than once a week. The details of one or more embodiments of the invention are set forth herein. Other features, objects, and advantages of the invention will be apparent from the embodiments, drawings, examples, and examples.

定義 如本文中所使用,術語「投與(administer/administering/administration)」如本文中所使用指代植入、吸收、攝取、注入、吸入或以其他方式引入本發明化合物或其醫藥組合物。 如本文中所使用,術語「促效劑」可用於指代其存在、位準、水準、類型或形式與另一藥劑(亦即,受促效的藥劑)之增加的位準或活性相關之藥劑、狀況或事件。大體而言,促效劑可為或包括任何化學物質類別之藥劑,包括例如小分子、多肽、核酸、碳水化合物、脂、金屬及/或展示相關活化活性之任何其他實體。在一些實施例中,促效劑可為直接的(在此情況下,其直接對其靶標發揮其影響);在一些實施例中,促效劑可為間接的(在此情況下,其藉由除與其靶標結合外,例如藉由與靶標之調節子相互作用以使得更改靶標之位準或活性來發揮其影響)。 術語「生物樣本」指代包括以下之任何樣本:組織樣本(諸如組織切片及組織之穿刺活檢);細胞樣本(例如,細胞學抹片(諸如子宮頸抹片(Pap smear)或血液抹片)或藉由顯微切割獲得之細胞之樣本);整個有機體之樣本(諸如酵母或細菌之樣本);或細胞碎片、片段或細胞器(諸如藉由裂解細胞及以離心或以其他方式分離其組分而獲得)。生物樣本之其他實例包括血液、血清、尿液、精液、糞便物質、腦脊髓液、間質液、黏液、淚液、汗液、膿、活檢組織(例如,藉由外科活檢或穿刺活檢獲得)、乳頭抽出物、乳汁、陰道液、唾液、拭子(諸如,口腔拭子)或含有源自第一生物樣本之生物分子的任何物質。生物樣本亦包括轉殖基因的彼等生物樣本,諸如轉殖基因卵母細胞、精子細胞、囊胚、胚胎、胎兒、供體細胞或細胞核。在一些態樣中,來自患有AML、MDS或MM之個體的生物樣本為骨髓抽出物。 如本文中所使用,術語「生物標記」指代其存在、位準或形式與所關注之特定生物學事件或狀態相關的實體,因此將其視為該事件或狀態之「標記」。僅給出少許實例,在一些實施例中,生物標記可為或包含特定疾病病況或階段的或可罹患特定疾病、病症或病狀之可能性的標記。在一些實施例中,生物標記可為或包含特定疾病或治療結果或其可能性之標記。因此,對於所關注之相關生物學事件或狀態,在一些實施例中,生物標記為預測性的,在一些實施例中,生物標記為預後的,在一些實施例中,生物標記為診斷性的。生物標記可為任何化學物質類別之實體。舉例而言,在一些實施例中,生物標記可為或包含核酸、多肽、脂質、碳水化合物、小分子、無機試劑(例如,金屬或離子)或其組合。在一些實施例中,生物標記為細胞表面標記。在一些實施例中,生物標記為胞內生物標記。在一些實施例中,生物標記發現於細胞外部 (例如,在細胞外部分泌或以其他方式產生或存在,例如在諸如血液、尿液、淚液、唾液、腦脊髓液等之體液中)。在一些實施例中,術語指代具有特定腫瘤、腫瘤子類別、腫瘤階段等之特徵的基因表現產物。替代地或另外,在一些實施例中,特定標記之存在或位準與例如可具有特定類別之腫瘤之特徵的特定信號傳導路徑之活性(或活性位準)相關。存在或不存在生物標記之統計顯著性可視特定生物標記而變化。在一些實施例中,生物標記之偵測具有高度特異性,此係由於其反映腫瘤屬於特定子類別之高機率。此特異性可能以敏感性為代價(例如,即使腫瘤為將預期表現生物標記之腫瘤仍可能出現陰性結果)。在一些實施例中,生物標記包含RARA生物標記(例如,一或多種RARA生物標記(例如,一或多種RARA 基因組分或產物之存在、位準、形式及/或活性,包括例如RARA 超強化子強度、序數分級或盛行分級及RARA mRNA位準或盛行分級))。在一些實施例中,生物標記包含IRF8生物標記(例如,一或多種IRF8 基因組分或產物之存在、位準、形式及/或活性,包括例如IRF8 超強化子強度、序數分級或盛行分級及IRF8 mRNA位準或盛行分級)。在一些實施例中,生物標記指代一或多種生物標記(諸如RARA生物標記或IRF8生物標記)之組合。 如本文中所使用之術語「共投與(co-administer/co-administering)」在投與療法(例如,RARA促效劑及CD38特異性抗體)之情形下指示,一種療法可在個體患病過程期間與另外一或多種療法組合使用。在一些實施例中,療法之投與係同時存在的或同時發生的,意謂一種治療之傳遞在第二種治療之傳遞開始時仍在進行。在其他實施例中,一種治療之傳遞在另一種治療之傳遞開始之前結束。在任一情況之一些實施例中,治療由於組合投與而更有效。舉例而言,第二種治療更有效,例如,在第二種治療較少進行之情況下發現等效作用,或第二種治療比在無在第一種治療存在下投與第二種治療時將發現的更大程度地減輕症狀,或在第一種治療中發現類似情況。在一些實施例中,傳遞使得症狀減輕,或與病症相關之其他參數大於在無另一治療存在下傳遞的一種治療所將觀測到的參數。兩種治療之作用可為部分相加的,完全相加的或大於相加的。傳遞可使得所傳遞之第一治療之作用在傳遞第二治療時仍可偵測。此處,可在相同或分開的組合物中同時或依次投與RARA促效劑及CD38特異性抗體。對於依次投與,首先可投與RARA促效劑且其次可投與CD38特異性抗體或可顛倒投與次序。 如本文中所使用,術語「病狀」、「疾病」及「病症」可互換使用。 本文中所描述之化合物(諸如RARA促效劑及/或CD38特異性抗體)之「有效量」指代足以引出所要生物反應(亦即,治療病狀)之量。如一般技術者將瞭解,本文中所描述之化合物(諸如RARA促效劑及/或CD38特異性抗體)之有效量可視如所要生物學端點、化合物之藥物動力學、正經治療之病狀、投藥模式及個體之年齡及健康狀況等此類因素而變化。在一些實施例中,有效量涵蓋治療性及預防性治療。在其他實施例中,有效量僅涵蓋治療性治療。舉例而言,在治療癌症中,有效量之本發明化合物或組合物可減小腫瘤負荷或停止腫瘤之生長或擴散。 預期投與之「個體」包括但不限於人類(亦即,任何年齡組之男性或女性,例如兒科個體(例如,嬰兒、兒童、青年)或成人個體(例如,年輕人、中年人或老年人))及/或其他非人類動物,例如哺乳動物(例如,靈長類動物(例如,食蟹獼猴、恆河猴);商業相關的哺乳動物,諸如牛、豬、馬、羊、山羊、貓及/或狗)及鳥類(例如,商業相關的鳥類,諸如雞、鴨、鵝及/或火雞)。在某些實施例中,動物為哺乳動物。動物可為雄性或雌性且處於任何發育階段。非人類動物可為轉殖基因動物。在某些實施例中,個體為人類。 本文中所描述之化合物(諸如RARA促效劑及/或CD38特異性抗體)之「治療有效量」為足以在治療病狀中提供治療效益或足以延緩或最小化與該病狀相關聯之一或多種症狀的量。在一些實施例中,治療有效量為足以在治療病狀中提供治療效益或足以最小化與該病狀相關聯之一或多種症狀的量。治療有效量之化合物意謂單獨或與其他療法組合,在治療病狀中提供治療效益之一定量之治療劑。術語「治療有效量」可涵蓋改良整個療法、減輕或避免病狀之症狀或病因,或增強另一治療劑之治療功效的量。 如本文中所使用,術語「治療(treatment/treat/treating)」指代逆轉、減緩、延緩本文中所描述之「病理性病狀」(例如,疾病、病症或病狀,或其一或多種病徵或症狀)之發作,或抑制該「病理性病狀」之進程。在一些實施例中,「治療(treatment/treat/treating)」要求疾病、病症或病狀之病徵或症狀已發展或已觀測到。在其他實施例中,可在無疾病或病狀之病徵或症狀存在下投與治療。舉例而言,可在症狀發作之前向易感個體投與治療(例如,依據症狀歷史及/或依據遺傳性因素或其他易感性因素)。亦可在症狀已消退之後繼續治療,例如以延緩或預防復發。 術語「RARA 基因」指代編碼官能性視黃酸受體-α基因且特定言之不包括包含RARA 基因之全部或部分的基因融合之基因組DNA序列。在一些實施例中,RARA 基因位於基因組結構hg19中之chr17:38458152-38516681。 術語「IRF8 基因」指代編碼干擾素共同序列結合蛋白質或其剪接變異體且特定言之不包括包含IRF8 基因之全部或部分的基因融合之基因組DNA序列。在一些實施例中,IRF8 基因位於基因組結構hg19中之chr16:85862582-85990086。 術語「強化子」指代基因組DNA之用於調節距多達1 Mbp遠之基因之區域。強化子可與基因編碼區域重疊,但通常不由其組成。強化子通常由轉錄因子結合且藉由特定組織蛋白標記指定。 術語「超強化子」指代含有組織蛋白標記及/或轉錄蛋白質相對於特定細胞中之其他強化子的不對稱份額之強化子之子組。由於此,預測由超強化子調節的基因對該細胞之功能具有高度重要性。超強化子通常係藉由基於強度來排序細胞中之全部強化子及使用諸如ROSE (https://bitbucket.org/young_computation/rose)之可用軟體來測定具有比細胞中之中位強化子顯著更高強度的強化子之子集來測定(參見例如,美國專利9,181,580,其以引用之方式併入本文中)。 如本文中所使用之術語「強度」在提及強化子或超強化子之部分時意謂針對經分析之基因組DNA片段之長度繪製的H3K27Ac或其他基因組標記讀段之數目之曲線下的面積。因此,「強度」為由量測給定鹼基對處之標記而產生的信號在定義經選擇以量測之區域之鹼基對跨度內的積分。 術語指定值(例如,與RARA 基因相關聯之超強化子之強度)之「盛行分級」意謂等於或大於特定值之群體百分比。舉例而言,與測試細胞中之RARA 基因相關聯之超強化子的強度之35%盛行分級意謂群體之35%具有強度等於或大於測試細胞之RARA 基因強化子。 術語指定值(例如,與RARA 基因相關聯之超強化子之強度)之「盛行截止」意謂定義群體之兩個子集(例如,反應者及無反應者)之間的分隔線之盛行分級。因此,等於或高於(亦即,較低百分比值)盛行截止之盛行分級定義群體的一個子集;並且低於(例如,較高百分比值)盛行截止之盛行分級定義群體之另一子集。 術語「截止」及「截止值」意謂分析法中所量測之定義群體之兩個子集(例如,反應者及無反應者)之間的分隔線之值。因此,等於或高於截止值之值定義群體的一個子集;並且低於截止值之值定義群體的另一子集。 術語「臨限值」及「臨限值位準」意謂定義群體之兩個子集(例如,反應者及無反應者)之間的分隔線之位準。臨限值位準可為盛行截止或截止值。 術語「群體」或「樣本之群體」意謂合理地反映在較大群組中量測之值的分佈之不同樣本之足夠數目(例如,至少30、40、50或更多)。樣本群體中之各樣本可為細胞株、獲自生物之生物樣本(例如,活檢或體液樣本)或獲自異種移植之樣本(例如,藉由植入細胞株或病患樣本在小鼠中生長之腫瘤),其中各樣本來自患有相同疾病、病狀或病症之生物或來自呈現相同疾病、病狀或病症之細胞株或異種移植物。 術語指定值之「序數分級」意謂該值相比於其他值之集合之排序。舉例而言,就與測試細胞中之RARA基因相關聯之超強化子相比於測試細胞中之其他超強化子之強度而言,100之序數分級意謂測試細胞中之其他99種超強化子具有比與RARA基因相關聯之超強化子更大的強度。 術語「排序」意謂各值自最高至最低或自最低至最高的排序。RARA IRF8 本發明提供用於例如藉由以下來用與CD38特異性抗體組合之RARA促效劑治療患有癌症(例如,非APL AML、MDS或MM)之個體的方法:測定RARA生物標記及/或IRF8生物標記之盛行率(例如,測定一或多種RARAIRF8 基因組分或產物之位準、強度、序數分級、盛行分級及/或活性,包括例如RARAIRF8 超強化子強度、序數分級或盛行分級及RARA或IRF8 mRNA位準或盛行分級),以及因此共投與RARA促效劑及CD38特異性抗體。 視黃酸受體亞型α (RARA)為在未結合或由拮抗劑結合時充當轉錄抑制子及充當促效劑結合狀態下之基因活化劑的核激素受體。RARA之天然配位體為由維生素A產生之視黃酸,亦稱為全反視黃酸(ATRA)。 超強化子(SE)為調節關鍵細胞識別基因(包括惡性細胞中之致癌基因)之大型高度活性染色質區域。使用基因控制平台,SE在60個原發性AML病患樣本中經鑑別為能夠發現新的腫瘤弱點之全基因組。在病患樣本之中顯現差異存在的SE中之一者與編碼RARA之RARA 基因相關聯。 使用預先分析RARA 強化子之強度及序數之不同AML細胞株及病患樣本,發現干擾素反應因子8 (IRF8) mRNA位準與RARA類似在病患群體中上調。IRF8為已知對造血至關重要且其信號傳導缺失造成未成熟骨髓細胞之異常增殖的干擾素反應性轉錄因子。在AML中,觀測到IRF8過度表現且其可與不佳臨床結果相關。不管此上調如何,IRF8信號傳導實際上藉由抑制的轉錄輔因子及在IRF8信號傳導處於SE驅動抑制狀態中時潛在地RARA來削弱。此外,干擾素-α自身(IRF之上游信號傳導配位體)在AML中呈現促分化作用及與RARA路徑之信號傳導串話(cross-talk)。 本發明描述RARA促效劑(例如,他米巴羅汀)可以RARA或IRF8 SE依賴性方式在非APL AML、MDS或MM中誘發CD38上調之最新發現。另外,其提供尤其適用於基於對共投與他米巴羅汀及CD38特異性抗體之可能反應而特徵化、鑑別、選擇或分層病患的各種組合物及方法。RARA IRF8 超強化子鑑別及臨限值 位準之測定 本文中所描述的係藉由在測定RARA生物標記及/或IRF8生物標記之存在(例如,測定一或多種RARAIRF8 基因組分或產物之盛行率、位準、形式及/或活性,包括例如RARAIRF8 超強化子強度、序數分級或盛行分級及RARA或IRF8 mRNA位準或盛行分級)後投與RARA促效劑(例如,他米巴羅汀)及CD38特異性抗體來治療癌症之方法。 強化子或超強化子之鑑別可藉由此項技術中已知之各種方法來達成,例如,如Cell 2013, 155, 934-947及PCT/US2013/066957中所描述,兩者皆以引用之方式併入本文中。在一些實施例中,超強化子之鑑別藉由自病患中之癌症樣本(例如,自活檢)獲得細胞材料及DNA來達成。強化子量測之重要量度以二維形式進行:在其內連續地偵測基因組標記(例如,H3K27Ac)之DNA長度,以及基因組標記在沿構成量值之該DNA跨度上之各鹼基對處的編譯發生率。由長度及量值分析之積分產生的曲線下面積(「AUC」)之量測測定強化子之強度。係IRF8RARA 超強化子相對於用於本發明之一個態樣中之對照組的強度來測定個體是否將對RARA促效劑(例如,他米巴羅汀)及CD38特異性抗體起反應。對熟習此項技術者將容易地顯而易見的係,若在其內偵測基因組標記之DNA長度對RARAIRF8 與對照組兩者而言係相同的,則RARAIRF8 超強化子相對於對照組之量值的比率將等效於強度,且亦可用於測定個體是否將對RARA促效劑及CD38特異性抗體起反應。在一些實施例中,在與其他樣本進行比較之前將細胞中之RARAIRF8 強化子之強度歸一化。歸一化係藉由與已知包含在全部細胞中以類似位準呈現之普遍存在的超強化子或強化子之同一細胞中之區域進行比較來達成。此類普遍存在的超強化子區域之一個實例為MALAT1超強化子基因座(chr11:65263724-65266724) (基因組結構hg19)。 已經由H3K27Ac ChIP-seq方法測定存在與位於chr17:38458152-38516681 (基因組結構hg19)處之RARA 基因相關聯之超強化子基因座。ChIP定序(亦稱為ChIP-seq)用於分析蛋白質與DNA之相互作用。ChIP-seq將染色質免疫沈澱(ChIP)與大規模並行DNA定序組合以鑑別DNA相關蛋白質之結合位點。其可用以精確定位所關注之任何蛋白質之全局結合位點。此前,晶片上ChIP (ChIP-on-chip)為用以研究此等蛋白質-DNA關係之最常見技術。成功的ChIP-seq取決於許多因素,包括音波處理強度及方法、緩衝液組成、抗體品質及細胞數目;參見例如,T. Furey, Nature Reviews Genetics 13, 840-852 (2012年12月);M.L. Metzker, Nature Reviews Genetics 11, 31-46 (2010年1月);以及P.J Park, Nature Reviews Genetics 10, 669-680 (2009年10月)。除H3K27Ac外之可用以使用ChIP-seq鑑別超強化子之基因組標記包括P300、CBP、BRD2、BRD3、BRD4、介質複合物之組分(J Loven等人,Cell, 153(2):320-334, 2013)、組織蛋白3離胺酸4單甲基化(H3K4me1)或其他組織特異性強化子連接轉錄因子(E Smith及A Shilatifard, Nat Struct Mol Biol, 21(3):210-219, 2014) (S Pott及Jason Lieb, Nature Genetics, 47(1):8-12, 2015)。 在一些實施例中,細胞株或病患樣本之全部基因組之H3K27ac或其他標記ChIP-seq資料超強化子定位已存在。在此等實施例中,吾人將僅判定位於chr17:38458152-38516681 (基因組結構hg19)基因座處之此類定位中之強化子或超強化子之強度或序數分級是否等於或大於預定臨限值位準。 應理解,RARAIRF8MALAT1 之特定染色體位置對於不同基因組結構及/或對於不同細胞類型可不同。然而,鑒於本說明書,熟習此項技術者可藉由定位於對應於基因組結構hg 19中之RARA 及/或MALAT1 基因座的此類其他基因組結構特異性序列中而測定此類不同位置。 鑑別超強化子之其他方法包括染色質免疫沈澱(JE Delmore等人,Cell, 146(6)904-917, 2011)及晶片陣列(ChIP-chip),及使用相同免疫沈澱基因組標記及與chr17:38458152-38516681 (基因組結構hg19)RARA 基因座或chr16:85862582-85990086 (基因組結構hg19)IRF8 基因座雜交之寡核苷酸序列的染色質免疫沈澱後接qPCR (ChIP-qPCR)。在ChIP-chip之情況下,如同其他基於陣列之技術一樣,通常藉由由探針與輸入分析樣本之雜交產生的螢光強度偵測信號。對於ChIP-qPCR,將僅在嵌入PCR反應中所產生的雙鏈DNA之後會發出螢光的染料用於量測模版之擴增。 在一些實施例中,細胞是否具有大於必要臨限值位準之RARAIRF8 超強化子之測定藉由將測試細胞中之RARAIRF8 強化子強度與已知對RARA或IRF8不起反應的細胞(「對照細胞」)中之對應RARAIRF8 強度進行比較來達成。對照細胞較佳為與測試細胞相同之細胞類型。在此等實施例之一個態樣中,對照細胞為HCC1143中之此類細胞。 在一些實施例中,細胞是否具有大於必要臨限值位準之RARAIRF8 超強化子強度之測定藉由將測試細胞中之RARAIRF8 強化子強度與細胞樣本群體中之對應RARAIRF8 強度進行比較來達成,其中細胞樣本中之各者獲自不同來源(亦即,不同個體、不同細胞株、不同異種移植)。在此等實施例之一些態樣中,僅來自個體之原發腫瘤細胞樣本用於測定臨限值位準。在此等實施例之一些態樣中,群體中之樣本中之至少一些已進行對特異性RARA促效劑及CD38特異性抗體之反應測試,以便建立:a)對特異性RARA促效劑及CD38特異性抗體起反應的群體中之樣本之最低RARA 強化子強度(「最低反應者」);及/或b)對特異性RARA促效劑及CD38特異性抗體起反應的群體中之樣本之最低IRF8 強化子強度(「最低反應者」);及視情況c)對特異性RARA促效劑及CD38特異性抗體不起反應的群體中之樣本之最高RARA 強化子強度(「最高無反應者」)及/或d)對特異性RARA促效劑及CD38特異性抗體不起反應的群體樣本中之之最高IRF8 強化子強度(「最高無反應者」)。在此等實施例中,RARAIRF8 強化子強度(大於該強度時測試細胞將視為對該特異性RARA促效劑及CD38特異性抗體起反應)之截止設定為:i)等於或至多大於群體中最低反應者之RARAIRF8 強化子強度5%;或ii)等於或至多大於群體中最高無反應者之RARAIRF8 強化子強度5%;或iii)群體中最低反應者與最高無反應者之RARAIRF8 強化子強度之間的值。 應理解,在上述實施例中,通常並非群體中之所有樣本均需要對RARA促效劑及CD38特異性抗體之反應進行測試,但均量測所有樣本之RARAIRF8 強化子強度。在一些實施例中,樣本基於RARAIRF8 強化子強度排序。上文所闡述之用以建立截止之三種方法之樣本的選擇將視群體中之最低反應者與最高無反應者之間的RARAIRF8 強化子強度差及目標為最小化偽陽性之數目抑或為最小化遺漏潛在反應樣本或個體之機率而定。當最低反應者與最高無反應者之間的差較大時(例如,當許多樣本未進行測試落在RARAIRF8 強化子強度之排序之最低反應者與最高無反應者之間的反應時),截止通常設定為等於或至多大於群體中最低反應者之RARAIRF8 強化子強度5%。此截止最大化潛在反應者之數目。當此差較小時(例如,當少許樣本或無樣本未進行測試落在RARAIRF8 強化子強度之排序之最低反應者與最高無反應者之間的反應時),截止通常設定為最低反應者與最高無反應者之RARAIRF8 強化子強度之間的值。此截止最小化偽陽性之數目。當最高無反應者之RARAIRF8 強化子強度高於最低反應者時,截止通常設定為等於或至多大於群體中最高無反應者之RARAIRF8 強化子強度5%之值。此方法亦最小化偽陽性之數目。 在一些實施例中,細胞是否具有大於必要臨限值位準之RARAIRF8 超強化子強度之測定藉由將測試細胞中之RARA 強化子強度的序數與細胞樣本群體中之RARAIRF8 強化子強度之序數進行比較來達成,其中細胞樣本中之各者獲自不同來源(亦即,不同個體、不同細胞株、不同異種移植)。在此等實施例之中,群體中之樣本中之至少一些已進行對特異性RARA促效劑之反應測試,以便建立:a)對特異性RARA促效劑及CD38特異性抗體起反應的群體中之樣本之最低RARA 強化子強度(「最低序數反應者」);b)對特異性RARA促效劑及CD38特異性抗體起反應的群體中之樣本之最低IRF8 強化子強度序數(「最低序數反應者」);及視情況c)對特異性RARA促效劑及CD38特異性抗體不起反應的群體中之樣本之最高RARA 強化子強度序數(「最高序數無反應者」);及/或d)對特異性RARA促效劑及CD38特異性抗體不起反應的群體中之樣本之最高RARA 強化子強度序數(「最高序數無反應者」)。在此等實施例中,RARAIRF8 強化子強度(大於該強度時測試細胞將視為對該特異性RARA促效劑及CD38特異性抗體起反應)之截止設定為:i)等於或至多大於群體中最低序數反應者之RARAIRF8 強化子強度5%;或ii)等於或至多大於群體中最高序數無反應者之RARAIRF8 強化子強度5%;或iii)群體中最低序數反應者與最高序數無反應者之RARAIRF8 強化子強度之間的值。 應理解,在上述實施例中,通常並非群體中之所有樣本均需要進行對RARA促效劑及CD38特異性抗體之反應測試,但均量測所有樣本之RARAIRF8 強化子強度,且建立RARAIRF8 強化子相比於同一樣本中之其他強化子之序數。序數通常藉由量測細胞中之所有其他強化子之強度及測定RARAIRF8 強化子相比於其他強化子在強度方面具有何種排序(亦即,序數)來獲得。 在一些實施例中,樣本基於RARA 強化子強度之序數來排序。在一些實施例中,樣本基於IRF8 強化子強度之序數來排序。上文所闡述之用以建立截止之方法之樣本的選擇將視群體中之最低序數反應者與最高序數無反應者之間的RARAIRF8 強化子強度序數差及截止經設計以最小化偽陽性抑或最大化反應者之數目而定。當此差較大時(例如,當許多樣本未進行測試落在RARAIRF8 強化子強度序數之排序最低序數反應者與最高序數無反應者之間的反應時),截止通常設定為等於或至多大於群體中最低序數反應者之RARAIRF8 強化子強度序數5%。當此差較小時(例如,當少數樣本或無樣本未進行測試落在RARAIRF8 強化子強度序數之排序最低序數反應者與最高序數無反應者之間的反應時),截止通常設定為最低序數反應者與最高序數無反應者之RARAIRF8 強化子強度序數之間的值。當最高序數無反應者之RARAIRF8 強化子強度序數高於最低反應者之RARAIRF8 強化子強度序數時,截止通常設定為等於或至多大於群體中最高序數無反應者之RARAIRF8 強化子強度序數5%之值。 在將測試細胞或樣本與群體進行比較之實施例的一些態樣中,將針對群體所獲得之一或多個截止值(例如,RARA 強化子強度或RARA 強化子序數及/或IRF8 強化子強度或IRF8 強化子序數)轉換成盛行分級,且將截止表示為具有截止值或更高(亦即,盛行截止)的群體之百分比。在不受理論限制之情況下,申請人認為測試樣本之盛行分級將類似,無論用於測定RARAIRF8 強化子強度之方法如何。因此,針對一個參數(例如,RARA 強化子強度序數或IRF8 強化子強度序數)所測定之盛行截止是便攜型的且可應用於另一參數(例如,RARA mRNA位準或IRF8 mRNA位準)以測定該另一參數之截止值。此允許測定任何參數之截止值而不必以實驗方式測定此類參數之位準與對RARA促效劑及CD38特異性抗體之反應之間的相關性。RARA IRF8 mRNA 位準測定 本發明提供用於在RARA生物標記及/或IRF8生物標記存在時用RARA促效劑(例如,他米巴羅汀)及CD38特異性抗體治療癌症之方法。已測定RARA 及/或IRF8 超強化子基因座之鑑別允許吾人使用RNA轉錄物來測定敏感性而非超強化子位準以測定對RARA促效劑及CD38特異性抗體之敏感性。來自超強化子基因座自身之RNA轉錄物可經定量且與該基因座處之超強化子位準極其良好相關。亦已展示,編碼RARA之mRNA轉錄物亦與對單獨RARA促效劑或與CD38特異性抗體組合之敏感性相關,且因此mRNA位準可用以鑑別將對此療法組合起反應之細胞。因此,在一些實施例中,RARA或IFR8 mRNA位準而非超強化子強度或序數分級可用於測定對RARA促效劑及CD38特異性抗體之敏感性。 在一些實施例中,來自超強化子基因座之RNA轉錄位準使用定量技術來定量,該等定量技術將樣本中之RARAIRF8 強化子RNA轉錄位準與已知對RARA促效劑及CD38特異性抗體不起反應的細胞或細胞株中之對應RARAIRF8 強化子RNA轉錄位準進行比較。此類方法包括用於讀段與強化子通讀相關聯之eRNA (N Hah等人,PNAS, 112(3):E297-302, 2015)的基於RNA陣列或定序之方法,以及RNA qPCR。 在替代實施例中,使用RNA-Seq或RNA-qPCR技術將個體中(亦即,腫瘤樣本中、癌細胞樣本中、血液樣本中等)之RARA或IRF8 mRNA位準與患有相同疾病或病狀之個體群體中之RARA或IRF8 mRNA位準進行比較以鑑別對RARA促效劑及CD38特異性抗體的反應者。在此等實施例中,群體中之樣本中之至少一些已進行對特異性RARA促效劑及CD38特異性抗體之反應測試,以便建立:a)對特異性RARA促效劑及CD38特異性抗體起反應的群體中之樣本之最低RARA mRNA位準(「最低mRNA反應者」);及/或b)對特異性RARA促效劑及CD38特異性抗體起反應的群體中之樣本之最低IRF8 mRNA位準(「最低mRNA反應者」);及視情況c)對特異性RARA促效劑及CD38特異性抗體不起反應的群體中之樣本之最高RARA mRNA位準(「最高mRNA無反應者」);及/或d)對特異性RARA促效劑及CD38特異性抗體不起反應的群體中之樣本之IRF8 mRNA最高位準(「最高mRNA無反應者」)。在此等實施例中,RARA或IRF8 mRNA位準(大於該位準時測試細胞將視為對該特異性RARA促效劑及CD38特異性抗體起反應)之截止設定為:i)等於或至多大於群體中最低mRNA反應者之RARA或IRF8 mRNA位準5%;或ii)等於或至多大於群體中最高mRNA無反應者之RARA或IRF8 mRNA位準5%;或iii)群體中最低mRNA反應者與最高mRNA無反應者之RARA或IRF8 mRNA位準之間的值。 在一些實施例中,並非群體中之所有樣本均需要進行對RARA促效劑及CD38特異性抗體之反應測試,但均量測所有樣本之RARA或IRF8 mRNA位準。在一些實施例中,樣本基於RARA mRNA位準排序。在一些實施例中,樣本基於IRF8 mRNA位準排序。上文所闡述之用以建立截止之三種方法之樣本的選擇將視群體中之最低mRNA反應者與最高mRNA無反應者之間的RARA或IRF8 mRNA位準差及截止經設計以最小化偽陽性抑或最大化潛在反應者之數目而定。當此差較大時(例如,當許多樣本未進行測試落在RARA或IRF8 mRNA位準之排序最低mRNA反應者與最高mRNA無反應者之間的反應時),截止通常設定為等於或至多大於群體中最低mRNA反應者之RARA或IRF8 mRNA位準5%。當此差較小時(例如,當少數樣本或無樣本未進行測試落在RARA或IRF8 mRNA位準之排序最低mRNA反應者與最高mRNA無反應者之間的反應時),截止通常設定為最低mRNA反應者與最高mRNA無反應者之RARA或IRF8 mRNA位準之間的值。當最高mRNA無反應者之RARA或IRF8 mRNA位準高於最低mRNA反應者時,截止通常設定為等於或至多大於群體中最高mRNA無反應者之RARA或IRF8 mRNA位準5%之值。 在一些實施例中,群體基於RARA mRNA位準排序。在一些實施例中,群體基於IRF8 mRNA位準排序。在此等實施例中,量測各樣本中之RARA或IRF8 mRNA位準且將其與細胞中之所有其他mRNA之mRNA位準進行比較以獲得RARA或IRF8 mRNA位準之序數分級。隨後,基於RARA或IRF8 mRNA序數分級之截止基於群體中之樣本而測定,以與先前針對測定RARAIFR8 超強化子強度序數截止所描述的相同之方式測試該等樣本對RARA促效劑之反應。隨後,直接使用所測定的RARA或IRF8 mRNA序數截止或將其用於測定盛行截止,隨後使用其中之任一者來分層對RARA促效劑及CD38特異性抗體有潛在反應的額外樣本。 在一些實施例中,使用基於如上文所描述之RARA 強化子強度或RARA 強化子強度序數、或IRF8 強化子強度或IRF8 強化子強度序數而建立的盛行截止來測定RARA或IRF8 mRNA位準之截止。在此等實施例之一些態樣中,量測群體之mRNA位準,且將事先測定之盛行截止應用於該群體以測定mRNA截止位準。在此等實施例之一些態樣中,建立群體之RARA或IRF8 mRNA位準之排序標準曲線,且將預定盛行截止應用於該標準曲線以測定RARA或IRF8 mRNA截止位準。 在將測試細胞或樣本與群體進行比較之實施例的一些態樣中,將所獲得之群體之截止mRNA位準值轉換成盛行分級,且將mRNA位準截止表示為具有截止值或更高(亦即,盛行截止)的群體之百分比。 在不受理論限制之情況下,申請人認為測試樣本之盛行分級及群體之盛行截止將類似,無論用於測定IRF8或RARA mRNA位準之方法如何。 在此等實施例之一些態樣中,若個體之RARA或IRF8 mRNA位準對應於如藉由群體中之RARA或IRF8 mRNA位準所測定之79%、78%、77%、76%、75%、74%、73%、72%、71%、70%、69%、68%、67%、66%、65%、64%、63%、62%、61%、60%、59%、58%、57%、56%、55%、54%、53%、52%、51%、50%、49%、48%、47%、46%、45%、44%、43%、42%、41%、40%、39%、38%、37%、36%、35%、34%、33%、32%、31%、30%、29%、28%、27%、26%、25%、24%、23%、22%、21%或20%的群體之盛行分級,則該個體經鑑別為RARA促效劑反應者。在此等實施例之一個態樣中,基於針對RARAIRF8 強化子強度建立的盛行截止而建立截止值。在此等實施例之一替代態樣中,基於針對RARAIRF8 強化子強度序數建立的盛行截止而建立截止值。在此等實施例之另一替代態樣中,基於RARA或IRF8 mRNA位準而建立截止值。在此等實施例之其他更特定態樣中,基於針對RARAIRF8 強化子強度序數測定的盛行值而建立AML病患之截止值,且該盛行值用於測定RARA或IRF8 mRNA位準之截止值。在此等實施例之甚至更特定態樣中,使用在25至45%之間(例如,在25至30%、25至35%、25至40%、30至35%、30至40%、35至45%、35至40%、31至35%、32至35%、33至35%、34至35%、31至36%、32至36%、33至36%、34至36%或35至36%之間)的盛行截止來測定AML病患之截止值。在此等實施例之其他甚至更特定態樣中,使用36%之盛行值來測定AML病患之截止值。在此等實施例之又其他甚至更特定態樣中,使用25%之盛行值來測定AML病患之截止值。 在另其他實施例中,可將群體分為三個群組--反應者、部分反應者及無反應者,且設定兩個截止值或盛行截止值。部分反應者群組可包括反應者及無反應者,以及對RARA促效劑及CD38特異性抗體之反應不如反應者群組高之彼等群體成員。在此等實施例中,測定兩個截止值或盛行截止值。當在群體中最高RARA或IRF8 mRNA無反應者之RARA或IRF8 mRNA位準高於最低RARA mRNA反應者時,此分層類型可尤其適用。在此情境下,反應者與部分反應者之間的截止位準或盛行截止設定為等於或至多大於最高RARA或IRF8 mRNA無反應者之RARA或IRF8 mRNA位準5%;並且部分反應者與無反應者之間的截止位準或盛行截止設定為等於或至多大於最低RARA或IRF8 mRNA反應者之RARA或IRF8 mRNA位準5%。是否應向部分反應者投與RARA促效劑及CD38特異性抗體之判定將視治療醫師之判斷及/或管制機構之核准而定。 定量細胞或生物樣本中之特異性RNA序列之方法為此項技術中已知,且包括但不限於諸如用於由NanoString Technologies提供的服務及產品之螢光雜交、基於陣列之技術(Affymetrix)、如用SYBR® Green (Life Technologies)或TaqMan®技術(Life Technologies)之逆轉錄酶qPCR、RNA定序(例如,RNA-seq)、如與RNAscope® (Advanced Cell Diagnostics)一起使用之RNA雜交及信號放大,或北方墨點法(northern blot)。 在此等實施例之一些態樣中,在進行比較之前將測試細胞及對照細胞兩者或群體之所有成員中之RNA轉錄物(mRNA或另一RARAIRF8 轉錄物)之位準歸一化。歸一化涉及藉由在兩者細胞中之等效位準下與原生於及其親本之另一RNA轉錄物(例如,GADPH mRNA、18S RNA),或與在超強化子強度測定之前「外加」至細胞中之每一者之樣本中的固定位準之外源RNA比較來調整RARAIRF8 RNA轉錄物之所測定位準(J Lovén等人,Cell, 151(3):476-82 (2012); J Kanno等人,BMC Genomics 7:64 (2006); J Van de Peppel等人,EMBO Rep 4:387-93 (2003))。RARA 促效劑及靶向 CD38 抗體 本發明提供例如在鑑別出RARA或IRF8生物標記時用與CD38特異性抗體組合之RARA促效劑治療患有癌症(例如,非APL AML、MDS或MM)之個體之方法。 RARA 促效劑 用以治療鑑別為具有與RARA 基因相關聯之超強化子之病患的RARA促效劑之選擇可自此項技術中已知之任何RARA促效劑中作出。較佳地為,用於本發明方法中之RARA促效劑對RARA 具有特異性且針對RaR之其他形式(例如,RaR-ß及RaR-γ)具有顯著較低(至少低10×、至少低100×、至少低1,000×、至少低10,000×、至少低100,000×)促效活性。 在一些實施例中,RARA促效劑係選自揭示於以下美國專利中之任一者或屬於該等專利中之任一者中所闡述的類別內之任何化合物:US 4,703,110、US 5,081,271、US 5,089,509、US 5,455,265、US 5,759,785、US 5,856,490、US 5,965,606、US 6,063,797、US 6,071,924、US 6,075,032、US 6,187,950、US 6,355,669、US 6,358,995及US 6,387,950,其中之每一者以引用之方式併入本文中。 在一些實施例中,RARA促效劑係選自以下表1中所闡述的已知RARA促效劑中之任一者,或其醫藥學上可接受之鹽,或前述之溶劑合物或水合物: 表1.適用於本發明之例示性RARA促效劑。 在一些實施例中,RARA促效劑為他米巴羅汀。在一些實施例中,RARA促效劑為(AGN-195183)。 CD38 特異性抗體 CD38為涉及許多代謝功能(包括細胞外核苷酸之分解代謝、受體介導之黏著、遷移調節及各種信號傳導事件)之跨膜糖蛋白。靶向CD38之抗體可調節此等功能中之任一者,由此促進RARA促效劑在癌症治療中之療效。在一些實施例中,CD38特異性抗體可識別及/或結合至CD38片段之任何部分。CD38特異性抗體可包含單株抗體、人類化抗體或人類抗體。針對CD38具有特異性之例示性抗體包括伊薩土西單抗(isatuximab)、達土木單抗、MOR202、Ab79、Ab19及EPR4106。在一些實施例中,CD38特異性抗體為達土木單抗。治療方案 本發明之方法理論上適用於治療表徵為與RARA生物標記或IRF8生物標記(例如,一或多種RARAIRF8 基因組分或產品之存在、位準、形式及/或活性,包括例如RARAIRF8 超強化子強度、序數分級或盛行分級及RARA 或IRF8 mRNA位準或盛行分級)相關聯之癌症。超強化子相關RARAIRF8 基因可在某些類型之癌症中比其他類型之癌症中更普遍。本發明特定言之係關於AML (例如,不表徵為涉及RARA 基因之染色體易位的非APL AML及呈其他形式之AML)以及MDS及MM之治療。在一些實施例中,待治療之疾病為不表徵為涉及RARA 基因之染色體易位的非APL AML、MDS或MM。在一些實施例中,待治療之疾病為不表徵為涉及IRF8 基因之染色體易位的非APL AML、MDS或MM。 在一些實施例中,待用RARA促效劑(例如,他米巴羅汀)及CD38特異性抗體治療之個體患有復發性或難治性非APL AML。若個體:a)並未在誘導化學療法之第一週期之後顯示部分反應;或b)並未在誘導化學療法之第二週期之後顯示完全反應;或c)在習知化學療法之後復發;或d)在進行單一幹細胞移植後復發,則將其歸類為患有復發性或難治性非APL AML。在一些實施例中,待用RARA促效劑(例如,他米巴羅汀)及CD38特異性抗體治療之個體患有難治性MM。 在其他實施例中,待用RARA促效劑(例如,他米巴羅汀)治療之個體為不合適的老年人個體。如本文中所使用之術語「不合適的老年人」意謂個體為至少60歲且由醫師判定為非標準誘導療法之候選者的人類。 在一些實施例中,向個體共投與RARA促效劑(例如,他米巴羅汀)及CD38特異性抗體(例如,達土木單抗)。在一些實施例中,RARA促效劑(例如,他米巴羅汀)與CD38特異性抗體(例如,達土木單抗)同時投與。在一些實施例中,RARA促效劑(例如,他米巴羅汀)及CD38特異性抗體(例如,達土木單抗)彼此在約1小時至約48小時內投與。在一些實施例中,RARA促效劑(例如,他米巴羅汀)及CD38特異性抗體(例如,達土木單抗)彼此在以下時間內投與:約1分鐘、約2分鐘、約5分鐘、約10分鐘、約15分鐘、約20分鐘、約30分鐘、約45分鐘、約1小時、約1.5小時、約2小時、約3小時、約4小時、約5小時、約6小時、約8小時、約10小時、約12小時、約16小時、約20小時、約24小時、約36小時或約48小時。 在一些實施例中,病患群體包括接受癌症(例如,非APL AML、MDS或MM)治療之先前療法的一或多個個體(例如,包含多個個體或由多個個體組成)。在一些實施例中,病患群體包括未接受癌症(例如,非APL AML、MDS或MM)治療之先前療法的一或多個個體(例如,包含多個個體或由多個個體組成)。在一些實施例中,病患群體包含未接受非APL AML、MDS或MM治療之先前療法的病患或由該等病患組成。 在一些實施例中,接受先前療法之病患可能已接受選自由化學療法、免疫療法、放射療法、姑息療護、手術及其組合組成之群的先前療法。在一些實施例中,病患已接受移植。在一些實施例中,病患已接受標準細胞毒性化學療法。在一些實施例中,標準細胞毒性化學療法包括阿糖胞苷及/或蒽環黴素。在一些實施例中,標準細胞毒性化學療法可包括額外化學療法及/或造血幹細胞移植(HSCT)。在一些實施例中,病患已接受低甲基化藥劑。在一些實施例中,病患已接受來那度胺(lenalidomide)。 在一些實施例中,病患群體包括已接受及/或正接受其他療法例如以使得除了CD38特異性抗體外與其他療法(例如化學療法藥劑)組合投與RARA促效劑療法(例如,他米巴羅汀)組合物的一或多個個體(例如,包含多個個體或由多個個體組成)。在一些實施例中,此類其他療法可包含癌症(例如,如本文中所描述)、疼痛、噁心、便秘等之療法、與癌症療法相關聯之一或多個副作用(例如,搔癢症、掉髮、失眠等)之療法或其任何組合,或由其組成。本發明提供一種治療非APL AML、MDS或MM之方法,其包含用治療有效量之RARA促效劑療法(例如,他米巴羅汀)或其醫藥學上可接受之鹽及CD38特異性抗體治療鑑別為患有非APL AML、MDS或MM之病患。 在一些實施例中,本發明提供一種治療先前以包含化學療法之治療方案治療的病患之非APL AML、MDS或MM之方法,該治療藉由向此類病患投與治療有效量之RARA促效劑(例如,他米巴羅汀)及CD38特異性抗體進行。在一些實施例中,本發明提供一種用於無標準療法存在之治療病患之非APL AML、MDS或MM的方法。在一些實施例中,本發明提供一種用於治療不適合於標準療法之病患的方法。 在一些實施例中,病患或病患群體可不為(例如,可不包括)有對他米巴羅汀之成份過敏的先前歷史之病患。在一些實施例中,病患或病患群體可不為(例如,可不包括)正接受維生素A調配物之病患。在一些實施例中,病患或病患群體可不為(例如,可不包括)患有維生素A過多症之病患。 在一些實施例中,病患或病患群體可不為(例如,可不包括)老年病患。在一些實施例中,病患或病患群體可為或包括一或多個老年病患。在一些實施例中,當與一或多個較年輕病患進行比較時,老年病患可經更頻繁監測以偵測可能不良事件(包括例如,低位準之血清白蛋白及/或血漿中之較高濃度之游離藥物等)。在一些實施例中,可對經測定呈現此類不良事件之一或多個症狀的老年病患減少、暫停及/或終止RARA促效劑及/或CD38特異性抗體之投與。劑量形式及給藥方案 大體而言,以治療有效量使用與良好醫學實踐一致且適合於相關藥劑及個體之醫藥組合物及給藥方案來調配、給藥及投與根據本發明使用之各活性劑(例如,RARA促效劑或CD38特異性抗體)。原則上,可藉由此項技術中已知之任何適當方法投與治療性組合物,投與包含但不限於經口、經黏膜、吸入、局部、經頰、經鼻、經直腸或非經腸投與(例如,靜脈內、輸注、瘤內、結節內、皮下、腹膜內、肌肉內、皮內、經皮或投與之其他類型)。在一些實施例中,RARA促效劑(例如,他米巴羅汀)將經口投與。在一些實施例中,CD38特異性抗體將經靜脈內投與。 在一些實施例中,特定活性劑之給藥方案可涉及間歇或連續投與,例如以在正接受療法之個體中所關注之一或多種組織或體液中達成特定所要藥物動力學圖譜或其他暴露模式。 在一些實施例中,以組合投與之不同藥劑可經由不同傳遞途徑及/或根據不同時程投與。替代地或另外,在一些實施例中,一或多種劑量之第一活性劑大致上與一或多種其他活性劑同時投與,且在一些實施例中,該一或多種劑量之第一活性劑經由常見路線及/或作為單一組合物之部分與一或多種其他活性劑一起投與。 在最佳化給定治療方案之途徑及/或給藥時程時考慮之因素可包括例如:正經治療之特定適應症、個體之臨床病狀(例如,年齡、整體健康狀況、所接受之先前療法及/或對其之反應等)、藥劑之傳遞位點、藥劑性質、藥劑投與模式及/或途徑、存在或不存在組合療法及開業醫師已知之其他因素。舉例而言,在癌症治療中,正經治療之適應症之相關特徵可尤其包括癌症類型、階段、部位等中之一或多者。 在一些實施例中,特定醫藥組合物及/或所用給藥方案之一或多個特徵可隨時間推移而調節(例如,增加或減小任何個體劑量中之有效量,增加或減小給藥之間的時間間隔等),例如以便最佳化所要治療效果或反應。 大體而言,根據本發明之活性劑之給藥之類型、量及頻率由在向哺乳動物(較佳人類)投與一或多種相關藥劑時應用之安全及療效要求來控制。大體而言,與未觀測到療法相比,此類給藥特徵經選擇以提供特定且通常可偵測之治療反應。 在本發明之上下文中,例示性期望治療反應可包括但不限於:腫瘤生長、腫瘤大小、轉移、與腫瘤相關聯之症狀及副作用中之一或多者之抑制及/或減小,以及腫瘤細胞凋亡增加、一或多種細胞標記或循環標記之治療相關的減少或增加及類似者。此類準則可藉由各種免疫學、細胞學及揭示於文獻中之其他方法中之任一者容易地評定。 在一些實施例中,基於誘導性標記之時序及/或臨限值表現量,可能需要調適給藥方案,且特定言之需要設計連續給藥方案,無論針對特定類型之腫瘤、特定腫瘤、特定病患群體(例如,攜載遺傳標記之病患群體)及/或特定病患。在一些此類實施例中,治療性給藥方案可與在療法之前及/或期間評估一或多種誘導性標記之表現的偵測方法組合或依據該偵測方法進行調整。 在一些實施例中,RARA促效劑(例如,他米巴羅汀)療法方案包含約1 mg/m2 、2 mg/m2 、3 mg/m2 、4 mg/m2 、5 mg/m2 、6 mg/m2 、7 mg/m2 、8 mg/m2 、9 mg/m2 、10 mg/m2 、11 mg/m2 、12 mg/m2 、13 mg/m2 、14 mg/m2 、15 mg/m2 、16 mg/m2 之至少一個(或包括一個或恰好由一個組成)劑量,或RARA促效劑(例如,他米巴羅汀)之此等值中之任兩者之間的劑量。在一些實施例中,RARA促效劑(例如,他米巴羅汀)療法方案包含在1 mg/m2 與50 mg/m2 之間的劑量。在一些實施例中,RARA促效劑(例如,他米巴羅汀)療法方案包含在5 mg/m2 與25 mg/m2 之間的劑量。在一些實施例中,RARA促效劑(例如,他米巴羅汀)療法方案包含在5 mg/m2 與15 mg/m2 之間的劑量。在一些實施例中,RARA促效劑(例如,他米巴羅汀)療法方案包含12 mg/m2 之劑量。在一些實施例中,RARA促效劑(例如,他米巴羅汀)療法方案包含6 mg/m2 之劑量。 在一些實施例中,RARA促效劑(例如,他米巴羅汀)療法方案包含他米巴羅汀組合物之複數個劑量。在一些此類實施例中,他米巴羅汀療法方案包含例如2個、5個、10個、20個、30個、60個、90個、180個、365個劑量或此等值中之任兩者之間的多個劑量,及/或包含重複劑量模式(例如,至少一個兩個日劑量之週期,該週期可重複,視情況用替代投與或視情況不投與一段時間來隔開不同週期)。在一些實施例中,他米巴羅汀療法方案一日投與兩次。在一些實施例中,他米巴羅汀療法方案一日投與一次。在一些實施例中,他米巴羅汀療法方案包含6 mg/m2 至12 mg/m2 之總劑量,分為每日兩次經口給藥。 在一些實施例中,CD38特異性抗體(例如,達土木單抗)療法方案包含約0.5 mg/kg、1 mg/kg、2 mg/kg、5 mg/kg、7.5 mg/kg、10 mg/kg、11 mg/kg、12 mg/kg、13 mg/kg、14 mg/kg、15 mg/kg、16 mg/kg、17 mg/kg、18 mg/kg、19 mg/kg、20 mg/kg、25 mg/kg、30 mg/kg、35 mg/kg、40 mg/kg、45 mg/kg、50 mg/kg之至少一個(或包括一個或恰好由一個組成)劑量,或CD38特異性抗體(例如,達土木單抗)之此等值中之任兩者之間的劑量。在一些實施例中,CD38特異性抗體(例如,達土木單抗)療法方案包含在1 mg/kg與100 mg/kg之間的劑量。在一些實施例中,CD38特異性抗體(例如,達土木單抗)療法方案包含在5 mg/kg與50 mg/kg之間的劑量。在一些實施例中,CD38特異性抗體(例如,達土木單抗)療法方案包含在10 mg/kg與20 mg/kg之間的劑量。 在一些實施例中,CD38特異性抗體(例如,達土木單抗)療法方案包含達土木單抗組合物之複數個劑量。在一些此類實施例中,達土木單抗療法方案包含例如2個、5個、10個、20個、30個、60個、90個、180個、365個劑量或此等值中之任兩者之間的多個劑量,及/或包含重複劑量模式(例如,至少一個兩個日劑量之週期,該週期可重複,視情況用替代投與或視情況不投與一段時間來隔開不同週期)。在一些實施例中,達土木單抗療法方案一週投與一次。在一些實施例中,達土木單抗療法方案一週至多投與一次。在一些實施例中,達土木單抗療法方案每兩週投與一次。在一些實施例中,達土木單抗療法方案包含10 mg/kg至20 mg/kg之總劑量,一週至多一次。調配物 如本文中所使用,醫藥組合物係指化合物(諸如他米巴羅汀或CD38特異性抗體)與其他化學組分(諸如載劑、穩定劑、稀釋劑、分散劑、懸浮劑、增稠劑及/或賦形劑)的混合物。醫藥組合物有助於向有機體投與化合物。含有化合物之醫藥組合物可藉由此項技術中已知之包括但不限於以下之任何習知形式及路徑以治療有效量投與:靜脈內、經口、經直腸、氣霧劑、非經腸、經眼、經肺、經皮、經陰道、經耳、經鼻及局部投與。 對於經口投與,化合物可藉由組合活性化合物與此項技術中熟知的醫藥學上可接受之載劑或賦形劑來容易地調配。此類載劑准許本文中所描述之化合物調配成用於待治療之個體口服攝取之錠劑、散劑、丸劑、糖衣藥丸、膠囊、液體、凝膠、糖漿劑、酏劑、漿液、懸浮液及其類似物。大體而言,賦形劑(諸如填充劑、崩解劑、滑動劑、界面活性劑、再結晶抑制劑、潤滑劑、顏料、結合劑、調味劑等)可出於慣例目的且以不影響組合物之特性之典型的量使用。在一些實施例中,賦形劑為乳糖水合物、玉米澱粉、羥基丙基纖維素及/或硬脂酸鎂中之一或多者。在一些實施例中,他米巴羅汀可用乳糖水合物、玉米澱粉、羥基丙基纖維素及/或硬脂酸鎂中之一或多者調配。 他米巴羅汀之可接受調配物之鑑別可藉由此項技術中已知之各種方法來達成,例如,如US 20100048708中所描述,其以引用之方式併入本文中。經包裝醫藥組合物 本發明之經包裝醫藥組合物包含寫入插入物或標記物,其包含在患有癌症且已經測定具有與強度或序數分級等於或大於臨限值位準,或RARA mRNA位準等於或大於臨限值位準之RARA 基因相關聯之超強化子的個體中使用RARA促效劑及靶向CD38之抗體的說明。如上文中所詳細描述,在來自診斷為患有相同疾病之個體或與該疾病(其中醫藥組合物經指示用於治療該疾病)相同之疾病的細胞株或異種移植模型之樣本群體中測定臨限值位準。說明可黏附或以其他方式附著至包含RARA促效劑及靶向CD38之抗體的容器。替代地,說明及包含RARA促效劑之容器將彼此分開,但一起存在於單個包裝、盒子或其他類型之容器中。 經包裝醫藥組合物之說明將通常由批准RARA促效劑及靶向CD38之抗體之治療使用的政府機構授權或推薦。說明可包含測定超強化子是否與RARAIRF8 基因相關聯之特定方法,以及測定與RARAIRF8 基因相關聯之強化子是否為超強化子之定量方法,測定RARA或IRF8 mRNA位準之定量方法;及/或超強化子或RARA或IRF8 mRNA之臨限值位準,在該臨限值位準下用經包裝RARA促效劑及靶向CD38之抗體進行之治療經推薦及/或假定治療有效。在一些態樣中,說明指導向RARA或IRF8 mRNA位準落在已量測RARA或IRF8 mRNA位準之群體的至少第30個百分位中之個體投與組合物。在一些態樣中,若個體之RARA或IRF8 mRNA位準盛行分級為已量測RARA或IRF8 mRNA位準之群體的79%、78%、77%、76%、75%、74%、73%、72%、71%、70%、69%、68%、67%、66%、65%、64%、63%、62%、61%、60%、59%、58%、57%、56%、55%、54%、53%、52%、51%、50%、49%、48%、47%、46%、45%、44%、43%、42%、41%、40%、39%、38%、37%、36%、35%、34%、33%、32%、31%、30%、29%、28%、27%、26%、25%、24%、23%、22%、21%或20%,則該個體經鑑別為RARA促效劑反應者。在一些態樣中,說明指導,向RARA或IRF8 mRNA位準已藉由特定檢定來量測之個體投與組合物。 說明可視情況包含給藥資訊,經批准用RARA促效劑及/或靶向CD38之抗體治療之癌症類型,關於RARA促效劑及/或靶向CD38之抗體之物理化學資訊;關於RARA促效劑及/或靶向CD38之抗體之藥物動力學資訊;或藥物-藥物相互作用資訊。在一些態樣中,說明指導向診斷為患有非APL AML之個體投與組合物。在一些態樣中,說明指導向診斷為患有非APL MM之個體投與組合物。在一些態樣中,醫藥組合物包含他米巴羅汀。在一些態樣中,醫藥組合物包含AGN-195183。在一些態樣中,醫藥組合物包含達土木單抗。在一些實施例中,醫藥組合物包含他米巴羅汀及達土木單抗兩者。 實例 為了能更全面地理解本文中所描述之發明,闡述以下實例。提供本申請案中所描述之合成及生物實例以說明本文提供之化合物、醫藥組合物及方法,且並不應理解為以任何方式限制其範疇。實例 1 . RARA RNA 及蛋白質表現量之量測 採用表現量量測來確定所標記之RARA 基因的mRNA位準。mRNA位準與強化子位準良好相關且因此亦對RARA促效劑具預測敏感性。吾人使用如下文所闡述之量測RNA之各種手段。I . 基於陣列之技術。 用三等份批次之1x106 個細胞評估HCC1143及AU565中之表現量。遵循製造商之方案,使用Trizol®自細胞提取RNA並使用mirVanaTM RNA純化套組純化RNA (兩者均來自Life Technologies)。在Dana Farber Cancer Institute Microarray Core (http://mbcf.dfci.harvard.edu/)之Affymetrix PrimeViewTM 陣列上讀出RNA位準。 圖1展示使用以上方案來量測之他米巴羅汀反應性(Au565)及非反應性(HCC1143)細胞株中之各種RAR亞型的mRNA表現量。在反應性細胞株中RARA mRNA之表現比非反應性細胞株高8倍,而RaR-ß及RaR-γ之表現在該等細胞株之間並無顯著差異。此確定RARA mRNA表現分析與RARA 超強化子強度及對RARA促效劑之敏感性相關,以及表明RARA mRNA位準可用以預測對此類促效劑之敏感性。II . RNA - Seq 。RARA表現量藉由RNA-Seq定量。執行Poly-A RNA-Seq且使用rsem v1.2.21軟體(rsem-calculate-expression;參數 = -p 4 --samtools-sort-mem 3G --ci-memory 3072 --bowtie-chunkmbs 1024 --quiet --output-genome-bam --bowtie2 --bowtie2-path /data/devtools/bowtie2-2.0.5 --strand-specific)將讀段與HG19轉錄組進行比對,且隨後使用相同rsem程式組(rsem-parse-alignments, rsem-build-read-index, rsem-run-em)進行並以每百萬轉錄物(TPM)報告mRNA定量,其表示對所偵測之每百萬個轉錄物指定基因的轉錄物數目之估計。隨後針對各樣本提取所有蛋白質編碼基因且將其記分一起進行分位數歸一化。吾人處理所有來自PDX模型之AML病患樣本、原發性AML病患樣本及AML細胞株並將其一起歸一化以產生通用mRNA記分。標繪值展示48個原發性AML病患的RARA之log2 (TPM+1)位準(y軸)對於超強化子強度(RARA /MALAT1)(圖1)。實例 2 . AML 中之 RARA 超強化子強度序數分級截止 使用H3K27Ac及ChIP-Seq來分析95個AML樣本(病患樣本及AML細胞株(包括SigM5、MV411、HEL及Kasumi1)兩者)之整體強化子/超強化子圖譜。在樣本中之每一者中,測定RARA 相關強化子相比於相同細胞中之其他強化子及超強化子在強度(如藉由H3K27Ac所量測)方面之序數分級,且所測定的序數分級標繪在排序條形圖上(圖2)。在MV411中,測定RARA 相關強化子為第133最強強化子。MV411為經確認具有最低超強化子強度序數之他米巴羅汀反應性細胞株。在HEL中,測定RARA 相關強化子為第155最強強化子。HEL為經確認具有最高超強化子強度序數之他米巴羅汀非反應性細胞株。根據此等值,吾人設定RARA 強化子強度序數截止為150 (在HEL序數與MV411序數之間的值)。 如自吾人對來自人類個體之70個原發性AML細胞樣本之分析所測定,36%之彼等樣本具有至少在彼等細胞中為第150最強的RARA 超強化子(圖3)。因此,盛行截止設定為36%。當基於RARA或IRF8 mRNA量測而鑑別對他米巴羅汀之潛在AML反應者時,亦將相同盛行截止用作RARA mRNA盛行截止。 吾人亦藉由RARA 強化子與MALAT1強化子之比率(「歸一化強化子強度」)來定量擴增之AML細胞組的強化子。繪製此歸一化強化子強度值對於對他米巴羅汀之敏感性,吾人確認RARA 強化子強度比率大於1的6個細胞株中之5個係反應性的,而強化子小於此位準之7個細胞株中僅4個係反應性的(圖5)。 當截止移動至1.4或更高之歸一化強化子強度時,所有細胞株(4個中之4個)均係反應性的。實例 3 . AML 中之 RARA 超強化子強度序數分級截止與 RARA mRNA 位準相關 將用於測定36%RARA 超強化子強度序數盛行截止之AML病患樣本分成兩組--盛行分級為36%或更高(亦即,較低%值)之彼等樣本,及盛行分級低於36% (亦即,較高%值)之彼等樣本--並使用如實例1中所描述之RNA-seq來分析該等樣本之RARA mRNA位準。結果展示於圖5A中。在RARA 超強化子強度序數之36%盛行分級或更高之群組比低於該盛行分級之群組具有統計顯著較高位準之RARA mRNA (p<0.001)。此再次確認在超強化子位準方面測定之盛行截止亦可用作mRNA位準之盛行截止。 吾人亦使用RNA-seq來測定11個不同AML細胞株中之RARA mRNA位準且將mRNA位準與對他米巴羅汀之敏感性進行比較。將所測試之AML細胞株基於其對他米巴羅汀之敏感性或不敏感性分成兩個不同群組。對他米巴羅汀敏感之細胞株全部均具有由RNAseq量測之>10 TPM之RARA mRNA,而三個不敏感的細胞株具有低於此截止位準之位準(圖5B)。實例 4 APL AML 細胞株中之 IRF8 mRNA 位準與對 RARA 促效劑之反應相關 吾人使用Affymetrix GeneChip® PrimeView 人類基因表現陣列來檢查此等AML細胞株中之七個(四個對他米巴羅汀敏感--NOMO-1、AML3、MV-4-11及Sig-M5;及三個對他米巴羅汀不敏感--KG1a、OCI-M1及Kasumi-1)之其他mRNA,該其他mRNA可能在他米巴羅汀敏感細胞株中尤其較高,且將IRF8 mRNA鑑別為潛在候選者。吾人接著定量此等七個AML細胞株以及藉由執行如下文所闡述之RNA-seq分析進行對他米巴羅汀之敏感性測試的若干其他AML細胞株中之每一者之IRF8 mRNA位準。前七個細胞株之結果展示於圖6中。有趣的是,NOMO-1確實不具有高RARA mRNA位準,但對他米巴羅汀起反應。NOMO-1具有較高IRF8 mRNA位準之事實幫助說明此表面上的不一致性且進一步驗證IRF8 mRNA位準預測對他米巴羅汀之反應之用途。以與描述於實例1中之方式類似之方式進行RNA分離、製備及RNA-seq資料處理。吾人接著將對他米巴羅汀之敏感性與IRF8 mRNA位準進行比較,如圖6及表2中所展示。 表2:AML細胞株IRF8 mRNA位準及他米巴羅汀抗增殖效力 *HL60為APL細胞株。 如自上表可見,除HL60之外的所有他米巴羅汀反應性細胞株均具有在分析中高於190 TPM (log2 (7.57))之IRF8 mRNA位準,而所有非反應性細胞株均具有小於16.5 TPM (log2 (4.03))之IRF8 mRNA位準。在無IRF8 mRNA之相應的高位準(6.73 TPM)之情況下HL60對他米巴羅汀之反應表明,IRF8 mRNA位準與他米巴羅汀敏感性之間的相關性可能對APL無效,且因此可更好地適合於分層患有非APL AML之個體。圖7移除HL60之資料點。 表3展示除如log2 值所闡述之IRF8 mRNA值以外的與表2類似之資料,且展示Sig-M5及THP-1細胞株之額外資料。 表3.表示為log2 值之AML細胞株mRNA位準及他米巴羅汀抗增殖效力 *HL60為APL細胞株。實例 5 RARA 促效劑治療之 IRF8 mRNA 臨限值之測定 AML細胞株結果表明RNA-Seq分析中之截止值在15.5與190 TPM之間(亦即,在log2 (4.03)與log2 (7.57)之間)。吾人選擇AML病患樣本(由Stanford University惠贈)之群體以便檢查IRF mRNA位準之分佈及基於該截止值測定盛行截止值。吾人添加該AML細胞株群體且隨後產生排序圖。圖8展示組合的病患樣本/AML細胞株群體中之IRF8 mRNA位準之排序分佈。吾人測定25%之盛行截止對應於近似log2 (7)之IRF8 mRNA值。實例 6 IRF8 mRNA RARA mRNA 位準之相關性 吾人接著將AML細胞株及病患群體中之IRF8與RARA mRNA位準進行比較以測定相關性。圖9展示對他米巴羅汀起反應的一些細胞株具有相對低RARA mRNA,但具有高位準之IRF8 mRNA。圖10展示病患子組亦展現高IRF8 mRNA位準,但展現相對低RARA mRNA位準,且反之亦然。此支撐若任一mRNA位準大於臨限值,則量測病患中之IRF8及RARA mRNA兩者及選擇該病患用於用RARA促效劑(諸如他米巴羅汀)治療可最佳化可治療病患群體之想法。實例 7 由他米巴羅汀在 AML 細胞株中進行之 RARA mRNA 依賴性 CD38 誘導 吾人藉由量測由CD38-FITC抗體染色之細胞表面平均螢光強度(MFI)來在他米巴羅汀治療之後監測細胞表面CD38表現之誘導。圖11A展示以50 nmol/L之濃度進行72 h他米巴羅汀治療並不誘導RARA mRNA低細胞株Kasumi中之CD38表現(CD38- )。圖11B表明在他米巴羅汀治療RARA mRNA高細胞株MV411 72 h之後,整個細胞群體表現高位準之CD38 (CD38HI )。此外,在圖11C中,OCI-AML3 (RARA mRNA高)在基線處具有低CD38 MFI (CD38DIM )。在他米巴羅汀治療後,進一步誘導CD38表現且細胞群體變成CD38HI 。圖11D展示72h他米巴羅汀治療並不誘導另一RARA mRNA低細胞株OCI-M1中之CD38表現(CD38- )。此等資料指示由他米巴羅汀進行之CD38HI 誘導可由RARA mRNA位準預測。此等結果在圖11E中描繪為條形圖,其展示在治療之前及之後在各細胞株中偵測之CD38 mRNA表現之位準。圖11F展示,在他米巴羅汀治療之前及之後展示之基於FACS之CD38HI 細胞之百分比。在圖11E及圖11F中,亦展示APL細胞株NB4。實例 8 RARA mRNA 預測在他米巴羅汀及達土木單抗組合療法之後之 AML 細胞株 MV411 NK 細胞 介導之細胞毒性 為了功能性地評估他米巴羅汀及達土木單抗組合之療效,用他米巴羅汀治療具有不同RARA mRNA位準之AML細胞株72小時,接著用人類NK細胞與達土木單抗或對照抗體共培養。接著在38 h共培養時程期間,NK細胞增殖及腫瘤細胞死亡以相差進行成像。圖12A至圖12D為在以下治療條件下共培養分析中之RARA mRNA高MV411細胞株之相差影像的代表性影像:圖12A) 72 h DMSO MV411細胞株治療前及38 h共培養對照抗體治療。圖12B) 72 h SY1425 (50nM) MV411細胞株治療前及38 h共培養對照抗體治療。圖12C) 72 h DMSO MV411細胞株治療前及38 h共培養達土木單抗治療。圖12D) 72 h SY1425 (50nM) MV411細胞株治療前及38 h共培養達土木單抗治療。實例 9 RARA mRNA 位準預測在他米巴羅汀及達土木單抗組合療法之後之 AML 細胞株之 NK 細胞 介導之細胞毒性 藉由磷脂結合蛋白V染色腫瘤細胞來進行的NK細胞介導之腫瘤細胞死亡的動力學量測支撐以下發現:他米巴羅汀及達土木單抗治療組合相比於單一藥劑治療使得在僅一些AML細胞株(圖13A至圖13C)中腫瘤細胞死亡增加。與RARA mRNA高AML細胞株(亦即,OCI-AML3及MV411)而非RARA mRNA低AML細胞株(亦即,OCI-M1) (圖11B至圖11D)中之CD38表現之誘導增加的組合之此等資料支撐以下結論:他米巴羅汀以RARA mRNA依賴性方式誘導CD38HI 表現型,此為在達土木單抗治療之後有效的NK細胞介導之腫瘤細胞死亡所需的。實例 10 共培養分析中之 NK 細胞 活化僅在他米巴羅汀及達土木單抗組合治療之後發生 如藉由 NK 細胞 IFN γ 分泌所測定。 干擾素γ (IFNγ)分泌為NK細胞活化之指示。在AML細胞株及NK細胞共培養38 h之後遵循所指示之治療條件定量IFNγ分泌(圖14)。相比於單一藥劑治療條件,在用他米巴羅汀及達土木單抗組合治療之後僅在RARA mRNA高AML細胞株(MV411及OCI-AML3)及NK細胞共培養分析中觀測到IFNγ位準顯著增加。另外,此等為在他米巴羅汀治療之後顯示CD38HI 表現型的唯一細胞株。此等資料進一步支撐指示有效NK細胞活化之磷脂結合蛋白V定量(圖12A至圖12D及圖13)需要組合治療。實例 11 他米巴羅汀增加多發性骨髓瘤細胞中之 CD38 表現之強度。 達土木單抗臨床上經批准用於治療多發性骨髓瘤(MM)。然而,歸因於低位準之CD38表現,僅一部分此等病患反應。吾人在此處證明他米巴羅汀增加已CD38HI 之多發性骨髓瘤細胞株(MM1S)中之CD38HI 表現型(圖15A)且可增加CD38- 多發性骨髓瘤細胞株至中等狀態(HUNS1) (圖15B)。基於先前的AML實驗,吾人預測此將可能增加CD38HI 高個體對CD38治療性抗體針對免疫介導之死亡之反應。實例 12 CD38HI 表現型中之他米巴羅汀增加進一步使 MM 細胞株對達土木單抗依賴性 NK 細胞 介導之細胞毒性敏感。 為了研究用他米巴羅汀治療MM細胞株是否會增加對CD38抗體治療之敏感性,吾人重複先前所描述之與HUNS1及MM1S MM細胞株之NK細胞共培養分析。圖16表明在他米巴羅汀及達土木單抗組合治療之後,相比於單一藥劑抗CD38抗體治療增加MM1S腫瘤細胞死亡,如藉由磷脂結合蛋白V染色所定量。吾人藉由直接監測在此分析中治療38 h之後由IFNγ分泌之NK細胞活化來進一步確認此(圖17)。達成高CD38強度之多發性骨髓瘤細胞株MM1S展示回應於組合治療之強力細胞殺滅。實例 13 他米巴羅汀誘導原發性非 APL AML MDS 病患樣本中之 CD38HI 表現型。 為了確認AML細胞株中之CD38HI 位準之他米巴羅汀誘導可趨近於AML及MDS病患樣本,吾人將獲自15個不同AML或MDS病患之PBMC與50 nM他米巴羅汀或與作為陰性對照的DMSO一起培養。吾人測試治療24 h及48 h後之成活力及CD38誘導,如藉由流動式細胞測量術所量測。分析具有至少10%成活力之樣本之CD38位準。結果展示於圖18A中。大部分病患樣本(11/15)展示治療24小時後CD38HI 細胞之增加。一個病患樣本展示無反應(病患4)且三個病患樣本在24 h之後不滿足成活力標準(病患2、6及10)。48 h小時之後,已在48 h之後展示CD38誘導之四個額外病患樣本不再滿足成活力標準且經排除(病患3、7、9及14)。除無反應者病患4之外的所有剩餘病患樣本在48 h後展示CD38位準之進一步誘導。 吾人接著量測此等病患子組(病患1、5、11、12及13)中之RARA及IRF8 mRNA位準兩者,該病患子組表示展示一些反應的病患,在48 h之後仍存活,且表示AML及MDS兩者。如圖18B中所展示,展示如藉由RNA qPCR所量測之RARA mRNA及/或IRF8 mRNA之較高位準(dCq值愈低,mRNA位準愈高)的彼等病患樣本相比於具有較低RARA mRNA及/或IRF8 mRNA之樣本在他米巴羅汀治療後顯示更大百分比的CD38HI 。 針對AML病患樣本所觀測到之CD38誘導位準與獲自高RARA RNA AML細胞株之結果良好相關(例如,比較圖19C至圖19D中之病患樣本AML_1與AML細胞株MV411中他米巴羅汀誘導之CD38 MFI)。對於兩者而言,他米巴羅汀造成與達土木單抗敏感性多發性骨髓瘤細胞株及多發性骨髓瘤病患中所觀測到的類似,表現型CD38HI 表現型增加(例如,比較圖19A至圖19B中之病患樣本MM_1與多發性骨髓瘤細胞株MM1S)。基於此相關性,本發明人認為高RARA mRNA AML病患將受益於RARA特異性促效劑(諸如他米巴羅汀)及抗CD38抗體(諸如達土木單抗)之組合治療。實例 15 他米巴羅汀造成比具有高 RARA IRF8 位準之 AML 細胞株異種移植中之 ATRA 更大的 CD38 誘導之增加。 已報告ATRA造成比HL-60細胞(通常表徵為APL之AML細胞株)中之他米巴羅汀更大的CD38位準之誘導(A Uruno等人,2011, J Leuk Biol, 90,第235至247頁)。吾人意欲比較具有RARA或IRF8 mRNA之不同位準的非APL AML細胞株中之ATRA與他米巴羅汀之CD38誘導效果。在此實驗中,吾人使用MV411、THP-1或Kasumi-1之小鼠異種移植。MV411具有高位準之RARA mRNA及IRF8 mRNA (亦即,大於臨限值),而THP-1具有高位準之IRF-8 mRNA。認為Kasumi-1細胞具有低於臨限值之IRF8及RARA位準。 細胞株中之每一者在活體外在37℃下在含5% CO2 之空氣氛圍中保持為適當介質(THP-1細胞:補充有10%加熱不活化胎牛血清及0.05 mM β-巰基乙醇之RPMI1640介質;MV4-11細胞:補充有10%加熱不活化胎牛血清之IMDM介質;Kasumi-1細胞:補充有20%加熱不活化胎牛血清之RPMI1640介質)中之懸浮培養物。為腫瘤接種收集在指數生長期中生長之細胞且計數。 所有小鼠在腫瘤細胞接種之前經γ輻射(200 rad) 24 h。各小鼠在右側腹區域用適當腫瘤細胞株(THP-1細胞:含1×107 個細胞之0.1 ml PBS (1:1基質膠);MV4-11細胞:含5×106 個細胞之0.1 ml PBS (1:1基質膠);Kasumi-1細胞:含1×107 個細胞之0.1 ml PBS (1:1基質膠))進行皮下接種以用於腫瘤發育。當平均腫瘤大小達至近似100至200 mm3 時,開始用他米巴羅汀、ATRA或媒劑治療。將小鼠分成3個群組,每群組九隻,且每一個群組經口投與藥物(ATRA 4 mg/kg;他米巴羅汀3 mg/kg)或僅媒劑BID持續至多28天。腫瘤細胞接種之日期表示為第0天。使用測徑規每週兩次以二維量測腫瘤體積,且使用以下式以mm3 為單位表示體積:V = 0.5a ×b 2 ,其中ab 分別為腫瘤之長度及寬度。在分組之後,在第7天、第14天及第21天每群組殺死三隻小鼠(總共研究4週)。收集腫瘤之一半以用於嵌入至FFPE嵌段中以用於免疫組織化學(IHC)染色,且收集腫瘤之另一半以用於對腫瘤細胞之CD38 FACS分析。利用BOND RX自動染色儀使用針對CD38之C端之抗CD38抗體執行IHC染色(clone SP149; Abcam; Cat. No. ab183326)。在pH 9.0的EDTA緩衝液中以1:100稀釋抗體,且隨後與腫瘤切片一起培育20分鐘。使用DAB及製造商之結合聚合物精細偵測套組產生信號。 如圖20A中所見,7日之後,經他米巴羅汀治療之MV4-11異種移植小鼠展示大於200之平均螢光強度(MFI),其中藉由FACS分析超過80%腫瘤細胞為CD38HI 。經ATRA治療之MV4-11異種移植小鼠展示略微大於100之MFI,其中近似70%腫瘤細胞為CD38HI 。用抗CD38抗體進行之腫瘤切片的免疫組織化學染色確認在此異種移植模型中他米巴羅汀優於ATRA之出人意料及未預期的優越性(圖20B)。此趨勢在3週時變得甚至更明顯。如圖20C中所展示,在三週時,在經他米巴羅汀治療之MV4-11異種移植中超過60%腫瘤細胞保持CD38HI ,其中MFI約為110,而在經ATRA治療之異種移植物中少於20%腫瘤細胞保持CD38HI ,其中MFI約為50。圖20D展示三週之後腫瘤細胞之免疫組織化學染色,此確認他米巴羅汀優於ATRA之優越性。 如圖21中所展示,亦在THP-1細胞中觀測到7日後他米巴羅汀在誘導CD38中之優越性。儘管經ATRA治療之異種移植展示相比於他米巴羅汀類似的CD38HI 細胞百分比,但他米巴羅汀治療相比於ATRA (~175)產生更高的MFI (大於200)。對比而言,Kasumi-1細胞展示用任一治療之極少量CD38誘導(圖22)。一週後各類型之異種移植與各類型之治療之免疫組織化學比較展示於圖23中。等效物及範疇 在實施例中,除非相反地指示或另外自上下文顯而易見,否則諸如「一(a/an)」及「該(the)」之冠詞可意謂一或大於一。除非相反地指示或另外自上下文顯而易見,否則在群組之一或多個成員之間包括「或」之實施例或描述視為滿足一個、大於一個或所有群組成員存在於、用於給定產物或方法中或另外與給定產物或方法相關之情況。本發明包括群組中恰好一個成員存在於、用於給定產物或方法中或另外與給定產物或方法相關之實施例。本發明包括多於一個或所有的群組成員存在於、用於給定產物或方法中或另外與給定產物或方法相關之實施例。 此外,本發明涵蓋其中來自所列實施例中之一或多者之一或多個限制、要素、條款及描述性術語引入另一實施例中的所有變化、組合及排列。舉例而言,視另一實施例而定之任何實施例可經修改成包括在視同一基礎實施例而定的任何其他實施例中發現之一或多個限制。在要素如所列,例如呈馬庫什(Markush)組格式呈現之情況下,亦揭示要素之各子組,且可自該組移除任何要素。應瞭解,大體而言,在本發明或本發明之態樣稱為包含具體要素及/或特徵時,本發明或本發明態樣之某些實施例由此類要素及/或特徵組成或主要由此類要素及/或特徵組成。出於簡單的目的,彼等實施例尚未具體地以詞語闡述在本文中。亦應注意,術語「包含」及「含有」意欲為開放性的且容許包括額外要素或步驟。當給出範圍時,包括端點。此外,除非另外指示或另外自上下文及一般技術者的理解顯而易見,否則表示為範圍之值可在本發明之不同實施例中採用所陳述範圍內之任何特定值或子範圍,除非上下文另外明確規定,否則達到該範圍下限之+分之一單位。 本申請案係關於各種頒予之專利、公開之專利申請案、期刊文章及其他出版物,以上所有者均以引用之方式併入本文中。若任何併入之參考文獻與本說明書之間存在衝突,則應以本說明書為準。另外,本發明之屬於先前技術之任何特定實施例可明確地自實施例中之任何一或多者排除。因為此類實施例被認為係一般技術者所已知的,所以其可經排除,即使未在本文中明確地闡述該排除。本發明之任何特定實施例可出於任何原因自任何實施例排除,無論是否與先前技術之存在相關。 熟習此項技術者僅使用常規實驗將認識到或能夠確定本文中所描述之特定實施例之許多等效物。本文中所描述之本發明實施例之範疇並不意欲限於以上描述,而實際上如隨附實施例中所闡述。一般技術者將瞭解,可在不脫離如以下實施例所定義之本發明之精神或範疇的情況下對本說明書作出各種改變及修改。Definitions As used herein, the term "administer / administering / administration" as used herein refers to implantation, absorption, ingestion, infusion, inhalation, or other introduction of the compound of the present invention or its pharmaceutical composition. As used herein, the term "agonist" can be used to refer to its presence, level, level, type, or form related to the increased level or activity of another agent (ie, an agent that is agonized) Medicine, condition or event. In general, an agonist may be or include an agent of any chemical class, including, for example, small molecules, polypeptides, nucleic acids, carbohydrates, lipids, metals, and / or any other entities that exhibit related activation activities. In some embodiments, the agonist may be direct (in this case, it directly exerts its influence on its target); in some embodiments, the agonist may be indirect (in this case, it borrows In addition to binding to its target, for example, by interacting with the regulator of the target so as to change the level or activity of the target to exert its influence). The term "biological sample" refers to any sample that includes the following: tissue samples (such as tissue sections and tissue biopsies); cell samples (eg, cytological smears (such as Pap smear or blood smears) Or samples of cells obtained by microdissection); samples of whole organisms (such as samples of yeast or bacteria); or cell fragments, fragments, or organelles (such as by lysing cells and centrifuging or otherwise separating their groups) Points). Other examples of biological samples include blood, serum, urine, semen, fecal matter, cerebrospinal fluid, interstitial fluid, mucus, tears, sweat, pus, biopsy tissue (eg, obtained by surgical biopsy or biopsy), nipples Aspirates, milk, vaginal fluid, saliva, swabs (such as oral swabs), or any substance containing biomolecules derived from the first biological sample. Biological samples also include biological samples of transgenic genes, such as transgenic gene oocytes, sperm cells, blastocysts, embryos, fetuses, donor cells or nuclei. In some aspects, the biological sample from an individual with AML, MDS, or MM is a bone marrow aspirate. As used herein, the term "biomarker" refers to an entity whose existence, level, or form is related to a specific biological event or state of interest, and therefore is considered a "marker" of that event or state. To give only a few examples, in some embodiments, the biomarker may be or include a marker of a specific disease condition or stage or the possibility of suffering from a specific disease, disorder or condition. In some embodiments, the biomarker may be or include a marker for a specific disease or treatment outcome or its likelihood. Therefore, for related biological events or states of interest, in some embodiments, the biomarker is predictive, in some embodiments, the biomarker is prognostic, and in some embodiments, the biomarker is diagnostic . Biomarkers can be entities of any chemical substance category. For example, in some embodiments, the biomarker may be or include a nucleic acid, polypeptide, lipid, carbohydrate, small molecule, inorganic agent (eg, metal or ion), or a combination thereof. In some embodiments, the biomarker is a cell surface marker. In some embodiments, the biomarker is an intracellular biomarker. In some embodiments, biomarkers are found outside the cell (eg, secreted or otherwise produced or present outside the cell, for example, in body fluids such as blood, urine, tears, saliva, cerebrospinal fluid, etc.). In some embodiments, the term refers to a gene expression product characterized by a specific tumor, tumor subclass, tumor stage, and the like. Alternatively or additionally, in some embodiments, the presence or level of a specific marker is related to the activity (or active level) of a specific signaling pathway that may be characteristic of a tumor of a specific class, for example. The statistical significance of the presence or absence of biomarkers can vary depending on the specific biomarkers. In some embodiments, the detection of biomarkers is highly specific because it reflects a high probability that the tumor belongs to a specific sub-category. This specificity may come at the expense of sensitivity (for example, even if the tumor is a tumor that will be expected to exhibit a biomarker, a negative result may still occur). In some embodiments, the biomarkers comprise RARA biomarkers (eg, one or more RARA biomarkers (eg, one or moreRARA The presence, level, form and / or activity of genetic components or products, including for exampleRARA Super enhancer intensity, ordinal grade or prevalent grade and RARA mRNA level or prevalent grade)). In some embodiments, the biomarker comprises an IRF8 biomarker (eg, one or moreIRF8 The presence, level, form and / or activity of genetic components or products, including for exampleIRF8 (Super-enhancer intensity, ordinal grade or prevalent grade and IRF8 mRNA level or prevalent grade). In some embodiments, biomarkers refer to a combination of one or more biomarkers, such as RARA biomarkers or IRF8 biomarkers. As used herein, the term "co-administer (co-administer / co-administering)" in the case of administration therapy (eg, RARA agonist and CD38-specific antibody) indicates that a therapy can cause disease in an individual Use in combination with one or more therapies during the procedure. In some embodiments, the administration of therapy is simultaneous or simultaneous, meaning that the delivery of one treatment is still ongoing at the beginning of the delivery of the second treatment. In other embodiments, the delivery of one treatment ends before the delivery of another treatment begins. In some embodiments of either case, the treatment is more effective due to combined administration. For example, the second treatment is more effective, for example, an equivalent effect is found when the second treatment is performed less frequently, or the second treatment is more effective than the second treatment in the absence of the first treatment Will be found to relieve symptoms to a greater extent, or a similar condition is found in the first treatment. In some embodiments, the delivery reduces symptoms, or other parameters related to the condition are greater than the parameters that would be observed for one treatment delivered in the absence of another treatment. The effects of the two treatments can be partially additive, fully additive or greater than additive. The delivery can make the effect of the delivered first treatment still detectable when delivering the second treatment. Here, the RARA agonist and CD38 specific antibody may be administered simultaneously or sequentially in the same or separate compositions. For sequential administration, the RARA agonist can be administered first and the CD38 specific antibody can be administered second or the order of administration can be reversed. As used herein, the terms "disease", "disease" and "disease" are used interchangeably. The "effective amount" of the compounds described herein (such as RARA agonists and / or CD38 specific antibodies) refers to an amount sufficient to elicit the desired biological response (ie, to treat the condition). As one of ordinary skill will appreciate, the effective amount of compounds described herein (such as RARA agonists and / or CD38-specific antibodies) can depend on the desired biological endpoint, the pharmacokinetics of the compound, the pathological condition being treated, Such factors as the dosage pattern and the individual's age and health status vary. In some embodiments, the effective amount encompasses both therapeutic and prophylactic treatment. In other embodiments, the effective amount covers only therapeutic treatment. For example, in the treatment of cancer, an effective amount of a compound or composition of the invention can reduce tumor burden or stop tumor growth or spread. "Individuals" expected to be administered include, but are not limited to, humans (ie, men or women of any age group, such as pediatric individuals (eg, infants, children, youth) or adult individuals (eg, young, middle-aged, or elderly Human)) and / or other non-human animals, such as mammals (eg, primates (eg, crab-eating macaques, rhesus monkeys); commercially relevant mammals, such as cattle, pigs, horses, sheep, goats, Cats and / or dogs) and birds (eg, commercially relevant birds such as chickens, ducks, geese and / or turkeys). In some embodiments, the animal is a mammal. Animals can be male or female and at any stage of development. The non-human animal may be a transgenic animal. In some embodiments, the individual is a human. The "therapeutically effective amount" of the compounds described herein (such as RARA agonists and / or CD38 specific antibodies) is one that is sufficient to provide a therapeutic benefit in the treatment of the condition or sufficient to delay or minimize the association with the condition Or the amount of multiple symptoms. In some embodiments, the therapeutically effective amount is an amount sufficient to provide a therapeutic benefit in treating the condition or sufficient to minimize one or more symptoms associated with the condition. A therapeutically effective amount of a compound means a therapeutic agent that alone or in combination with other therapies, provides a quantitative amount of therapeutic benefit in treating the condition. The term "therapeutically effective amount" may encompass an amount that improves the entire therapy, reduces or avoids the symptoms or causes of the condition, or enhances the therapeutic efficacy of another therapeutic agent. As used herein, the term "treatment (treat / treating)" refers to reversing, slowing, or delaying the "pathological conditions" described herein (e.g., a disease, disorder or condition, or one or more symptoms Or symptom), or inhibit the progression of the "pathological condition". In some embodiments, "treatment (treat / treating)" requires that the signs or symptoms of the disease, disorder, or condition have developed or have been observed. In other embodiments, the treatment can be administered in the absence of signs or symptoms of the disease or condition. For example, treatment may be administered to susceptible individuals prior to the onset of symptoms (eg, based on the history of symptoms and / or based on genetic factors or other susceptibility factors). It is also possible to continue treatment after the symptoms have subsided, for example to delay or prevent relapse. the term"RARA "Gene" refers to a gene encoding a functional retinoic acid receptor-α and specifically excludesRARA The genomic DNA sequence of the gene fusion of all or part of the gene. In some embodiments,RARA The gene is located in chr17: 38458152-38516681 in the genome structure hg19. the term"IRF8 "Gene" refers to an interferon-encoding common sequence binding protein or its splice variant and specifically excludes inclusionIRF8 The genomic DNA sequence of the gene fusion of all or part of the gene. In some embodiments,IRF8 The gene is located in chr16: 85862582-85990086 in genome structure hg19. The term "enhancer" refers to the region of genomic DNA used to regulate genes up to 1 Mbp away. The enhancer may overlap with the gene coding region, but usually does not consist of it. Enhancers are usually bound by transcription factors and designated by specific tissue protein markers. The term "superenhancer" refers to a subset of enhancers that contain an asymmetric share of tissue protein markers and / or transcribed proteins relative to other enhancers in a particular cell. Because of this, it is predicted that genes regulated by superenhancers are highly important for the function of the cell. Superenhancers are usually determined by sorting all enhancers in the cell based on intensity and using available software such as ROSE (https://bitbucket.org/young_computation/rose) to determine that the A subset of high-strength enhancers is determined (see, for example, US Patent 9,181,580, which is incorporated herein by reference). The term "strength" as used herein when referring to the part of an enhancer or super-enhancer means the area under the curve of the number of reads of H3K27Ac or other genomic markers plotted against the length of the analyzed genomic DNA fragments. Therefore, "intensity" is the integral of the signal generated by measuring the marker at a given base pair over the span of base pairs defining the area selected for measurement. The term specifies the value (for example, withRARA The "prevalence rating" of the intensity of super-enhancers associated with genes means the percentage of groups that are equal to or greater than a certain value. For example, with the test cellRARA The prevalence of 35% of the intensity of gene-associated superenhancers means that 35% of the population has an intensity equal to or greater than that of the test cellRARA Gene enhancer. The term specifies the value (for example, withRARA The "prevalence cutoff" of the intensity of super-enhancers associated with genes means the prevalence of the dividing line that defines the dividing line between two subsets of a population (eg, responders and non-responders). Thus, a prevalence rating that is equal to or higher than (ie, a lower percentage value) a subset of the prevalence rating group that defines the prevalence cutoff; and a subset of the prevalence rating that is lower than (eg, a higher percentage value) prevalence definition community . The terms "cutoff" and "cutoff value" mean the value of the dividing line measured between two subsets of defined populations (eg, responders and non-responders) measured in the analysis method. Therefore, a value equal to or higher than the cut-off value defines a subset of the population; and a value lower than the cut-off value defines another subset of the population. The terms "threshold value" and "threshold value level" are meant to define the level of the dividing line between two subsets of a population (eg, responders and non-responders). The threshold level may be the prevailing cut-off or cut-off value. The term "population" or "population of samples" means a sufficient number of different samples (eg, at least 30, 40, 50, or more) that reasonably reflect the distribution of measured values in a larger group. Each sample in the sample population may be a cell line, a biological sample obtained from an organism (eg, a biopsy or body fluid sample), or a sample obtained from a xenotransplantation (eg, grown in a mouse by implanting a cell line or a patient sample Tumors), where each sample is from an organism with the same disease, condition or condition or from a cell line or xenograft showing the same disease, condition or condition. The term "ordinal ranking" of a specified value means that the value is ordered compared to the set of other values. For example, in terms of the intensity of the superenhancer associated with the RARA gene in the test cell compared to other superenhancers in the test cell, an ordinal rating of 100 means 99 other superenhancers in the test cell It has a greater intensity than the superenhancer associated with the RARA gene. The term "sorting" means sorting values from highest to lowest or from lowest to highest.RARA and IRF8 The present invention provides methods for treating individuals with cancer (eg, non-APL AML, MDS, or MM) with RARA agonists combined with CD38-specific antibodies, for example, by measuring RARA biomarkers and / or IRF8 The prevalence of biomarkers (for example, to determine one or moreRARA orIRF8 Gene component or product level, strength, ordinal ranking, prevalent ranking, and / or activity, including for exampleRARA orIRF8 Super-enhancer intensity, ordinal or prevalent grade and RARA or IRF8 mRNA level or prevalent grade), and therefore co-administered RARA agonist and CD38 specific antibody. Retinoic acid receptor subtype α (RARA) is a nuclear hormone receptor that acts as a transcription repressor when unbound or bound by an antagonist and as a gene activator in the agonist-bound state. The natural ligand of RARA is retinoic acid produced by vitamin A, also known as all-trans retinoic acid (ATRA). Super-enhancers (SE) are large, highly active chromatin regions that regulate key cell recognition genes (including oncogenes in malignant cells). Using a genetic control platform, SE was identified in 60 primary AML patient samples as a whole genome capable of discovering new tumor weaknesses. One of the SEs showing differences in the patient sample and the code RARARARA Genes are related. Use pre-analysisRARA Different AML cell lines and patient samples from the intensity and ordinal of the enhancer were found to have upregulated the level of interferon response factor 8 (IRF8) mRNA similar to RARA in the patient population. IRF8 is an interferon-reactive transcription factor known to be essential for hematopoiesis and its lack of signal transduction causes abnormal proliferation of immature bone marrow cells. In AML, IRF8 overexpression is observed and it may be correlated with poor clinical outcome. Regardless of this upregulation, IRF8 signaling is actually attenuated by suppressed transcription cofactors and potentially RARA when IRF8 signaling is in the SE-driven inhibition state. In addition, interferon-α itself (the upstream signaling ligand of IRF) exhibits pro-differentiation and cross-talk with the RARA pathway in AML. The present invention describes the latest discovery that RARA agonists (eg, Tamibarotene) can induce CD38 upregulation in non-APL AML, MDS, or MM in a RARA or IRF8 SE dependent manner. In addition, it provides various compositions and methods that are particularly suitable for characterizing, identifying, selecting, or stratifying patients based on a possible response to co-administration of Tamibarotene and CD38-specific antibodies.RARA and IRF8 Super enhancer identification and threshold Level determination Described herein is by determining the presence of RARA biomarkers and / or IRF8 biomarkers (eg, measuring one or moreRARA orIRF8 The prevalence, level, form and / or activity of genetic components or products, including for exampleRARA orIRF8 Super-enhancer intensity, ordinal or prevalent grading, and RARA or IRF8 mRNA level or prevalent grading), a method of treating cancer by administering RARA agonist (eg, Tamibarotene) and CD38 specific antibody. The identification of enhancers or super enhancers can be achieved by various methods known in the art, for example, as described in Cell 2013, 155, 934-947 and PCT / US2013 / 066957, both of which are cited by reference Incorporated in this article. In some embodiments, the identification of superenhancers is achieved by obtaining cellular material and DNA from cancer samples in patients (eg, from biopsies). The important measurement of enhancer measurement is performed in two dimensions: the DNA length of the genomic marker (eg, H3K27Ac) is continuously detected within it, and the genomic marker is located at each base pair along the DNA span that constitutes the measure Compile rate. The measurement of the area under the curve ("AUC") resulting from the integration of length and magnitude analysis determines the strength of the enhancer. systemIRF8 orRARA The intensity of the superenhancer relative to the control group used in one aspect of the invention determines whether the individual will respond to RARA agonists (eg, Tamibarotene) and CD38 specific antibodies. It will be easy for those familiar with this technique to detect the length of DNA within the genome markerRARA orIRF8 The same as the control group, thenRARA orIRF8 The ratio of the amount of superenhancer to the control group will be equivalent to intensity, and can also be used to determine whether an individual will respond to RARA agonists and CD38 specific antibodies. In some embodiments, prior to comparison with other samplesRARA orIRF8 The intensity of the enhancer is normalized. Normalization is achieved by comparison with regions in the same cell that are known to contain ubiquitous superenhancers or enhancers that appear at similar levels in all cells. An example of such a ubiquitous superenhancer region is the MALAT1 superenhancer locus (chr11: 65263724-65266724) (genomic structure hg19). It has been determined by the H3K27Ac ChIP-seq method to exist and be located at chr17: 38458152-38516681 (genomic structure hg19)RARA Gene-linked super-enhancer locus. ChIP sequencing (also called ChIP-seq) is used to analyze the interaction of proteins and DNA. ChIP-seq combines chromatin immunoprecipitation (ChIP) with massively parallel DNA sequencing to identify binding sites for DNA-related proteins. It can be used to pinpoint the global binding site of any protein of interest. Previously, ChIP-on-chip (ChIP-on-chip) was the most common technique used to study these protein-DNA relationships. Successful ChIP-seq depends on many factors, including the intensity and method of sonic processing, buffer composition, antibody quality, and cell number; see, for example, T. Furey, Nature Reviews Genetics 13, 840-852 (December 2012); ML Metzker, Nature Reviews Genetics 11, 31-46 (January 2010); and PJ Park, Nature Reviews Genetics 10, 669-680 (October 2009). Genomic markers other than H3K27Ac that can be used to identify super-enhancers using ChIP-seq include components of P300, CBP, BRD2, BRD3, BRD4, and media complex (J Loven et al., Cell, 153 (2): 320-334 , 2013), tissue protein 3 lysine 4 monomethylation (H3K4me1) or other tissue-specific enhancer-linked transcription factors (E Smith and A Shilatifard, Nat Struct Mol Biol, 21 (3): 210-219, 2014 ) (S Pott and Jason Lieb, Nature Genetics, 47 (1): 8-12, 2015). In some embodiments, the H3K27ac or other marker ChIP-seq data super-enhancer localization of the entire genome of the cell line or patient sample already exists. In these examples, we will only determine whether the intensity or ordinal rank of the enhancer or super-enhancer in such a location at the locus of chr17: 38458152-38516681 (genomic structure hg19) is equal to or greater than the predetermined threshold Level. It should be understood thatRARA ,IRF8 andMALAT1 The specific chromosomal location may be different for different genome structures and / or for different cell types. However, in view of this specification, those skilled in the art can locate theRARA And / orMALAT1 Such different positions are determined in such other genome-specific sequences of loci. Other methods for identifying superenhancers include chromatin immunoprecipitation (JE Delmore et al., Cell, 146 (6) 904-917, 2011) and chip array (ChIP-chip), and the use of the same immunoprecipitation genomic markers and chr17: 38458152-38516681 (genomic structure hg19)RARA Locus or chr16: 85862582-85990086 (genomic structure hg19)IRF8 Chromatin immunoprecipitation of oligonucleotide sequences hybridized to loci followed by qPCR (ChIP-qPCR). In the case of ChIP-chip, like other array-based technologies, the fluorescence intensity detection signal is usually generated by the hybridization of the probe and the input analysis sample. For ChIP-qPCR, dyes that emit fluorescence only after the double-stranded DNA generated in the PCR reaction is embedded are used to measure template amplification. In some embodiments, whether the cell has a threshold level greater than necessaryRARA orIRF8 The determination of superenhancers is achieved byRARA orIRF8 The strength of the enhancer corresponds to that of cells that are known not to respond to RARA or IRF8 ("control cells")RARA orIRF8 The intensity is compared to achieve. The control cell is preferably the same cell type as the test cell. In one aspect of these embodiments, the control cells are such cells in HCC1143. In some embodiments, whether the cell has a threshold level greater than necessaryRARA orIRF8 The determination of super-autron intensity is achieved byRARA orIRF8 Enhancer intensity and correspondence in cell sample populationRARA orIRF8 The intensity is compared by comparison, where each of the cell samples is obtained from different sources (ie, different individuals, different cell lines, different xenotransplantations). In some aspects of these embodiments, only the primary tumor cell sample from the individual is used to determine the threshold level. In some aspects of these embodiments, at least some of the samples in the population have been tested for response to specific RARA agonists and CD38 specific antibodies in order to establish: a) for specific RARA agonists and The sample with the lowest CD38 specific antibody responseRARA Enhancer intensity ("lowest responder"); and / or b) the lowest sample in the population that responds to specific RARA agonists and CD38 specific antibodiesIRF8 Enhancer intensity ("lowest responder"); and as appropriate c) the highest sample in the population that does not respond to specific RARA agonists and CD38 specific antibodiesRARA Enhancer intensity ("highest non-responder") and / or d) highest in population samples that do not respond to specific RARA agonists and CD38 specific antibodiesIRF8 Enhance sub-intensity ("the highest non-responder"). In these embodiments,RARA orIRF8 The intensity of the enhancer (the test cell will be regarded as reacting to the specific RARA agonist and CD38 specific antibody) when the intensity is greater than: i) equal to or at most greater than the lowest responder in the populationRARA orIRF8 Enhancer strength 5%; or ii) equal to or at most greater than the highest non-responder in the groupRARA orIRF8 Enhancer strength 5%; or iii) the lowest responder and highest non-responder in the groupRARA orIRF8 Value between enhancer intensities. It should be understood that in the above embodiments, generally not all samples in the population need to be tested for the reaction of RARA agonist and CD38 specific antibody, but all samples are measuredRARA orIRF8 Enhance sub-intensity. In some embodiments, the sample is based onRARA orIRF8 Enhancer intensity ranking. The selection of the three methods used to establish the cutoff described above will depend on the difference between the lowest responder and the highest non-responder in the group.RARA orIRF8 The intensities of the enhancers and the goal are to minimize the number of false positives or to minimize the chance of missing potential reaction samples or individuals. When the difference between the lowest responder and the highest non-responder is large (for example, when many samples are not tested and fallRARA orIRF8 (In the case of the reaction between the lowest responder and the highest non-responder in the order of enhancer intensity), the cutoff is usually set to be equal to or at most greater than the lowest responder in the groupRARA orIRF8 Strengthen sub-strength 5%. This cutoff maximizes the number of potential responders. When the difference is small (for example, when a few samples or no samples are not tested and fallRARA orIRF8 (In the order of strengthening the intensity of the reaction between the lowest responder and the highest non-responder), the cutoff is usually set to the lowest responder and the highest non-responderRARA orIRF8 Value between enhancer intensities. This cut-off minimizes the number of false positives. When the highest non-responderRARA orIRF8 When the intensifier intensity is higher than the lowest responder, the cutoff is usually set to be equal to or at most greater than the highest non-responderRARA orIRF8 Strengthen the value of 5%. This method also minimizes the number of false positives. In some embodiments, whether the cell has a threshold level greater than necessaryRARA orIRF8 The determination of super-autron intensity is achieved byRARA The number of enhancer intensities and the number of cell sample populationsRARA orIRF8 The ordinal number of enhancer intensity is compared by comparison, where each of the cell samples is obtained from different sources (ie, different individuals, different cell lines, different xenotransplantations). In these embodiments, at least some of the samples in the population have been tested for response to specific RARA agonists in order to establish: a) a population that responds to specific RARA agonists and CD38 specific antibodies The lowest sampleRARA Enhancer strength ("lowest ordinal responder"); b) The lowest sample in the population that responded to specific RARA agonists and CD38 specific antibodiesIRF8 Enhancer intensity ordinal number ("lowest ordinal responder"); and as appropriate c) the highest sample in the group that does not respond to specific RARA agonists and CD38 specific antibodiesRARA Enhancer intensity ordinal number ("the highest ordinal non-responder"); and / or d) the highest sample in the population that does not respond to specific RARA agonists and CD38 specific antibodiesRARA Strengthen the sub-intensity number ("the highest number without responders"). In these embodiments,RARA orIRF8 The intensity of the enhancer (the test cell will be regarded as reacting to the specific RARA agonist and CD38 specific antibody) when the intensity is greater than: i) is equal to or at most greater than the lowest ordinal responderRARA orIRF8 The intensity of the enhancer is 5%; or ii) equal to or at most greater than the highest number of non-responders in the groupRARA orIRF8 Enhancer intensity 5%; or iii) The lowest ordinal responder in the group and the highest ordinal non-responderRARA orIRF8 Value between enhancer intensities. It should be understood that in the above embodiments, not all samples in the population usually need to be tested for the response to RARA agonist and CD38 specific antibody, but all samples are measured.RARA orIRF8 Enhance substrength and establishRARA orIRF8 The number of enhancers compared to other enhancers in the same sample. The ordinal number is usually determined by measuring the strength of all other enhancers in the cell and determiningRARA orIRF8 Compared with other enhancers, what kind of order does the enhancer have in terms of strength (ie, ordinal number). In some embodiments, the sample is based onRARA Sort by the ordinal number of enhancer intensity. In some embodiments, the sample is based onIRF8 Sort by the ordinal number of enhancer intensity. The selection of the sample used to establish the cutoff method described above will depend on the difference between the lowest ordinal responder and the highest ordinal responder in the population.RARA orIRF8 The sequence number difference and cutoff of enhancer intensity are designed to minimize the number of false positives or maximize the number of responders. When this difference is large (for example, when many samples are not tested and fallRARA orIRF8 (Strength of the ordinal substrength ordinal number is the reaction between the lowest ordinal responder and the highest ordinal non-responder).RARA orIRF8 The intensity number of the enhancer is 5%. When this difference is small (for example, when a few samples or no samples are not tested and fallRARA orIRF8 (Strength of substrength ordinal number is the reaction between the lowest ordinal responder and the highest ordinal non-responder), the cutoff is usually set to the lowest ordinal responder and the highest ordinal non-responderRARA orIRF8 Values between ordinal intensity intensities. When the highest number of non-respondersRARA orIRF8 The sequence number of the enhancer intensity is higher than the lowest responderRARA orIRF8 When strengthening the ordinal intensity number, the cutoff is usually set to be equal to or at most greater than the highest number in the group of non-respondersRARA orIRF8 The value of 5% of the ordinal intensity. In some aspects of embodiments in which test cells or samples are compared to a population, one or more cut-off values will be obtained for the population (eg,RARA Enhancer strength orRARA Strengthen sub-sequence and / orIRF8 Enhancer strength orIRF8 The enhanced sub-sequence number) is converted into a prevalence ranking, and the cut-off is expressed as the percentage of the population having a cut-off value or higher (ie, prevalence cut-off). Without being limited by theory, the applicant believes that the prevalence of test samples will be similar, regardless of whether it is used for determinationRARA orIRF8 How to strengthen the sub-intensity. Therefore, for a parameter (for example,RARA Enhancer intensity ordinalIRF8 The prevalence cutoff of the enhancer intensity number) is portable and can be applied to another parameter (eg, RARA mRNA level or IRF8 mRNA level) to determine the cutoff value of the other parameter. This allows the cut-off value of any parameter to be determined without having to experimentally determine the correlation between the level of such parameter and the response to the RARA agonist and CD38 specific antibody.RARA and IRF8 mRNA Level determination The present invention provides methods for treating cancer with RARA agonists (eg, Tamibarotene) and CD38 specific antibodies in the presence of RARA biomarkers and / or IRF8 biomarkers. DeterminedRARA And / orIRF8 The identification of super-enhancer loci allows us to use RNA transcripts to measure sensitivity rather than super-enhancer levels to determine sensitivity to RARA agonists and CD38 specific antibodies. The RNA transcript from the super-enhancer locus itself can be quantified and is extremely well correlated with the super-enhancer level at that locus. It has also been shown that mRNA transcripts encoding RARA are also associated with sensitivity to RARA agonists alone or in combination with CD38-specific antibodies, and therefore mRNA levels can be used to identify cells that will respond to this therapy combination. Therefore, in some embodiments, the RARA or IFR8 mRNA level rather than superenhancer intensity or ordinal ranking can be used to determine sensitivity to RARA agonists and CD38 specific antibodies. In some embodiments, RNA transcription levels from super-enhancer loci are quantified using quantitative techniques, which quantify theRARA orIRF8 Enhancer RNA transcription levels correspond to cells or cell lines known not to respond to RARA agonists and CD38 specific antibodiesRARA orIRF8 Enhancer RNA transcription levels are compared. Such methods include RNA array-based or sequencing-based methods for eRNA (N Hah et al., PNAS, 112 (3): E297-302, 2015) associated with read-through and enhancer read-through, and RNA qPCR. In an alternative embodiment, the RNA-Seq or RNA-qPCR technology is used to place the RARA or IRF8 mRNA levels in the individual (ie, in the tumor sample, cancer cell sample, blood sample, etc.) as if they had the same disease or condition RARA or IRF8 mRNA levels in a population of individuals are compared to identify responders to RARA agonists and CD38 specific antibodies. In these embodiments, at least some of the samples in the population have been tested for response to specific RARA agonists and CD38 specific antibodies in order to establish: a) specific RARA agonists and CD38 specific antibodies The lowest RARA mRNA level of the sample in the responding group ("the lowest mRNA responder"); and / or b) the lowest IRF8 mRNA of the sample in the group responding to the specific RARA agonist and CD38 specific antibody Level ("Minimum mRNA Responder"); and optionally c) The highest RARA mRNA level of samples in a population that does not respond to specific RARA agonists and CD38-specific antibodies ("Highest mRNA Unresponder") ); And / or d) the highest level of IRF8 mRNA in samples from groups that do not respond to specific RARA agonists and CD38 specific antibodies ("the highest mRNA non-responders"). In these embodiments, the RARA or IRF8 mRNA level (a test cell greater than this level will be considered to react to the specific RARA agonist and CD38 specific antibody) is set to: i) equal to or at most The RARA or IRF8 mRNA level of the lowest mRNA responder in the population is 5%; or ii) is equal to or at most greater than the RARA or IRF8 mRNA level of the highest mRNA non-responder in the population 5%; or iii) the lowest mRNA responder in the population is The value between RARA or IRF8 mRNA level of the highest mRNA non-responder. In some embodiments, not all samples in a population need to be tested for response to RARA agonists and CD38 specific antibodies, but all samples are measured for RARA or IRF8 mRNA levels. In some embodiments, the samples are ordered based on RARA mRNA levels. In some embodiments, the samples are ordered based on IRF8 mRNA levels. The selection of samples for the three methods described above to establish the cut-off will depend on the RARA or IRF8 mRNA level difference between the lowest mRNA responder and the highest mRNA non-responder in the population and the cut-off is designed to minimize false positives Or to maximize the number of potential responders. When this difference is large (for example, when many samples are not tested for the reaction between the lowest mRNA responder and the highest mRNA non-responder in the order of RARA or IRF8 mRNA level), the cutoff is usually set to be equal to or at most The RARA or IRF8 mRNA level of the lowest mRNA responder in the population was 5%. When the difference is small (for example, when a small number of samples or no samples are not tested, the reaction falls between the lowest mRNA responder and the highest mRNA non-responder in the ranking of RARA or IRF8 mRNA level), the cutoff is usually set to the lowest The value between the RARA or IRF8 mRNA level of the mRNA responder and the highest mRNA non-responder. When the RARA or IRF8 mRNA level of the highest mRNA non-responder is higher than the lowest mRNA responder, the cutoff is usually set to a value equal to or at most greater than 5% of the RARA or IRF8 mRNA level of the highest mRNA non-responder in the population. In some embodiments, the population is ranked based on RARA mRNA levels. In some embodiments, the population is ranked based on IRF8 mRNA levels. In these embodiments, the RARA or IRF8 mRNA levels in each sample are measured and compared to the mRNA levels of all other mRNAs in the cell to obtain the ordinal ranking of the RARA or IRF8 mRNA levels. Subsequently, the cut-off based on RARA or IRF8 mRNA ordinal ranking is determined based on the samples in the population toRARA orIFR8 The response of the samples to the RARA agonist was tested in the same manner as described in the Super Enhancer Intensity Number Cutoff. Subsequently, the measured RARA or IRF8 mRNA number cutoff is used directly or used to determine the prevalence cutoff, and then either of them is used to stratify additional samples that potentially respond to RARA agonists and CD38 specific antibodies. In some embodiments, the use is based onRARA Enhancer strength orRARA Enhancer intensity number, orIRF8 Enhancer strength orIRF8 The prevalent cut-off established by the intensity number of the enhancer was used to determine the cut-off of the RARA or IRF8 mRNA level. In some aspects of these embodiments, the mRNA level of the population is measured, and the pre-determined prevalence cutoff is applied to the population to determine the mRNA cutoff level. In some aspects of these embodiments, a ranking standard curve for the RARA or IRF8 mRNA level of the population is established, and a predetermined prevalent cutoff is applied to the standard curve to determine the RARA or IRF8 mRNA cutoff level. In some aspects of the embodiment where the test cell or sample is compared with the population, the cut-off mRNA level value of the obtained population is converted into a prevalent grade, and the mRNA level cut-off is expressed as having a cut-off value or higher ( That is, the percentage of groups that are prevalently cut off. Without being limited by theory, the applicant believes that the prevalence of test samples and the prevalence of population will be similar, regardless of the method used to determine IRF8 or RARA mRNA levels. In some aspects of these embodiments, if the individual's RARA or IRF8 mRNA level corresponds to 79%, 78%, 77%, 76%, 75 as determined by the RARA or IRF8 mRNA level in the population %, 74%, 73%, 72%, 71%, 70%, 69%, 68%, 67%, 66%, 65%, 64%, 63%, 62%, 61%, 60%, 59%, 58%, 57%, 56%, 55%, 54%, 53%, 52%, 51%, 50%, 49%, 48%, 47%, 46%, 45%, 44%, 43%, 42% , 41%, 40%, 39%, 38%, 37%, 36%, 35%, 34%, 33%, 32%, 31%, 30%, 29%, 28%, 27%, 26%, 25 %, 24%, 23%, 22%, 21%, or 20% of the prevalent groups, the individual is identified as a RARA agonist responder. In one aspect of these embodiments, based onRARA orIRF8 The prevalence of strengthening substrength is established and the cutoff value is established. In one alternative form of these embodiments, based onRARA orIRF8 The prevalence of the establishment of the substrength ordinal number cutoff establishes the cutoff value. In another alternative aspect of these embodiments, a cut-off value is established based on the RARA or IRF8 mRNA level. In other more specific aspects of these embodiments, based onRARA orIRF8 The prevalence value of the enhancer intensity ordinal number determination establishes the cut-off value of AML patients, and the prevalence value is used to determine the cut-off value of RARA or IRF8 mRNA level. In even more specific aspects of these embodiments, use is between 25 to 45% (eg, between 25 to 30%, 25 to 35%, 25 to 40%, 30 to 35%, 30 to 40%, 35 to 45%, 35 to 40%, 31 to 35%, 32 to 35%, 33 to 35%, 34 to 35%, 31 to 36%, 32 to 36%, 33 to 36%, 34 to 36% or Between 35 and 36%) to determine the cut-off value of AML patients. In other even more specific aspects of these embodiments, a prevalence value of 36% is used to determine the cutoff value for AML patients. In yet other even more specific aspects of these embodiments, a prevalence value of 25% is used to determine the cutoff value for AML patients. In still other embodiments, the group may be divided into three groups-responders, partial responders, and non-responders, and two cut-off values or prevalent cut-off values are set. Part of the responder group may include responders and non-responders, as well as members of other groups whose response to RARA agonists and CD38 specific antibodies is not as high as the responder group. In these embodiments, two cutoff values or prevailing cutoff values are determined. This type of stratification may be particularly applicable when the RARA or IRF8 mRNA level of the highest RARA or IRF8 mRNA non-responder in the population is higher than the lowest RARA mRNA responder. In this situation, the cutoff level or prevalence cutoff between the responders and some of the responders is set to be equal to or at most greater than 5% of the RARA or IRF8 mRNA level of the highest RARA or IRF8 mRNA non-responders; The cutoff level or prevalence cutoff between responders is set to be equal to or at most greater than 5% of the RARA or IRF8 mRNA level of the lowest RARA or IRF8 mRNA responder. The decision whether to administer RARA agonists and CD38 specific antibodies to some responders will depend on the judgment of the treating physician and / or the approval of the regulatory agency. Methods for quantifying specific RNA sequences in cells or biological samples are known in the art, and include, but are not limited to, such as fluorescent hybridization for services and products provided by NanoString Technologies, array-based technology (Affymetrix), Reverse transcriptase qPCR using SYBR® Green (Life Technologies) or TaqMan® technology (Life Technologies), RNA sequencing (e.g., RNA-seq), RNA hybridization and signals as used with RNAscope® (Advanced Cell Diagnostics) Zoom in, or northern blot. In some aspects of these embodiments, the RNA transcript (mRNA or anotherRARA orIRF8 Transcripts) are normalized. Normalization involves the use of another RNA transcript (eg, GADPH mRNA, 18S RNA) that is native to its parents at the equivalent level in both cells, or prior to the determination of superaugrator intensity. "Apply" to the fixed level in the sample of each of the cells to adjust for comparisonRARA orIRF8 The measured levels of RNA transcripts (J Lovén et al., Cell, 151 (3): 476-82 (2012); J Kanno et al., BMC Genomics 7:64 (2006); J Van de Peppel et al., EMBO Rep 4: 387-93 (2003)).RARA Agonists and targeting CD38 Of Antibodies The present invention provides methods for treating individuals with cancer (eg, non-APL AML, MDS, or MM) with RARA agonists in combination with CD38-specific antibodies when, for example, RARA or IRF8 biomarkers are identified. RARA Agonist Used for treatment identification as havingRARA The selection of RARA agonists for patients with gene-associated superenhancers can be made from any RARA agonist known in the art. Preferably, the RARA agonist pair used in the method of the inventionRARA Other forms specific for RaR (eg, RaR-ß and RaR-γ) are significantly lower (at least 10 ×, at least 100 ×, at least 1,000 ×, at least 10,000 ×, at least 100,000 ×) Promoting activity. In some embodiments, the RARA agonist is selected from any compound disclosed in any of the following U.S. patents or within the class set forth in any of these patents: US 4,703,110, US 5,081,271, US 5,089,509, US 5,455,265, US 5,759,785, US 5,856,490, US 5,965,606, US 6,063,797, US 6,071,924, US 6,075,032, US 6,187,950, US 6,355,669, US 6,358,995 and US 6,387,950, each of which is incorporated herein by reference. In some embodiments, the RARA agonist is selected from any one of the known RARA agonists described in Table 1 below, or a pharmaceutically acceptable salt thereof, or a solvate or hydrate of the foregoing Materials: Table 1. Exemplary RARA agonists suitable for the present invention. In some embodiments, the RARA agonist is Tamibarotene. In some embodiments, the RARA agonist is(AGN-195183). CD38 Specific antibody CD38 is a transmembrane glycoprotein involved in many metabolic functions (including catabolism of extracellular nucleotides, receptor-mediated adhesion, migration regulation, and various signaling events). Antibodies targeting CD38 can modulate any of these functions, thereby promoting the efficacy of RARA agonists in cancer treatment. In some embodiments, the CD38 specific antibody can recognize and / or bind to any part of the CD38 fragment. CD38-specific antibodies may comprise monoclonal antibodies, humanized antibodies, or human antibodies. Exemplary antibodies specific for CD38 include isatuximab, daratumumab, MOR202, Ab79, Ab19 and EPR4106. In some embodiments, the CD38 specific antibody is daclizumab.treatment solutions The method of the present invention is theoretically applicable to treatments characterized by biomarkers with RARA or IRF8 (eg, one or moreRARA orIRF8 The presence, level, form and / or activity of genetic components or products, including for exampleRARA orIRF8 Super enhancer intensity, ordinal classification or prevalent classification andRARA Or IRF8 mRNA level or prevalence grade) associated with cancer. SuperenhancerRARA orIRF8 Genes can be more common in certain types of cancer than others. The present invention is specifically related to AML (for example, not characterized as involvingRARA Gene chromosomal translocation (non-APL AML and other forms of AML) and the treatment of MDS and MM. In some embodiments, the disease to be treated is not characterized as involvingRARA Non-APL AML, MDS or MM of chromosomal translocation of genes. In some embodiments, the disease to be treated is not characterized as involvingIRF8 Non-APL AML, MDS or MM of chromosomal translocation of genes. In some embodiments, individuals to be treated with RARA agonists (eg, Tamibarotene) and CD38-specific antibodies have relapsed or refractory non-APL AML. If the individual: a) does not show a partial response after the first cycle of induction chemotherapy; or b) does not show a complete response after the second cycle of induction chemotherapy; or c) relapses after conventional chemotherapy; or d) Relapse after a single stem cell transplantation is classified as having relapsed or refractory non-APL AML. In some embodiments, individuals to be treated with RARA agonists (eg, Tamibarotene) and CD38 specific antibodies have refractory MM. In other embodiments, the individual to be treated with a RARA agonist (eg, Tamibarotene) is an inappropriate elderly individual. The term "inappropriate elderly person" as used herein means a person who is at least 60 years old and is determined by a physician as a candidate for non-standard induction therapy. In some embodiments, the individual is co-administered with an RARA agonist (eg, Tamibarotene) and a CD38-specific antibody (eg, darumumab). In some embodiments, the RARA agonist (eg, Tamibarotene) and CD38-specific antibody (eg, darumumab) are administered simultaneously. In some embodiments, the RARA agonist (eg, Tamibarotene) and the CD38-specific antibody (eg, darumumab) are administered to each other within about 1 hour to about 48 hours. In some embodiments, the RARA agonist (eg, Tamibarotene) and the CD38-specific antibody (eg, darumumab) are administered to each other within the following time: about 1 minute, about 2 minutes, about 5 Minutes, about 10 minutes, about 15 minutes, about 20 minutes, about 30 minutes, about 45 minutes, about 1 hour, about 1.5 hours, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, About 8 hours, about 10 hours, about 12 hours, about 16 hours, about 20 hours, about 24 hours, about 36 hours, or about 48 hours. In some embodiments, the patient population includes one or more individuals (e.g., comprising or consisting of multiple individuals) undergoing previous therapy for cancer (eg, non-APL AML, MDS, or MM). In some embodiments, the patient population includes one or more individuals (eg, comprising or consisting of multiple individuals) who have not received previous therapy for cancer (eg, non-APL AML, MDS, or MM). In some embodiments, the patient population includes or consists of patients who have not received prior therapy with non-APL AML, MDS, or MM. In some embodiments, the patient receiving the previous therapy may have received a previous therapy selected from the group consisting of chemotherapy, immunotherapy, radiation therapy, palliative care, surgery, and combinations thereof. In some embodiments, the patient has undergone transplantation. In some embodiments, the patient has received standard cytotoxic chemotherapy. In some embodiments, standard cytotoxic chemotherapy includes cytarabine and / or anthracycline. In some embodiments, standard cytotoxic chemotherapy may include additional chemotherapy and / or hematopoietic stem cell transplantation (HSCT). In some embodiments, the patient has received a hypomethylated agent. In some embodiments, the patient has received lenalidomide. In some embodiments, the patient population includes those who have received and / or are receiving other therapies such as to allow administration of RARA agonist therapy (e.g., tamil) in combination with other therapies (eg, chemotherapy agents) in addition to CD38 specific antibodies (Barorotene) One or more individuals of the composition (e.g., comprising or consisting of multiple individuals). In some embodiments, such other therapies may include cancer (eg, as described herein), therapy for pain, nausea, constipation, etc., one or more side effects associated with cancer therapy (eg, pruritus, off Hair, insomnia, etc.) or any combination thereof, or consist of it. The present invention provides a method for treating non-APL AML, MDS, or MM, which includes therapy with a therapeutically effective amount of RARA agonist (eg, Tamibarotene) or a pharmaceutically acceptable salt thereof and a CD38-specific antibody Treatment is identified as patients with non-APL AML, MDS, or MM. In some embodiments, the present invention provides a method of treating non-APL AML, MDS, or MM of a patient previously treated with a treatment regimen including chemotherapy by administering a therapeutically effective amount of RARA to such patient Agonists (eg, Tamibarotene) and CD38 specific antibodies are performed. In some embodiments, the present invention provides a method for treating non-APL AML, MDS, or MM of patients without the presence of standard therapies. In some embodiments, the present invention provides a method for treating patients who are not suitable for standard therapy. In some embodiments, the patient or patient group may not be (eg, may not include) a patient with a previous history of allergy to the ingredients of Tamibarotene. In some embodiments, the patient or group of patients may not be (eg, may not include) patients who are receiving vitamin A formulations. In some embodiments, the patient or group of patients may not be (eg, may not include) patients suffering from hypervitaminosis A. In some embodiments, the patient or group of patients may not be (eg, may not include) elderly patients. In some embodiments, the patient or group of patients may be or include one or more elderly patients. In some embodiments, when compared to one or more younger patients, older patients may be monitored more frequently to detect possible adverse events (including, for example, low levels of serum albumin and / or plasma Higher concentration of free drugs, etc.). In some embodiments, administration of RARA agonists and / or CD38 specific antibodies may be reduced, suspended, and / or terminated in elderly patients determined to exhibit one or more symptoms of such adverse events.Dosage form and dosing regimen In general, each therapeutic agent used in accordance with the present invention is formulated, administered, and administered in a therapeutically effective amount in accordance with good medical practice and in a pharmaceutical composition and administration regimen suitable for the relevant agent and individual (e.g. Agents or CD38 specific antibodies). In principle, the therapeutic composition can be administered by any suitable method known in the art, including but not limited to oral, transmucosal, inhalation, topical, buccal, nasal, transrectal or parenteral Administration (eg, intravenous, infusion, intratumor, intranodular, subcutaneous, intraperitoneal, intramuscular, intradermal, transdermal, or other types of administration). In some embodiments, the RARA agonist (eg, Tamibarotene) will be administered orally. In some embodiments, the CD38 specific antibody will be administered intravenously. In some embodiments, the dosing regimen of a specific active agent may involve intermittent or continuous administration, for example, to achieve a specific desired pharmacokinetic profile or other exposure in one or more tissues or body fluids of interest in the individual receiving therapy mode. In some embodiments, different agents administered in combination may be administered via different delivery routes and / or according to different time courses. Alternatively or additionally, in some embodiments, one or more doses of the first active agent are administered substantially simultaneously with one or more other active agents, and in some embodiments, the one or more doses of the first active agent It is administered with one or more other active agents via common routes and / or as part of a single composition. Factors considered in optimizing the route and / or dosing schedule of a given treatment regimen may include, for example: the specific indication being treated, the individual ’s clinical condition (eg, age, overall health status, previous received Therapy and / or its response, etc.), the site of delivery of the agent, the nature of the agent, the mode and / or route of administration of the agent, the presence or absence of combination therapy and other factors known to medical practitioners. For example, in the treatment of cancer, the relevant characteristics of the indication being treated may include, among other things, one or more of the type, stage, location, etc. of the cancer. In some embodiments, one or more characteristics of a particular pharmaceutical composition and / or dosing regimen used can be adjusted over time (eg, increase or decrease the effective amount in any individual dose, increase or decrease administration Time interval, etc.), for example, in order to optimize the desired therapeutic effect or response. In general, the type, amount, and frequency of administration of the active agent according to the present invention are controlled by the safety and efficacy requirements for the application of one or more related agents to mammals (preferably humans). In general, such administration characteristics are selected to provide specific and usually detectable treatment responses compared to unobserved therapies. In the context of the present invention, exemplary desired therapeutic responses may include, but are not limited to, inhibition and / or reduction of one or more of tumor growth, tumor size, metastasis, symptoms and side effects associated with the tumor, and tumor Increased apoptosis, treatment-related decrease or increase in one or more cell markers or circulating markers, and the like. Such criteria can be easily assessed by any of various immunology, cytology, and other methods disclosed in the literature. In some embodiments, based on the timing and / or threshold performance of inducible markers, it may be necessary to adapt the dosing regimen, and in particular, a continuous dosing regimen needs to be designed, Patient groups (eg, patient groups carrying genetic markers) and / or specific patients. In some such embodiments, the therapeutic dosing regimen can be combined with or adjusted according to a detection method that evaluates the performance of one or more inducible markers before and / or during therapy. In some embodiments, the RARA agonist (eg, Tamibarotene) therapy regimen contains about 1 mg / m2 , 2 mg / m2 , 3 mg / m2 , 4 mg / m2 , 5 mg / m2 , 6 mg / m2 , 7 mg / m2 , 8 mg / m2 , 9 mg / m2 , 10 mg / m2 , 11 mg / m2 , 12 mg / m2 , 13 mg / m2 , 14 mg / m2 , 15 mg / m2 , 16 mg / m2 At least one (or include one or consist of exactly one) dose, or a dose between any of these equivalents of a RARA agonist (eg, Tamibarotene). In some embodiments, the RARA agonist (eg, Tamibarotene) therapy regimen is included at 1 mg / m2 With 50 mg / m2 Between doses. In some embodiments, the RARA agonist (eg, Tamibarotene) therapy regimen is contained at 5 mg / m2 With 25 mg / m2 Between doses. In some embodiments, the RARA agonist (eg, Tamibarotene) therapy regimen is contained at 5 mg / m2 With 15 mg / m2 Between doses. In some embodiments, the RARA agonist (eg, Tamibarotene) therapy regimen comprises 12 mg / m2 Of dosage. In some embodiments, the RARA agonist (eg, Tamibarotene) therapy regimen contains 6 mg / m2 Of dosage. In some embodiments, the RARA agonist (eg, Tamibarotene) therapy regimen includes multiple doses of the Tamibarotene composition. In some such embodiments, the Tamibarotene therapy regimen includes, for example, 2, 5, 10, 20, 30, 60, 90, 180, 365 doses or one of these values Multiple doses between any two, and / or include repeated dose patterns (eg, at least one cycle of two daily doses, which can be repeated, with alternate administration as appropriate or without administration for a period of time Open different cycles). In some embodiments, the Tamibarotene therapy regimen is administered twice a day. In some embodiments, the Tamibarotene therapy regimen is administered once a day. In some embodiments, the Tamibarotene therapy regimen contains 6 mg / m2 Up to 12 mg / m2 The total dose is divided into oral administration twice a day. In some embodiments, the CD38 specific antibody (eg, daclizumab) therapy regimen comprises about 0.5 mg / kg, 1 mg / kg, 2 mg / kg, 5 mg / kg, 7.5 mg / kg, 10 mg / kg, 11 mg / kg, 12 mg / kg, 13 mg / kg, 14 mg / kg, 15 mg / kg, 16 mg / kg, 17 mg / kg, 18 mg / kg, 19 mg / kg, 20 mg / At least one (or include one or consist of exactly one) dose of kg, 25 mg / kg, 30 mg / kg, 35 mg / kg, 40 mg / kg, 45 mg / kg, 50 mg / kg, or CD38 specific The dose between any two of these equivalent values of antibody (eg, daclizumab). In some embodiments, the CD38 specific antibody (eg, daclizumab) therapy regimen contains a dose between 1 mg / kg and 100 mg / kg. In some embodiments, a CD38 specific antibody (eg, daclizumab) therapy regimen contains a dose between 5 mg / kg and 50 mg / kg. In some embodiments, the CD38 specific antibody (eg, daclizumab) therapy regimen contains a dose between 10 mg / kg and 20 mg / kg. In some embodiments, the CD38 specific antibody (eg, daclizumab) therapy regimen comprises multiple doses of the daclizumab composition. In some such embodiments, the daclizumab therapy regimen includes, for example, 2, 5, 10, 20, 30, 60, 90, 180, 365 doses, or any of these values Multiple doses between the two, and / or include a repeated dose pattern (eg, at least one cycle of two daily doses, which can be repeated, separated by alternative or non-administrative periods as appropriate Different cycles). In some embodiments, the daclizumab therapy regimen is administered once a week. In some embodiments, daclizumab therapy regimen is administered at most once a week. In some embodiments, the daclizumab therapy regimen is administered every two weeks. In some embodiments, the daclizumab therapy regimen contains a total dose of 10 mg / kg to 20 mg / kg, at most once a week.Formulation As used herein, a pharmaceutical composition refers to a compound (such as a Tamibarotene or CD38 specific antibody) and other chemical components (such as a carrier, stabilizer, diluent, dispersant, suspending agent, thickener) And / or excipients). The pharmaceutical composition helps to administer the compound to the organism. The pharmaceutical composition containing the compound can be administered in a therapeutically effective amount by any known form and route known in the art including but not limited to the following: intravenous, oral, rectal, aerosol, parenteral , Eye, lung, skin, vagina, ear, nose and local administration. For oral administration, the compound can be easily formulated by combining the active compound with pharmaceutically acceptable carriers or excipients well known in the art. Such carriers allow the compounds described herein to be formulated into tablets, powders, pills, sugar-coated pills, capsules, liquids, gels, syrups, elixirs, slurries, suspensions and Its analogs. In general, excipients (such as fillers, disintegrants, slip agents, surfactants, recrystallization inhibitors, lubricants, pigments, binders, flavoring agents, etc.) can be used for routine purposes and without affecting the combination Typical amounts of physical properties are used. In some embodiments, the excipient is one or more of lactose hydrate, corn starch, hydroxypropyl cellulose, and / or magnesium stearate. In some embodiments, Tamibarotene can be formulated with one or more of lactose hydrate, corn starch, hydroxypropyl cellulose, and / or magnesium stearate. The identification of acceptable formulations of Tamibarotene can be achieved by various methods known in the art, for example, as described in US 20100048708, which is incorporated herein by reference.Packaged pharmaceutical composition The packaged pharmaceutical composition of the present invention comprises a written insert or marker, which is included in cancer and has been determined to have an intensity or ordinal grade equal to or greater than the threshold level, or the RARA mRNA level is equal to or greater than the Limit levelRARA Instructions for the use of RARA agonists and CD38-targeting antibodies in individuals with gene-associated superenhancers. As described in detail above, the threshold is determined in a sample population from a cell line or xenograft model from an individual diagnosed with the same disease or the same disease as the disease in which the pharmaceutical composition is indicated to treat the disease Level. Description Can be adhered or otherwise attached to a container containing RARA agonist and CD38-targeting antibody. Alternatively, the instructions and the containers containing the RARA agonist will be separate from each other, but together exist in a single package, box, or other type of container. Instructions for packaged pharmaceutical compositions will usually be authorized or recommended by government agencies that approve the therapeutic use of RARA agonists and antibodies targeting CD38. The description may include determining whether the superenhancer is compatible withRARA orIRF8 Specific methods associated with genes, and determination andRARA orIRF8 Quantitative method to determine whether the gene-related enhancer is a super-enhancer, and the quantitative method to determine the RARA or IRF8 mRNA level; and / or the threshold level of the super-enhancer or RARA or IRF8 mRNA, at the threshold level Treatment with packaged RARA agonists and antibodies targeting CD38 is recommended and / or presumed to be effective. In some aspects, the instructions instruct to administer the composition to an individual whose RARA or IRF8 mRNA level falls in at least the 30th percentile of a population whose RARA or IRF8 mRNA level has been measured. In some aspects, if the individual ’s RARA or IRF8 mRNA level is prevalently classified as 79%, 78%, 77%, 76%, 75%, 74%, 73% of the group whose RARA or IRF8 mRNA level has been measured , 72%, 71%, 70%, 69%, 68%, 67%, 66%, 65%, 64%, 63%, 62%, 61%, 60%, 59%, 58%, 57%, 56 %, 55%, 54%, 53%, 52%, 51%, 50%, 49%, 48%, 47%, 46%, 45%, 44%, 43%, 42%, 41%, 40%, 39%, 38%, 37%, 36%, 35%, 34%, 33%, 32%, 31%, 30%, 29%, 28%, 27%, 26%, 25%, 24%, 23% , 22%, 21%, or 20%, the individual is identified as a RARA agonist responder. In some aspects, instructions are given to administer the composition to individuals whose RARA or IRF8 mRNA levels have been measured by specific tests. Instructions may include information on administration, the type of cancer approved for treatment with RARA agonists and / or CD38-targeting antibodies, physicochemical information on RARA agonists and / or CD38-targeting antibodies; about RARA agonists Information on the pharmacokinetics of the agent and / or antibody targeting CD38; or information on drug-drug interactions. In some aspects, the instructions direct administration of the composition to individuals diagnosed with non-APL AML. In some aspects, the instructions direct administration of the composition to individuals diagnosed with non-APL MM. In some aspects, the pharmaceutical composition comprises Tamibarotene. In some aspects, the pharmaceutical composition comprises AGN-195183. In some aspects, the pharmaceutical composition comprises daclizumab. In some embodiments, the pharmaceutical composition includes both Tamibarotene and Daclizumab. Examples For a more complete understanding of the invention described herein, the following examples are illustrated. The synthetic and biological examples described in this application are provided to illustrate the compounds, pharmaceutical compositions and methods provided herein, and should not be construed as limiting its scope in any way.Examples 1 . RARA RNA And measurement of protein expression Use performance measurement to determine the markedRARA Gene mRNA level. The mRNA level is well correlated with the enhancer level and therefore also has predictive sensitivity to RARA agonists. We use various methods for measuring RNA as described below.I . Array-based technology. Use 1x10 in three equal batches6 Cells to evaluate the performance in HCC1143 and AU565. Follow manufacturer's protocol, use Trizol® to extract RNA from cells and use mirVanaTM RNA purification kits purify RNA (both from Life Technologies). Affymetrix PrimeView at Dana Farber Cancer Institute Microarray Core (http://mbcf.dfci.harvard.edu/)TM Read the RNA level on the array. Figure 1 shows the mRNA expression levels of various RAR subtypes measured in the Tamibarotene reactive (Au565) and non-reactive (HCC1143) cell lines using the above protocol. The expression of RARA mRNA in reactive cell lines was 8 times higher than that of non-reactive cell lines, and the performance of RaR-ß and RaR-γ did not differ significantly between these cell lines. This confirms RARA mRNA performance analysis andRARA Superenhancer strength is related to sensitivity to RARA agonists, and indicates that RARA mRNA levels can be used to predict sensitivity to such agonists.II . RNA - Seq . RARA performance was quantified by RNA-Seq. Run Poly-A RNA-Seq and use rsem v1.2.21 software (rsem-calculate-expression; parameter = -p 4 --samtools-sort-mem 3G --ci-memory 3072 --bowtie-chunkmbs 1024 --quiet- -output-genome-bam --bowtie2 --bowtie2-path /data/devtools/bowtie2-2.0.5 --strand-specific) compare reads to HG19 transcriptome, and then use the same rsem program set (rsem -parse-alignments, rsem-build-read-index, rsem-run-em) and report mRNA quantification per million transcripts (TPM), which represents the specified gene for each million transcripts detected Estimate of the number of transcripts. Subsequently, all protein-coding genes were extracted for each sample and scored together for quantile normalization. We process all AML patient samples, primary AML patient samples and AML cell lines from the PDX model and normalize them together to generate universal mRNA scores. Plotted values show the log2 (TPM + 1) level (y-axis) of RARA for 48 primary AML patientsRARA / MALAT1) (Figure 1).Examples 2 . AML Nakano RARA Super enhancer intensity ordinal number cutoff H3K27Ac and ChIP-Seq were used to analyze the overall enhancer / super enhancer maps of 95 AML samples (both patient samples and AML cell lines (including SigM5, MV411, HEL, and Kasumi1)). In each of the samples, determineRARA The relative enhancers are ranked in terms of strength (as measured by H3K27Ac) compared to other enhancers and superenhancers in the same cell, and the measured ordinal ranks are plotted on the sorting bar graph (Figure 2 ). In MV411, determineRARA The related enhancer is the 133rd strongest enhancer. MV411 is the Tamibarotene-reactive cell line confirmed to have the lowest superenhancer intensity number. In HEL, determineRARA The related enhancer is the 155th strongest enhancer. HEL is a non-reactive cell line of Tamibarotene that has been confirmed to have the highest superenhancer intensity number. Based on these values, we setRARA The cut-off of the intensifier ordinal number is 150 (value between HEL ordinal number and MV411 ordinal number). As determined by the analysis of 70 primary AML cell samples from human individuals, 36% of their samples had the strongest 150th among them.RARA Super enhancer (Figure 3). Therefore, the prevailing cut-off is set at 36%. When identifying potential AML responders to Tamibarotene based on RARA or IRF8 mRNA measurements, the same prevalence cutoff is also used as the RARA mRNA prevalence cutoff. I also useRARA The ratio of enhancer to MALAT1 enhancer ("normalized enhancer intensity") is used to quantitatively expand the enhancer of the AML cell group. Plotting the sensitivity of this normalized enhancer intensity value to Tamibarotene, I confirmRARA Five of the 6 cell lines with an enhancer intensity ratio greater than 1 were reactive, while only 4 of the 7 cell lines with an enhancer less than this level were reactive (Figure 5). When the cutoff moves to a normalized enhancer intensity of 1.4 or higher, all cell lines (4 out of 4) are reactive.Examples 3 . AML Nakano RARA Super-enhancer intensity ordinal grade cutoff and RARA mRNA Level related Will be used to determine 36%RARA Samples of AML patients with cut-off pre-emphasis of super-enhanced intensity are divided into two groups-their prevalence is 36% or higher (i.e., lower% value) and their prevalence is less than 36% (i.e., Higher% values) of their samples-and use RNA-seq as described in Example 1 to analyze the RARA mRNA levels of these samples. The results are shown in Figure 5A. inRARA The 36% of the super-enhancer intensity sequence with a prevalence rating or higher has a statistically significantly higher level of RARA mRNA than the group with a lower prevalence rating (p <0.001). This again confirms that the prevalence cutoff measured in terms of super-enhanced sublevels can also be used as the prevalence cutoff for mRNA levels. We also used RNA-seq to determine the RARA mRNA level in 11 different AML cell lines and compared the mRNA level with the sensitivity to Tamibarotene. The AML cell lines tested were divided into two different groups based on their sensitivity or insensitivity to Tamibarotene. All cell lines sensitive to Tamibarotene had RARA mRNA> 10 TPM measured by RNAseq, while three insensitive cell lines had levels below this cut-off level (Figure 5B).Examples 4 : non- APL AML Of the cell lines IRF8 mRNA Level and Right RARA Agonist response I use Affymetrix GeneChip® PrimeView Human Gene Expression Array to examine seven of these AML cell lines (four are sensitive to Tamibarotene-NOMO-1, AML3, MV-4-11 and Sig-M5; and three are Tamiba Rotin is insensitive-other mRNAs of KG1a, OCI-M1 and Kasumi-1), which may be particularly high in Tamibarotene sensitive cell lines, and IRF8 mRNA is identified as a potential candidate. We then quantified the IRF8 mRNA levels of each of these seven AML cell lines and each of several other AML cell lines that were tested for sensitivity to Tamibarotene by performing RNA-seq analysis as described below . The results of the first seven cell lines are shown in Figure 6. Interestingly, NOMO-1 does not have a high RARA mRNA level, but responds to Tamibarotene. The fact that NOMO-1 has a higher IRF8 mRNA level helps to explain this apparent inconsistency and further validates the use of IRF8 mRNA level to predict the response to Tamibarotene. RNA isolation, preparation, and RNA-seq data processing were performed in a manner similar to that described in Example 1. We will then compare the sensitivity of Tamibarotene to the IRF8 mRNA level, as shown in Figure 6 and Table 2. Table 2: IRF8 mRNA level of AML cell line and anti-proliferative efficacy of Tamibarotene * HL60 is an APL cell line. As can be seen from the table above, all Tamibarotene-reactive cell lines except HL60 have a higher than 190 TPM (log2 (7.57)) IRF8 mRNA level, and all non-reactive cell lines have less than 16.5 TPM (log2 (4.03)) IRF8 mRNA level. In the absence of the corresponding high level of IRF8 mRNA (6.73 TPM), the response of HL60 to Tamibarotene indicates that the correlation between IRF8 mRNA level and Tamibarotene sensitivity may not be effective for APL, and Therefore, it can be better suited for stratifying individuals with non-APL AML. Figure 7 removes the data points of HL60. Table 3 shows the division log2 The values are similar to Table 2 except for the IRF8 mRNA values described in the values, and show additional data for Sig-M5 and THP-1 cell lines. Table 3. Expressed as log2 Value of AML cell line mRNA level and anti-proliferative efficacy of Tamibarotene * HL60 is an APL cell line.Examples 5 : RARA Agonist treatment IRF8 mRNA Determination of threshold AML cell line results indicate that the cut-off value in RNA-Seq analysis is between 15.5 and 190 TPM (ie, in the log2 (4.03) and log2 (Between 7.57)). We selected a population of AML patient samples (gift from Stanford University) to check the distribution of IRF mRNA levels and determine the prevailing cut-off value based on this cut-off value. We added this population of AML cell lines and then generated a sorted map. Figure 8 shows the sequence distribution of IRF8 mRNA levels in the combined patient sample / AML cell line population. We have determined that the prevailing cut-off of 25% corresponds to approximate log2 (7) IRF8 mRNA value.Examples 6 : IRF8 mRNA versus RARA mRNA Level correlation We then compared IRF8 and RARA mRNA levels in AML cell lines and patient populations to determine the correlation. Figure 9 shows that some cell lines that respond to Tamibarotene have relatively low RARA mRNA, but have high level IRF8 mRNA. Figure 10 shows that the patient subgroup also exhibits high IRF8 mRNA levels, but exhibits relatively low RARA mRNA levels, and vice versa. If any mRNA level is greater than the threshold, measuring both IRF8 and RARA mRNA in the patient and selecting the patient for treatment with RARA agonists (such as Tamibarotene) may be optimal The idea of treating a patient group.Examples 7 : By him Mibarotene AML Cell line RARA mRNA Dependence CD38 Induce We monitored the induction of CD38 expression on the cell surface after treatment with Tamibarotene by measuring the mean fluorescence intensity (MFI) of the cell surface stained with the CD38-FITC antibody. FIG. 11A shows that treatment with Tamibarotene at a concentration of 50 nmol / L for 72 h does not induce CD38 expression in the RARA mRNA low cell line Kasumi (CD38- ). Figure 11B shows that after Tamibarotene treatment of the RARA mRNA high cell line MV411 for 72 h, the entire cell population showed a high level of CD38 (CD38HI ). In addition, in Figure 11C, OCI-AML3 (RARA mRNA high) has a low CD38 MFI (CD38DIM ). After Tamibarotene treatment, CD38 performance was further induced and the cell population became CD38HI . Figure 11D shows that 72h Tamibarotene treatment did not induce CD38 expression in another RARA mRNA low cell line OCI-M1 (CD38- ). This information indicates CD38 performed by TamibaroteneHI Induction can be predicted by RARA mRNA level. These results are depicted in FIG. 11E as a bar graph showing the level of CD38 mRNA expression detected in each cell line before and after treatment. Figure 11F shows the FACS-based CD38 before and after treatment with TamibaroteneHI The percentage of cells. In FIGS. 11E and 11F, the APL cell line NB4 is also shown.Examples 8 : RARA mRNA Predicted after the combination therapy of Tamibarotene and Dacirumab AML Cell line MV411 Of NK cell Cytotoxicity In order to functionally evaluate the efficacy of the combination of Tamibarotene and dalmatumab, AML cell lines with different RARA mRNA levels were treated with Tamibarotene for 72 hours, followed by human NK cells and dalmatumab or Control antibody co-culture. Then during the 38-hour co-culture time course, NK cell proliferation and tumor cell death were imaged with a phase difference. Figures 12A-12D are representative images of phase-contrast images of RARA mRNA high MV411 cell lines in the co-culture analysis under the following treatment conditions: Figure 12A) 72 h DMSO MV411 cell line before treatment and 38 h co-culture control antibody treatment. Figure 12B) 72 h SY1425 (50nM) MV411 cell line before treatment and 38 h co-culture control antibody treatment. Figure 12C) 72 hours of DMSO MV411 cell line before treatment and 38 hours of co-cultivation of damuci monoclonal antibody treatment. Figure 12D) 72 hours of SY1425 (50nM) MV411 cell line before treatment and 38 hours of co-cultivation of damuci monoclonal antibody treatment.Examples 9 : RARA mRNA The level is predicted to be after the combination therapy of tamibarrotin and daclitumab AML Cell line NK cell Cytotoxicity The NK cell-mediated kinetic measurement of tumor cell death by phospholipid binding protein V staining of tumor cells supports the following findings: the combination of Tamibarotene and dalmatumab treatment compared to single agent treatment Tumor cell death increased in some AML cell lines (Figures 13A to 13C). Combination with increased induction of CD38 expression in RARA mRNA high AML cell lines (ie, OCI-AML3 and MV411) but not RARA mRNA low AML cell lines (ie, OCI-M1) (Figures 11B to 11D) These data support the conclusion that Tamibarotene induces CD38 in a RARA mRNA-dependent mannerHI Phenotype, which is required for effective NK cell-mediated tumor cell death after dalmatumab treatment.Examples 10 : Co-cultivation analysis NK cell Activation only occurs after combination therapy with Tamibarotene and dalmatumab , By NK cell IFN γ Determined by secretion. Interferon gamma (IFNγ) secretion is an indicator of NK cell activation. After co-culture of AML cell lines and NK cells for 38 h, IFNγ secretion was quantified following the indicated treatment conditions (Figure 14). Compared to single agent treatment conditions, IFNγ levels were only observed in RARA mRNA high AML cell lines (MV411 and OCI-AML3) and NK cell co-culture analysis after combination treatment with Tamibarotene and dalmatumab Significant increase. In addition, this is shown as CD38 after Tamibarotene treatmentHI Phenotype is the only cell line. These data further support the indication that effective NK cell activation of phospholipid binding protein V quantification (Figures 12A-12D and 13) requires combination therapy.Examples 11 : Tamibarotene increases the number of multiple myeloma cells CD38 The intensity of performance. Datulimumab is clinically approved for the treatment of multiple myeloma (MM). However, due to the low level of CD38 performance, only some of these patients responded. I hereby prove that his Mibarotene increase has been CD38HI CD38 in multiple myeloma cell line (MM1S)HI Phenotype (Figure 15A) and can increase CD38- Multiple myeloma cell line to medium state (HUNS1) (Figure 15B). Based on previous AML experiments, we predict that this may increase CD38HI High individual response to CD38 therapeutic antibody against immune-mediated death.Examples 12 : CD38 HI The increase in the phenotype of Tamibarotene further makes MM Darmuzumab-dependent NK cell Mediated cytotoxicity is sensitive. To investigate whether treatment of MM cell lines with Tamibarotene would increase the sensitivity to CD38 antibody treatment, we repeated the previously described NK cell co-culture analysis with HUNS1 and MM1S MM cell lines. Figure 16 shows that after combination therapy with Tamibarotene and daltelimumab, MM1S tumor cell death was increased compared to single agent anti-CD38 antibody treatment, as quantified by phospholipid binding protein V staining. We further confirmed this by directly monitoring the activation of NK cells secreted by IFNγ after 38 h of treatment in this analysis (Figure 17). Multiple myeloma cell lines MM1S that achieved high CD38 intensity exhibited potent cell killing in response to combination therapy.Examples 13 : Tamibarotene induces primary non- APL AML and MDS Among the patient samples CD38 HI Phenotype. To confirm CD38 in AML cell linesHI Levels of Tamibarotene induction can approach AML and MDS patient samples. We will obtain PBMC from 15 different AML or MDS patients with 50 nM Tamibarotene or with DMSO as a negative control to cultivate. We tested the viability and CD38 induction after 24 and 48 hours of treatment, as measured by flow cytometry. Analyze the CD38 level of samples with at least 10% viability. The results are shown in Figure 18A. Most patient samples (11/15) showed CD38 after 24 hours of treatmentHI The increase in cells. One patient sample showed no response (patient 4) and three patient samples did not meet the vitality criteria (patients 2, 6, and 10) after 24 h. After 48 h, four additional patient samples that had been shown to induce CD38 after 48 h no longer met the viability criteria and were excluded (patients 3, 7, 9 and 14). All remaining patient samples except non-responder patient 4 showed further induction of CD38 level after 48 h. We then measured both the RARA and IRF8 mRNA levels in these subgroups of patients (Patients 1, 5, 11, 12, and 13). The subgroup of patients indicated that patients who showed some responses at 48 h After that, he is still alive and indicates both AML and MDS. As shown in FIG. 18B, the patient samples showing higher levels of RARA mRNA and / or IRF8 mRNA as measured by RNA qPCR (the lower the dCq value, the higher the mRNA level) were compared to those with Samples with lower RARA mRNA and / or IRF8 mRNA showed a greater percentage of CD38 after Tamibarotene treatmentHI . The observed CD38 induction level for AML patient samples correlates well with the results obtained from high RARA RNA AML cell lines (eg, comparing the patient samples AML_1 in Figures 19C to 19D with the Tamiba in AML cell line MV411 Rotin-induced CD38 MFI). For both, Tamibarotene caused a similarity to that observed in multiple myeloma cell lines and multiple myeloma patients with daclizumab sensitivity, phenotype CD38HI Increased phenotype (for example, comparing the patient sample MM_1 in FIG. 19A to FIG. 19B with the multiple myeloma cell line MM1S). Based on this correlation, the present inventors believe that patients with high RARA mRNA AML will benefit from the combined treatment of RARA-specific agonists (such as Tamibarotene) and anti-CD38 antibodies (such as daratumumab).Examples 15 : Tamibarotene caused higher than RARA or IRF8 Of the level AML Cell line xenotransplantation ATRA bigger CD38 Induced increase. ATRA has been reported to cause greater induction of the CD38 level than tamibarotene in HL-60 cells (usually characterized as AML cell lines of APL) (A Uruno et al., 2011, J Leuk Biol, 90, 235 To page 247). We intend to compare the CD38-inducing effects of ATRA and Tamibarotene in non-APL AML cell lines with different levels of RARA or IRF8 mRNA. In this experiment, we used MV411, THP-1 or Kasumi-1 mouse xenotransplantation. MV411 has high-level RARA mRNA and IRF8 mRNA (ie, greater than the threshold), while THP-1 has high-level IRF-8 mRNA. It is believed that Kasumi-1 cells have IRF8 and RARA levels below the threshold. Each of the cell lines is 5% CO at 37 ° C in vitro2 Maintain the appropriate medium in the air atmosphere (THP-1 cells: supplemented with 10% heating-inactivated fetal bovine serum and 0.05 mM β-mercaptoethanol in RPMI1640 medium; MV4-11 cells: supplemented with 10% heating inactivated fetal bovine serum IMDM medium; Kasumi-1 cells: suspension culture in RPMI1640 medium supplemented with 20% heat-inactivated fetal bovine serum). Cells grown in the exponential growth phase were collected for tumor inoculation and counted. All mice were irradiated with gamma (200 rad) for 24 h before tumor cell inoculation. Use appropriate tumor cell lines (THP-1 cells: containing 1 × 10 in the right ventral area of each mouse)7 0.1 ml PBS (1: 1 Matrigel) per cell; MV4-11 cells: containing 5 × 106 0.1 ml PBS (1: 1 Matrigel) per cell; Kasumi-1 cells: containing 1 × 107 Each cell of 0.1 ml PBS (1: 1 Matrigel) was inoculated subcutaneously for tumor development. When the average tumor size reaches approximately 100 to 200 mm3 At the time, began treatment with Tamibarotene, ATRA or vehicle. The mice were divided into 3 groups, nine in each group, and each group was orally administered a drug (ATRA 4 mg / kg; Tamibarotene 3 mg / kg) or vehicle BID lasted up to 28 day. The date of tumor cell inoculation is indicated as day 0. Use a caliper gauge to measure the tumor volume in two dimensions twice a week, and use the following formula in mm3 Express volume as a unit: V = 0.5a ×b 2 ,among thema andb Respectively, the length and width of the tumor. After grouping, three mice were killed per group on days 7, 14, and 21 (a total of 4 weeks of study). One half of the tumor was collected for embedding into the FFPE block for immunohistochemistry (IHC) staining, and the other half of the tumor was collected for CD38 FACS analysis of tumor cells. IHC staining was performed using a BOND RX automatic staining instrument using anti-CD38 antibodies against the C-terminus of CD38 (clone SP149; Abcam; Cat. No. ab183326). The antibody was diluted 1: 100 in EDTA buffer at pH 9.0 and then incubated with tumor sections for 20 minutes. Use DAB and manufacturer's combined polymer fine detection kit to generate signals. As seen in Figure 20A, after 7 days, MV4-11 xenograft mice treated with Tamibarotene exhibited an average fluorescence intensity (MFI) greater than 200, where more than 80% of tumor cells were CD38 by FACS analysisHI . ATRA-treated MV4-11 xenograft mice display a MFI slightly greater than 100, of which approximately 70% of tumor cells are CD38HI . Immunohistochemical staining of tumor sections with anti-CD38 antibody confirmed the unexpected and unexpected superiority of Tamibarotene over ATRA in this xenograft model (Figure 20B). This trend became even more pronounced at 3 weeks. As shown in Figure 20C, at three weeks, more than 60% of tumor cells maintained CD38 in MV4-11 xenografts treated with TamibaroteneHI , Of which MFI is about 110, and less than 20% of tumor cells maintain CD38 in ATRA-treated xenograftsHI , Where MFI is about 50. Figure 20D shows immunohistochemical staining of tumor cells after three weeks, which confirms the superiority of Tamibarotene over ATRA. As shown in Figure 21, the superiority of Tamibarotene in inducing CD38 was also observed in THP-1 cells after 7 days. Although xenotransplants treated with ATRA showed similar CD38 compared to TamibaroteneHI Cell percentage, but Tamibarotene treatment produced a higher MFI (greater than 200) compared to ATRA (~ 175). In contrast, Kasumi-1 cells demonstrated minimal CD38 induction with either treatment (Figure 22). One week later, the immunohistochemical comparison of various types of xenotransplantation and various types of treatment is shown in FIG. 23.Equivalents and categories In embodiments, articles such as "a / an" and "the" can mean one or greater than one unless indicated to the contrary or otherwise obvious from the context. Unless indicated to the contrary or otherwise obvious from the context, an embodiment or description including "or" between one or more members of a group is deemed to satisfy one, more than one, or all group members present in, for a given A condition in a product or method or otherwise related to a given product or method. The invention includes embodiments where exactly one member of the group is present in, used in, or otherwise related to a given product or method. The invention includes embodiments where more than one or all group members are present in, used in, or otherwise related to a given product or method. Furthermore, the present invention covers all variations, combinations, and arrangements in which one or more limitations, elements, clauses, and descriptive terms from one or more of the listed embodiments are introduced into another embodiment. For example, any embodiment depending on another embodiment may be modified to include one or more limitations found in any other embodiment depending on the same base embodiment. In the case where the elements are listed, for example, in the Markush group format, each sub-group of the elements is also revealed, and any elements can be removed from the group. It should be understood that, generally speaking, when the invention or aspects of the invention are referred to as including specific elements and / or features, certain embodiments of the invention or aspects of the invention are composed of such elements and / or features or mainly Consist of such elements and / or features. For simplicity, their embodiments have not been specifically described in words herein. It should also be noted that the terms "comprising" and "containing" are intended to be open and allow additional elements or steps to be included. When a range is given, the endpoint is included. In addition, unless otherwise indicated or otherwise obvious from the context and understanding of one of ordinary skill, values expressed as ranges may employ any particular value or subrange within the stated range in different embodiments of the invention, unless the context clearly dictates otherwise , Otherwise it reaches + 1 / unit of the lower limit of the range. This application is about various granted patents, published patent applications, journal articles and other publications, and the above owners are incorporated by reference. If there is a conflict between any incorporated reference and this specification, this specification shall prevail. In addition, any specific embodiment of the present invention belonging to the prior art may be explicitly excluded from any one or more of the embodiments. Because such embodiments are considered to be known to those of ordinary skill, they can be excluded even if the exclusion is not explicitly set forth herein. Any particular embodiment of the present invention may be excluded from any embodiment for any reason, whether related to the existence of the prior art or not. Those skilled in the art using only routine experimentation will recognize or be able to determine many equivalents of the specific embodiments described herein. The scope of the embodiments of the present invention described herein is not intended to be limited to the above description, but is actually as set forth in the accompanying embodiments. A person of ordinary skill will understand that various changes and modifications can be made to this specification without departing from the spirit or scope of the invention as defined in the following embodiments.

圖1展示使用RNA-Seq針對48個不同AML病患樣本的RARA mRNA表現(log2 (1+TPM))與RARA SE強度(RARA /MALAT1倍富集)之間的相關性。 圖2描繪94 AML樣本中之RARA 超強化子強度序數之log10 排序圖,該等樣本包括四個AML細胞株--Sig-M5、MV411、HEL及Kasumi。 圖3描繪70個AML病患樣本中之RARA 超強化子強度序數之log10 排序圖。較淺色條柱表示其RARA 超強化子強度序數等於或大於盛行截止之樣本。較深色條柱表示其RARA 超強化子強度序數小於盛行截止之樣本。 圖4描繪11個不同AML細胞株中RARA 強化子強度與他米巴羅汀敏感性之間的相關性。 圖5A描繪來自70個AML病患樣本及如實例2中所描述之根據其RARA 超強化子強度序數是否大於(或等於)盛行截止(「高RARA」)或小於盛行截止(「低RARA」)而分組之RARA mRNA位準。圖5B描繪11個不同AML細胞株中RARA mRNA位準與他米巴羅汀敏感性之間的相關性。 圖6描繪七個不同AML細胞株中之IRF8 mRNA位準。圖表左側上所展示之四個細胞株表明對他米巴羅汀治療之實質性反應。圖表右側上所展示之三個細胞株表明對他米巴羅汀治療之極小反應或無反應。 圖7展示他米巴羅汀抗增殖效力(EC50 值,nM)與IRF8 mRNA位準之相關性,如藉由RNA-Seq所量測。應注意,IRF8 mRNA位準 = 1 (log10)及他米巴羅汀EC50 值估算為50 μM (非反應性)之左上角點表示來自具有低IRF8 mRNA位準且對他米巴羅汀無抗增殖反應的2個AML細胞株之資料。他米巴羅汀敏感性與IRF8 mRNA位準之相關性高度顯著(p = 0.0001,斯皮爾曼相關性(Spearman's correlation),雙尾)。 圖8描繪個別病患AML樣本及AML細胞株中之IRF8 mRNA位準的排序圖,如藉由RNA-Seq所量測。指示以下AML細胞株:PL21,其為具有最低IRF8 mRNA位準之對他米巴羅汀起反應的細胞株;及Kasumi,其為具有最高IRF8 mRNA位準之對他米巴羅汀無反應的細胞株。在此群體中,25%盛行截止等於近似log2 (7)之RNA-Seq TPM值。 圖9描繪經測試對他米巴羅汀起反應之非APL AML細胞株中之IRF8 mRNA位準與RARA mRNA位準之間的相關性。 圖10描繪AML病患樣本之群體中之IRF8 mRNA位準與RARA mRNA位準之間的相關性。虛線表示各mRNA之25%盛行截止。 圖11A至圖11F描繪他米巴羅汀(「SY1425」)對不同AML細胞株中CD38位準之作用。圖11A至圖11D表明他米巴羅汀對不同細胞株的作用,如藉由FITC細胞分類所量測。圖11E為在用他米巴羅汀治療之前及之後不同細胞株中之CD38 mRNA位準之圖形表示。圖11F為如藉由FITC細胞分類所測定之CD38陽性的細胞%之圖形表示。 圖12A至圖12D描繪單獨或組合形式之他米巴羅汀及達土木單抗(「Dara」)對不同NK細胞/AML細胞株共培養基中之NK細胞增殖及腫瘤細胞死亡的作用,如藉由位相差顯微鏡所觀測到。 圖13A至圖13C描繪對照抗體或Dara單獨或與他米巴羅汀組合對不同AML細胞株之NK細胞/AML共培養物中之凋亡細胞之數目的作用。 圖14描繪對照抗體或Dara單獨或與他米巴羅汀組合對圖13A至圖13C中所描繪之不同AML細胞株的NK細胞/AML共培養物中之干擾素-γ分泌之作用。 圖15A至圖15B描繪對照抗體或Dara單獨或與他米巴羅汀組合對兩個不同多發性骨髓瘤細胞株(MM1S;圖15A) (HUNS1;圖15B)中之CD38位準的作用,如藉由FITC細胞分類所量測。 圖16描繪對照抗體或Dara單獨或與他米巴羅汀組合對與NK細胞共培養之兩個不同多發性骨髓瘤細胞株中之凋亡細胞之數目的作用。 圖17描繪對照抗體或Dara單獨或與他米巴羅汀組合對圖15A至圖15B及圖16中所描繪之多個不同細胞株的NK細胞/多發性骨髓瘤共培養物中之干擾素-γ分泌之作用。 圖18A描繪24或48小時之後他米巴羅汀治療對不同AML及MDS病患樣本中之CD38表現之作用。圖18B描繪24或48小時之後他米巴羅汀治療對不同AML及MDS病患樣本中之如藉由FACS所量測之CD38表現以及如藉由qPCR所量測之RARA及IRF8 mRNA表現的作用。 圖19A至圖19D比較他米巴羅汀治療對AML細胞株、AML病患樣本及多發性骨髓瘤細胞株中之CD38位準的作用以及對CD38HI 多發性骨髓瘤病患樣本中之基線CD38位準的作用。 圖20A至圖20D比較一週(圖20A及圖20B)及三週(圖20C及圖20D)之後他米巴羅汀及全反視黃酸(ATRA)對小鼠MV4-11異種移植模型中之CD38位準的作用。藉由FACS (圖20A及圖20C)及免疫組織化學(IHC)染色(圖20B及圖20D)兩者測試腫瘤細胞中CD38之存在。 圖21比較一週之後他米巴羅汀及全反視黃酸(ATRA)對小鼠THP-1異種移植模型中之CD38位準的作用,如藉由FACS所量測。 圖22比較一週之後他米巴羅汀及全反視黃酸(ATRA)對小鼠Kasumi-1異種移植模型中之CD38位準的作用,如藉由FACS所量測。 圖23比較一週之後他米巴羅汀及全反視黃酸(ATRA)對小鼠THP-1、Kasumi-1及MV4-11異種移植模型中之CD38位準的作用,如藉由IHC所量測。Figure 1 shows the correlation between RARA mRNA expression (log 2 (1 + TPM)) and RARA SE intensity ( RARA / MALAT 1-fold enrichment) for 48 different AML patient samples using RNA-Seq. Figure 2 depicts a log 10 ranking chart of the RARA superenhancer intensity numbers in 94 AML samples, which include four AML cell lines-Sig-M5, MV411, HEL, and Kasumi. Figure 3 depicts a log 10 ranking chart of RARA superenhancer intensity numbers in 70 AML patient samples. Lighter bars indicate samples whose RARA superenhancer intensity number is equal to or greater than the prevalent cutoff. The darker bar indicates that the RARA super-enhancer intensity number is less than the prevalent cut-off sample. Figure 4 depicts the correlation between RARA enhancer intensity and tamibarotene sensitivity in 11 different AML cell lines. Figure 5A depicts samples from 70 AML patients and according to whether their RARA superenhancer intensity number is greater than (or equal to) the prevalence cutoff ("High RARA") or less than the prevalence cutoff ("Low RARA") as described in Example The RARA mRNA level of the group. Figure 5B depicts the correlation between RARA mRNA levels and the sensitivity of Tamibarotene in 11 different AML cell lines. Figure 6 depicts IRF8 mRNA levels in seven different AML cell lines. The four cell lines shown on the left side of the graph indicate a substantial response to Tamibarotene treatment. The three cell lines shown on the right side of the graph indicate minimal or no response to Tamibarotene treatment. Figure 7 shows the correlation between the antiproliferative efficacy (EC 50 value, nM) of Tamibarotene and the IRF8 mRNA level, as measured by RNA-Seq. It should be noted that the IRF8 mRNA level = 1 (log10) and the Tamibarotene EC 50 value is estimated to be 50 μM (non-reactive). The point in the upper left corner is from a low IRF8 mRNA level and has no effect on Tamibarotene Data of 2 AML cell lines with anti-proliferative response. The correlation between Tamibarotene sensitivity and IRF8 mRNA level is highly significant (p = 0.0001, Spearman's correlation, two-tailed). FIG. 8 depicts a ranking chart of IRF8 mRNA levels in AML samples and AML cell lines of individual patients, as measured by RNA-Seq. The following AML cell lines are indicated: PL21, which is the cell line with the lowest IRF8 mRNA level that responds to Tamibarotene; and Kasumi, which is the one with the highest IRF8 mRNA level that does not respond to Tamibarotene Cell line. In this group, the 25% prevalence cutoff is equal to the RNA-Seq TPM value of approximately log 2 (7). Figure 9 depicts the correlation between IRF8 mRNA level and RARA mRNA level in non-APL AML cell lines tested to respond to Tamibarotene. Figure 10 depicts the correlation between IRF8 mRNA level and RARA mRNA level in a population of AML patient samples. The dotted line indicates the prevalence of 25% of each mRNA. Figures 11A-11F depict the effect of Tamibarotene ("SY1425") on the CD38 level in different AML cell lines. 11A to 11D show the effect of Tamibarotene on different cell lines, as measured by FITC cell classification. Figure 11E is a graphical representation of CD38 mRNA levels in different cell lines before and after treatment with Tamibarotene. Figure 11F is a graphical representation of% CD38 positive cells as determined by FITC cell classification. Figures 12A to 12D depict the effects of tamibarotene and darumumab ("Dara") alone or in combination on NK cell proliferation and tumor cell death in different NK cell / AML cell line co-culture media, such as Observed by phase contrast microscope. 13A to 13C depict the effect of control antibody or Dara alone or in combination with Tamibarotene on the number of apoptotic cells in NK cell / AML co-cultures of different AML cell lines. Figure 14 depicts the effect of control antibody or Dara alone or in combination with Tamibarotene on IFN-γ secretion in NK cell / AML co-cultures of different AML cell lines depicted in Figures 13A to 13C. 15A to 15B depict the effect of control antibody or Dara alone or in combination with Tamibarotene on the CD38 level in two different multiple myeloma cell lines (MM1S; FIG. 15A) (HUNS1; FIG. 15B), such as Measured by FITC cell classification. Figure 16 depicts the effect of control antibody or Dara alone or in combination with Tamibarotene on the number of apoptotic cells in two different multiple myeloma cell lines co-cultured with NK cells. Figure 17 depicts interferon in a NK cell / multiple myeloma co-culture of control antibody or Dara alone or in combination with Tamibarotene on multiple different cell lines depicted in Figures 15A to 15B and 16 The role of γ secretion. Figure 18A depicts the effect of Tamibarotene treatment on CD38 performance in different AML and MDS patient samples after 24 or 48 hours. Figure 18B depicts the effect of Tamibarotene treatment on CD38 performance as measured by FACS and RARA and IRF8 mRNA performance as measured by qPCR in different AML and MDS patient samples after 24 or 48 hours . Figures 19A to 19D compare the effect of Tamibarotene treatment on the CD38 level in AML cell lines, AML patient samples and multiple myeloma cell lines, and the baseline CD38 in CD38 HI multiple myeloma patient samples The role of level. Figures 20A to 20D compare one of the mice MV4-11 xenograft model with Tamibarotene and all-trans retinoic acid (ATRA) after one week (Figures 20A and 20B) and three weeks (Figures 20C and 20D). The role of CD38 level. The presence of CD38 in tumor cells was tested by both FACS (Figures 20A and 20C) and immunohistochemistry (IHC) staining (Figures 20B and 20D). FIG. 21 compares the effects of Tamibarotene and ATRA on CD38 level in a mouse THP-1 xenograft model one week later, as measured by FACS. Figure 22 compares the effects of tamibarrotin and all-trans retinoic acid (ATRA) on the CD38 level in the mouse Kasumi-1 xenograft model one week later, as measured by FACS. Figure 23 compares the effect of tamibarrotin and all-trans retinoic acid (ATRA) on the CD38 level in mouse THP-1, Kasumi-1 and MV4-11 xenograft models after one week, as measured by IHC Measurement.

Claims (16)

一種診斷人類個體是否患有選自非APL AML、多發性骨髓瘤及MDS之他米巴羅汀(tamibarotene)敏感型疾病之方法,其包含診斷該個體是否具有: i.預先測定為等於或大於預定臨限值之來自該個體的患病細胞之樣本中之視黃酸受體α mRNA位準;及/或 ii.預先測定為等於或大於預定臨限值之來自該個體的患病細胞之樣本中之IRF8 mRNA位準。A method for diagnosing whether a human individual has a tamibarotene sensitive disease selected from non-APL AML, multiple myeloma, and MDS, which includes diagnosing whether the individual has: i. Pre-determined as equal to or greater than The retinoic acid receptor α mRNA level in a sample of diseased cells from the individual with a predetermined threshold; and / or ii. Of the diseased cells from the individual previously determined to be equal to or greater than the predetermined threshold IRF8 mRNA level in the sample. 一種他米巴羅汀在製造用於治療患有選自非APL AML、多發性骨髓瘤或MDS之疾病的人類個體之藥劑中之用途,其中患病細胞中之視黃酸受體α mRNA位準及/或IRF8 mRNA位準已經測定具有等於或大於預定臨限值之視黃酸受體α mRNA位準,或等於或大於預定臨限值之IRF8 mRNA位準,其中該藥劑進一步包含CD38特異性抗體或與CD38特異性抗體組合使用。A use of Tamibarotene in the manufacture of a medicament for treating a human subject suffering from a disease selected from non-APL AML, multiple myeloma or MDS, wherein the retinoic acid receptor alpha mRNA position in the diseased cells And / or IRF8 mRNA level has been determined to have a retinoic acid receptor alpha mRNA level equal to or greater than a predetermined threshold, or an IRF8 mRNA level equal to or greater than a predetermined threshold, wherein the agent further contains CD38 specificity Sex antibodies may be used in combination with CD38 specific antibodies. 如請求項2之用途,其中該CD38特異性抗體為達土木單抗(daratumumab)。The use according to claim 2, wherein the CD38 specific antibody is daratumumab. 如請求項3之用途,其中該藥劑用於在投與該CD38特異性抗體之前投與他米巴羅汀一段時間;且在開始投與他米巴羅汀之後,僅在該個體中之CD38位準經測定為CD38hi 時共投與該CD38特異性抗體。The use according to claim 3, wherein the agent is used to administer Tamibarotene for a period of time before administering the CD38-specific antibody; and after starting Tamibarotene, only CD38 in the individual The CD38 specific antibody was co-administered when the level was determined to be CD38 hi . 如請求項4之用途,其中在開始投與他米巴羅汀之後的6至72小時之間測定該個體中之該CD38位準。The use as in claim 4, wherein the CD38 level in the individual is determined between 6 and 72 hours after the start of administration of Tamibarotene. 如請求項2至5中任一項之用途,其中他米巴羅汀用於經口投與。The use as claimed in any one of claims 2 to 5, wherein Tamibarotene is used for oral administration. 如請求項2至5中任一項之用途,其中該藥劑用於以6 mg/m2 /天與12 mg/m2 /天之間的劑量投與他米巴羅汀,其中該劑量分為兩個劑量。The use according to any one of claims 2 to 5, wherein the medicament is for administration of Tamibarotene at a dose between 6 mg / m 2 / day and 12 mg / m 2 / day, wherein the dose is divided For two doses. 如請求項2至5中任一項之用途,其中該抗CD38抗體用於一週不多於一次以10至20 mg/kg個體體重之間的劑量投與。The use according to any one of claims 2 to 5, wherein the anti-CD38 antibody is used for administration not more than once a week at a dose between 10 and 20 mg / kg of the body weight of the individual. 一種測定用於患有非APL AML、多發性骨髓瘤或MDS之人類個體之合適的療法之方法,其包含基於視黃酸受體α mRNA位準及/或IRF8 mRNA位準而診斷該個體是否患有他米巴羅汀敏感型疾病,該等位準中之任一者或兩者預先測定為存在於來自該個體之患病細胞樣本中;且其中: i.該個體適合於包含以下之療法:若該位準指示該疾病對他米巴羅汀敏感,則投與有效治療該疾病之一定量之他米巴羅汀及共投與CD38特異性抗體;且 ii.該個體適合於包含以下之療法:若該位準指示該疾病對他米巴羅汀不敏感,則投與除他米巴羅汀以外的藥劑。A method of determining a suitable therapy for a human individual with non-APL AML, multiple myeloma, or MDS, which includes diagnosing whether the individual is based on the retinoic acid receptor alpha mRNA level and / or IRF8 mRNA level Suffering from Tamibarotene sensitive disease, either or both of these levels are pre-determined to be present in a sample of diseased cells from the individual; and wherein: i. The individual is suitable to include the following Therapy: If the level indicates that the disease is sensitive to Tamibarotene, then administer a quantitative amount of Tamibarotene effective to treat the disease and co-administer CD38-specific antibodies; The following therapy: If the level indicates that the disease is not sensitive to Tamibarotene, then administer drugs other than Tamibarotene. 如請求項9之方法,其中該視黃酸受體α mRNA及/或該IRF8 mRNA位準指示若該位準大於預定臨限值,則該疾病對他米巴羅汀敏感,及若該位準小於該預定臨限值,則該疾病對他米巴羅汀不敏感。The method of claim 9, wherein the retinoic acid receptor alpha mRNA and / or the IRF8 mRNA level indicates that if the level is greater than a predetermined threshold, the disease is sensitive to Tamibarotene, and if the bit If it is less than the predetermined threshold, the disease is not sensitive to Tamibarotene. 如請求項9之方法,其中該CD38特異性抗體為達土木單抗。The method of claim 9, wherein the CD38-specific antibody is daclizumab. 如請求項9之方法,其中若該視黃酸受體α mRNA或該IRF8 mRNA位準指示該疾病對他米巴羅汀敏感,則該療法包含在投與該CD38特異性抗體之前向該個體投與他米巴羅汀一段時間;以及在開始投與他米巴羅汀之後,僅在該個體中之該CD38位準經測定為CD38hi 時向該個體共投與該CD38特異性抗體。The method of claim 9, wherein if the retinoic acid receptor alpha mRNA or the IRF8 mRNA level indicates that the disease is sensitive to Tamibarotene, then the therapy comprises administering to the individual before administering the CD38-specific antibody Tamibarotene was administered for a period of time; and after starting Tamibarotene, the CD38 specific antibody was co-administered to the individual only when the CD38 level in the individual was determined to be CD38 hi . 如請求項12之方法,其中在開始投與他米巴羅汀之後的6至72小時之間測定該個體中之該CD38位準。The method of claim 12, wherein the CD38 level in the individual is determined between 6 and 72 hours after the start of administration of Tamibarotene. 如請求項9至13中任一項之方法,其中他米巴羅汀經口投與。The method of any one of claims 9 to 13, wherein Tamibarotene is administered orally. 如請求項9至13中任一項之方法,其中以6 mg/m2 /天與12 mg/m2 /天之間的劑量向該個體投與他米巴羅汀,其中該劑量分為兩個劑量。The method according to any one of claims 9 to 13, wherein the subject is administered Tamibarotene at a dose between 6 mg / m 2 / day and 12 mg / m 2 / day, wherein the dose is Two doses. 如請求項9至13中任一項之方法,其中一週不多於一次以10至20 mg/kg個體體重之間的劑量投與該抗CD38抗體。The method of any one of claims 9 to 13, wherein the anti-CD38 antibody is administered at a dose between 10 and 20 mg / kg of the body weight of the individual no more than once a week.
TW106134613A 2016-10-06 2017-10-06 Methods of treating patients with a retinoic acid receptor-[alpha] agonist and an anti-CD38 antibody TW201823471A (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201662405164P 2016-10-06 2016-10-06
US62/405,164 2016-10-06
US201762449450P 2017-01-23 2017-01-23
US62/449,450 2017-01-23
US201762564690P 2017-09-28 2017-09-28
US62/564,690 2017-09-28

Publications (1)

Publication Number Publication Date
TW201823471A true TW201823471A (en) 2018-07-01

Family

ID=61831262

Family Applications (1)

Application Number Title Priority Date Filing Date
TW106134613A TW201823471A (en) 2016-10-06 2017-10-06 Methods of treating patients with a retinoic acid receptor-[alpha] agonist and an anti-CD38 antibody

Country Status (2)

Country Link
TW (1) TW201823471A (en)
WO (1) WO2018067946A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2016243702B2 (en) 2015-03-31 2021-10-07 Syros Pharmaceuticals, Inc. Methods of stratifying patients for treatment with retinoic acid receptor-alpha agonists
US9868994B2 (en) 2016-04-08 2018-01-16 Syros Pharmaceuticals, Inc. Methods of stratifying patients for treatment with retinoic acid receptor-α agonists
US20200046737A1 (en) * 2018-08-09 2020-02-13 Notable Labs, Inc. Methods for treating cancer, and compositions therefor

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2691493T3 (en) * 2012-06-07 2018-11-27 Childrens Hospital Los Angeles Methods to treat neutropenia using retinoid agonists
MY192918A (en) * 2014-09-09 2022-09-15 Janssen Biotech Inc Combination therapies with anti-cd38 antibodies
EP3267969A1 (en) * 2015-03-09 2018-01-17 King's College London Combination therapy with rar alpha agonists for enhancing th1 response
AU2016243702B2 (en) * 2015-03-31 2021-10-07 Syros Pharmaceuticals, Inc. Methods of stratifying patients for treatment with retinoic acid receptor-alpha agonists

Also Published As

Publication number Publication date
WO2018067946A1 (en) 2018-04-12
WO2018067946A8 (en) 2019-03-21

Similar Documents

Publication Publication Date Title
JP7401710B2 (en) System and method for identifying cancer treatment from normalized biomarker scores
US11447831B2 (en) Methods of stratifying patients for treatment with retinoic acid receptor-alpha agonists
WO2019094392A1 (en) Cancer biomarkers and methods of use thereof
US20210285059A1 (en) Methods of stratifying patients for treatment with retinoic acid receptor-alpha agonists
US20240102106A1 (en) Methods of stratifying patients for treatment with retinoic acid receptor-alpha agonists
Hussain et al. CXCL13 signaling in the tumor microenvironment
TW201823471A (en) Methods of treating patients with a retinoic acid receptor-[alpha] agonist and an anti-CD38 antibody
JP2022519649A (en) How to diagnose and treat cancer
Zhang et al. HHLA2 promotes tumor progression by long non‑coding RNA H19 in human gallbladder cancer
Bruno et al. β3-adrenergic receptor on tumor-infiltrating lymphocytes sustains IFN-γ-dependent PD-L1 expression and impairs anti-tumor immunity in neuroblastoma
CN105189786A (en) FALZ for use as a target for therapies to treat cancer
WO2018136961A1 (en) METHODS OF TREATING PATIENTS WITH A RETINOIC ACID RECEPTOR-α AGONIST AND AN ANTI-CD38 ANTIBODY
Taghi Khani et al. Isoform-specific knockdown of long and intermediate prolactin receptors interferes with evolution of B-cell neoplasms
RU2799789C2 (en) Rara agonists for the treatment of aml and mds
CN114908164A (en) Application of ILT5 in colorectal cancer diagnosis and treatment