TW202237858A - Precision drug screening for personalized cancer therapy - Google Patents

Precision drug screening for personalized cancer therapy Download PDF

Info

Publication number
TW202237858A
TW202237858A TW110144981A TW110144981A TW202237858A TW 202237858 A TW202237858 A TW 202237858A TW 110144981 A TW110144981 A TW 110144981A TW 110144981 A TW110144981 A TW 110144981A TW 202237858 A TW202237858 A TW 202237858A
Authority
TW
Taiwan
Prior art keywords
organoid
patient
spheroids
cells
derived
Prior art date
Application number
TW110144981A
Other languages
Chinese (zh)
Inventor
西凌 沈
大衛 徐
丹尼爾 德魯巴克
王朝暉
Original Assignee
美國公爵大學
美商西利斯股份有限公司
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 美國公爵大學, 美商西利斯股份有限公司 filed Critical 美國公爵大學
Publication of TW202237858A publication Critical patent/TW202237858A/en

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5076Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving cell organelles, e.g. Golgi complex, endoplasmic reticulum
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M23/00Constructional details, e.g. recesses, hinges
    • C12M23/02Form or structure of the vessel
    • C12M23/16Microfluidic devices; Capillary tubes
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/502Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures
    • B01L3/5027Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M41/00Means for regulation, monitoring, measurement or control, e.g. flow regulation
    • C12M41/12Means for regulation, monitoring, measurement or control, e.g. flow regulation of temperature
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M41/00Means for regulation, monitoring, measurement or control, e.g. flow regulation
    • C12M41/40Means for regulation, monitoring, measurement or control, e.g. flow regulation of pressure
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M41/00Means for regulation, monitoring, measurement or control, e.g. flow regulation
    • C12M41/44Means for regulation, monitoring, measurement or control, e.g. flow regulation of volume or liquid level
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B30/00Methods of screening libraries
    • C40B30/06Methods of screening libraries by measuring effects on living organisms, tissues or cells
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/02Libraries contained in or displayed by microorganisms, e.g. bacteria or animal cells; Libraries contained in or displayed by vectors, e.g. plasmids; Libraries containing only microorganisms or vectors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2200/00Solutions for specific problems relating to chemical or physical laboratory apparatus
    • B01L2200/10Integrating sample preparation and analysis in single entity, e.g. lab-on-a-chip concept
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/08Geometry, shape and general structure
    • B01L2300/0861Configuration of multiple channels and/or chambers in a single devices
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/502Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures
    • B01L3/5027Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip
    • B01L3/502769Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip characterised by multiphase flow arrangements
    • B01L3/502784Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip characterised by multiphase flow arrangements specially adapted for droplet or plug flow, e.g. digital microfluidics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Abstract

Precision drug screening methods and apparatuses for personalized cancer therapies include the formation of a library of mature Micro-Organospheres, including Patient-Derived Micro-Organospheres (PMOSs), from a single patient tissue sample, such as from a tumor sample, are described. Also described herein are methods and systems for screening a patient using these Patient-Derived Micro-Organospheres, including personalized therapies.

Description

用於個人化癌症療法之精準藥物篩選Precision Drug Screening for Personalized Cancer Therapy

本文中所描述之方法、設備及物質組合物係關於患者衍生之微型類器官球體(organosphere) (PMOS)、及用於形成PMOS之方法及設備、及使用PMOS之方法及設備。具體而言,本文中描述用於鑑別可有效治療特定患者之一或多種藥物調配物的方法及設備。The methods, devices and compositions of matter described herein relate to patient-derived micro organospheres (PMOSs), and methods and devices for forming PMOSs, and methods and devices for using PMOSs. In particular, described herein are methods and apparatus for identifying one or more pharmaceutical formulations that are effective in treating a particular patient.

模型細胞及組織系統適用於生物學及醫學研究。最常見實踐為自組織衍生永生化細胞株且將其等培養在二維(2D)條件中(例如,在皮氏培養皿(Petri dish)或孔盤中)。然而,儘管非常適用於基礎研究,但2D細胞株與個別患者對療法之反應並不充分相關。特定而言,三維細胞培養模型證實尤其有助於發生生物學、疾病病理學、再生醫學、藥物毒性及功效測試及個人化醫療。舉例而言,球體及類器官為已經研究的三維細胞聚集體。然而,類器官及球體均具有降低其功效之限制。Model cell and tissue systems are suitable for biological and medical research. The most common practice is to derive immortalized cell lines from tissues and culture them in two-dimensional (2D) conditions (eg, in Petri dishes or well plates). However, while well suited for basic research, 2D cell lines are not well correlated with individual patient responses to therapy. In particular, three-dimensional cell culture models prove particularly useful in developmental biology, disease pathology, regenerative medicine, drug toxicity and efficacy testing, and personalized medicine. For example, spheroids and organoids are three-dimensional cell aggregates that have been studied. However, both organoids and spheroids have limitations that reduce their efficacy.

多細胞腫瘤球體首次描述於70年代早期,且藉由在非黏著條件下培養癌細胞株而獲得。球體通常由作為超低附著盤中之自由浮動的細胞聚集體的癌細胞株形成。與2D細胞培養相比,球體已展示維持更多的幹細胞相關特性。Multicellular tumor spheroids were first described in the early 1970s and were obtained by culturing cancer cell lines under non-adherent conditions. Spheroids are typically formed from cancer cell lines as free-floating cell aggregates in ultra-low attachment dishes. Spheroids have been shown to maintain more stem cell-related properties compared to 2D cell cultures.

類器官為活體外衍生的細胞聚集體,其包括可分化為主要細胞譜系的細胞的幹細胞群體。類器官通常具有多於一毫米直徑之直徑,且經由繼代進行培養。與2D細胞培養相比,生長及擴展類器官培養通常較慢。為了自臨床樣本產生類器官,需要足夠數目的活細胞(例如,數百至數千)開始,因此自諸如生檢的較小體積樣本衍生類器官通常具有挑戰性,且—即使成功—擴展培養以用於諸如藥物測試之應用將耗費大量時間。另外,類器官大小、形狀及細胞數目存在大量變化性。類器官可需要生長因子之複合混合物及培養條件以便生長及表現所需細胞類型。Organoids are in vitro derived aggregates of cells that include populations of stem cells that can differentiate into cells of major cell lineages. Organoids typically have a diameter of more than one millimeter in diameter and are cultured through passage. Organoid cultures are generally slower to grow and expand than 2D cell cultures. To generate organoids from clinical samples, sufficient numbers of viable cells (e.g., hundreds to thousands) are required to start with, so deriving organoids from smaller volume samples such as biopsies is often challenging and—even if successful—expanded in culture For applications such as drug testing would be time consuming. In addition, organoid size, shape, and cell number exhibit substantial variability. Organoids may require complex mixtures of growth factors and culture conditions in order to grow and express desired cell types.

對於快速及可靠篩選,尤其對於個人化醫療,諸如執行藥物反應的活體外測試,腫瘤球體或類器官皆非最佳的。舉例而言,除平衡相對益處與風險以達成最有利的結果之外,腫瘤學之實踐不斷地面對使合宜的治療方案與合宜的患者匹配的巨大挑戰。患者衍生之癌症模型(PDMC)可包括使用類器官(包括患者衍生之類器官)以促進較多個別化治療目標之鑑別及發展。然而,儘管回溯性研究已展示衍生自切除或生檢之患者腫瘤的類器官與患者對療法之反應相關,但使用類器官導引療法存在主要限制。如上文所提及,自腫瘤樣本衍生及擴展類器官(且特定而言,患者衍生之類器官)以用於藥物敏感性測試會耗費數月,其降低臨床適用性,此係由於患者無法等待那麼久才接受治療。另外,目前無法在臨床上可實行的時間範圍自核心生檢試樣中獲得用超過幾十種化合物執行藥物篩選所需要的類器官之數目,其通常為患有轉移性或不能手術的癌症的患者組織之唯一可用形式。自生檢衍生類器官的較大失敗率亦防止其用作可靠診斷分析。此外,類器官之大小(及潛在地反應)可存在高度變化性,尤其在培養時間較長之情況下,且因此可存在許多繼代。Neither tumor spheroids nor organoids are optimal for rapid and reliable screening, especially for personalized medicine, such as performing in vitro tests of drug response. For example, in addition to balancing relative benefits and risks to achieve the most favorable outcome, the practice of oncology is constantly faced with the enormous challenge of matching appropriate treatment regimens to appropriate patients. Patient-derived cancer models (PDMC) can include the use of organoids, including patient-derived organoids, to facilitate the identification and development of more individualized therapeutic targets. However, while retrospective studies have shown that organoids derived from resected or biopsied patient tumors correlate with patient response to therapy, there are major limitations to using organoid-guided therapy. As mentioned above, deriving and expanding organoids (and patient-derived organoids in particular) from tumor samples for drug susceptibility testing can take months, which reduces clinical applicability because patients cannot wait It took so long to receive treatment. Additionally, the number of organoids needed to perform drug screens with more than a few dozen compounds, typically in patients with metastatic or inoperable cancer, is currently not available in a clinically feasible time frame from core biopsy samples The only available form of organization. The high failure rate of biopsy-derived organoids also prevents their use as reliable diagnostic assays. Furthermore, there can be a high degree of variability in the size (and potentially response) of organoids, especially over longer periods of time in culture, and thus many passages.

歸因於其與患者結果之較佳相關性,亦採用PDMC代替2D細胞株作為用於藥物發現的高通量篩選平台,諸如RNAi、CRISPR及藥用小分子篩網。然而,與細胞株相比,此等PDMC模型(包括球體及類器官)通常就擴展及操縱而言慢得多,使得高通量應用具有挑戰性且成本高。擴展此等模型以擴增細胞數目所需要的較長時間亦傾向於允許塑膠中之最快增長株系占主導且勝過其他株系,因此使得模型較均質且損失原始組織組合物及株系多樣性。此外,其相對較大及異質大小及有限的擴散性使其對於許多基於自動化螢光及成像的讀數分析具有挑戰性。Due to their better correlation with patient outcomes, PDMCs have also been adopted instead of 2D cell lines as high-throughput screening platforms for drug discovery, such as RNAi, CRISPR, and pharmaceutical small molecule screens. However, such PDMC models, including spheroids and organoids, are generally much slower to expand and manipulate than cell lines, making high-throughput applications challenging and costly. The longer time required to expand these models to expand cell numbers also tends to allow the fastest growing strain in the plastic to dominate and outperform the others, thus making the model more homogeneous and losing the original tissue composition and strain diversity. Furthermore, its relatively large and heterogeneous size and limited diffusivity make it challenging for many automated fluorescence- and imaging-based readouts.

因此,需要用於自切除或生檢產生患者衍生之組織模型(例如,腫瘤模型及/或非腫瘤組織模型)的方法、組合物及設備。特定而言,提供方法及設備將為有用的,該方法及設備可使得大量患者衍生之組織模型能夠具有單個生檢(諸如18規格之核心生檢)之可預測及臨床上地相關特性,其可例如在獲得生檢後7至10天內完成。此將准許穩固及可靠測試且將導引患者特定療法中之延遲降至最低。此外,產生患者衍生之模型亦將為有用的,該模型可以高度平行方式快速擴展,從而產生具有較小及較均勻大小、每單位細胞數目的較佳可控性及高通量篩選應用的較佳擴散性(例如,經由增加表面積與體積之比)之單元。Accordingly, there is a need for methods, compositions and devices for generating patient-derived tissue models (eg, tumor models and/or non-tumor tissue models) from resections or biopsies. In particular, it would be useful to provide methods and apparatus that enable a large number of patient-derived tissue models to have predictable and clinically relevant properties of a single biopsy, such as an 18-scale core biopsy, which This can be done, for example, within 7 to 10 days of obtaining a biopsy. This will allow robust and reliable testing and minimize delays in leading to patient-specific therapy. In addition, it would also be useful to generate patient-derived models that can be rapidly scaled up in a highly parallel fashion, resulting in better models with smaller and more uniform size, better controllability of the number of cells per unit, and high-throughput screening applications. Cells with better diffusivity (eg, by increasing the surface area to volume ratio).

本文中描述患者衍生之微型類器官球體(PMOS)、製備PMOS之設備及方法以及使用PMOS之設備及方法。本文中亦描述用於使用此等患者衍生之微型類器官球體篩選患者的方法及系統,包括個人化治療方法。Described herein are patient-derived micro-organoid spheroids (PMOS), devices and methods for making PMOS, and devices and methods for using PMOS. Also described herein are methods and systems for screening patients, including personalized therapy approaches, using these patient-derived mini-organoid spheroids.

一般而言,本文中描述形成及生長患者衍生之微型類器官球體的方法及設備,該微型類器官球體含有來源於患者的細胞,例如自較小的患者生檢提取(例如,用於快速診斷以導引療法);自經切除之患者組織,包括經切除之原發性腫瘤或功能異常器官之部分(例如,用於高通量篩選)提取;及/或自已經建立之PDMC提取,包括患者衍生之異種移植物(PDX)及類器官(例如,產生用於高通量篩選之微型類器官球體)。In general, described herein are methods and apparatus for forming and growing patient-derived miniature organoid spheroids containing patient-derived cells, e.g., extracted from smaller patient biopsies (e.g., for rapid diagnostics) to guide therapy); extraction from resected patient tissue, including resected primary tumors or fractions of dysfunctional organs (e.g., for high-throughput screening); and/or extraction from established PDMCs, including Patient-derived xenografts (PDX) and organoids (e.g., to generate miniature organoid spheroids for high-throughput screening).

此等PMOS可自正常的(例如,正常器官組織)或來自腫瘤組織的初代細胞形成。舉例而言,在一些變化形式中,此等方法及設備可自癌腫瘤生檢組織形成PMOS,進而實現可使用經檢驗的特定腫瘤組織選擇定製之治療。出人意料地,此等方法及設備准許在自患者移除生檢的幾小時內自單個組織生檢形成數百、數千或甚至數萬(例如,500、750、1000、2000、5000、10,000或更多)個PMOS。來自患者生檢的經解離之初代細胞可與流體基質材料(諸如受質基底膜基質(例如,基質膠))合併,以形成微型類器官球體。所得到的複數個患者衍生之微型類器官球體可具有預定義的尺寸範圍(諸如直徑,例如10 µm至700 µm及其內的任何子範圍),及初始數目之初代細胞(例如,1至1000個,且尤其較低數目之細胞,諸如1至200個)。細胞數目及/或直徑可控制在例如+/-5%、10%、15%、20%、25%、30%等內。當如本文中所描述形成時,此等PMOS具有特別高的存活率(>75%、>80%、>85%、>90%、>95%)且在極短時間段內使用及測試係穩定的,包括在形成之後的前1至10天內(例如,1天內、2天內、3天內、4天內、5天內、6天內、7天內、8天內、9天內、10天內等)。此允許對潛在大量的患者特定的及生物學相關的PMOS進行快速測試,其可節省產生及展開患者療法(諸如癌症治療計劃)的關鍵時間。本文中所描述之PMOS快速形成3D細胞結構,該3D細胞結構複製且對應於其等進行生檢的組織環境,諸如三維(3D)腫瘤微環境。本文中所描述之PMOS亦可稱作「小滴」。各PMOS可包括,例如作為流體基質材料之部分,可模擬原始組織(例如,腫瘤)環境的生長因子及結構蛋白質(例如,膠原蛋白、層黏連蛋白、巢蛋白等)。可使用幾乎任何初代細胞組織,包括幾乎任何腫瘤組織。These PMOSs can be formed from primary cells that are normal (eg, normal organ tissue) or from tumor tissue. For example, in some variations, these methods and devices can form PMOSs from cancerous tumor biopsy tissue, thereby enabling the selection of customized treatments using the specific tumor tissue tested. Surprisingly, such methods and devices permit the formation of hundreds, thousands, or even tens of thousands (e.g., 500, 750, 1000, 2000, 5000, 10,000, or more) PMOS. Dissociated primary cells from patient biopsies can be combined with a fluid matrix material, such as a substrate basement membrane matrix (eg, Matrigel), to form miniature organoid spheroids. The resulting plurality of patient-derived micro-organoid spheroids can have a predefined size range (such as diameter, e.g., 10 µm to 700 µm and any subrange therein), and an initial number of primary cells (e.g., 1 to 1000 , and especially lower numbers of cells, such as 1 to 200). Cell number and/or diameter can be controlled within, eg, +/- 5%, 10%, 15%, 20%, 25%, 30%, etc. These PMOSs had exceptionally high survival rates (>75%, >80%, >85%, >90%, >95%) when formed as described herein and were used and tested in extremely short periods of time. Stable, including within the first 1 to 10 days after formation (e.g., within 1 day, within 2 days, within 3 days, within 4 days, within 5 days, within 6 days, within 7 days, within 8 days, within 9 days days, within 10 days, etc.). This allows rapid testing of potentially large numbers of patient-specific and biologically relevant PMOSs, which can save critical time in generating and launching patient therapies, such as cancer treatment plans. The PMOS described herein rapidly forms 3D cellular structures that replicate and correspond to the tissue environment in which they are biopsied, such as a three-dimensional (3D) tumor microenvironment. The PMOS described herein may also be referred to as a "droplet." Each PMOS can include, for example, as part of a fluid matrix material, growth factors and structural proteins (eg, collagen, laminin, nestin, etc.) that can mimic the original tissue (eg, tumor) environment. Almost any primary cell tissue can be used, including almost any tumor tissue.

舉例而言,迄今為止,經測試的所有腫瘤類型及部位已成功地產生PMOS (例如,目前成功率為100%,n=32,包括來自包括肝臟、腸網膜及振動膜的原始部位或轉移性部位的結腸癌、食道癌、皮膚癌(黑色素瘤)、子宮癌、骨癌(肉瘤)、腎癌、卵巢癌、肺癌及乳癌)。用於成功產生微型類器官球體的組織類型可自其他位置轉移。在一些變化形式中,本文中所描述之PMOS可自細針抽出物(FNA)或自循環腫瘤細胞(CTC)生長,例如自液體生檢生長。增殖及生長通常地見於少至3至4天中,且PMOS可維持及繼代數月,或其可低溫保存及/或立即用於分析(例如,在前7至10天內)。For example, all tumor types and sites tested to date have successfully produced PMOS (eg, 100% success rate to date, n=32, including from original sites including liver, omentum, and vibrating membrane) or metastatic tumors. colon cancer, esophageal cancer, skin cancer (melanoma), uterine cancer, bone cancer (sarcoma), kidney cancer, ovarian cancer, lung cancer and breast cancer). Tissue types used to successfully generate miniature organoid spheroids can be transferred from other locations. In some variations, the PMOS described herein can be grown from fine needle aspirate (FNA) or from circulating tumor cells (CTC), eg, from liquid biopsy. Proliferation and growth are typically seen in as few as 3-4 days, and PMOSs can be maintained and subcultured for months, or they can be cryopreserved and/or used immediately for analysis (eg, within the first 7-10 days).

特定而言,本文中描述形成患者衍生之微型類器官球體之方法。一般而言,此等方法包括將解離之初代組織細胞(包括但不限於癌/異常組織、正常組織等)與液體基質材料合併以形成未聚合材料,且接著將未聚合材料聚合以形成通常直徑小於約1000 µm (例如,小於約900 µm、小於約800 µm、小於約700 µm、小於約600 µm,且尤其,小於約500 µm)的微型類器官球體,其中分佈有解離之初代組織細胞。解離之細胞的數目可在預定範圍內,如上文所提及(例如,約1至約500個細胞、約1至200個細胞、約1至150個細胞、約1至100個細胞之間、約1至75個細胞、約1至50個細胞、約1至30個細胞、約1至20個細胞、約1至10個細胞、約5至15個細胞、約20至30個細胞、約30至50個細胞、約40至60個細胞、約50至70個細胞、約60至80個細胞、約70至90個細胞、約80至100個細胞、約90至110個細胞等,包括約1個細胞、約10個細胞、約20個細胞、約30個細胞、約40個細胞、約50個細胞、約60個細胞、約70個細胞等)。此等方法中之任一者可如本文中所描述經結構設計以產生可重複大小(例如,具有較窄大小分佈)之微型類器官球體。In particular, methods for forming patient-derived mini-organoid spheroids are described herein. Generally, these methods involve combining dissociated primary tissue cells (including but not limited to cancer/abnormal tissue, normal tissue, etc.) with a liquid matrix material to form an unpolymerized material, and then polymerizing the unpolymerized material to form a typical diameter Miniature organoid spheroids of less than about 1000 µm (e.g., less than about 900 µm, less than about 800 µm, less than about 700 µm, less than about 600 µm, and especially, less than about 500 µm) having dissociated primary tissue cells distributed therein. The number of dissociated cells can be within a predetermined range, as mentioned above (e.g., between about 1 to about 500 cells, about 1 to 200 cells, about 1 to 150 cells, about 1 to 100 cells, about 1 to 75 cells, about 1 to 50 cells, about 1 to 30 cells, about 1 to 20 cells, about 1 to 10 cells, about 5 to 15 cells, about 20 to 30 cells, about 30 to 50 cells, about 40 to 60 cells, about 50 to 70 cells, about 60 to 80 cells, about 70 to 90 cells, about 80 to 100 cells, about 90 to 110 cells, etc., including about 1 cell, about 10 cells, about 20 cells, about 30 cells, about 40 cells, about 50 cells, about 60 cells, about 70 cells, etc.). Any of these methods can be structurally engineered as described herein to produce miniature organoid spheroids of reproducible size (eg, with a narrow size distribution).

解離之細胞可剛剛進行生檢且可以任何適當的方式解離,包括機械及/或化學解離(例如,藉由使用一或多種酶類的酶解聚,諸如膠原蛋白酶、胰蛋白酶等)。解離之細胞可視情況經處理、選擇及/或修飾。舉例而言,可分選或選擇細胞以鑑別及/或分離具有一或多個特徵(例如,大小、形態等)的細胞。可標記(例如,用一或多個標記)可用於輔助選擇的細胞。在一些變化形式中,可藉由已知細胞分選技術,包括但不限於微流體細胞分選、螢光激活細胞分選、磁激活細胞分選等。在一些實例中,可藉由諸如磁性珠粒或條碼狀粒子的非細胞物體分選PMOS。可用於分選的非細胞物體(例如,磁性珠粒、經螢光標記粒子等)可併入至PMOS中作為本文所描述方法中之任一者之部分。替代地,可在不進行分選之情況下使用細胞。Dissociated cells can be freshly biopsied and can be dissociated in any suitable manner, including mechanical and/or chemical dissociation (eg, by enzymatic disaggregation using one or more enzymes, such as collagenase, trypsin, etc.). Dissociated cells can optionally be treated, selected and/or modified. For example, cells can be sorted or selected to identify and/or isolate cells with one or more characteristics (eg, size, morphology, etc.). Cells that can be labeled (eg, with one or more markers) can be used to aid in selection. In some variations, known cell sorting techniques can be used, including but not limited to microfluidic cell sorting, fluorescence activated cell sorting, magnetic activated cell sorting, and the like. In some examples, PMOS can be sorted by acellular objects such as magnetic beads or barcode-like particles. Acellular objects useful for sorting (eg, magnetic beads, fluorescently labeled particles, etc.) can be incorporated into the PMOS as part of any of the methods described herein. Alternatively, cells can be used without sorting.

在一些變化形式中,可藉由用一或多種試劑處理來修飾解離之細胞。舉例而言,細胞可經基因修飾。在一些變化形式中,可使用CRISPR-Cas9或其他基因編輯技術修飾細胞。在一些變化形式中,可藉由任何適當的方法(例如,電穿孔、細胞擠壓、奈米粒子注射、磁轉染、化學轉染、病毒轉染等)轉染細胞,包括用質體、RNA、siRNA等轉染。替代地,可在不進行修飾之情況下使用細胞。In some variations, dissociated cells can be modified by treatment with one or more reagents. For example, cells can be genetically modified. In some variations, cells can be modified using CRISPR-Cas9 or other gene editing techniques. In some variations, cells can be transfected by any suitable method (e.g., electroporation, cell extrusion, nanoparticle injection, magnetofection, chemical transfection, viral transfection, etc.), including with plastids, RNA, siRNA, etc. transfection. Alternatively, cells can be used without modification.

一或多種額外材料可與解離之細胞及流體(例如,液體)基質材料合併以形成未聚合之混合物。舉例而言,未聚合之混合物可包括額外細胞或組織類型,包括支援細胞。額外細胞或組織可來源於不同生檢(例如,來自不同解離組織的初代細胞)及/或經培養的細胞。額外細胞可為例如免疫細胞、基質細胞、內皮細胞等。額外材料可包括介質(例如,生長介質、冷凍介質等)、生長因子、支援網狀分子(例如,膠原蛋白、糖蛋白、細胞外基質等)或其類似者。在一些變化形式中,額外材料可包括藥物組合物。在一些變化形式中,未聚合之混合物僅包括解離組織樣本(例如,初代細胞)及流體基質材料。One or more additional materials can be combined with dissociated cells and fluid (eg, liquid) matrix material to form an unpolymerized mixture. For example, the unpolymerized mixture can include additional cell or tissue types, including supporting cells. Additional cells or tissues may be derived from different biopsies (eg, primary cells from different dissociated tissues) and/or cultured cells. Additional cells can be, for example, immune cells, stromal cells, endothelial cells, and the like. Additional materials may include media (eg, growth media, freezing media, etc.), growth factors, support network molecules (eg, collagen, glycoproteins, extracellular matrix, etc.), or the like. In some variations, the additional material can include a pharmaceutical composition. In some variations, the unpolymerized mixture includes only a dissociated tissue sample (eg, primary cells) and a fluid matrix material.

方法可自單個組織生檢快速形成複數個患者衍生之微型類器官球體,以使得自每個生檢形成大於約500個患者衍生之微型類器官球體(例如,大於約600個、大於約700個、大於約800個、大於約900個、大於約1000個、大於約2000個、大於約2500個、大於約3000個、大於約4000個、大於約5000個、大於約6000個、大於約7000個、大於約8000個、大於約9000個、大於約10,000個、大於約11,000個、大於約12,000個等)。生檢可為標準大小生檢,諸如18G (例如,14G、16G、18G等)核心生檢。舉例而言,藉由生檢移除的及用於形成複數個患者衍生之微型類器官球體的組織體積可為約1/32至1/8吋直徑及約¾吋至¼吋長之較小圓柱(用生檢針獲取),諸如約1/16吋直徑×½吋長之圓柱。生檢可藉由針生檢例如藉由芯針生檢獲取。在一些變化形式中,生檢可藉由細針抽吸獲取。可使用的其他生檢類型包括剃削生檢、穿刺生檢、切取生檢、切除生檢及其類似者。通常來自單個患者生檢的材料可用於產生複數個(例如,大於約2000個、大於約5000個、大於約7500個、大於約10,000個等)如上文所描述之患者衍生之微型類器官球體。可使用可經結構設計以產生此大量高度有規律的(大小、細胞數目等)如本文中所描述之微型類器官球體的設備(如本文中所描述)形成複數個患者衍生之微型類器官球體。在一些變化形式中,此等方法及設備可以快速速率(例如,每分鐘大於約1個微型類器官球體、每10秒大於約1個微型類器官球體、每5秒大於約1個微型類器官球體、每2秒大於約1個微型類器官球體、每秒大於約1個微型類器官球體、每秒大於約2個微型類器官球體、每秒大於約3個微型類器官球體、每秒大於約4個微型類器官球體、每秒大於約5個微型類器官球體、每秒大於約10個微型類器官球體、每秒大於50個微型類器官球體、每秒大於100個微型類器官球體、每秒大於125個微型類器官球體等)產生複數個微型類器官球體。The method can rapidly form a plurality of patient-derived micro-organoid spheroids from a single tissue biopsy, such that greater than about 500 patient-derived micro-organoid spheroids (e.g., greater than about 600, greater than about 700) are formed from each biopsy , greater than about 800, greater than about 900, greater than about 1000, greater than about 2000, greater than about 2500, greater than about 3000, greater than about 4000, greater than about 5000, greater than about 6000, greater than about 7000 , greater than about 8000, greater than about 9000, greater than about 10,000, greater than about 11,000, greater than about 12,000, etc.). The biopsy may be a standard size biopsy, such as an 18G (eg, 14G, 16G, 18G, etc.) core biopsy. For example, the volume of tissue removed by biopsy and used to form the plurality of patient-derived micro-organoid spheroids can be as small as about 1/32 to 1/8 inch in diameter and about ¾ inch to ¼ inch long. Cylinder (obtained with a biopsy needle), such as a cylinder approximately 1/16 inch diameter by ½ inch long. The biopsy may be obtained by needle biopsy, for example by core needle biopsy. In some variations, biopsies may be obtained by fine needle aspiration. Other types of biopsies that may be used include shaving biopsies, punch biopsies, excisional biopsies, excisional biopsies, and the like. Typically material from a single patient biopsy can be used to generate a plurality (e.g., greater than about 2000, greater than about 5000, greater than about 7500, greater than about 10,000, etc.) of patient-derived mini-organoid spheroids as described above. A plurality of patient-derived mini-organoid spheroids can be formed using equipment (as described herein) that can be structurally designed to produce such large numbers of highly regular (size, cell number, etc.) mini-organoid spheroids as described herein . In some variations, these methods and devices can be used at rapid rates (e.g., greater than about 1 micro-organoid spheroid per minute, greater than about 1 micro-organoid spheroid per 10 seconds, greater than about 1 micro-organoid per 5 seconds Spheroids, greater than about 1 micro-organoid spheroid per 2 seconds, greater than about 1 micro-organoid spheroid per second, greater than about 2 micro-organoid spheroids per second, greater than about 3 micro-organoid spheroids per second, greater than about 4 micro-organoid spheroids, greater than about 5 micro-organoid spheroids per second, greater than about 10 micro-organoid spheroids per second, greater than 50 micro-organoid spheroids per second, greater than 100 micro-organoid spheroids per second, More than 125 micro-organoid spheroids per second, etc.) generate multiple micro-organoid spheroids.

舉例而言,在一些變化形式中,可藉由用與未聚合材料不混溶的材料(例如,液體材料)梳理未聚合混合物而執行此等方法。方法及設備可藉由至少部分地控制未聚合混合物(及/或解離組織及流體基質)及與未聚合混合物不混溶的材料(例如,疏水性材料、油等)中之一或多者之流量而控制微型類器官球體之大小及/或細胞密度。舉例而言,在一些變化形式中,可使用微流體設備執行此等方法。在一些變化形式中,可以並行地(例如,2個並行、3個並行、4個並行等)形成多個微型類器官球體。相同設備可因此包括多個並行通道,其可耦接至未聚合材料之相同源極,或解離初代組織之相同源極及/或流體基質之源極。For example, in some variations, the methods may be performed by combing the unpolymerized mixture with a material that is immiscible with the unpolymerized material (eg, a liquid material). Methods and apparatus may be controlled by at least partially controlling one or more of an unpolymerized mixture (and/or dissociated tissue and fluid matrix) and a material immiscible with the unpolymerized mixture (e.g., hydrophobic material, oil, etc.) Flow rate to control the size and/or cell density of the micro-organoid spheroids. For example, in some variations, microfluidic devices may be used to perform such methods. In some variations, multiple micro-organoid spheroids can be formed in parallel (eg, 2 in parallel, 3 in parallel, 4 in parallel, etc.). The same device may thus include multiple parallel channels, which may be coupled to the same source of unpolymerized material, or the same source of dissociated primary tissue and/or source of fluid matrix.

未聚合材料可經聚合以便以多種不同方式形成患者衍生之微型類器官球體。在一些變化形式中,方法可包括藉由改變溫度(例如,將溫度升高超過臨限值,諸如(例如)高於約20℃、高於約25℃、高於約30℃、高於約35℃等)而聚合微型類器官球體。Unpolymerized materials can be polymerized to form patient-derived micro-organoid spheroids in a number of different ways. In some variations, the method may include changing the temperature (e.g., raising the temperature above a threshold value, such as, for example, above about 20°C, above about 25°C, above about 30°C, above about 35°C, etc.) to aggregate micro-organoid spheroids.

一旦聚合,則患者衍生之微型類器官球體可允許例如藉由培養而生長及/或可在培養之前或之後分析及/或可在培養之前或之後低溫保存。患者衍生之微型類器官球體可培養任何適當的時長,但特定而言,可培養1天至10天(例如,1天至9天、1天至8天、1天至7天、1天至6天、3天至9天、3天至8天、3天至7天等)。在一些變化形式中,可在六次繼代之前低溫保存或分析患者衍生之微型類器官球體,其可將細胞之異質性保存在患者衍生之微型類器官球體內;限制繼代之數目可防止較快分裂細胞超過較緩分裂細胞。Once aggregated, the patient-derived micro-organoid spheroids can be allowed to grow, for example, by culture and/or can be analyzed before or after culture and/or can be cryopreserved before or after culture. Patient-derived mini-organoid spheroids can be cultured for any suitable length of time, but in particular can be cultured for 1 day to 10 days (e.g., 1 day to 9 days, 1 day to 8 days, 1 day to 7 days, 1 day to 6 days, 3 to 9 days, 3 to 8 days, 3 to 7 days, etc.). In some variations, patient-derived micro-organoid spheroids can be cryopreserved or analyzed prior to six passages, which preserves cellular heterogeneity within patient-derived micro-organoid spheroids; limiting the number of passages prevents Faster dividing cells outnumber slower dividing cells.

一般而言,由於相同患者生檢可提供較高數目的(例如,大於2,000個、大於3,000個、大於4,000個、大於5,000個、大於6,000個、大於7,000個、大於8,000個、大於9,000個、大於10,000個等)細胞,因此患者衍生之微型類器官球體中之一些可低溫保存(例如,超過一半),而一些經培養及/或分析。如在本文中將更詳細地描述,低溫保存的患者衍生之微型類器官球體可儲備且供稍後使用(例如,分析、繼代等)。In general, because the same patient biopsy can provide a higher number (eg, greater than 2,000, greater than 3,000, greater than 4,000, greater than 5,000, greater than 6,000, greater than 7,000, greater than 8,000, greater than 9,000, Greater than 10,000, etc.) cells, therefore some of the patient-derived micro-organoid spheroids can be cryopreserved (eg, more than half), while some are cultured and/or analyzed. As will be described in more detail herein, cryopreserved patient-derived mini-organoid spheroids can be stored and used later (eg, analysis, passage, etc.).

因此,本文中描述方法,包括形成複數個患者衍生之微型類器官球體之方法。舉例而言,一種形成複數個患者衍生之微型類器官球體之方法可包括:將解離之組織樣本及流體基質材料合併以形成未聚合之混合物;形成複數個未聚合之混合物小滴;及使小滴聚合以形成各自直徑介於50 µm與500 µm之間的複數個患者來源之微型類器官球體,其中分佈有1至200個解離之細胞。Accordingly, methods are described herein, including methods for forming a plurality of patient-derived micro-organoid spheroids. For example, a method of forming a plurality of patient-derived micro-organoid spheroids can include: combining a dissociated tissue sample and a fluid matrix material to form an unpolymerized mixture; forming a plurality of droplets of the unpolymerized mixture; and making the small Droplets aggregate to form patient-derived micro-organoid spheroids, each between 50 µm and 500 µm in diameter, distributed with 1 to 200 dissociated cells.

一種例如形成複數個患者衍生之微型類器官球體之方法可包括將解離之組織樣本及流體基質材料合併以形成未聚合之混合物;自未聚合之混合物之連續流形成複數個小滴,其中小滴具有小於25%的大小變化;及藉由升溫使小滴聚合以形成複數個患者衍生之微型類器官球體,各自具有分佈於各患者衍生之微型類器官球體內的1至200個解離之細胞。A method of, for example, forming a plurality of patient-derived micro-organoid spheroids may comprise combining a dissociated tissue sample and a fluid matrix material to form an unpolymerized mixture; forming a plurality of droplets from the continuous flow of the unpolymerized mixture, wherein the droplets having a size change of less than 25%; and polymerizing the droplets by increasing the temperature to form a plurality of patient-derived micro-organoid spheroids, each having 1 to 200 dissociated cells distributed within each patient-derived micro-organoid spheroid.

在一些變化形式中,如本文中所描述的用於形成複數個患者衍生之微型類器官球體之方法可包括:將解離之組織樣本及流體基質材料合併以形成未聚合之混合物;藉由使未聚合之混合物流同與未聚合之混合物不混溶的流體之一或多個流會聚而形成複數個小滴且小滴之大小變化小於25%的;使小滴聚合以形成直徑介於50 µm與500 µm之間的複數個患者衍生之微型類器官球體,其中分佈有1至200個解離之細胞;及將複數個患者衍生之微型類器官球體與不混溶的流體分離。In some variations, a method for forming a plurality of patient-derived micro-organoid spheroids as described herein may comprise: combining a dissociated tissue sample and a fluid matrix material to form an unpolymerized mixture; Convergence of a stream of a polymerized mixture with one or more streams of a fluid immiscible with an unpolymerized mixture to form a plurality of droplets that vary in size by less than 25%; causing droplets to coalesce to form diameters between 50 µm and patient-derived micro-organoid spheroids between 500 µm with 1 to 200 dissociated cells distributed therein; and separating the patient-derived micro-organoid spheroids from immiscible fluids.

此等方法中之任一者可包括在形成小滴之前修飾解離之組織樣本內的細胞。Any of these methods can include modifying the cells within the dissociated tissue sample prior to forming the droplets.

形成複數個小滴可包含形成具有小於約25%的大小變化(例如,小於約20%的大小變化、小於約15%的大小變化、小於約10%的大小變化、小於約8%的大小變化、小於約5%的大小變化等)的均勻大小的複數個未聚合混合物之小滴。大小變化亦可描述為大小變化之較窄分佈。舉例而言,大小分佈可包括具有低標準差(例如,15%或更小之標準差、12%或更小之標準差、10%或更小之標準差、8%或更小之標準差、6%或更小之標準差、5%或更小之標準差等)的患者衍生之微型類器官球體尺寸分佈(例如,微型類器官球體直徑與所形成的微型類器官球體之數目)。Forming a plurality of droplets can comprise forming a droplet having a size change of less than about 25% (e.g., a size change of less than about 20%, a size change of less than about 15%, a size change of less than about 10%, a size change of less than about 8%) , less than about 5% size variation, etc.) of uniformly sized droplets of a plurality of unpolymerized mixtures. Size changes can also be described as a narrower distribution of size changes. For example, the size distribution can include samples with a low standard deviation (e.g., 15% or less standard deviation, 12% or less standard deviation, 10% or less standard deviation, 8% or less standard deviation , 6% or less standard deviation, 5% or less standard deviation, etc.) patient-derived micro-organoid spheroid size distribution (eg, micro-organoid diameter and number of micro-organoid spheroids formed).

此等方法中之任一者亦可包括塗鋪或分佈患者衍生之微型類器官球體。舉例而言,在一些變化形式中,方法可包括在分析之前將來自不同來源的患者衍生之微型類器官球體合併至容器中。舉例而言,微型類器官球體可置放於多孔盤中。因此,此等方法中之任一者可包括在分析患者衍生之微型類器官球體之前將患者衍生之微型類器官球體施配至多孔盤中。每孔可包括一或多個(或在一些變化形式中,等量的)患者衍生之微型類器官球體。在一些變化形式中,將患者衍生之微型類器官球體塗覆至容器中可包括將類器官球體置放至藉由至少部分滲透膜間隔開的複數個腔室中,以准許上清液材料在腔室之間循環。此可允許患者衍生之微型類器官球體共用同一上清液。Any of these methods may also include plating or distributing patient-derived micro-organoid spheroids. For example, in some variations, methods can include combining patient-derived micro-organoid spheroids from different sources into a container prior to analysis. For example, miniature organoid spheroids can be placed in porous dishes. Accordingly, any of these methods may comprise dispensing the patient-derived micro-organoid spheroids into a multi-well disc prior to analyzing the patient-derived micro-organoid spheroids. Each well can include one or more (or, in some variations, equivalent amounts) of patient-derived mini-organoid spheroids. In some variations, coating the patient-derived micro-organoid spheroids into the container may include placing the organoid spheroids into a plurality of chambers separated by at least partially permeable membranes to allow the supernatant material to Circulation between chambers. This allows patient-derived micro-organoid spheroids to share the same supernatant.

在此等方法中之任一者中,可分析患者衍生之微型類器官球體。分析可通常包括於條件(例如,藥物組合物)下暴露或處理個別患者衍生之微型類器官球體以判定藥物組合物對患者衍生之微型類器官球體的細胞是否具有作用(且在一些情況下,其具有何種作用)。分析可包括將患者衍生之微型類器官球體子集(個別地或以群組形式)暴露於一或多種濃度之藥物組合物,及允許患者衍生之微型類器官球體保持暴露預定時段(數分鐘、數小時、數天等)及/或暴露及移除藥物組合物,接著將患者衍生之微型類器官球體培養預定時段。之後可檢驗患者衍生之微型類器官球體以鑑別任何作用,尤其包括對患者衍生之微型類器官球體中的細胞的毒性,或患者衍生之微型類器官球體中的細胞之形態及/或生長的變化。在一些變化形式中,分析可包括標記(例如,藉由免疫組織化學)患者衍生之微型類器官球體內的活細胞或固定細胞。可手動或自動分析(例如,檢驗)細胞。舉例而言,可使用自動讀取器設備檢驗細胞以測定任何毒性(細胞死亡)。在一些變化形式中,分析複數個患者衍生之微型類器官球體可包括對患者衍生之微型類器官球體之上清液、環境及微環境中之一或多者進行取樣,以用於分析分泌因子及其他效果。在此等變化形式中之任一者中,患者衍生之微型類器官球體可在分析之後回收以用於進一步分析、擴展或保存(例如,低溫保存、凝固等)以供後續檢驗。In any of these methods, patient-derived micro-organoid spheroids can be analyzed. Assays can generally involve exposing or treating individual patient-derived mini-organoid spheroids to conditions (e.g., pharmaceutical compositions) to determine whether the pharmaceutical composition has an effect on the cells of the patient-derived mini-organoid spheroids (and in some cases, what effect does it have). Assays may include exposing a subset of patient-derived micro-organoid spheroids (individually or in groups) to one or more concentrations of the pharmaceutical composition, and allowing the patient-derived micro-organoid spheroids to remain exposed for a predetermined period of time (minutes, minutes, Hours, days, etc.) and/or exposure and removal of the pharmaceutical composition, followed by culturing the patient-derived micro-organoid spheroids for a predetermined period of time. The patient-derived micro-organoid spheroids can then be assayed to identify any effects including, inter alia, toxicity to cells in the patient-derived micro-organoid spheroids, or changes in the morphology and/or growth of cells in the patient-derived micro-organoid spheroids . In some variations, the analysis can include labeling (eg, by immunohistochemistry) living or fixed cells within the patient-derived mini-organoid spheroids. Cells can be analyzed (eg, inspected) manually or automatically. For example, cells can be assayed using an automated reader device to determine any toxicity (cell death). In some variations, analyzing the plurality of patient-derived micro-organoid spheroids can include sampling one or more of the supernatant, the environment, and the microenvironment of the patient-derived micro-organoid spheroids for analysis of secreted factors and other effects. In any of these variations, the patient-derived micro-organoid spheroids can be recovered after analysis for further analysis, expanded, or stored (eg, cryopreserved, frozen, etc.) for subsequent examination.

如所提及,可使用幾乎任何分析。舉例而言,可使用本文中所描述之PMOS分析基因體、轉錄組、蛋白質體學或間-基因體標記(諸如甲基化)。因此,本文中所描述之此等組合物及方法中之任一者可用於鑑別或檢驗一或多個標記物及生物學/生理學路徑,包括(例如)胞外體,其可幫助鑑別用於患者治療的藥物及/或療法。As mentioned, almost any analysis can be used. For example, gene body, transcriptome, proteomic or inter-genome markers such as methylation can be analyzed using the PMOS described herein. Accordingly, any of the compositions and methods described herein can be used to identify or test one or more markers and biological/physiological pathways, including, for example, exosomes, which can aid in the identification of Medications and/or therapies used to treat the patient.

可使用任何適當的組織樣本。在一些變化形式中,組織樣本可包含來自轉移性腫瘤的生檢樣本。舉例而言,組織樣本可包含臨床腫瘤樣本;臨床腫瘤樣本可包含癌細胞及基質細胞兩者。在一些變化形式中,組織樣本包含腫瘤細胞及以下一或多者:間葉細胞、內皮細胞及免疫細胞。Any suitable tissue sample can be used. In some variations, the tissue sample may comprise a biopsy sample from a metastatic tumor. For example, a tissue sample can include a clinical tumor sample; a clinical tumor sample can include both cancer cells and stromal cells. In some variations, the tissue sample includes tumor cells and one or more of: mesenchymal cells, endothelial cells, and immune cells.

本文中所描述之方法中之任一者可包括最初將來自組織生檢的解離細胞以任何適當的濃度均勻地(或在一些變化形式中不均勻地)分佈在整個流體基質材料中。舉例而言,在一些變化形式中,本文中所描述之方法可包括將解離之組織樣本及流體基質材料合併,以使得解離之組織細胞在流體基質材料內分佈至小於1×10 7個細胞/毫升(例如,小於9×10 6個細胞/毫升、7×10 6個細胞/毫升、5×10 6個細胞/毫升、3×10 6個細胞/毫升、1×10 6個細胞/毫升、9×10 5個細胞/毫升、7×10 5個細胞/毫升、5×10 5個細胞/毫升等)之密度。 Any of the methods described herein may comprise initially dissociated cells from a tissue biopsy distributed uniformly (or in some variations non-uniformly) throughout the fluid matrix material at any suitable concentration. For example, in some variations, the methods described herein can include combining a dissociated tissue sample and a fluid matrix material such that the dissociated tissue cells are distributed within the fluid matrix material to less than 1 x 107 cells/ mL (e.g., less than 9×10 6 cells/ml, 7×10 6 cells/ml, 5×10 6 cells/ml, 3×10 6 cells/ml, 1×10 6 cells/ml, 9×10 5 cells/ml, 7×10 5 cells/ml, 5×10 5 cells/ml, etc.).

一般而言,形成小滴可包含自未聚合之混合物的連續流形成小滴。舉例而言,形成小滴可包含將一或多個與未聚合之混合物不混溶的流體之會聚流施用至未聚合之混合物流。該等流可在微流體裝置中合併,例如具有複數個會聚通道的裝置,未聚合混合物及不混溶的流體於其中相互作用以形成具有精確控制體積的小滴。在一些變化形式中,在過量之不混溶的材料中形成(例如,夾斷)小滴,且小滴可同時及/或隨後經聚合以形成患者衍生之微型類器官球體。舉例而言,流會聚的區域可經結構設計以在形成小滴之後例如藉由加熱而聚合未聚合之混合物,及/或下游的區域可經結構設計以在形成小滴已之後聚合未聚合之混合物且由不混溶的材料圍繞。在一些變化形式中,將不混溶的材料加熱(或替代地冷卻)至促進未聚合材料之聚合的溫度,從而形成患者衍生之微型類器官球體。舉例而言,聚合可包含將小滴加熱至高於35℃。In general, forming droplets may comprise forming droplets from a continuous stream of unpolymerized mixture. For example, forming droplets can include applying a converging stream of one or more fluids immiscible with the unpolymerized mixture to the unpolymerized mixture stream. These streams can be combined in a microfluidic device, such as a device with a plurality of converging channels in which unpolymerized mixtures and immiscible fluids interact to form droplets with precisely controlled volumes. In some variations, droplets are formed (eg, pinched off) in an excess of immiscible material, and the droplets can be simultaneously and/or subsequently polymerized to form patient-derived miniature organoid spheroids. For example, the region where the streams converge can be configured to polymerize the unpolymerized mixture after the droplets have been formed, for example by heating, and/or the region downstream can be configured to polymerize the unpolymerized mixture after the droplets have been formed. Mixed and surrounded by immiscible materials. In some variations, the immiscible material is heated (or alternatively cooled) to a temperature that promotes polymerization of the unpolymerized material, thereby forming patient-derived micro-organoid spheroids. For example, polymerizing may involve heating the droplets to above 35°C.

因此,在此等方法中之任一者中,形成小滴可包括在與未聚合之混合物不混溶的流體中形成小滴。此外,此等方法中之任一者可包括將不混溶的流體與患者衍生之微型類器官球體分離。舉例而言,任何此等方法可包括將不混溶的流體自患者衍生之微型類器官球體移除。一般而言,不混溶的流體可包括液體(例如,油、聚合物等),尤其包括疏水性材料或與未聚合(例如,水性)材料不混溶的的其他材料。Thus, in any of these methods, forming the droplets may comprise forming the droplets in a fluid that is immiscible with the unpolymerized mixture. Additionally, any of these methods can include separating the immiscible fluid from the patient-derived micro-organoid spheroids. For example, any of these methods can include removing immiscible fluid from the patient-derived micro-organoid spheroids. In general, immiscible fluids may include liquids (eg, oils, polymers, etc.), including hydrophobic materials or other materials that are immiscible with unpolymerized (eg, aqueous) materials, among others.

流體基質材料可為合成或非合成未聚合之基底膜材料。在一些變化形式中,未聚合基底材料可包含聚合水凝膠。在一些變化形式中,流體基質材料可包含基質膠。因此,將解離之組織樣本及流體基質材料合併可包含將解離之組織樣本與基底膜基質合併。The fluid matrix material may be a synthetic or non-synthetic unpolymerized basement membrane material. In some variations, the unpolymerized base material may comprise a polymerized hydrogel. In some variations, the fluid matrix material may comprise Matrigel. Thus, combining the dissociated tissue sample and the fluid matrix material may comprise combining the dissociated tissue sample with the basement membrane matrix.

組織樣本可在將組織樣本自患者移除的六個小時內或更早(例如,約5小時內、約4小時內、約3小時內、約2小時內、約1小時內等)與流體基質材料合併。The tissue sample can be mixed with the fluid within six hours or earlier (e.g., within about 5 hours, within about 4 hours, within about 3 hours, within about 2 hours, within about 1 hour, etc.) of removing the tissue sample from the patient. Matrix material combined.

本文中亦描述分析或保存患者衍生之微型類器官球體之方法。舉例而言,方法可包括:將解離之組織樣本及流體基質材料合併以形成未聚合之混合物;形成複數個未聚合之混合物小滴且小滴之大小變化小於25%;使小滴聚合以形成直徑介於50 µm與700 µm之間的複數個患者衍生之微型類器官球體,其中分佈有1至1000個解離之細胞;及分析或低溫保存複數個患者衍生之微型類器官球體。Also described herein are methods of analyzing or preserving patient-derived micro-organoid spheroids. For example, the method may include: combining a dissociated tissue sample and a fluid matrix material to form an unpolymerized mixture; forming a plurality of droplets of the unpolymerized mixture and the size of the droplets vary by less than 25%; polymerizing the droplets to form Multiple patient-derived micro-organoid spheroids between 50 µm and 700 µm in diameter with 1 to 1000 dissociated cells distributed therein; and analysis or cryopreservation of multiple patient-derived micro-organoid spheroids.

在一些變化形式中,方法可包括:將解離之組織樣本及流體基質材料合併以形成未聚合之混合物;形成複數個未聚合之混合物小滴;使小滴聚合以形成各自直徑介於50 µm與500 µm之間的複數個患者衍生之微型類器官球體,其中分佈有1至200個解離細胞;及在15天內低溫保存或分析複數個患者衍生之微型類器官球體,其中分析微型類器官以測定一或多種試劑對患者衍生之微型類器官球體內的細胞的作用。In some variations, the method may include: combining the dissociated tissue sample and the fluid matrix material to form an unpolymerized mixture; forming a plurality of droplets of the unpolymerized mixture; Multiple patient-derived micro-organoid spheroids between 500 µm with 1 to 200 dissociated cells distributed; The effect of one or more reagents on cells within the patient-derived mini-organoid spheroids is determined.

舉例而言,方法可包括:將解離之組織樣本及流體基質材料合併以形成未聚合混合物;藉由使未聚合之混合物流之同與未聚合之混合物不混溶的流體之一或多個流會聚而形成複數個小滴且小滴大小變化小於25%;藉由升溫使小滴聚合以形成各自直徑介於50 µm與500 µm之間的患者衍生之微型類器官球體,其中分佈有1至200個解離細胞;及在六次繼代之前分析或低溫保存患者衍生之微型類器官球體,由此維持患者衍生之微型類器官球體內的細胞之異質性,此外其中分析包含便於測定一或多種試劑對患者衍生之微型類器官球體內的細胞的作用的分析。For example, the method may comprise: combining a dissociated tissue sample and a fluid matrix material to form an unpolymerized mixture; Converge to form multiple droplets with a droplet size variation of less than 25%; the droplets are aggregated by heating to form patient-derived micro-organoid spheroids with individual diameters between 50 µm and 500 µm, distributed with 1 to 200 dissociated cells; and analyzing or cryopreserving the patient-derived micro-organoid spheroids prior to six passages, thereby maintaining the heterogeneity of cells within the patient-derived micro-organoid spheroids, further wherein the analysis includes facilitating the determination of one or more Analysis of the effect of reagents on cells within patient-derived micro-organoid spheroids.

在此等方法中之任一者中,可在六次繼代之前低溫保存或分析複數個患者衍生之微型類器官球體,由此維持患者衍生之微型類器官球體內的細胞之異質性。此等方法中之任一者可進一步包括在形成小滴之前修飾解離之組織樣本內的細胞。In either of these methods, the plurality of patient-derived micro-organoid spheroids can be cryopreserved or analyzed prior to six passages, thereby maintaining the heterogeneity of the cells within the patient-derived micro-organoid spheroids. Any of these methods can further include modifying the cells within the dissociated tissue sample prior to forming the droplets.

形成小滴可包括形成具有小於約25%的大小變化(例如,小於約20%、小於約15%、小於約10%、小於約7%、小於約5%等)的均勻大小的複數個未聚合之混合物小滴。Forming the droplets can include forming a plurality of uniformly sized droplets having a size variation of less than about 25% (e.g., less than about 20%, less than about 15%, less than about 10%, less than about 7%, less than about 5%, etc.). Polymerized mixture droplets.

此等方法中之任一者可包括將患者衍生之微型類器官球體培養適當的時長,如上文所提及(例如,在分析之前將患者衍生之微型類器官球體培養2至14天)。舉例而言,此等方法可包括在培養之前將不混溶的流體自患者衍生之微型類器官球體移除。在一些變化形式中,培養患者衍生之微型類器官球體包含在懸浮液中培養患者衍生之微型類器官球體。Any of these methods may include culturing the patient-derived mini-organoid spheroids for an appropriate length of time, as mentioned above (eg, culturing the patient-derived mini-organoid spheroids for 2 to 14 days prior to analysis). For example, such methods can include removing immiscible fluids from patient-derived micro-organoid spheroids prior to culturing. In some variations, culturing the patient-derived micro-organoid spheroids comprises culturing the patient-derived micro-organoid spheroids in suspension.

在本文中所描述之方法之任一者中,患者衍生之微型類器官球體可一起大量培養。在一些變化形式中,例如當形成庫時,藉由大小及/或細胞之數目分選的患者衍生之微型類器官球體可一起培養。患者衍生之微型類器官球體可在懸浮液中或在層中(例如,在培養皿、腔室、管內等)培養。In any of the methods described herein, patient-derived mini-organoid spheroids can be cultured together in large quantities. In some variations, for example when forming a bank, patient-derived mini-organoid spheroids sorted by size and/or number of cells can be cultured together. Patient-derived micro-organoid spheroids can be cultured in suspension or in layers (eg, in dishes, chambers, tubes, etc.).

一般而言,分析患者衍生之微型類器官球體可包含在分析或低溫保存患者衍生之微型類器官球體之前及/或之後在基因體學上、轉錄組學上、表現基因組學上及/或代謝上分析患者衍生之微型類器官球體中的細胞。此等方法中之任一者可包括藉由將患者衍生之微型類器官球體暴露於藥物(例如,藥物組合物)而分析患者衍生之微型類器官球體。In general, analyzing patient-derived micro-organoid spheroids may comprise genomic, transcriptomic, epigenomic and/or metabolic testing prior to and/or after analysis or cryopreservation of patient-derived micro-organoid spheroids. Analysis of cells in patient-derived micro-organoid spheroids. Any of these methods can comprise analyzing the patient-derived micro-organoid spheroids by exposing the patient-derived micro-organoid spheroids to a drug (eg, a pharmaceutical composition).

在此等方法中之任一者中,分析可包含手動及/或自動地在視覺上分析一或多種試劑對患者衍生之微型類器官球體中的細胞的作用。此等方法中之任一者可包括標記或標註患者衍生之微型類器官球體中的細胞以用於可視化。舉例而言,分析可包括螢光分析一或多種試劑對細胞的作用。In any of these methods, analyzing can comprise manually and/or automatically visually analyzing the effect of one or more reagents on the cells in the patient-derived mini-organoid spheroids. Any of these methods can include labeling or labeling the cells in the patient-derived mini-organoid spheroids for visualization. For example, analysis can include fluorometric analysis of the effect of one or more reagents on cells.

本文中所描述之患者衍生之微型類器官球體本身為新穎的且可表徵為物質組合物。舉例而言,物質組合物可包含複數個低溫保存的患者衍生之微型類器官球體,其中各患者衍生之微型類器官球體具有直徑介於50 µm與500 µm之間的球形且包含聚合基底材料,及分佈於基底材料內的繼代少於六次的約1至1000個解離之初代細胞,由此維持患者衍生之微型類器官球體內的細胞之異質性。The patient-derived micro-organoid spheroids described herein are novel per se and can be characterized as compositions of matter. For example, the composition of matter may comprise a plurality of cryopreserved patient-derived micro-organoid spheroids, wherein each patient-derived micro-organoid spheroid has a spherical shape with a diameter between 50 µm and 500 µm and comprises a polymeric base material, and about 1 to 1000 dissociated primary cells of less than six passages distributed within the base material, thereby maintaining the heterogeneity of the cells within the patient-derived micro-organoid spheroids.

本文中亦描述包含複數個低溫保存的患者衍生之微型類器官球體的物質組合物,其中各患者衍生之微型類器官球體具有直徑介於50 µm與500 µm之間的球形,其中患者衍生之微型類器官球體具有小於25%的大小變化,且其中各患者衍生之微型類器官球體包含聚合基底材料,及分佈於基底材料內的繼代少於六次的約1至500個解離之初代細胞,由此維持患者衍生之微型類器官球體內的細胞之異質性。Also described herein are compositions of matter comprising a plurality of cryopreserved patient-derived micro-organoid spheroids, wherein each patient-derived micro-organoid spheroid has a spherical shape with a diameter between 50 µm and 500 µm, wherein the patient-derived micro-organoid The organoid spheroids have a size variation of less than 25%, and wherein each patient-derived micro-organoid spheroid comprises a polymeric substrate material and about 1 to 500 dissociated primary cells of less than six passages distributed within the substrate material, The heterogeneity of the cells within the patient-derived mini-organoid spheroids is thereby maintained.

初代細胞可為初代腫瘤細胞。舉例而言,解離之初代細胞可已經基因或生物化學修飾。複數個低溫保存的患者衍生之微型類器官球體可具有均勻大小且大小變化小於25%。在一些變化形式中,複數個低溫保存的患者衍生之微型類器官球體可包含來自不同來源的患者衍生之微型類器官球體。在此等微型類器官球體中之任一者中,各微型類器官球體中的大多數細胞可包含並非幹細胞的細胞。在一些變化形式中,初代細胞包含轉移性腫瘤細胞。初代細胞可包含癌細胞及基質細胞兩者。在一些變化形式中,初代細胞包含腫瘤細胞及以下一或多者:間葉細胞、內皮細胞及免疫細胞。Primary cells may be primary tumor cells. For example, dissociated primary cells may have been genetically or biochemically modified. The plurality of cryopreserved patient-derived micro-organoid spheroids can be of uniform size with less than 25% variation in size. In some variations, the plurality of cryopreserved patient-derived micro-organoid spheroids may comprise patient-derived micro-organoid spheroids from different sources. In any of these mini-organoid spheroids, the majority of cells in each mini-organoid spheroid may comprise cells that are not stem cells. In some variations, the primary cells comprise metastatic tumor cells. Primary cells can include both cancer cells and stromal cells. In some variations, the primary cells comprise tumor cells and one or more of: mesenchymal cells, endothelial cells, and immune cells.

初代細胞可以小於例如5×10 7個細胞/毫升、1×10 7個細胞/毫升、9×10 6個細胞/毫升、7×10 6個細胞/毫升、5×10 6個細胞/毫升、1×10 6個細胞/毫升、9×10 5個細胞/毫升、7×10 5個細胞/毫升、5×10 5個細胞/毫升、1×10 5個細胞/毫升等之密度分佈於聚合基底材料內。 Primary cells can be less than, for example, 5 x 107 cells/ml, 1 x 107 cells/ml, 9 x 106 cells/ml, 7 x 106 cells/ml, 5 x 106 cells/ml, The density distribution of 1×10 6 cells/ml, 9×10 5 cells/ml, 7×10 5 cells/ml, 5×10 5 cells/ml, 1×10 5 cells/ml, etc. in aggregation in the base material.

一般而言,聚合基底材料可包含基底膜基質(例如,基質膠)。在一些變化形式中,聚合基底材料包含合成材料。Generally, the polymeric base material can comprise a basement membrane matrix (eg, Matrigel). In some variations, the polymeric base material comprises a synthetic material.

微型類器官可具有介於50 µm與1000 µm之間的直徑,或更佳50 µm與700 µm之間、或更佳50 µm與500 µm之間、或50 µm與400 µm之間、或50 µm與300 µm之間、或50 µm與250 µm之間等(例如,小於約500 µm、小於約400 µm、小於約300 µm、小於約250 µm、小於約200 µm等)。The micro-organoids may have a diameter between 50 µm and 1000 µm, or more preferably between 50 µm and 700 µm, or more preferably between 50 µm and 500 µm, or between 50 µm and 400 µm, or 50 Between µm and 300 µm, or between 50 µm and 250 µm, etc. (eg, less than about 500 µm, less than about 400 µm, less than about 300 µm, less than about 250 µm, less than about 200 µm, etc.).

如所提及,本文中所描述之患者衍生之微型類器官球體可包括最初在各患者衍生之微型類器官球體中的任何適當數目之初代組織細胞,例如少於約200個初代細胞、或更佳少於約150個初代細胞、或更佳少於約100個初代細胞、或更佳少於約75個初代細胞、或少於約50個細胞、或少於約30個細胞、或少於約25個細胞、或少於約20個細胞、或少於約10個細胞、或少於約5個細胞等)。在一些變化形式中,各患者衍生之微型類器官球體包括約1至500個細胞、約1至400個細胞、約1至300個細胞、約1至200個細胞、約1至150個細胞、約1至100個細胞、約1至75個細胞、約1至50個細胞、約1至30個細胞、約1至25個細胞、約1至20個細胞等。As mentioned, the patient-derived micro-organoid spheroids described herein can include any suitable number of primary tissue cells initially in each patient-derived micro-organoid spheroid, such as less than about 200 primary cells, or more Preferably less than about 150 primary cells, or more preferably less than about 100 primary cells, or more preferably less than about 75 primary cells, or less than about 50 cells, or less than about 30 cells, or less than about 25 cells, or less than about 20 cells, or less than about 10 cells, or less than about 5 cells, etc.). In some variations, each patient-derived micro-organoid spheroid comprises about 1 to 500 cells, about 1 to 400 cells, about 1 to 300 cells, about 1 to 200 cells, about 1 to 150 cells, About 1 to 100 cells, about 1 to 75 cells, about 1 to 50 cells, about 1 to 30 cells, about 1 to 25 cells, about 1 to 20 cells, etc.

本文中亦描述用於形成患者衍生之微型類器官球體的設備,及操作此等設備以形成患者衍生之微型類器官球體的方法。舉例而言,本文中描述操作患者衍生之微型類器官球體形成設備的方法,其包含:在第一孔口中接收包含解離之組織樣本及第一流體基質材料之冷卻混合物的未聚合之混合物;在第二孔口中接收與未聚合之混合物不混溶的第二流體;將未聚合之混合物流與第二流體中之一或多個流合併以形成均勻大小且變化小於25%的未聚合之混合物小滴;及使未聚合之混合物小滴聚合以形成複數個患者衍生之微型類器官球體。Also described herein are devices for forming patient-derived micro-organoid spheroids, and methods of operating such devices to form patient-derived micro-organoid spheroids. For example, described herein is a method of operating a patient-derived micro-organoid spheroid forming device comprising: receiving in a first port an unpolymerized mixture comprising a dissociated tissue sample and a cooled mixture of a first fluid matrix material; Receiving a second fluid immiscible with the unpolymerized mixture in the second orifice; combining the stream of the unpolymerized mixture with one or more streams of the second fluid to form an unpolymerized mixture of uniform size and variation less than 25% droplets; and polymerizing the unpolymerized mixture droplets to form a plurality of patient-derived micro-organoid spheroids.

一種操作患者衍生之微型類器官球體形成設備的方法可包括:在第一孔口中接收包含解離之組織樣本及第一流體基質材料之冷卻混合物的未聚合之混合物;在第二孔口中接收與未聚合之混合物不混溶的第二流體;將處於第一速率的未聚合之混合物流與處於第二速率的第二流體中之一或多個流合併以形成均勻大小且變化小於25%的未聚合之混合物小滴,其中小滴的直徑介於50 µm與500 µm之間;及使未聚合之混合物小滴聚合以形成複數個患者衍生之微型類器官球體。A method of operating a patient-derived micro-organoid spheroid forming device may include: receiving an unpolymerized mixture comprising a dissociated tissue sample and a cooled mixture of a first fluid matrix material in a first port; receiving an unpolymerized mixture in a second port; A second fluid in which the polymerized mixture is immiscible; combining the stream of the unpolymerized mixture at the first rate with one or more streams of the second fluid at the second rate to form unpolymerized streams of uniform size and less than 25% variation polymerizing the mixture droplets, wherein the diameter of the droplets is between 50 µm and 500 µm; and polymerizing the unpolymerized mixture droplets to form a plurality of patient-derived micro-organoid spheroids.

此等方法中之任一者可包括將含有未聚合之混合物的第一儲集器與第一孔口以流體連通形式耦接。舉例而言,方法可包括將解離之組織樣本及第一流體基質材料合併以形成未聚合之混合物。在一些變化形式中,方法包括將未聚合之混合物添加至與第一孔口流體連通的第一儲集器。此等方法可包括將含有第二流體的第二儲集器與第二孔口以流體連通形式耦接。此等方法中之任一者可包括將第二流體添加至與第二孔口流體連通的第二儲集器。在一些變化形式中,接收第二流體包含接收油。Any of these methods can include coupling a first reservoir containing the unpolymerized mixture in fluid communication with the first orifice. For example, a method can include combining a dissociated tissue sample and a first fluid matrix material to form an unpolymerized mixture. In some variations, the method includes adding the unpolymerized mixture to a first reservoir in fluid communication with the first orifice. Such methods may include coupling a second reservoir containing a second fluid in fluid communication with a second orifice. Any of these methods can include adding the second fluid to a second reservoir in fluid communication with the second orifice. In some variations, receiving the second fluid includes receiving oil.

一般而言,此等方法可包括將第二流體(例如,不混溶的流體)與複數個患者衍生之微型類器官球體分離。可手動或自動分離此流體。舉例而言,可藉由洗滌、過濾或任何其他適當的方法移除第二(不混溶的)流體。Generally, such methods can include separating a second fluid (eg, an immiscible fluid) from the plurality of patient-derived micro-organoid spheroids. This fluid can be separated manually or automatically. For example, the second (immiscible) fluid may be removed by washing, filtering or any other suitable method.

將流合併可包含驅動處於第一流動速率的未聚合之混合物流穿過以第二流動速率移動的第二流體中之一或多個流。在一些變化形式中,第一流動速率大於第二流動速率。材料(例如,未聚合之混合物)之流動速率及/或量中的任一者或兩者可以比第二流體較小的量存在,以使得未聚合之混合物囊封於精確控制的小滴中,如本文中所描述,該小滴可接著例如在第二流體內聚合。Combining the streams may comprise driving the unpolymerized mixture stream at the first flow rate through one or more streams of the second fluid moving at the second flow rate. In some variations, the first flow rate is greater than the second flow rate. Either or both of the flow rate and/or amount of material (e.g., unpolymerized mixture) may be present in a smaller amount than the second fluid so that the unpolymerized mixture is encapsulated in precisely controlled droplets , as described herein, the droplets may then polymerize, for example, within a second fluid.

在一些變化形式中,將流合併包含驅動未聚合之混合物流穿過第二流體中之一或多個流亦會聚至其中的接合處。聚合小滴可包含將小滴加熱至高於未聚合材料聚合的溫度(例如,高於約25℃、高於約30℃、高於約35℃等)。In some variations, combining the streams includes driving the stream of the unpolymerized mixture through a junction into which one or more streams of the second fluid also converge. Polymerizing the droplets can comprise heating the droplets to a temperature above which the unpolymerized material polymerizes (eg, above about 25°C, above about 30°C, above about 35°C, etc.).

此等方法中之任一者可包括將複數個患者衍生之微型類器官球體等分。舉例而言,等分至多孔培養皿中。Any of these methods may comprise aliquoting a plurality of patient-derived mini-organoid spheroids. For example, aliquot into multi-well Petri dishes.

本文中亦描述使用此等患者衍生之微型類器官球體治療患者的方法及分析其等的方法。舉例而言,方法可包括:接收來自腫瘤的患者生檢;在獲取生檢的2週內藉由以下操作判定腫瘤將對藥物調配物有反應:自患者生檢形成直徑介於50 µm與500 µm之間的複數個微型類器官球體,其中1至200個解離之腫瘤細胞分佈於整個聚合基底材料中,及在解離之腫瘤細胞已進行多於五次繼代之前將患者衍生之微型類器官球體中之至少一些暴露於藥物調配物;及量測藥物調配物對微型類器官球體中之至少一些內的細胞的作用,以基於所判定之作用判定藥物是否將治療腫瘤。Also described herein are methods of treating patients using these patient-derived mini-organoid spheroids and methods of analyzing the same. For example, the method may comprise: receiving a patient biopsy from the tumor; determining that the tumor will respond to the drug formulation within 2 weeks of obtaining the biopsy by: forming a tumor from the patient biopsy between 50 µm and 500 Multiple micro-organoid spheroids between µm with 1 to 200 dissociated tumor cells distributed throughout the polymeric substrate material, and patient-derived micro-organoids before the dissociated tumor cells have been passaged for more than five passages exposing at least some of the spheroids to the drug formulation; and measuring the effect of the drug formulation on cells within at least some of the mini-organoid spheroids to determine whether the drug will treat the tumor based on the determined effect.

在一些變化形式中,此等方法可包括藉由以下操作判定在一或多次投與藥物之後腫瘤仍對藥物調配物有反應:在患者已用藥物調配物治療之後接收第二患者生檢及自第二患者生檢形成第二複數個患者衍生之微型類器官球體,將第二複數個患者衍生之微型類器官球體中之至少一些暴露於藥物調配物,及量測藥物調配物對第二複數個微型類器官球體中之至少一些內的細胞的作用。In some variations, the methods may include determining that the tumor remains responsive to the drug formulation after one or more administrations of the drug by receiving a second patient biopsy after the patient has been treated with the drug formulation and Forming a second plurality of patient-derived micro-organoid spheroids from a second patient biopsy, exposing at least some of the second plurality of patient-derived micro-organoid spheroids to the drug formulation, and measuring the effect of the drug formulation on the second The role of cells within at least some of the plurality of miniature organoid spheroids.

判定腫瘤將對藥物調配物有反應可包括將患者衍生之微型類器官球體中之至少一些暴露於複數種藥物調配物,及報導藥物調配物中之每一者的量測作用。在一些變化形式中,判定進一步包含在分析患者衍生之微型類器官球體之前將微型類器官球體施配至多孔盤中。Determining that a tumor will respond to a drug formulation can include exposing at least some of the patient-derived mini-organoid spheroids to a plurality of drug formulations, and reporting a measured effect of each of the drug formulations. In some variations, determining further comprises dispensing the micro-organoid spheroids into the porous disc prior to analyzing the patient-derived micro-organoid spheroids.

此等方法中之任一者可包括對患者進行生檢以收集患者生檢(或以其他方式獲取來自患者的組織樣本或患者衍生之組織或細胞的樣本),及/或用藥物調配物治療患者,或幫助醫師治療患者(例如,建議醫師何種藥物調配物將有效)。一般而言,接收生檢與報導之間的時間可少於約21天(例如,少於約15天、少於約14天、少於約13天、少於約12天、少於約11天、少於約10天、少於約9天、少於約8天、少於約7天等)。Any of these methods may include taking a biopsy of the patient to collect the patient biopsy (or otherwise obtain a tissue sample from the patient or a sample of patient-derived tissue or cells), and/or treat with the drug formulation patient, or assisting a physician in treating a patient (eg, advising the physician which drug formulation will be effective). Generally, the time between receipt of a biopsy and reporting can be less than about 21 days (e.g., less than about 15 days, less than about 14 days, less than about 13 days, less than about 12 days, less than about 11 days days, less than about 10 days, less than about 9 days, less than about 8 days, less than about 7 days, etc.).

一般而言,所描述之方法及設備經結構設計以在處理期間達成較高囊封效率及樣本/細胞之最低損耗。因此,如本文中所描述,可自單個經切除或生檢的腫瘤樣本高效地形成大量患者衍生之微型類器官球體。此等方法及系統可克服與其他系統相關之問題,其中較小臨床樣本將在處理中完全損耗,且較大臨床樣本無法達至足夠建立類器官的密度。In general, the methods and apparatus described are structurally designed to achieve high encapsulation efficiency and minimal loss of sample/cells during processing. Thus, as described herein, large numbers of patient-derived mini-organoid spheroids can be efficiently formed from a single resected or biopsied tumor sample. These methods and systems can overcome problems associated with other systems in which smaller clinical samples are completely lost in processing and larger clinical samples cannot reach sufficient densities for organoid establishment.

特定而言,此等方法及設備可提供接收來自患者的腫瘤之解離樣本(例如,單一腫瘤,儘管在一些實例中可提供多個腫瘤樣本)的微流體系統,且控制黏度及/或系統內的流動速率。可控制路徑長度及直徑以及扭曲率(例如,曲線)以防止堵塞及/或損壞解離細胞。In particular, such methods and apparatus can provide microfluidic systems that receive a dissociated sample of a tumor from a patient (e.g., a single tumor, although in some examples multiple tumor samples can be provided), and control the viscosity and/or flow rate. Path length and diameter and twist rate (eg, curve) can be controlled to prevent clogging and/or damage to dissociated cells.

舉例而言,此等方法及設備可藉由在微流體系統(在一些實例中其可經結構設計為可移除濾筒)之通道內維持恆定的或接近恆定的壓力而控制壓力、流動速率(流速)或壓力及流動速率。濾筒可為單次用的或可再用的。因此可經由具有恆定或幾乎恆定壓力的微流體系統泵送流體(例如,解離細胞溶液及未聚合基質材料)。一般而言,經由微流體系統的材料之流可為層狀的,尤其解離細胞之流。可藉由將流動速率維持在預定範圍(例如,0.01 ml/min與100 ml/min之間、0.01 ml/min與50 ml/min之間、0.01 ml/min與20 ml/min之間、0.01 ml/min與10 ml/min之間、0.01 ml/min與5 ml/min之間、0.05 ml/min與100 ml/min之間、0.05 ml/min與50 ml/min之間、0.05 ml/min與10 ml/min之間、0.05 ml/min與5 ml/min之間、0.1 ml/min與100 ml/min之間、0.1 ml/min與50 ml/min之間、0.1 ml/min與10 ml/min之間、0.1 ml/min與5 ml/min之間、1 ml/min與100 ml/min之間、1 ml/min與50 ml/min之間、1 ml/min與20 ml/min之間、1 ml/min與10 ml/min之間、1 ml/min與5 ml/min之間、5 ml/min與100 ml/min之間、5 ml/min與50 ml/min之間、5 ml/min與10 ml/min之間等)內來維持層流。For example, the methods and apparatus can control pressure, flow rate by maintaining a constant or near constant pressure within the channels of a microfluidic system (which in some instances can be configured with a removable filter cartridge) (flow rate) or pressure and flow rate. Filter cartridges can be single use or reusable. Fluids (eg, solutions of dissociated cells and unpolymerized matrix material) can thus be pumped through the microfluidic system with constant or nearly constant pressure. In general, the flow of materials through a microfluidic system can be laminar, especially the flow of dissociated cells. By maintaining the flow rate within a predetermined range (for example, between 0.01 ml/min and 100 ml/min, between 0.01 ml/min and 50 ml/min, between 0.01 ml/min and 20 ml/min, between 0.01 Between ml/min and 10 ml/min, between 0.01 ml/min and 5 ml/min, between 0.05 ml/min and 100 ml/min, between 0.05 ml/min and 50 ml/min, between 0.05 ml/min between min and 10 ml/min, between 0.05 ml/min and 5 ml/min, between 0.1 ml/min and 100 ml/min, between 0.1 ml/min and 50 ml/min, between 0.1 ml/min and Between 10 ml/min, between 0.1 ml/min and 5 ml/min, between 1 ml/min and 100 ml/min, between 1 ml/min and 50 ml/min, between 1 ml/min and 20 ml /min, between 1 ml/min and 10 ml/min, between 1 ml/min and 5 ml/min, between 5 ml/min and 100 ml/min, between 5 ml/min and 50 ml/min between 5 ml/min and 10 ml/min, etc.) to maintain laminar flow.

微流體系統內的自解離細胞之輸入端至形成未聚合基質之小滴(及解離細胞)的區域的路徑長度可小於例如10 cm、小於9 cm、小於8 cm、小於7 cm、小於6 cm、小於5 cm、小於4 cm、小於3 cm、小於2 cm、小於1 cm等。The path length within the microfluidic system from the input of the dissociated cells to the region where the unpolymerized matrix is formed (and the dissociated cells) may be less than, for example, 10 cm, less than 9 cm, less than 8 cm, less than 7 cm, less than 6 cm , less than 5 cm, less than 4 cm, less than 3 cm, less than 2 cm, less than 1 cm, etc.

解離細胞通過的一或多個通道之直徑可經結構設計以防止堵塞。舉例而言,通道之直徑可大於80 µm、大於100 µm、大於120 µm、大於150 µm、大於200 µm、大於250 µm、大於300 µm、大於400 µm、大於450 µm、大於500 µm等(例如,80 µm至500 µm之間、100 µm至500 µm之間、120 µm至500 µm之間、150 µm至500 µm之間、200 µm至500 µm之間等)。The diameter of the channel or channels through which dissociated cells pass can be engineered to prevent clogging. For example, the diameter of the channel can be greater than 80 µm, greater than 100 µm, greater than 120 µm, greater than 150 µm, greater than 200 µm, greater than 250 µm, greater than 300 µm, greater than 400 µm, greater than 450 µm, greater than 500 µm, etc. (e.g. , between 80 µm and 500 µm, between 100 µm and 500 µm, between 120 µm and 500 µm, between 150 µm and 500 µm, between 200 µm and 500 µm, etc.).

解離細胞可在以流體方式耦接至微流體系統之輸入端的孔口或腔室、容器等處藉由短的或非常短的導管(例如,小於20 cm、小於15 cm、小於10 cm、小於7.5 cm、小於5 cm等)添加至微流體系統,以將「死亡」區域減至最少。Dissociated cells can be passed through short or very short conduits (e.g., less than 20 cm, less than 15 cm, less than 10 cm, less than 7.5 cm, less than 5 cm, etc.) are added to the microfluidic system to minimize the "dead" area.

一般而言,微流體系統之通道之幾何形狀可經結構設計以避免堵塞。舉例而言,一或多個通道可具有大於1 mm (大於2 mm、大於3 mm、大於5 mm、大於10 mm、大於15 mm、大於20 mm、大於25 mm、大於30 mm、大於40 mm、大於50 mm等)之曲率半徑。路徑內的所有或實質上所有邊角可為平滑的、彎曲的或切成圓角的。In general, the geometry of the channels of a microfluidic system can be engineered to avoid clogging. For example, one or more channels may have a diameter greater than 1 mm (greater than 2 mm, greater than 3 mm, greater than 5 mm, greater than 10 mm, greater than 15 mm, greater than 20 mm, greater than 25 mm, greater than 30 mm, greater than 40 mm , greater than 50 mm, etc.) radius of curvature. All or substantially all corners within the path may be smooth, curved or rounded.

微流體系統可包括一或多個氣泡移除腔室(例如,用於移除氣泡的透氣但流體不可滲透的膜及/或真空孔口等)。A microfluidic system can include one or more air bubble removal chambers (eg, gas-permeable but fluid-impermeable membranes and/or vacuum ports, etc. for removing air bubbles).

一般而言,微流體系統可控制一或多個通道內的材料之溫度(且因此黏度)。如所提及,攜載解離細胞及未聚合基質的通道可經冷卻且維持(在+/-1℃內或較佳)至20℃或更低(例如,18℃或更低、15℃或更低、12℃或更低、10℃或更低、7℃或更低、5℃或更低、4℃或更低、1℃與20℃之間、1℃至15℃之間、1℃至12℃之間、1℃至10℃之間、1℃至5℃之間、1℃至4℃之間等)的溫度。舉例而言,系統可包括整合式珀爾貼(Peltier)元件用以在處理期間維持材料之黏度。在一些實例中,整個微流體系統中的多個溫度區域可僅在囊封已成功之後控制基質材料之聚合。In general, microfluidic systems can control the temperature (and thus viscosity) of materials within one or more channels. As mentioned, channels carrying dissociated cells and unpolymerized matrix can be cooled and maintained (within +/- 1°C or preferably) to 20°C or lower (e.g., 18°C or lower, 15°C or Lower, 12°C or lower, 10°C or lower, 7°C or lower, 5°C or lower, 4°C or lower, between 1°C and 20°C, between 1°C and 15°C, 1 °C to 12 °C, 1 °C to 10 °C, 1 °C to 5 °C, 1 °C to 4 °C, etc.). For example, the system may include integrated Peltier elements to maintain the viscosity of the material during processing. In some examples, multiple temperature zones throughout the microfluidic system can control polymerization of the matrix material only after encapsulation has been successful.

舉例而言,本文中描述用於個人化癌症治療之精準藥物篩選的方法,該方法包含:接收來自患者腫瘤的單一組織樣本;使生檢組織樣本解離以形成解離之組織樣本;藉由以下操作自解離之組織樣本形成多於1000個患者衍生之微型類器官球體:驅動解離之組織樣本及未聚合流體基質材料穿過微流體設備之一或多個通道,其中微流體設備控制通道內的壓力、流動速率或壓力及流動速率且將解離之組織樣本及未聚合流體基質材料之溫度維持在20℃或更低的溫度,其中解離之組織樣本穿過層流之下的一或多個通道,將微流體設備內的解離之組織樣本及未聚合流體基質材料合併以形成複數個未聚合之混合物小滴;將未聚合之混合物小滴暴露於高於25℃之溫度以聚合流體基質材料且形成患者衍生之微型類器官球體,其中患者衍生之微型類器官球體各自具有50 µm與500 µm之間的直徑,其中分佈有1至200個解離細胞;將患者衍生之微型類器官球體培養2至14天以形成成熟的患者衍生之微型類器官球體,該成熟的患者衍生之微型類器官球體具有複製其等經生檢或切除的腫瘤之結構的結構化細胞群集;及藉由將成熟的患者衍生之微型類器官球體中之一或多者暴露於各藥物療法而並行地分析複數種藥物療法。For example, described herein is a method for precision drug screening for personalized cancer therapy comprising: receiving a single tissue sample from a patient's tumor; dissociated the biopsy tissue sample to form a dissociated tissue sample; by Formation of more than 1000 patient-derived micro-organoid spheroids from dissociated tissue samples: driving dissociated tissue samples and unpolymerized fluid matrix material through one or more channels of a microfluidic device, where the microfluidic device controls the pressure within the channel , flow rate or pressure and flow rate and maintaining the temperature of the dissociated tissue sample and the unpolymerized fluid matrix material at a temperature of 20° C. or lower, wherein the dissociated tissue sample passes through one or more channels under laminar flow, combining the dissociated tissue sample and unpolymerized fluid matrix material within the microfluidic device to form a plurality of unpolymerized mixture droplets; exposing the unpolymerized mixture droplets to a temperature above 25° C. to polymerize the fluid matrix material and form Patient-derived micro-organoid spheroids, wherein the patient-derived micro-organoid spheroids each have a diameter between 50 µm and 500 µm and have 1 to 200 dissociated cells distributed therein; the patient-derived micro-organoid spheroids are cultured for 2 to 14 to form mature patient-derived mini-organoid spheroids with structured cell clusters replicating the structure of their biopsied or resected tumors; and by deriving the mature patient-derived One or more of the miniature organoid spheroids were exposed to each drug therapy and multiple drug therapies were analyzed in parallel.

在本文中所描述的方法中之任一者中,可以任何適當的分析法分析患者衍生之微型類器官球體,包括但不限於:3D存活率分析、成像、分析基因及/或蛋白質表現(例如,DNA、RNA、蛋白質體學、胞外體等)。可對細胞、患者衍生之微型類器官球體、形成為患者衍生之微型類器官球體之部分的細胞外基質及/或培養患者衍生之微型類器官球體的上清液等進行分析。In any of the methods described herein, patient-derived micro-organoid spheroids can be analyzed by any suitable assay, including but not limited to: 3D viability analysis, imaging, analysis of gene and/or protein expression (e.g. , DNA, RNA, proteomics, exosomes, etc.). Cells, patient-derived micro-organoid spheroids, extracellular matrix forming part of the patient-derived micro-organoid spheroids, and/or supernatants from culturing the patient-derived micro-organoid spheroids, etc. can be assayed.

包括精準藥物篩選分析的本文中所描述的方法中之任一者可篩選以下一或多者:化學療法、靶向劑及免疫療法。舉例而言,一種用於個人化癌症療法之精準藥物篩選的方法可包括篩選包括化學療法、靶向劑及免疫療法中之一或多者的藥物療法。化學療法或化學治療劑係指用細胞生長抑制或細胞毒性劑(亦即化合物)治療以減少或消除非所要細胞(例如癌細胞)之生長或增殖。因此,如本文中所使用,化學療法或化學治療劑係指用於治療增生性病症(例如癌)的細胞毒性或細胞生長抑制劑。如本文中所使用,免疫療法或免疫治療劑通常促進免疫反應,例如該藥劑可為免疫刺激劑或免疫抑制劑之抑制劑(亦即抗免疫抑制劑)。術語因此包括免疫原性組合物及疫苗。免疫治療劑亦可包括檢查點阻斷劑或抑制劑、嵌合抗原受體(CAR)及授受T細胞療法。Any of the methods described herein, including precision drug screening assays, can screen for one or more of: chemotherapy, targeting agents, and immunotherapy. For example, a method for precision drug screening for personalized cancer therapy can include screening drug therapies that include one or more of chemotherapy, targeting agents, and immunotherapy. Chemotherapy or chemotherapeutic agents refer to treatment with cytostatic or cytotoxic agents (ie, compounds) to reduce or eliminate the growth or proliferation of unwanted cells, such as cancer cells. Thus, as used herein, chemotherapy or chemotherapeutic agents refer to cytotoxic or cytostatic agents used in the treatment of proliferative disorders such as cancer. As used herein, an immunotherapy or immunotherapeutic agent generally promotes an immune response, eg, the agent may be an immunostimulator or an inhibitor of an immunosuppressant (ie, an anti-immunosuppressant). The term thus includes immunogenic compositions as well as vaccines. Immunotherapeutics may also include checkpoint blockers or inhibitors, chimeric antigen receptors (CARs), and T cell therapy.

微流體設備可將全部或一部分微流體設備之溫度維持在經冷卻的目標溫度範圍(例如,約20℃或更低的,例如,約18℃或更低、約17℃或更低、約16℃或更低、約15℃或更低、約14℃或更低、約13℃或更低、約12℃或更低、約11℃或更低、約10℃或更低、約9℃或更低、約8℃或更低、約7℃或更低、約6℃或更低、約5℃或更低、約1℃與約20℃之間、約2℃與約15℃之間、約2℃與約10℃之間等)。微流體設備可包括例如用於冷卻的珀爾貼(Peltier)裝置。微流體設備可冷卻全部或一部分微流體設備。舉例而言,微流體設備可經結構設計以冷卻輸入樣本、導管及/或微流體設備晶片,以使得輸入材料(例如組織樣本)仍冷卻。未聚合流體基質材料亦可保持冷卻。微流體設備可包括可將來自冷卻源(例如,珀爾貼)之冷卻劑分配至此等區域中之任一者或所有的導熱材料(例如,鋁等)。微流體設備可包括用於感測此等區域中之溫度的一或多個熱感測器(例如,熱敏電阻等),且可包括使用感測器輸入調節溫度的控制回饋。The microfluidic device can maintain the temperature of all or a portion of the microfluidic device within a cooled target temperature range (e.g., about 20°C or less, e.g., about 18°C or less, about 17°C or less, about 16°C °C or lower, about 15 °C or lower, about 14 °C or lower, about 13 °C or lower, about 12 °C or lower, about 11 °C or lower, about 10 °C or lower, about 9 °C or lower, about 8°C or lower, about 7°C or lower, about 6°C or lower, about 5°C or lower, between about 1°C and about 20°C, between about 2°C and about 15°C between, between about 2°C and about 10°C, etc.). Microfluidic devices may include, for example, Peltier devices for cooling. The microfluidic device can cool all or a portion of the microfluidic device. For example, the microfluidic device can be structurally designed to cool the input sample, the catheter, and/or the microfluidic device wafer so that the input material (eg, tissue sample) remains cool. The unpolymerized fluid matrix material can also be kept cool. The microfluidic device can include a thermally conductive material (eg, aluminum, etc.) that can distribute coolant from a cooling source (eg, Peltier) to any or all of these regions. Microfluidic devices may include one or more thermal sensors (eg, thermistors, etc.) for sensing temperature in these regions, and may include control feedback to adjust temperature using sensor input.

組織樣本可為生檢樣本(包括經切除的),及/或可為細針抽出物,及/或循環的腫瘤細胞組織樣本。可急性地獲取(例如,在執行方法(例如,包括形成患者衍生之微型類器官球體的精準藥物篩選方法)的幾分鐘或數小時內獲取)組織樣本。舉例而言,組織樣本可為「新鮮的」且可在形成患者衍生之微型類器官球體的約12小時內、約8小時內、4小時、約3小時內、約2小時內、約1小時內等自患者獲取。組織樣本可低溫(例如,在約0℃與約20℃之間、約1℃與15℃之間、低於約15℃、低於10℃等)儲存。The tissue sample can be a biopsy sample (including resected), and/or can be a fine needle aspirate, and/or a tissue sample of circulating tumor cells. Tissue samples can be obtained acutely (eg, within minutes or hours of performing a method, eg, a precision drug screening method involving the formation of patient-derived micro-organoid spheroids). For example, the tissue sample can be "fresh" and can be within about 12 hours, within about 8 hours, 4 hours, within about 3 hours, within about 2 hours, within about 1 hour of formation of the patient-derived micro-organoid spheroids Nether is obtained from the patient. Tissue samples can be stored at low temperatures (eg, between about 0°C and about 20°C, between about 1°C and 15°C, below about 15°C, below 10°C, etc.).

方法可包括基於對患者衍生之微型類器官球體的反應表徵藥物療法之反應。形成患者衍生之微型類器官球體可進一步包含基於細胞數目及/或小滴大小分選患者衍生之微型類器官球體。舉例而言,形成患者衍生之微型類器官球體可包括光學分選患者衍生之微型類器官球體或基於細胞數目及/或小滴大小。可在聚合之前或之後進行分選。分選可包括導引流體流以基於其等分選特徵將特定PDMO沈積至特定倉(例如,腔室)中。The method can include characterizing the response to a drug therapy based on the response to the patient-derived miniature organoid spheroids. Forming the patient-derived micro-organoid spheroids may further comprise sorting the patient-derived micro-organoid spheroids based on cell number and/or droplet size. For example, forming the patient-derived micro-organoid spheroids can include optically sorting the patient-derived micro-organoid spheroids or based on cell number and/or droplet size. Sorting can be performed before or after polymerization. Sorting can include directing fluid flow to deposit specific PDMOs into specific bins (eg, chambers) based on their sorting characteristics.

分析可包括分析多於5種不同藥物療法(多於10種、多於15種、多於20種、多於30種、多於35種、多於40種、多於50種、多於75種、多於100種、多於150種、多於200種、多於250種、多於300種、多於500種、多於1000種等)。如本文中所使用,藥物療法可包括不同條件(濃度、載劑等)、或不同給藥方案(例如,重複劑量、給藥持續時間等)中的單一藥物及/或不同組合及/或條件及/或給藥方案中的多種藥物。舉例而言,不同藥物療法可包括一或多種藥物之不同濃度、三種或更多種藥物之不同組合、兩種或更多種藥物之不同比率、一或多種藥物的不同載劑及/或一或多種藥物的不同給藥時間。Analysis may include analysis of more than 5 different drug therapies (more than 10, more than 15, more than 20, more than 30, more than 35, more than 40, more than 50, more than 75 species, more than 100 species, more than 150 species, more than 200 species, more than 250 species, more than 300 species, more than 500 species, more than 1000 species, etc.). As used herein, drug therapy may include a single drug and/or different combinations and/or conditions in different conditions (concentration, vehicle, etc.), or in different dosing regimens (e.g., repeated doses, duration of administration, etc.) and/or multiple drugs in the dosing regimen. For example, different drug regimens may include different concentrations of one or more drugs, different combinations of three or more drugs, different ratios of two or more drugs, different carriers of one or more drugs, and/or a or different administration times of multiple drugs.

形成多於1000個患者衍生之微型類器官球體包含形成多於5000個患者衍生之微型類器官球體(多於10,000個等),如上文所提及。Forming more than 1000 patient-derived micro-organoid spheroids includes forming more than 5000 patient-derived micro-organoid spheroids (greater than 10,000 etc.), as mentioned above.

微流體系統可在形成小滴之前維持解離之組織樣本及未聚合流體基質材料之黏度。The microfluidic system can maintain the viscosity of the dissociated tissue sample and the unpolymerized fluid matrix material prior to the formation of droplets.

微流體系統可通常經結構設計以防止解離之組織樣本在一或多個通道內堵塞。舉例而言,微流體系統可經結構設計以藉由具有100 μm或更大之通道直徑來防止堵塞。微流體系統可經結構設計而維持一或多個通道內的近似恆定的壓力及/或流動速率。Microfluidic systems can typically be structurally designed to prevent clogging of dissociated tissue samples within one or more channels. For example, microfluidic systems can be engineered to prevent clogging by having channel diameters of 100 μm or greater. Microfluidic systems can be engineered to maintain an approximately constant pressure and/or flow rate within one or more channels.

將未聚合之混合物小滴暴露於高於25℃之溫度可包含將小滴暴露於30℃或更高(例如,31℃或更高、32℃或更高、33℃或更高、34℃或更高、35℃或更高、36℃或更高等)之溫度。Exposing the unpolymerized mixture droplets to a temperature above 25°C may comprise exposing the droplets to 30°C or higher (e.g., 31°C or higher, 32°C or higher, 33°C or higher, 34°C or higher, 35°C or higher, 36°C or higher, etc.).

將未聚合之混合物小滴暴露於高於25℃之溫度可包含使小滴流動至微流體設備之維持在高於25℃之溫度下的區域。在一些實例中,小滴可自微流體設備輸出,以便在經升溫之腔室中聚合。Exposing the unpolymerized mixture droplet to a temperature above 25°C may comprise flowing the droplet to a region of the microfluidic device maintained at a temperature above 25°C. In some examples, droplets can be output from a microfluidic device to polymerize in an elevated temperature chamber.

如所提及,微流體設備可在一或多個通道內維持恆定流動速率。As mentioned, a microfluidic device can maintain a constant flow rate within one or more channels.

本文中所描述之方法可包括量測各藥物療法對患者衍生之微型類器官球體內的細胞的作用以基於所量測之作用判定藥物療法是否將治療腫瘤。方法可包括藉由以下操作判定在一或多次投與藥物療法之後腫瘤仍對藥物療法有反應:在患者已用藥物療法治療之後接收第二患者腫瘤組織及自第二患者腫瘤組織形成第二複數個患者衍生之微型類器官球體,將第二複數個患者衍生之微型類器官球體中之至少一些暴露於藥物療法,及量測藥物療法對第二複數個患者衍生之微型類器官中之至少一些內的細胞的作用。The methods described herein can include measuring the effect of each drug therapy on cells within a patient-derived mini-organoid spheroid to determine whether the drug therapy will treat the tumor based on the measured effect. The method can include determining that the tumor remains responsive to the drug therapy after one or more administrations of the drug therapy by receiving a second patient tumor tissue after the patient has been treated with the drug therapy and forming a second patient tumor tissue from the second patient tumor tissue. The plurality of patient-derived micro-organoid spheroids, exposing at least some of the second plurality of patient-derived micro-organoid spheroids to a drug therapy, and measuring the effect of the drug therapy on at least some of the second plurality of patient-derived mini-organoids The role of some inner cells.

此等方法中之任一者可用於測試單一藥物療法,包括重複測試,如上文所描述。Any of these methods can be used to test monotherapy, including repeated testing, as described above.

方法可包括用來自複數種藥物療法的一種藥物療法治療患者。The method may comprise treating the patient with a drug therapy from a plurality of drug therapies.

接收腫瘤組織樣本與表徵反應之間的時間可少於21天(少於20天、少於19天、少於18天、少於17天、少於16天、少於15天等)。The time between receiving the tumor tissue sample and characterizing the response can be less than 21 days (less than 20 days, less than 19 days, less than 18 days, less than 17 days, less than 16 days, less than 15 days, etc.).

患者腫瘤組織可包含來自轉移性腫瘤的生檢樣本。一般而言,本文中所使用的腫瘤組織可為上皮腺癌組織(如本文中所提供之所有實例所見)。Patient tumor tissue may comprise a biopsy sample from a metastatic tumor. In general, tumor tissue as used herein may be epithelial adenocarcinoma tissue (as seen in all examples provided herein).

一般而言,此等方法可包括形成複數個微型類器官,包含形成具有小於25%的大小變化及/或細胞數目變化(例如,小於22%變化、小於20%變化、小於18%變化、小於15%變化、小於10%變化、小於6%變化等)的微型類器官。大小及/或細胞數目的較低變化可尤其有助於將不同藥物療法之間的反應歸一化。另外,可使用或比較多次。分析複數種藥物療法之步驟可包含分析具有類似大小及細胞數目的患者衍生之微型類器官球體。In general, such methods can include forming a plurality of mini-organoids, including forming mini-organoids with less than 25% variation in size and/or cell number variation (e.g., less than 22% variation, less than 20% variation, less than 18% variation, less than 15% variation, less than 10% variation, less than 6% variation, etc.) miniature organoids. Lower variation in size and/or cell number can be especially helpful in normalizing responses between different drug therapies. Also, multiple times can be used or compared. The step of analyzing the plurality of drug therapies may comprise analyzing patient-derived mini-organoid spheroids of similar size and cell number.

一般而言,患者衍生之微型類器官球體可重新塗鋪來自其等所來源之組織的組織結構,例如,其等可形成細胞之出芽群集及/或細胞之中空結構,該等細胞複製其等經生檢或切除的腫瘤之結構。In general, patient-derived micro-organoid spheroids can be recoated with tissue structures from the tissue from which they were derived, e.g., they can form sprouting clusters of cells and/or hollow structures of cells that replicate their The structure of the tumor after biopsy or resection.

舉例而言,一種用於個人化癌症療法之精準藥物篩選的方法,該方法可包含:接收來自患者腫瘤的單個生檢或經切除的組織樣本;使生檢組織樣本解離以形成解離之組織樣本;藉由以下操作自解離之組織樣本形成複數個患者衍生之微型類器官球體:驅動解離之組織樣本及未聚合流體基質材料穿過微流體設備之一或多個通道,其中微流體設備控制通道內的壓力、流動速率或壓力及流動速率且將解離之組織樣本及未聚合流體基質材料之溫度維持在20℃或更低的溫度,其中解離之組織樣本穿過層流之下的一或多個通道,將微流體設備內的解離之組織樣本及未聚合流體基質材料合併以形成複數個未聚合之混合物小滴;將未聚合之混合物小滴暴露於高於25℃之溫度以聚合流體基質材料且形成患者衍生之微型類器官球體,其中患者衍生之微型類器官球體各自具有50 µm與500 µm之間的直徑,其中分佈有1至200個解離之細胞,其中藉由大小、細胞的數目或大小及細胞的數目兩者分選患者衍生之微型類器官球體,將患者衍生之微型類器官球體培養2至14天以形成成熟的患者衍生之微型類器官球體,該成熟的患者衍生之微型類器官球體具有複製其等經生檢或切除的腫瘤之結構的結構化細胞群集;及藉由將各藥物療法暴露於成熟的患者衍生之微型類器官球體中之一或多者來測試藥物療法。For example, a method for precision drug screening for personalized cancer therapy, the method may comprise: receiving a single biopsy or resected tissue sample from a patient's tumor; dissociating the biopsy tissue sample to form a dissociated tissue sample ; forming a plurality of patient-derived micro-organoid spheroids from a dissociated tissue sample by driving the dissociated tissue sample and unpolymerized fluid matrix material through one or more channels of a microfluidic device, wherein the microfluidic device controls the channel pressure, flow rate or pressure and flow rate and maintain the temperature of the dissociated tissue sample and the unpolymerized fluid matrix material at a temperature of 20° C. or lower, wherein the dissociated tissue sample passes through one or more chambers under laminar flow channels for combining dissociated tissue samples and unpolymerized fluid matrix material within the microfluidic device to form a plurality of unpolymerized mixture droplets; exposing the unpolymerized mixture droplets to temperatures greater than 25°C to polymerize the fluid matrix material and form patient-derived micro-organoid spheroids, wherein the patient-derived micro-organoid spheroids each have a diameter between 50 µm and 500 µm, with 1 to 200 dissociated cells distributed therein, wherein by size, number of cells The patient-derived mini-organoid spheroids were sorted for both size and number of cells, and the patient-derived mini-organoid spheroids were cultured for 2 to 14 days to form mature patient-derived mini-organoid spheroids. Organoid spheroids have structured cell clusters that replicate the architecture of their biopsied or resected tumors; and drug therapies are tested by exposing each drug therapy to one or more of the mature patient-derived miniature organoid spheroids .

以引用之方式併入incorporated by reference

本說明書中所提及之所有公開案及專利申請案均以全文引用之方式併入本文中,其引用程度如同各個別公開案或專利申請案經特定及個別地指示以引用之方式併入一般。All publications and patent applications mentioned in this specification are herein incorporated by reference in their entirety to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. .

一般而言,本文中描述患者衍生之微型類器官球體、用於形成其等之方法及設備及使用其等例如分析組織(包括但不限於癌組織)反應之方法及設備。In general, described herein are patient-derived miniature organoid spheroids, methods and apparatus for forming them, and methods and apparatus for using them, for example, to analyze the response of tissues, including but not limited to cancerous tissues.

本文中所描述之患者衍生之微型類器官球體通常為自分佈於基底材料內的解離之初代細胞形成的球體。此等患者衍生之微型類器官球體(「PMOS」或「類器官球體」)可具有約50 µm與約500 µm之間的直徑(例如,約50 µm與約400 µm之間、約50 µm與約300 µm之間、約50 µm與約250 µm之間等),且可最初含有約1至1000個分佈於基底材料內的解離之初代細胞(例如,約1至750個、約1至500個、約1至400個、約1至300個、約1至200個、約1至150個、約1至100個、約1至75個、約1至50個、約1至40個、約1至30個、約1至20個等)。The patient-derived micro-organoid spheroids described herein are generally spheres formed from dissociated primary cells distributed within a substrate material. These patient-derived miniature organoid spheroids ("PMOS" or "organoid spheroids") can have a diameter between about 50 µm and about 500 µm (e.g., between about 50 µm and about 400 µm, between about 50 µm and between about 300 µm, between about 50 µm and about 250 µm, etc.), and may initially contain about 1 to 1000 dissociated primary cells (e.g., about 1 to 750, about 1 to 500 about 1 to 400, about 1 to 300, about 1 to 200, about 1 to 150, about 1 to 100, about 1 to 75, about 1 to 50, about 1 to 40, about 1 to 30, about 1 to 20, etc.).

出人意料地,儘管其等大小較小(通常介於約50 µm至250 µm)且細胞密度低(例如,通常少於每個微型類器官球體100個細胞),此等微型類器官球體可立即使用或培養極短暫的時間段(例如,14天或更短、10天或更短、7天或更短、5天或更短等)且可使得微型類器官球體內之細胞存活同時維持組織(包括其等經提取的腫瘤組織)之許多(若並非所有)特徵。微型類器官球體內的細胞之存活率顯著較高,且可經由多次繼代培養微型類器官球體數天(或數週),其中細胞將分裂、群集及形成與親本組織類似的結構。亦出人意料地,在一些變化形式中,微型類器官球體內的來自解離組織之細胞甚至在最小微型類器官球體內部形成形態結構;儘管在一些應用中,此類結構之存在對此等微型類器官球體(例如,可在出現實質性結構性重組之前使用其等)之效用並非必要的,但在一些變化形式中其等可尤其適用。Surprisingly, despite their small size (typically between about 50 µm and 250 µm) and low cell density (e.g., typically less than 100 cells per micro-organoid spheroid), these micro-organoid spheroids are ready-to-use Or cultured for a very short period of time (e.g., 14 days or less, 10 days or less, 7 days or less, 5 days or less, etc.) Including many, if not all, features of the extracted tumor tissue). The survival rate of the cells inside the micro-organoid spheroids is significantly higher, and the micro-organoid spheroids can be subcultured for several days (or weeks) through multiple subcultures, where the cells will divide, cluster and form structures similar to the parent tissue. Also surprisingly, in some variations, cells from dissociated tissue within micro-organoid spheroids form morphological structures inside even the smallest micro-organoid spheroids; The utility of organ spheroids (eg, they can be used before substantial structural reorganization occurs) is not essential, but in some variations they may be particularly useful.

用於形成及使用微型類器官球體的本文中所描述之方法及設備可用於自單個生檢產生許多(例如,大於10,000個)患者衍生之微型類器官球體。可使用此等微型類器官球體篩選藥物組合物,其可預測何種療法可有效地應用於獲取生檢的患者。此可例如適用於自健康正常組織及/或自癌(例如,腫瘤)組織毒性篩選藥物或其他化學組合物。特定而言,患者衍生之微型類器官球體、用於形成其等的方法及設備及用於測試其等的設備可用於篩選以在進行藥物療法之前鑑別可有效地治療患者(例如,癌症患者)的一或多種藥物組合物。此可允許在癌症患者將以其他方式進行數月可能不對其等有效的化學療法之前例如對癌症患者進行極快速篩選。The methods and apparatus described herein for forming and using miniature organoid spheroids can be used to generate many (eg, greater than 10,000) patient-derived miniature organoid spheroids from a single biopsy. Pharmaceutical compositions can be screened using these miniature organoid spheroids, which can predict which therapies can be effectively applied to patients who undergo biopsy. This can be applied, for example, to toxic screening of drugs or other chemical compositions from healthy normal tissue and/or from cancerous (eg, tumor) tissue. In particular, patient-derived micro-organoid spheroids, methods and devices for forming them, and devices for testing them can be used in screening to identify patients (e.g., cancer patients) who can be effectively treated prior to drug therapy one or more pharmaceutical compositions. This may allow, for example, very rapid screening of cancer patients before they would otherwise be subjected to chemotherapy for months which may not be equally effective.

因此,本文中描述使用單個患者特定的生檢(或其他適當的組織/細胞來源)的高通量藥物篩選方法(及用於執行此等方法的設備)。本文中描述小滴形成的患者衍生之微型類器官球體,其可自已解離且懸浮於基底基質(例如,基質膠)中的患者衍生之腫瘤樣本形成。微型類器官球體可圖案化至微流體微孔陣列上,以培育且給與藥物化合物。此微型化分析將腫瘤樣本之使用最大化,且能夠以每樣本顯著較低的成本自核心生檢篩選較多藥物化合物。Thus, described herein are high-throughput drug screening methods (and devices for performing such methods) using individual patient-specific biopsies (or other suitable tissue/cell sources). Described herein are droplet-forming patient-derived mini-organoid spheroids that can be formed from patient-derived tumor samples that have been dissociated and suspended in a base matrix (eg, Matrigel). Miniature organoid spheroids can be patterned onto microfluidic well arrays for incubation and administration of pharmaceutical compounds. This miniaturized assay maximizes the use of tumor samples and enables screening of more drug compounds from the core biopsy at a significantly lower cost per sample.

患者衍生之癌模型(PDMC) (諸如細胞株)、類器官及患者衍生之異種移植物(PDX)逐漸被接受為「標準」臨床前模型以促進新療法之鑑別及發展。舉例而言,由癌症患者衍生之細胞株及類器官之大規模藥物篩選已用於鑑別對大量潛在療法的敏感性。PDX亦用於預測藥物反應及鑑別新穎藥物組合。儘管經由研究此等不同的PDMC模型發展精準藥品策略,但對其等有效使用有實質性障壁。舉例而言,患者衍生之類器官(PDO)被認為在描繪患者腫瘤中最精確,此係由於研究已展示類器官之表現型及基因型剖析通常展示與原始患者腫瘤的高度相似性。令人遺憾地,至少兩個限制防礙使用PDO導引療法。首先,發展及測試類器官中的藥物敏感性會耗費數月,其降低臨床適用性。其次,自臨床上相關之18規格核心生檢獲得的類器官之數目不足以執行高通量藥物篩選。理想地,應在7至10天內自單個核心生檢執行分析。本文中所描述之微型類器官球體及製備及使用其等之方法可解決此等臨床限制。Patient-derived cancer models (PDMC) such as cell lines, organoids and patient-derived xenografts (PDX) are increasingly accepted as "standard" preclinical models to facilitate the identification and development of new therapies. For example, large-scale drug screens of cell lines and organoids derived from cancer patients have been used to identify sensitivities to a large number of potential therapies. PDX is also used to predict drug response and identify novel drug combinations. Despite the development of precision medicine strategies by studying these different PDMC models, there are substantial barriers to their effective use. For example, patient-derived organoids (PDOs) are considered the most accurate in delineating patient tumors because studies have shown that the phenotype and genotype profiles of organoids often exhibit a high degree of similarity to the original patient tumor. Unfortunately, at least two limitations prevent the use of PDO-guided therapy. First, developing and testing drug sensitivity in organoids can take months, which reduces clinical applicability. Second, the number of organoids obtained from clinically relevant 18-size core biopsies is insufficient to perform high-throughput drug screening. Ideally, the analysis should be performed from a single core biopsy within 7 to 10 days. The miniature organoid spheroids and methods of making and using them described herein can address these clinical limitations.

本發明所揭示之主題的一或多個實施例之細節闡述於此文檔中。對實施例的修改描述於此文檔中,且在研究此文檔中所提供的資訊之後,其他實施例將對一般熟習此項技術者為顯而易見的。此文檔中所提供的資訊及尤其描述的例示性實施例之特定細節主要為了理解之清晰度而提供,且不應理解為不必要的限制。在有衝突之情況下,將以此文檔之說明書(包括定義)為凖。The details of one or more embodiments of the presently disclosed subject matter are set forth in this document. Modifications to the embodiments are described in this document, and other embodiments will be apparent to those of ordinary skill in the art after studying the information provided in this document. The information provided in this document, and particularly specific details of the described exemplary embodiments, are provided primarily for clarity of understanding and should not be construed as unnecessary limitations. In case of conflict, the specification, including definitions, of this document will control.

雖然咸信本文中所使用之術語由一般熟習此項技術者充分理解,但本文中闡述定義以便於解釋本發明所揭示之主題。While the terms used herein are believed to be well understood by those of ordinary skill in the art, definitions are set forth herein in order to facilitate the explanation of the presently disclosed subject matter.

除非另外定義,否則本文中所使用之所有技術及科學術語具有與本發明所揭示之主題所屬領域之一般熟習此項技術者通常所理解相同之含義。儘管在本發明所揭示之主題之實踐或測試中可使用與本文中所描述之方法、裝置及材料類似或等效之任何方法、裝置及材料,但現描述代表性方法、裝置及材料。Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the presently disclosed subject matter belongs. Although any methods, devices and materials similar or equivalent to those described herein can be used in the practice or testing of the presently disclosed subject matter, representative methods, devices and materials are now described.

本文中使用術語「未聚合之混合物」以指代包含生物學相關材料的組合物,包括解離之組織樣本及第一流體基質材料。流體基質材料通常為可經聚合以形成其中分散有解離組織(細胞)的載劑或載劑網絡的材料。一旦聚合,則聚合材料可形成水凝膠且可由形成生物相容介質的蛋白質形成或及/或可包括形成生物相容介質的蛋白質,除細胞之外。根據本發明所揭示之主題使用的適合的生物相容介質可通常由為凝膠、半固體或液體(諸如室溫下(例如,25℃)的低黏度液體)的任何生物相容材料形成,且可用作細胞、組織、蛋白質及其他相關生物學材料的三維基底。根據本發明所揭示之主題可用於形成生物相容介質的例示性材料包括但不限於包含膠原蛋白、纖維蛋白、聚葡萄胺糖、MATRIGEL™ (BD Biosciences, San Jose, Calif.)、聚乙二醇、聚葡萄糖(包括化學可交聯或光可交聯的聚葡萄糖)及其類似者的聚合物及水凝膠,以及電紡生物學、合成或生物學合成的摻合物。在一些實施例中,生物相容介質由水凝膠構成。The term "unpolymerized mixture" is used herein to refer to a composition comprising biologically relevant material, including a dissociated tissue sample and a first fluid matrix material. A fluid matrix material is typically a material that can be polymerized to form a carrier or carrier network in which dissociated tissue (cells) are dispersed. Once polymerized, the polymeric material may form a hydrogel and may be formed from or and/or may include, in addition to cells, proteins that form a biocompatible medium. Suitable biocompatible media for use in accordance with the presently disclosed subject matter may generally be formed from any biocompatible material that is a gel, semi-solid, or liquid, such as a low-viscosity liquid at room temperature (e.g., 25° C.), And it can be used as a three-dimensional substrate for cells, tissues, proteins and other related biological materials. Exemplary materials that may be used to form biocompatible media in accordance with the presently disclosed subject matter include, but are not limited to, those comprising collagen, fibrin, polyglucosamine, MATRIGEL™ (BD Biosciences, San Jose, Calif.), polyethylene glycol Polymers and hydrogels of alcohols, polydextrose (including chemically or photocrosslinkable polydextrose) and the like, and electrospun biological, synthetic or biosynthetic blends. In some embodiments, the biocompatible medium is comprised of a hydrogel.

本文中使用術語「水凝膠」以指代包含聚合物鏈之三維網絡的兩種或多種組分的凝膠,其中水充當分散介質且填充聚合物鏈之間的空間。根據本發明所揭示之主題使用的水凝膠通常選擇用於基於結構之預期用途進行特定應用,考慮待使用以形成微型類器官球體的參數以及所選水凝膠將對併入至待置放於結構中的生物學懸浮液中的生物學材料(例如,細胞)之行為及活性的效果。本發明所揭示之主題的例示性水凝膠可由聚合材料構成,包括但不限於:海藻酸鹽、膠原蛋白(包括膠原蛋白I型及VI型)、彈性蛋白、角蛋白、纖維結合蛋白、蛋白聚糖、糖蛋白、聚乳酸交酯、聚乙二醇、聚己內酯、聚甘醇(polycolide)、聚二氧環己酮、聚丙烯酸酯、聚胺甲酸酯、聚碸、肽序列、蛋白質及衍生物、寡肽、明膠、彈性蛋白、纖維蛋白、層黏連蛋白、聚甲基丙烯酸酯、聚乙酸酯、聚酯、聚醯胺、聚碳酸酯、聚酸酐、聚胺基酸碳水化合物、多醣及修飾之多醣,及其衍生物及共聚物,以及無機材料,諸如玻璃(生物活性玻璃)、陶瓷、矽石、氧化鋁、方解石、羥基磷灰石、磷酸鈣、骨及前述所有之組合。The term "hydrogel" is used herein to refer to a gel of two or more components comprising a three-dimensional network of polymer chains, wherein water serves as the dispersion medium and fills the spaces between the polymer chains. Hydrogels used in accordance with the presently disclosed subject matter are generally selected for a particular application based on the intended use of the structure, considering the parameters to be used to form miniature organoid spheroids and the effect that the selected hydrogel will have on incorporation into the microspheres to be placed. Effects on the behavior and activity of biological material (eg, cells) in a biological suspension in a structure. Exemplary hydrogels of the presently disclosed subject matter can be composed of polymeric materials including, but not limited to: alginate, collagen (including collagen types I and VI), elastin, keratin, fibronectin, protein Glycans, glycoproteins, polylactides, polyethylene glycols, polycaprolactones, polyglycolides, polydioxanones, polyacrylates, polyurethanes, polyethylenes, peptide sequences , proteins and derivatives, oligopeptides, gelatin, elastin, fibrin, laminin, polymethacrylate, polyacetate, polyester, polyamide, polycarbonate, polyanhydride, polyamine Acid carbohydrates, polysaccharides and modified polysaccharides, their derivatives and copolymers, and inorganic materials such as glass (bioactive glass), ceramics, silica, alumina, calcite, hydroxyapatite, calcium phosphate, bone and All combinations of the foregoing.

另外關於用於產生本文中所描述之微型類器官球體的水凝膠,在一些實施例中,水凝膠係由選自以下所組成之群的材料構成:瓊脂糖、海藻酸鹽、膠原蛋白I型、聚氧乙烯-聚氧丙烯嵌段共聚物(例如,Pluronic® F127 (BASF Corporation, Mount Olive, N.J.))、聚矽氧、多醣、聚乙二醇及聚胺甲酸酯。在一些實施例中,水凝膠由海藻酸鹽構成。Additionally with respect to the hydrogels used to generate the micro-organoid spheroids described herein, in some embodiments, the hydrogels are composed of materials selected from the group consisting of: agarose, alginate, collagen Type I, polyoxyethylene-polyoxypropylene block copolymers (eg, Pluronic® F127 (BASF Corporation, Mount Olive, N.J.)), silicones, polysaccharides, polyethylene glycols, and polyurethanes. In some embodiments, the hydrogel is composed of alginate.

本文中所描述之微型類器官球體亦可包括生物學相關材料。片語「生物學相關材料」可描述能夠包括於如本文中所定義之生物相容介質中且隨後與生物系統相互作用及/或影響生物系統的材料。舉例而言,在一些實施中,生物學相關材料為磁性珠粒(亦即,本身為磁性的或含有對磁場起反應之材料的珠粒,諸如鐵粒子),該磁性珠粒可合併為未聚合材料之部分以產生可用於方法及組合物中(例如,用於微型類器官球體之分離及純化)的微型類器官球體。作為另一實例,在其他實施中,除解離之組織樣本(例如,生檢)材料之外,生物學相關材料可包括額外細胞。在未聚合之混合物中,解離組織樣本及額外生物學相關材料係在均勻混合物中或作為分佈混合物(例如,在微型類器官球體的剛好二分之一或其他部分上,包括剛好在核心中或剛好在形成的微型類器官球體之外部區域中)。在一些變化形式中,例如在聚合形成微型類器官球體的小滴之前,未聚合材料內的額外生物學相關材料可與解離之組織樣本懸浮於懸浮液中。The miniature organoid spheroids described herein can also include biologically relevant materials. The phrase "biologically relevant material" may describe a material capable of being included in a biocompatible medium as defined herein and subsequently interacting with and/or affecting a biological system. For example, in some implementations, the biologically relevant material is a magnetic bead (that is, a bead that is itself magnetic or contains a material that responds to a magnetic field, such as an iron particle), which can be incorporated into an Portions of the material are polymerized to produce micro-organoid spheroids that can be used in methods and compositions (eg, for the isolation and purification of micro-organoid spheroids). As another example, in other implementations, the biologically relevant material may include additional cells in addition to the dissociated tissue sample (eg, biopsy) material. In unpolymerized mixtures, the dissociated tissue sample and additional biologically relevant material are in a homogeneous mixture or as a distributed mixture (e.g., on just one half or other portion of a microorganoid spheroid, including just in the core or just in the outer region of the formed miniature organoid spheroids). In some variations, for example, additional biologically relevant material within the unpolymerized material may be suspended in suspension with the dissociated tissue sample prior to polymerization to form droplets of micro-organoid spheroids.

在一些變化形式中,可包括有解離之組織樣本(例如,生檢)材料的生物學相關材料可含有多個細胞類型,包括前脂肪細胞、間葉幹細胞(MSC)、內皮祖細胞、T細胞、B細胞、肥大細胞及脂肪組織巨噬細胞,以及在基質血管部分內發現的小血管或微血管片段。In some variations, biologically relevant materials, which can include dissociated tissue sample (e.g., biopsy) material, can contain multiple cell types, including preadipocytes, mesenchymal stem cells (MSCs), endothelial progenitor cells, T cells , B cells, mast cells, and adipose tissue macrophages, as well as small vessels or microvascular fragments found within the stromal vascular fraction.

一般而言,關於包括於本文中所描述之微型類器官球體中的解離之組織樣本(例如生檢)材料,此等組織可為來自患者的任何適當組織,通常自生檢獲取。儘管可使用非生檢組織,但一般而言,此等組織(及所得解離細胞)可為自如上文所描述之患者生檢(例如,藉由針生檢)獲取的初代細胞。組織可來自健康組織生檢或來自癌(例如,腫瘤)細胞生檢。可基於彼微型類器官球體之預期用途將解離細胞併入至本發明所揭示之主題的微型類器官球體中。舉例而言,相關組織(例如,解離之生檢組織)可通常包括通常在彼組織或器官(或腫瘤等)中發現的細胞。就此而言,可併入至本發明所揭示之主題的微型類器官球體中的例示性相關細胞包括神經元、心肌細胞、肌細胞、軟骨細胞、胰臟腺泡細胞、胰島(islets of Langerhans)、骨細胞、肝細胞、庫弗細胞(Kupffer cell)、纖維母細胞、肌母細胞、衛星細胞、內皮細胞、脂肪細胞、前脂肪細胞、膽上皮細胞及其類似者。可藉由此項技術中已知的習知技術解離此等類型之組織。適合生檢組織可衍生自:骨髓、皮膚軟骨、腱、骨、肌肉(包括心肌)、血管、角膜、神經、大腦、腸胃、腎臟、肝臟、胰臟(包括胰島細胞)、肺、垂體、甲狀腺、腎上腺、淋巴、唾液、卵巢、睪丸、子宮頸、膀胱、子宮內膜、前列腺、外陰及食道組織。可使用正常或病變(例如,癌)組織。在一些變化形式中,組織可自腫瘤組織,包括起源於此等正常組織中之任一者中的腫瘤產生。In general, with respect to dissociated tissue sample (eg, biopsy) material included in the miniature organoid spheroids described herein, such tissue may be any suitable tissue from a patient, typically obtained from a biopsy. Although non-biological tissues can be used, generally such tissues (and resulting dissociated cells) will be primary cells obtained from patient biopsies (eg, by needle biopsy) as described above. The tissue can be from a healthy tissue biopsy or from a cancer (eg, tumor) cell biopsy. Dissociated cells can be incorporated into the mini-organoid spheroids of the presently disclosed subject matter based on the intended use of that mini-organoid spheroid. For example, a tissue of interest (eg, dissociated biopsy tissue) can generally include cells normally found in that tissue or organ (or tumor, etc.). In this regard, exemplary related cells that can be incorporated into the miniature organoid spheroids of the presently disclosed subject matter include neurons, cardiomyocytes, myocytes, chondrocytes, pancreatic acinar cells, islets of Langerhans , bone cells, hepatocytes, Kupffer cells, fibroblasts, myoblasts, satellite cells, endothelial cells, adipocytes, preadipocytes, biliary epithelial cells, and the like. These types of tissues can be dissociated by conventional techniques known in the art. Suitable biopsy tissues can be derived from: bone marrow, skin cartilage, tendon, bone, muscle (including cardiac muscle), blood vessel, cornea, nerve, brain, stomach, kidney, liver, pancreas (including islet cells), lung, pituitary, thyroid , adrenal gland, lymph, saliva, ovary, testis, cervix, bladder, endometrium, prostate, vulva and esophagus tissue. Normal or diseased (eg, cancer) tissue can be used. In some variations, the tissue may arise from tumor tissue, including tumors originating in any of these normal tissues.

一旦形成,則可低溫保存及/或培養微型類器官球體。經培養的微型類器官球體可維持在懸浮液中,靜態(例如,在孔、瓶等中)或處於運動中(例如,滾動或攪動)。可使用已知培養技術培養微型類器官球體。例示性技術可見於以下各者以及其他地方:Freshney, Culture of Animal Cells, A Manual of Basic Techniques,第4版, Wiley Liss, John Wiley & Sons, 2000;Basic Cell Culture: A Practical Approach, Davis, 編, Oxford University Press, 2002;Animal Cell Culture: A Practical Approach,Masters, 編, 2000;及美國專利第5,516,681號及第5,559,022號。Once formed, the miniature organoid spheroids can be cryopreserved and/or cultured. Cultured micro-organoid spheroids can be maintained in suspension, static (eg, in wells, bottles, etc.) or in motion (eg, rolling or agitating). Micro-organoid spheroids can be grown using known culture techniques. Exemplary techniques can be found in, among other places: Freshney, Culture of Animal Cells, A Manual of Basic Techniques, 4th Edition, Wiley Liss, John Wiley & Sons, 2000; Basic Cell Culture: A Practical Approach, Davis, eds. , Oxford University Press, 2002; Animal Cell Culture: A Practical Approach, Masters, Ed., 2000; and US Patent Nos. 5,516,681 and 5,559,022.

在一些變化形式中,藉由在不混溶的材料(諸如流體疏水性材料(例如,油))中形成解離之組織樣本及流體基質材料的未聚合之混合物(例如,在一些變化形式中,冷卻混合物)小滴而形成微型類器官球體。舉例而言,可藉由將未聚合材料之流與不混溶的材料之一或多個流合併以形成小滴而形成微型類器官球體。可藉由稀釋未聚合材料中的解離材料(例如,細胞)測定存在於小滴中的細胞之密度。微型類器官球體之大小可與形成的小滴之大小相關。一般而言,微型類器官球體為具有穩定幾何形狀的球形結構。In some variations, by forming an unpolymerized mixture of a dissociated tissue sample and a fluid matrix material in an immiscible material such as a fluid hydrophobic material (e.g., oil) (e.g., in some variations, cooling mixture) droplets to form miniature organoid spheroids. For example, micro-organoid spheroids can be formed by combining a stream of unpolymerized material with one or more streams of immiscible material to form droplets. The density of cells present in a droplet can be determined by diluting the dissociated material (eg, cells) in the unpolymerized material. The size of the miniature organoid spheroids can be related to the size of the droplets formed. In general, micro-organoid spheroids are spherical structures with stable geometries.

除非另外指示,否則實施本發明所揭示之主題可採用細胞生物學、細胞培養、分子生物學、轉殖基因生物學、微生物學、重組DNA及免疫學之習知技術,此等習知技術在此項技術之技能範圍內。此類技術在文獻中已充分解釋。參見例如Molecular Cloning A Laboratory Manual (1989),第2版, Sambrook, Fritsch及Maniatis編, Cold Spring Harbor Laboratory Press, 第16及17章;美國專利第4,683,195號;DNA Cloning, 第I及II卷, Glover, 編, 1985;Oligonucleotide Synthesis, M.J.Gait, 編, 1984;Nucleic Acid Hybridization, D.Hames & S.J.Higgins, 編, 1984;Transcription and Translation, B.D.Hames & S.J.Higgins, 編, 1984;Culture Of Animal Cells, R.I.Freshney, Alan R.Liss, Inc., 1987;Immobilized Cells And Enzymes, IRL Press, 1986;Perbal (1984), A Practical Guide To Molecular Cloning;參見Methods In Enzymology (Academic Press, Inc., N.Y.);Gene Transfer Vectors For Mammalian Cells, J.H.Miller及M.P.Calos, 編, Cold Spring Harbor Laboratory, 1987;Methods In Enzymology, 第154及155卷, Wu等人, 編, Academic Press Inc., N.Y.;Immunochemical Methods In Cell And Molecular Biology (Mayer及Walker, 編, Academic Press, London, 1987);Handbook Of Experimental Immunology, 第I至IV卷, D.M.Weir及C.C.Blackwell, 編, 1986。Practice of the presently disclosed subject matter may employ, unless otherwise indicated, conventional techniques of cell biology, cell culture, molecular biology, transgenic biology, microbiology, recombinant DNA, and immunology, which are found in within the skill range of this technology. Such techniques are explained fully in the literature. See, e.g., Molecular Cloning A Laboratory Manual (1989), 2nd Edition, Eds. Sambrook, Fritsch and Maniatis, Cold Spring Harbor Laboratory Press, Chapters 16 and 17; U.S. Patent No. 4,683,195; DNA Cloning, Volumes I and II, Glover , ed., 1985; Oligonucleotide Synthesis, M.J.Gait, ed., 1984; Nucleic Acid Hybridization, D.Hames & S.J.Higgins, ed., 1984; Transcription and Translation, B.D.Hames & S.J.Higgins, ed., 1984; Culture Of Animal. Cells, R.I. Freshney, Alan R. Liss, Inc., 1987; Immobilized Cells And Enzymes, IRL Press, 1986; Perbal (1984), A Practical Guide To Molecular Cloning; see Methods In Enzymology (Academic Press, Inc., N.Y.); Gene Transfer Vectors For Mammalian Cells, J.H.Miller and M.P.Calos, eds., Cold Spring Harbor Laboratory, 1987; Methods In Enzymology, Volumes 154 and 155, Wu et al., eds., Academic Press Inc., N.Y.; Immunochemical Methods In Cell And Molecular Biology (Mayer and Walker, eds., Academic Press, London, 1987); Handbook Of Experimental Immunology, Volumes I to IV, D.M. Weir and C.C. Blackwell, eds., 1986.

如本文中所使用,藥物組合物可包括任何藥物、藥物稀釋、藥物調配物、包括多種藥物(例如,多種活性成分)之組合物、藥物調配物、藥物形式、藥物濃度、組合療法及其類似者。在一些變化形式中,藥物調配物係指包含藥物及一或多種非活性成分之混合物的調配物。As used herein, a pharmaceutical composition may include any drug, drug dilution, drug formulation, composition comprising multiple drugs (e.g., multiple active ingredients), drug formulation, drug form, drug concentration, combination therapy, and the like By. In some variations, a drug formulation refers to a formulation comprising a drug and a mixture of one or more inactive ingredients.

如此處所使用,術語「繼代」可指代微型類器官球體內的細胞的平均倍增數目。儘管傳統繼代數目係指細胞自一個培養容器至另一個的轉移或繼代培養,但微型類器官球體內的細胞可穩定保留在相同的微型類器官球體內,且可繼續生長及分裂。因此,如本文中所使用之繼代數目通常係指由微型類器官球體內的來自生檢組織的解離細胞進行的平均倍增數目。群體倍增數目為細胞群體自分離(例如,自由新鮮解離生檢組織形成微型類器官球體)已進行的大致倍增數目。一般而言,可相對於微型類器官球體內的一些或所有細胞的生長(例如倍增)培養本文中所描述之微型類器官球體較短時間段(例如,少於10次繼代、少於9次繼代、少於8次繼代、少於7次繼代、少於6次繼代、少於5次繼代、少於4次繼代、少於3次繼代等)。As used herein, the term "passaging" can refer to the average number of doublings of cells within a miniature organoid spheroid. While traditional passage numbers refer to the transfer or subculture of cells from one culture vessel to another, cells within micro-organoid spheroids are stably retained within the same micro-organoid spheroid and continue to grow and divide. Accordingly, passage number as used herein generally refers to the average number of doublings performed by dissociated cells from biopsy tissue within mini-organoid spheroids. The population doubling number is the approximate number of doublings a population of cells has undergone since isolation (eg, from freshly dissociated biopsy tissue to form miniature organoid spheroids). In general, the mini-organoid spheroids described herein can be cultured for relatively short periods of time (eg, less than 10 passages, less than 9 Second passage, less than 8 passages, less than 7 passages, less than 6 passages, less than 5 passages, less than 4 passages, less than 3 passages, etc.).

在培養期間,微型類器官球體內的來自解離生檢組織的細胞可聚集、群集或集合在微型類器官球體內。細胞之聚集體可經高度組織,且可形成所定義的形態或可為已群集或黏著在一起的細胞塊狀物。組織可反映來源組織。儘管在一些變化形式中微型類器官球體可含有單個細胞類型(同型),但更通常地此等微型類器官球體可含有多於一種細胞類型(異型)。Cells from dissociated biopsy tissue within the micro-organoid spheroid can aggregate, cluster, or collect within the micro-organoid spheroid during culture. Aggregates of cells may be highly organized and may form a defined morphology or may be clumps of cells that have clustered or stuck together. Organization can mirror the source organization. Although in some variations mini-organoid spheroids may contain a single cell type (homotype), more typically such mini-organoid spheroids may contain more than one cell type (heterotype).

如所提及,用於形成患者衍生之微型類器官球體(例如,解離組織)的(例如,生檢)組織可衍生自正常或健康生物組織,或衍生自罹患疾病或病痛的生物組織,諸如衍生自腫瘤的組織或流體。微型類器官球體中所使用的組織可包括免疫系統之細胞,諸如T淋巴細胞、B淋巴細胞、多形核白血球、巨噬細胞及樹突狀細胞。細胞可為幹細胞、祖細胞或體細胞。組織可為哺乳動物細胞,諸如人類細胞或來自動物(諸如小鼠、大鼠、兔及其類似者)的細胞。As mentioned, the (e.g., biopsy) tissue used to form the patient-derived mini-organoid spheroids (e.g., dissociated tissue) may be derived from normal or healthy biological tissue, or from biological tissue suffering from a disease or affliction, such as Tumor-derived tissue or fluid. The tissues used in the micro-organoid spheroids may include cells of the immune system, such as T lymphocytes, B lymphocytes, polymorphonuclear leukocytes, macrophages, and dendritic cells. Cells can be stem cells, progenitor cells or somatic cells. Tissues may be mammalian cells, such as human cells, or cells from animals such as mice, rats, rabbits, and the like.

一般而言,此等組織(及所得細胞)可通常自生檢獲取以形成微型類器官球體。因此,組織可衍生自生檢、手術試樣、抽吸、排出或含細胞流體中之任一者。適合的含細胞流體包括血液、淋巴液、皮脂流體、尿液、腦脊髓液或腹膜液中之任一者。舉例而言,在患有種植性轉移的患者中,卵巢或大腸癌細胞可自腹膜液分離。類似地,在患有子宮頸癌的患者中,可例如藉由大量切除轉換區域或藉由錐生檢自子宮頸獲取子宮頸癌細胞。通常,此類微型類器官球體將含有駐留在來源之組織或流體中的多個細胞類型。可在無繼代培養之中間步驟之情況下自個體直接獲得細胞,或其等可首先進行中間培養步驟以產生初代培養物。用於自含有生物組織及/或細胞的流體採集細胞之方法為此項技術中所熟知。舉例而言,用於自生物組織獲得細胞的技術包括由R.Mahesparan (Extracellular matrix -induced cell migration from glioblastoma biopsy specimens in vitro. Acta Neuropathol(1999) 97:231-239)描述的彼等技術。 In general, such tissues (and resulting cells) can be harvested typically from biopsies to form miniature organoid spheroids. Thus, tissue may be derived from any of a biopsy, surgical sample, aspiration, drain, or cell-containing fluid. Suitable cell-containing fluids include any of blood, lymph, sebum fluid, urine, cerebrospinal fluid, or peritoneal fluid. For example, in patients with implanted metastases, ovarian or colorectal cancer cells can be isolated from peritoneal fluid. Similarly, in patients with cervical cancer, cervical cancer cells can be harvested from the cervix, eg, by mass excision of the transition region or by cone biopsy. Typically, such miniature organoid spheroids will contain multiple cell types residing in the tissue or fluid of origin. Cells can be obtained directly from an individual without an intermediate step of subculturing, or they can first be subjected to an intermediate step of culturing to generate a primary culture. Methods for harvesting cells from fluids containing biological tissues and/or cells are well known in the art. For example, techniques for obtaining cells from biological tissues include those described by R. Mahesparan (Extracellular matrix-induced cell migration from glioblastoma biopsy specimens in vitro. Acta Neuropathol (1999) 97:231-239).

一般而言,在形成微型類器官球體之前細胞首先經解離或彼此分離。可藉由此項技術中已知的任何習知手段完成細胞之解離。較佳地,以機械方式及/或以化學方式處理細胞,諸如藉由用酶處理。藉由『以機械方式』,吾等包括例如使用刮刀或剪刀或藉由使用諸如均質機之機器破環相關細胞之間的連接之含義。藉由『以酶促方式』,吾等包括用破壞相關細胞之間的連接的一或多種酶處理細胞之含義,包括(例如)膠原蛋白酶、分散酶、DNA酶及/或玻尿酸酶中之任一者。一或多種酶可在不同反應條件下使用,諸如在37℃下在水浴中或在室溫下培育。Generally, cells are first dissociated or separated from each other prior to formation of miniature organoid spheroids. Dissociation of cells can be accomplished by any conventional means known in the art. Preferably, the cells are treated mechanically and/or chemically, such as by treatment with enzymes. By "mechanically" we include the meaning of disrupting the connections between the cells concerned, for example, using a scraper or scissors, or by using a machine such as a homogenizer. By "enzymatically" we include the meaning of treating cells with one or more enzymes that disrupt the junctions between the cells involved, including, for example, any of collagenase, dispase, DNase, and/or hyaluronidase. one. One or more enzymes can be used under different reaction conditions, such as incubation at 37°C in a water bath or at room temperature.

解離組織可經處理以移除死亡及/或垂死細胞及/或細胞碎片。可藉由熟習此項技術者已知的任何習知手段(例如,使用珠粒及/或抗體方法)完成此類死亡及/或垂死細胞之移除。舉例而言,已知磷脂醯絲胺酸在凋亡或死亡細胞中自內部質膜小葉重新分配至外部質膜小葉。使用磷脂結合蛋白V-生物素結合接著將該生物素結合至鏈黴抗生物素蛋白磁性珠粒使得凋亡細胞自活細胞分離。類似地,可藉由此項技術中任何適合的技術(包括(例如)過濾)來達成細胞碎片之移除。Dissociated tissue can be treated to remove dead and/or dying cells and/or cellular debris. Removal of such dead and/or dying cells can be accomplished by any conventional means known to those skilled in the art (eg, using beads and/or antibody methods). For example, phosphatidylserine is known to redistribute from the inner plasma membrane leaflet to the outer plasma membrane leaflet in apoptotic or dying cells. Apoptotic cells were isolated from viable cells using phospholipid binding protein V-biotin conjugation followed by conjugation of the biotin to streptavidin magnetic beads. Similarly, removal of cellular debris can be achieved by any suitable technique in the art, including, for example, filtration.

解離細胞在與流體基質材料合併之前可懸浮於載劑材料中,及/或流體基質材料可稱作載劑材料。在一些變化形式中,載劑材料可為在聚合及形成微型類器官球體之前具有延遲細胞在細胞懸浮液中沈降的黏度等級的材料。載劑材料可具有足夠的黏度以使得解離之生檢組織細胞在懸浮液中保持懸浮直至聚合。達成此所需之黏度可由熟練人員藉由監測不同黏度下的沈降速率及選擇一定黏度而優化,該選擇之黏度產生對於將細胞懸浮液負載至設備中與形成微型類器官球體(藉由將包括細胞的未聚合材料之小滴聚合而形成微類器官球體)之間的預期時間延遲而言適當的沈降速率。在一些變化形式中,即使使用較低黏度材料,未聚合材料仍可藉由設備流動或攪動,以便視需要將細胞保持在懸浮液中及/或保持細胞分佈。The dissociated cells may be suspended in a carrier material prior to combining with the fluid matrix material, and/or the fluid matrix material may be referred to as a carrier material. In some variations, the carrier material may be a material having a viscosity grade that retards the settling of cells in the cell suspension prior to polymerization and formation of micro-organoid spheroids. The carrier material may have sufficient viscosity to keep the dissociated biopsy cells suspended in suspension until polymerized. The viscosity required to achieve this can be optimized by the skilled person by monitoring the sedimentation rate at different viscosities and selecting a viscosity that yields a suitable viscosity for loading the cell suspension into the device and for forming miniature organoid spheroids (by including An appropriate settling rate in terms of the expected time delay between the aggregation of droplets of unpolymerized material of cells to form micro-organoid spheroids). In some variations, even with lower viscosity materials, the unpolymerized material can still be flowed or agitated by the device in order to keep cells in suspension and/or maintain cell distribution as desired.

如上文所提及,在一些變化形式中,未聚合混合物(包括解離之組織樣本及流體基質材料)可包括一或多種組分,例如生物學相關材料。舉例而言,可包括的生物學相關材料可包括以下任一者:細胞外基質蛋白質(例如,纖維結合蛋白)、藥物(例如,小分子)、肽或抗體(例如,以調節細胞存活、增殖或分化中之任一者);及/或特定細胞功能之抑制劑。可使用此類生物學相關材料,例如以藉由減少細胞死亡及/或降低細胞生長/複製之活化以提高細胞存活率或以其他方式模擬活體內環境。生物學相關材料可包括或可模擬以下組分中之一或多者:血清、介白素、趨化介素、生長因子、葡萄糖、生理鹽、胺基酸及激素。舉例而言,生物學相關材料可在流體基質材料中補充一或多種試劑。在一些變化形式中,流體基質材料為合成凝膠(水凝膠)且可藉由一或多種生物學相關材料補充。在一些變化形式中,流體基質為天然凝膠。因此,凝膠可由一或多種細胞外基質組分構成,諸如以下任一者:膠原蛋白、纖維蛋白原、層黏連蛋白、纖維結合蛋白、玻璃黏連蛋白、玻尿酸、纖維蛋白、海藻酸鹽、瓊脂糖及聚葡萄胺糖。舉例而言,基質膠包含對細胞存活率、增殖、發展及遷移重要的生物活性聚合物。舉例而言,基質材料可為包含膠原蛋白1型(諸如自大鼠尾部獲得的膠原蛋白1型)的凝膠。凝膠可為純膠原蛋白1型凝膠或可為含有除其他組分(諸如其他細胞外基質蛋白質)之外的膠原蛋白1型的凝膠。合成凝膠可指代自然界中並不天然地存在的凝膠。合成凝膠之實例包括衍生自以下任一者的凝膠:聚乙二醇(PEG)、甲基丙烯酸聚羥乙酯(PHEMA)、聚乙烯醇(PVA)、聚氧化乙烯(PEO)。 患者衍生之微型類器官球體 As mentioned above, in some variations, the unpolymerized mixture (including the dissociated tissue sample and the fluid matrix material) may include one or more components, such as biologically relevant materials. For example, biologically relevant materials that can be included can include any of the following: extracellular matrix proteins (e.g., fibronectin), drugs (e.g., small molecules), peptides, or antibodies (e.g., to regulate cell survival, proliferation or differentiation); and/or inhibitors of specific cellular functions. Such biologically relevant materials can be used, for example, to increase cell survival or otherwise mimic an in vivo environment by reducing cell death and/or reducing activation of cell growth/replication. Biologically relevant materials may include or may mimic one or more of the following components: serum, interleukins, chemokines, growth factors, glucose, physiological salts, amino acids, and hormones. For example, biologically relevant materials may be supplemented with one or more reagents in a fluid matrix material. In some variations, the fluid matrix material is a synthetic gel (hydrogel) and can be supplemented by one or more biologically relevant materials. In some variations, the fluid matrix is a natural gel. Thus, the gel may be composed of one or more extracellular matrix components, such as any of the following: collagen, fibrinogen, laminin, fibronectin, vitronectin, hyaluronic acid, fibrin, alginate , agarose and polyglucosamine. For example, Matrigel comprises bioactive polymers important for cell survival, proliferation, development and migration. For example, the matrix material may be a gel comprising collagen type 1, such as collagen type 1 obtained from a rat tail. The gel may be a pure collagen type 1 gel or may be a gel containing collagen type 1 among other components such as other extracellular matrix proteins. Synthetic gels may refer to gels that do not occur naturally in nature. Examples of synthetic gels include gels derived from any of the following: polyethylene glycol (PEG), polyhydroxyethyl methacrylate (PHEMA), polyvinyl alcohol (PVA), polyethylene oxide (PEO). Patient-derived micro-organoid spheroids

患者衍生之微型類器官球體之實例展示於圖1A至圖1C、圖2A至圖2C、圖3A至圖3C及圖4A至圖4E中。舉例而言,圖1A至圖1C說明所形成的微型類器官球體,每個微型類器官球體具有單個細胞。如所展示,微型類器官球體均為大致相同的大小,例如大致300 µm直徑。圖1B展示在培養3天之後在相同時間下形成的微型類器官球體。細胞在大小上已經擴展,在一些情況下,倍增及/或生長。藉由培養七天,如圖1C中所展示,細胞已倍增多次,展示細胞之群集或塊狀物。Examples of patient-derived micro-organoid spheroids are shown in Figures 1A-1C, 2A-2C, 3A-3C, and 4A-4E. By way of example, Figures 1A-1C illustrate the formation of micro-organoid spheroids, each micro-organoid spheroid having a single cell. As shown, the miniature organoid spheroids are all approximately the same size, eg approximately 300 µm diameter. Figure IB shows miniature organoid spheroids formed at the same time after 3 days of culture. Cells have expanded in size, and in some cases, doubled and/or grown. By seven days of culture, the cells had doubled multiple times, exhibiting clusters or clumps of cells, as shown in Figure 1C.

類似結果展示於圖2A至圖2C及圖3A至圖3C中,其等分別展示由每個微型類器官球體五個細胞或每個微型類器官球體20個細胞形成的微型類器官球體。在圖4A至圖4E中,微型類器官球體在形成之後立即示出,且培養五天,其中幾乎相同的微型類器官球體(例如,具有相同直徑)各自包括每個微型類器官球體10個細胞。在圖4A中,微型類器官球體在形成之後立即示出,在第0天仍由不混溶的流體(在此情況下,油)包圍。微型類器官球體自不混溶的流體移除且經洗滌,且培養五天。圖4B展示2天之後的微型類器官球體,圖4C展示3天之後的微型類器官球體,且圖4D及圖4E分別展示4天及5天的微型類器官球體。圖4A至圖4E展示微型類器官球體內的來自生檢的解離組織(細胞)具有活力且在幾乎所有微型類器官球體內以相當的速率生長。如此處將更詳細地描述,此等微型類器官球體可自單個平均大小的生檢大量形成,且可產生數百或數千個(例如,500個、750個、1000個、2000個、5000個、10,000個或更多個)包括大量活細胞的微型類器官球體,從而允許並行地執行多個快速分析。Similar results are shown in Figures 2A-2C and 3A-3C, which show micro-organoid spheroids formed from five cells per micro-organoid spheroid or 20 cells per micro-organoid spheroid, respectively. In FIGS. 4A-4E , mini-organoid spheroids are shown immediately after formation and cultured for five days, where nearly identical mini-organoid spheroids (e.g., with the same diameter) each included 10 cells per mini-organoid spheroid . In Figure 4A, miniature organoid spheroids are shown immediately after formation, still surrounded by immiscible fluid (in this case, oil) at day 0. The mini-organoid spheroids were removed from the immiscible fluid and washed, and cultured for five days. Figure 4B shows micro-organoid spheroids after 2 days, Figure 4C shows micro-organoid spheroids after 3 days, and Figure 4D and Figure 4E show micro-organoid spheroids at 4 days and 5 days, respectively. Figures 4A-4E demonstrate that dissociated tissue (cells) from biopsies within micro-organoid spheroids are viable and grow at comparable rates in almost all micro-organoid spheroids. As will be described in more detail herein, such miniature organoid spheroids can be formed in large numbers from a single average-sized biopsy, and hundreds or thousands (e.g., 500, 750, 1000, 2000, 5000 1, 10,000 or more) micro-organoid spheroids containing large numbers of living cells, allowing multiple rapid assays to be performed in parallel.

圖5A及圖5B說明如本文中所描述的自小鼠肝臟之解離生檢形成的微型類器官球體之實例,例如展示分佈於聚合流體基質材料(在此實例中,基質膠)內的小鼠肝細胞。各微型類器官球體包括形成為具有例如約300 µm之直徑的球體的聚合基質材料503,其中分散有設定數目的肝細胞507。在圖5A中,展示在生檢、解離及形成微型類器官球體之後一天的微型類器官球體。接著培養此等微型類器官10天,在此期間細胞(肝細胞)保持活力且生長,在多數情況下倍增多次以形成結構505,如圖5B中所展示。5A and 5B illustrate examples of miniature organoid spheroids formed from dissociated biopsies of mouse livers as described herein, such as showing mice distributed within a polymeric fluid matrix material (in this example, Matrigel). Hepatocyte. Each micro-organoid spheroid includes a polymeric matrix material 503 formed into a sphere having a diameter of, for example, about 300 µm, with a set number of hepatocytes 507 dispersed therein. In FIG. 5A , micro-organoid spheroids are shown one day after biopsy, dissociation, and formation of micro-organoid spheroids. These mini-organoids were then cultured for 10 days, during which time the cells (hepatocytes) remained viable and grew, in many cases doubling multiple times, to form structures 505, as shown in Figure 5B.

微型類器官球體可通常在微型類器官球體內包括呈固定或已知數目的細胞及/或濃度(個細胞/ml或細胞/mm 3)的解離(例如,生檢)組織(例如,細胞)。如上文所提及,此基質材料可為天然聚合物,諸如以下一或多者:海藻酸鹽、瓊脂糖、玻尿酸、膠原蛋白、明膠、纖維蛋白、彈性蛋白;或合成聚合物,諸如聚乙二醇(PEG)及聚丙烯醯胺中之一或多者。可使用有機及無機合成聚合物兩者。 Mini-organoid spheroids can typically include dissociated (e.g., biopsy) tissue (e.g., cells) at a fixed or known number and/or concentration (cells/ml or cells/mm 3 ) within a mini-organoid . As mentioned above, this matrix material can be a natural polymer, such as one or more of the following: alginate, agarose, hyaluronic acid, collagen, gelatin, fibrin, elastin; or a synthetic polymer, such as polyethylene One or more of glycol (PEG) and polyacrylamide. Both organic and inorganic synthetic polymers can be used.

在一些變化形式中,最初包括於微型類器官球體中的細胞之數目可選自1個細胞至多數百個。特定而言,在一些分析(例如,藥物毒性分析)中,包括約1至75個或約1至50個(例如,較低數目之細胞)可為有益的。每個微型類器官球體之細胞數目可由使用者設定或選擇。在一些變化形式中,如下文所描述,設備將包括一或多個控制件以設定來自初代組織的細胞數目以包括在各微型類器官球體中。可基於使用者欲如何使用微型類器官球體來選擇或設定細胞數目。舉例而言,具有極低細胞數目(例如,每個微型類器官球體1個細胞、每個微型類器官球體1至5個細胞等)的微型類器官球體可尤其適用於研究株系多樣性(例如,腫瘤異質性)。由於各微型類器官球體自單個細胞生長,因此吾等可觀測何種殖株具有藥物抗性且可檢驗(例如,藉由基因體定序)此等特定微型類器官球體以判定與特定株系有關的基因體(突變)多樣性。每個微型類器官球體有較低至適中數目的細胞(例如,約3至30個細胞、5至30個細胞、5至25個細胞、5至20個細胞、10至25個細胞等)可尤其適用於快速藥物測試,包括毒性測試,此係由於此等微型類器官球體通常快速生長。每個微型類器官球體有較大數目的細胞(例如,約20至100個細胞,例如,30至100個細胞、40至100個細胞、大於50個細胞等)可尤其適用於在各微型類器官球體中模擬組織組合物,此係由於微型類器官球體可含有不同譜系,潛在地包括上皮(或癌,等)及間葉(或基質、免疫、血管等)細胞。In some variations, the number of cells initially included in the miniature organoid spheroids can be selected from 1 cell up to several hundred. In particular, in some assays (eg, drug toxicity assays), it may be beneficial to include about 1 to 75 or about 1 to 50 (eg, lower numbers of cells). The number of cells per micro-organoid spheroid can be set or selected by the user. In some variations, as described below, the device will include one or more controls to set the number of cells from primary tissue to include in each micro-organoid spheroid. The number of cells can be selected or set based on how the user intends to use the micro-organoid spheroids. For example, mini-organoid spheroids with very low cell numbers (e.g., 1 cell per mini-organoid spheroid, 1 to 5 cells per mini-organoid spheroid, etc.) may be particularly useful for studying strain diversity ( For example, tumor heterogeneity). Since each mini-organoid is grown from a single cell, we can observe which strains are drug-resistant and can test (e.g., by genome sequencing) these specific mini-organoids to determine whether they are compatible with a particular strain. Associated gene body (mutation) diversity. A low to moderate number of cells (e.g., about 3 to 30 cells, 5 to 30 cells, 5 to 25 cells, 5 to 20 cells, 10 to 25 cells, etc.) per miniature organoid spheroid can It is especially suitable for rapid drug testing, including toxicity testing, since these miniature organoid spheroids usually grow rapidly. Larger numbers of cells per mini-organoid spheroid (e.g., about 20 to 100 cells, e.g., 30 to 100 cells, 40 to 100 cells, greater than 50 cells, etc.) Tissue composition is simulated in organoid spheroids, since miniature organoids can contain cells of different lineages, potentially including epithelial (or carcinoma, etc.) and mesenchymal (or stromal, immune, vascular, etc.) cells.

可以可匹配於待包括的細胞數目的任何適當大小形成微型類器官球體。舉例而言,大小可為直徑小至約20 µm,至多500 µm(例如,平均50 µm或100 µm,例如約100 µm至200 µm之間等)。在一些變化形式中,大小為約300 µm,其中約10至50個細胞(例如,約10至30個細胞)包括於各微型類器官球體中。細胞之數目及大小可變化及/或可受控制。在一些變化形式中,可藉由形成微型類器官球體的設備上的一或多個控制件設定細胞之數目及/或微型類器官球體之大小。舉例而言,可藉由調節解離之組織樣本(例如,來自生檢的細胞)之流動速率及/或濃度調節微型類器官球體之大小及/或微型類器官球體內的細胞之密度。Miniature organoid spheroids can be formed of any suitable size that can be matched to the number of cells to be included. For example, the size can be as small as about 20 µm in diameter, up to 500 µm (eg, on average 50 µm or 100 µm, eg, between about 100 µm and 200 µm, etc.). In some variations, the size is about 300 μm, with about 10 to 50 cells (eg, about 10 to 30 cells) included in each miniature organoid spheroid. The number and size of cells can be varied and/or controlled. In some variations, the number of cells and/or the size of the micro-organoid spheroids can be set by one or more controls on the apparatus for forming the micro-organoid spheroids. For example, the size of a micro-organoid spheroid and/or the density of cells within a micro-organoid spheroid can be adjusted by adjusting the flow rate and/or concentration of a dissociated tissue sample (eg, cells from a biopsy).

如圖1A至圖5B中所展示,即使在培養本文中所描述之微型類器官球體之後,亦允許有活力的及健康細胞穿過微型類器官球體之整個體積。可基於預期或意欲如何使用微型類器官球體中而選擇微型類器官球體之大小及/或包括於微型類器官球體中的細胞之數目。舉例而言,在微型類器官球體待用於檢驗生檢材料之細胞之間的關係的變化形式中,微型類器官球體可形成為具有多個細胞且可培養延長時段(例如,至多一週或更長時間)。As shown in Figures 1A-5B, even after culturing the mini-organoid spheroids described herein, viable and healthy cells were allowed to pass through the entire volume of the mini-organoid spheroids. The size of the micro-organoid spheroid and/or the number of cells included in the micro-organoid spheroid can be selected based on how the micro-organoid spheroid is intended or intended to be used. For example, in variations of micro-organoid spheroids to be used to examine relationships between cells of biopsy material, micro-organoid spheroids can be formed with multiple cells and can be cultured for extended periods of time (e.g., up to a week or more long time).

可藉由將解離之組織樣本(例如,生檢樣本)與可以受控方式聚合以形成微型類器官球體的流體基質合併製得本文中所描述之患者衍生之微型類器官球體。圖6說明一種形成患者衍生之微型類器官球體的方法。視情況而言,該方法可包括自患者獲取樣本,諸如自患者組織獲取生檢601。如上文所提及,可例如使用生檢針或穿孔獲取生檢。舉例而言,可用插入至患者組織中以移除生檢的14規格、16規格、18規格等的針而獲取生檢。在自患者移除組織之後,可以機械方式及/或以化學方式處理組織以解離材料。解離細胞可立即用於形成患者衍生之微型類器官球體,如所描述;在一些變化形式中,可諸如藉由基因修飾細胞603(例如,藉由轉染、電穿孔等)修飾所有或一些細胞。The patient-derived micro-organoid spheroids described herein can be prepared by combining a dissociated tissue sample (eg, a biopsy sample) with a fluid matrix that can polymerize in a controlled manner to form micro-organoid spheroids. Figure 6 illustrates a method for forming patient-derived micro-organoid spheroids. Optionally, the method may include obtaining a sample from the patient, such as obtaining a biopsy 601 from patient tissue. As mentioned above, a biopsy can be obtained, for example, using a biopsy needle or punch. For example, a biopsy may be obtained with a 14 gauge, 16 gauge, 18 gauge, etc. needle that is inserted into patient tissue to remove the biopsy. After the tissue is removed from the patient, the tissue may be treated mechanically and/or chemically to dissociate the material. The dissociated cells can be used immediately to form patient-derived mini-organoid spheroids, as described; in some variations, all or some of the cells can be modified, such as by genetically modifying the cells 603 (e.g., by transfection, electroporation, etc.) .

來自生檢材料的解離之組織樣本可與流體(例如,液體)基質材料合併以形成未聚合之混合物605。此未聚合之混合物可保持在未聚合狀態中,以使得來自解離組織的細胞可在混合物內保持懸浮。在一些變化形式中,可藉由例如在低於室溫下(例如,1℃至25℃之間)使其等冷卻而保持細胞懸浮及未聚合。The dissociated tissue sample from the biopsy material may be combined with a fluid (eg, liquid) matrix material to form an unpolymerized mixture 605 . This unpolymerized mixture can be maintained in an unpolymerized state so that cells from the dissociated tissue can remain suspended within the mixture. In some variations, cells can be kept in suspension and unaggregated, for example, by cooling them, eg, below room temperature (eg, between 1°C and 25°C).

未聚合之混合物可接著作為小滴以控制形成小滴之大小的方式施配至例如不混溶的材料(諸如油)中,且因此形成患者衍生之微型類器官球體之大小607。舉例而言,可藉由將未聚合材料之流合併至不混溶的材料(例如,油)之一或多個(例如,兩個會聚)流中形成均勻大小的小滴,以使得兩個流之流動速率及/或壓力可判定在其等與不混溶的材料相交時如何形成未聚合材料之小滴。可聚合小滴609以在不混溶的材料中形成患者衍生之微型類器官球體(PMOS)。在一些變化形式中,不混溶的材料可加熱或升溫至使未聚合之混合物(例如,未聚合材料中的流體基質材料)聚合的溫度。一旦形成,則患者衍生之微型類器官球體可自不混溶的流體分離,例如,PMOS可經洗滌以移除不混溶的流體611,且置放於培養介質中以允許患者衍生之微型類器官球體內的細胞生長。患者衍生之微型類器官球體可培養任何所需時間,或可立即低溫保存及/或分析。在一些變化形式中,患者衍生之微型類器官球體可培養短暫的時間段(例如,1至3天、1至4天、1至5天、1至6天、1至7天、1至8天、1至9天、1至10天、1至11天、1至14天等)。此可允許衍生自解離生檢組織的細胞生長及/或分裂(例如,倍增)至多五次或六次繼代。在培養之後,細胞可進行低溫保存615及/或分析617中的任一者或兩者。可使用的分析之實例亦描述於本文中。The unpolymerized mixture can then be dispensed as droplets into, for example, an immiscible material such as oil in a manner that controls the size of the formed droplets, and thus forms the size 607 of patient-derived micro-organoid spheroids. For example, uniformly sized droplets can be formed by combining a stream of unpolymerized material into one or more (e.g., two converging) streams of immiscible material (e.g., oil) such that both The flow rate and/or pressure of the stream can determine how droplets of unpolymerized material are formed when they intersect with immiscible materials. Droplets 609 can be polymerized to form patient-derived micro-organoid spheroids (PMOS) in an immiscible material. In some variations, the immiscible materials may be heated or raised to a temperature that polymerizes the unpolymerized mixture (eg, the fluid matrix material in the unpolymerized material). Once formed, patient-derived micro-organoid spheroids can be isolated from the immiscible fluid, e.g., PMOS can be washed to remove immiscible fluid 611, and placed in culture medium to allow patient-derived micro-organoid Cell growth within organ spheres. Patient-derived micro-organoid spheroids can be cultured for any desired time, or can be cryopreserved and/or analyzed immediately. In some variations, patient-derived mini-organoid spheroids can be cultured for brief periods of time (e.g., 1 to 3 days, 1 to 4 days, 1 to 5 days, 1 to 6 days, 1 to 7 days, 1 to 8 days). days, 1 to 9 days, 1 to 10 days, 1 to 11 days, 1 to 14 days, etc.). This can allow cells derived from the dissociated biopsy to grow and/or divide (eg, double) up to five or six passages. After culturing, the cells may be either or both cryopreserved 615 and/or analyzed 617 . Examples of assays that can be used are also described herein.

在本文中所描述之此等方法及設備中之任一者中,微型類器官球體可在聚合之後自不混溶的流體(例如,油)回收。舉例而言,在一些變化形式中,可藉由反乳化及/或去乳化回收微型類器官球體,例如藉由在形成小滴以移除任何油(及其他污染物)之後形成乳化小滴及回收微型類器官球體。此可允許細胞在不受不混溶的流體抑制之情況下在聚合小滴(微型類器官球體)內生長。In any of the methods and apparatus described herein, the micro-organoid spheroids can be recovered from an immiscible fluid (eg, oil) after polymerization. For example, in some variations, micro-organoid spheroids can be recovered by demulsification and/or de-emulsification, such as by forming emulsified droplets after forming droplets to remove any oil (and other contaminants) and Recover the miniature organoid spheroids. This can allow cells to grow within aggregated droplets (miniature organoid spheroids) without being inhibited by immiscible fluids.

儘管本文中所描述之方法及設備說明藉由使未聚合之混合物流動至不混溶的流體(諸如油或其他疏水性材料)之一或多個流中形成複數個小滴及因此複數個微型類器官球體之方法,但在一些變化形式中,可藉由允許如本文中所描述控制小滴大小的其他方法形成小滴。舉例而言,在一些變化形式中,可藉由列印形成小滴(例如,藉由將小滴列印至表面上)。此可減少或消除對乳化/去乳化之額外回收步驟的需要。舉例而言,小滴可列印至表面(諸如扁平或成型的表面)上且聚合。在此等變化形式中之任一者中,可使用壓力、音波、電荷等施配小滴。在一些變化形式中,可使用調適以將少量未聚合之混合物釋放至表面上、空氣中及/或液體介質(包括不混溶的流體)中的自動施配器(例如,移液裝置)形成小滴。Although the methods and apparatus described herein illustrate the formation of a plurality of droplets and thus a plurality of microscopic The method of organoid spheroids, but in some variations, droplets can be formed by other methods that allow control of droplet size as described herein. For example, in some variations, droplets may be formed by printing (eg, by printing droplets onto a surface). This can reduce or eliminate the need for an additional recovery step of emulsification/de-emulsification. For example, droplets can be printed onto a surface, such as a flat or shaped surface, and polymerized. In any of these variations, pressure, sound waves, electrical charges, etc. may be used to dispense the droplets. In some variations, automated dispensers (e.g., pipetting devices) adapted to release small amounts of unpolymerized mixture onto surfaces, into the air, and/or into liquid media (including immiscible fluids) can be used to form small drop.

用於形成患者衍生之微型類器官球體的方法可為自動的或使用一或多個設備執行。特定而言,可藉由允許選擇及/或控制患者衍生之微型類器官球體之大小(及因此細胞的數目之密度)的設備執行形成患者衍生之微型類器官球體的方法。舉例而言,圖7A說明用於形成如所描述之患者衍生之微型類器官球體的設備700的一個實例。Methods for forming patient-derived micro-organoid spheroids can be automated or performed using one or more devices. In particular, the method of forming patient-derived micro-organoid spheroids can be performed by a device that allows selection and/or control of the size (and thus the density of the number of cells) of the patient-derived micro-organoid spheroids. For example, FIG. 7A illustrates one example of an apparatus 700 for forming patient-derived micro-organoid spheroids as described.

在圖7A中,設備通常包括輸入端,其用於輸入解離之組織樣本的未聚合混合物及流體基質材料(已合併)或可單獨地接收解離之組織樣本(例如,在保持溶液中)及流體基質材料。在一些變化形式中,設備包括用於容納未聚合混合物的容納腔室706,及/或用於容納解離組織(例如,生檢)樣本及容納流體基質材料的容納腔室(未展示)。此等容納腔室中之任一者或所有可加壓以控制及/或加速流體自腔室流出及流至裝置中。設備可接收未聚合混合物或其可接收組分並將其混合。在一些變化形式中,設備可控制未聚合混合物中的細胞之濃度且可稀釋混合物(例如,藉由添加額外流體基質材料)以達成所需密度。舉例而言,設備可包括用於讀取未聚合混合物中的細胞之密度(例如,光學密度)的感測器(例如,光學讀取器) (未展示)。感測器亦可耦接至控制器724,其可自動地或半自動地(例如,藉由向使用者指示)控制未聚合混合物中的細胞之稀釋。設備亦可包括用於接收未聚合混合物的孔口。孔口可包括閥門或可耦接至閥門且閥門可由控制器724 (或各別控制器)控制。In FIG. 7A, the device generally includes an input for inputting an unpolymerized mixture of a dissociated tissue sample and a fluid matrix material (combined) or may receive a dissociated tissue sample (e.g., in a holding solution) and a fluid separately. matrix material. In some variations, the apparatus includes a holding chamber 706 for holding the unpolymerized mixture, and/or a holding chamber (not shown) for holding a dissociated tissue (eg, biopsy) sample and holding a fluid matrix material. Any or all of these containment chambers may be pressurized to control and/or accelerate fluid flow from the chamber and into the device. The equipment can receive the unpolymerized mixture or its receivable components and mix it. In some variations, the device can control the concentration of cells in the unpolymerized mixture and can dilute the mixture (eg, by adding additional fluid matrix material) to achieve a desired density. For example, an apparatus may include a sensor (eg, an optical reader) (not shown) for reading the density (eg, optical density) of cells in the unpolymerized mixture. The sensors can also be coupled to a controller 724, which can automatically or semi-automatically (eg, by indicating to the user) control the dilution of cells in the unaggregated mixture. The apparatus may also include an orifice for receiving the unpolymerized mixture. The orifices may include or be coupled to valves and the valves may be controlled by controller 724 (or a respective controller).

設備700可包括用於容納及/或接收不混溶的流體的腔室708及/或孔口。在一些變化形式中,不混溶的流體可容納在加壓腔室中以使得流動速率可受控制。加壓腔室中之任一者可由控制器724控制,該控制器可使用一或多個泵726控制壓力,且因此流體穿過設備。一或多個壓力及/或流動感測器可包括於系統中以監測穿過裝置的流體。Apparatus 700 may include a chamber 708 and/or an orifice for containing and/or receiving immiscible fluids. In some variations, immiscible fluids can be contained in a pressurized chamber such that the flow rate can be controlled. Either of the pressurized chambers may be controlled by a controller 724, which may use one or more pumps 726 to control the pressure, and thus the flow of fluid through the device. One or more pressure and/or flow sensors may be included in the system to monitor fluid passing through the device.

在圖7A中,整個設備700可包覆在殼體702中或設備的一部分704可包覆在殼體中。在一些變化形式中,殼體可包括例如用於添加不混溶的流體及/或未聚合混合物的位於裝置上的一或多個開口或入口部分。In FIG. 7A, the entire device 700 may be enclosed in a housing 702 or a portion 704 of the device may be enclosed in a housing. In some variations, the housing may include, for example, one or more openings or inlet portions on the device for adding immiscible fluid and/or unpolymerized mixture.

如所提及,此等設備700中之任一者亦可包括用於監測製造過程之所有或關鍵部分的一或多個感測器728。在一些變化形式中,感測器可包括光學感測器、機械感測器、電壓及/或電阻(或電容、或電感)感測器、力感測器等。此等感測器可用於監測組合件之正在進行的操作,包括患者衍生之微型類器官球體的形成。設備700亦可包括用於控制不混溶的流體及/或未聚合混合物(及/或流體基質材料)的任一者或兩者之溫度的一或多個熱/溫度調節器718。As mentioned, any of these apparatuses 700 may also include one or more sensors 728 for monitoring all or critical portions of the manufacturing process. In some variations, the sensors may include optical sensors, mechanical sensors, voltage and/or resistance (or capacitance, or inductance) sensors, force sensors, and the like. These sensors can be used to monitor the ongoing operation of the assembly, including the formation of patient-derived micro-organoid spheroids. Apparatus 700 may also include one or more heat/temperature regulators 718 for controlling the temperature of either or both of the immiscible fluid and/or the unpolymerized mixture (and/or the fluid matrix material).

此等設備中之任一者亦可包括如下文在圖7C及圖9中將說明的可(例如,使用一或多個感測器)監測一或多個小滴形成組合件720。小滴微型類器官球體形成組合件可包括分配器(例如PMOS分配器)722或可與分配器(例如PMOS分配器)722耦接。分配器噴嘴712可分配至例如多孔盤716中。Any of these apparatuses can also include one or more droplet forming assemblies 720 that can be monitored (eg, using one or more sensors) as will be explained below in FIGS. 7C and 9 . The droplet micro-organoid spheroid formation assembly can include or be coupled to a dispenser (eg, a PMOS dispenser) 722 . Distributor nozzle 712 may dispense into perforated disc 716, for example.

一般而言,小滴微型類器官球體形成組合件720可包括一或多個微流體晶片730或形成及控制未聚合混合物之流且形成實際小滴的結構。圖7B說明用於形成患者衍生之微型類器官球體的微流體晶片730的一個實例。在圖7B中,晶片730包括用於形成微型類器官球體的一對平行結構。圖7C說明用於形成PMOS之微流體晶片之小滴形成區域,包括未聚合通道出口741,其使「+」接合處或交叉區域737向通道出口741及不混溶的流體出口743、743'敞開(在此實例中,以直角形式)。在一些變化形式中,自不混溶的流體通道的輸入可處於相對於與未聚合材料之角度(及相交點)的一定角度。在圖7C中,如此說明書中展示尺寸的所有圖式中,展示的尺寸僅為例示性的,且並不意欲為限制性的,除非其以其他方式指明。In general, droplet micro-organoid spheroid formation assembly 720 may include one or more microfluidic chips 730 or structures that form and control the flow of unpolymerized mixture and form actual droplets. FIG. 7B illustrates an example of a microfluidic chip 730 used to form patient-derived micro-organoid spheroids. In FIG. 7B, wafer 730 includes a pair of parallel structures for forming miniature organoid spheroids. Figure 7C illustrates the droplet formation region of a microfluidic chip used to form a PMOS, including an unpolymerized channel outlet 741, which directs a "+" junction or intersection region 737 to the channel outlet 741 and immiscible fluid outlets 743, 743' open (in this example, at right angles). In some variations, the input from the immiscible fluid channel may be at an angle relative to the angle (and point of intersection) with the unpolymerized material. In FIG. 7C , in all figures in this specification where dimensions are shown, the dimensions shown are illustrative only and are not intended to be limiting unless otherwise indicated.

在圖7A中,微流體晶片730包括用於不混溶的流體(例如,自圖7A中所示的入口孔口或儲存腔室)進入晶片的入口(輸入孔口) 733。進入晶片的第二入口孔口735可經結構設計以接收未聚合材料且將其沿著半曲折的路徑輸送至接合區域。類似地,不混溶的流體的入口孔口可牢固地耦接至不混溶的流體腔室的出口或上文所描述之入口。In FIG. 7A, a microfluidic wafer 730 includes an inlet (input port) 733 for immiscible fluids to enter the wafer (eg, from an inlet port or storage chamber shown in FIG. 7A). The second inlet port 735 into the wafer can be structured to receive unpolymerized material and deliver it along a semi-tortuous path to the bonding area. Similarly, the immiscible fluid inlet port can be securely coupled to the outlet of the immiscible fluid chamber or the inlet described above.

未聚合材料進入晶片的入口孔口735可經由連接入口275的遞送路徑741耦接至接合區域(如圖7C中所展示)。類似地,不混溶的流體的入口733可將兩個(或更多)連接路徑743、743'連接至接合區域737。離開接合區域737的通道可使形成的微型類器官球體(在不混溶的流體中)沿該通道穿過達至出口731,該出口可連接至用於自微型類器官球體施配至一或多個腔室中例如用於培養及/或分析的施配器(未展示)。The inlet port 735 for unpolymerized material into the wafer can be coupled to the bonding region via a delivery path 741 connecting the inlet 275 (as shown in FIG. 7C ). Similarly, an inlet 733 for an immiscible fluid may connect two (or more) connection paths 743 , 743 ′ to the junction area 737 . A channel exiting the junction region 737 allows the formed micro-organoid spheroids (in an immiscible fluid) to pass along the channel to an outlet 731 which can be connected to a channel for dispensing from the micro-organoid spheroids to one or Dispensers (not shown) in multiple chambers, eg, for culture and/or analysis.

在圖7B及圖7C中所展示的實例中,一旦聚合可變為微型類器官球體的所形成的小滴可在自設備(未展示)施配之前沿著較長的溫度控制的微流體環境傳輸。舉例而言,圖8說明通道區域839 (例如,圖7B中的元件739)的一個實例,其繪示為透明的含有各自含有預定數目個細胞805的複數個微型類器官球體803。In the example shown in FIGS. 7B and 7C , the formed droplets that once aggregated can become miniature organoid spheroids can travel along a longer temperature-controlled microfluidic environment before being dispensed from the device (not shown). transmission. For example, FIG. 8 illustrates an example of a channel region 839 (eg, element 739 in FIG. 7B ), shown transparently containing a plurality of micro-organoid spheroids 803 each containing a predetermined number of cells 805 .

在圖8中,接合區域937如上文所描述成型,以使得攜載未聚合混合物的通道911與攜載與未聚合混合物不混溶的流體(諸如油)的一或多個(例如,兩個)通道909交叉。隨著未聚合混合物經加壓以第一速率流出第一通道911,交叉通道909、909'中流動的不混溶流體准許預定量的未聚合混合物在將其夾斷之前通過以形成傳遞至出口通道939中的小滴903。因此,在一些變化形式中,切碎(例如,解離)之臨床(例如,生檢或經切除的)組織樣本(諸如直徑<1 mm)可與溫度敏感性凝膠(亦即,基質膠,在4℃下)混合以形成未聚合之混合物。此未聚合混合物可置放於可產生體積及材料組成均勻的小滴(例如,油包水小滴)的微流體裝置中。同時,解離之腫瘤細胞可分割為此等小滴。未聚合材料中的凝膠可在加熱後(例如,在37℃下)凝固,且可形成所得到的患者衍生之微型類器官球體。在一些變化形式中,此方法可用於自組織(例如,生檢材料)產生超過10,000個(例如,超過20,000個、超過30,000個、超過40,000個、超過50,000個、超過60,000個、超過70,000個、超過80,000個、超過90,000個、超過100,000個)均勻小滴(患者衍生之微型類器官球體)。此等患者衍生之微型類器官球體與傳統3D細胞培養技術相容。圖10說明如上文所描述之複數個患者衍生之微型類器官球體1005,其懸浮於不混溶的材料1008 (例如,油)中。In FIG. 8 , junction region 937 is shaped as described above such that channel 911 carrying the unpolymerized mixture is in contact with one or more (eg, two) channels carrying a fluid immiscible with the unpolymerized mixture, such as oil. ) channel 909 crossed. As the unpolymerized mixture is pressurized to flow out of the first channel 911 at a first rate, the immiscible fluid flowing in the intersecting channels 909, 909' permits a predetermined amount of the unpolymerized mixture to pass before it is pinched off to form a pass to the outlet. Droplet 903 in channel 939. Thus, in some variations, minced (e.g., dissociated) clinical (e.g., biopsy or resected) tissue samples (such as <1 mm in diameter) can be combined with a temperature-sensitive gel (i.e., Matrigel, at 4°C) to form an unpolymerized mixture. This unpolymerized mixture can be placed in a microfluidic device that can generate droplets (eg, water-in-oil droplets) of uniform volume and material composition. At the same time, dissociated tumor cells can be divided into these droplets. The gel in the unpolymerized material can solidify upon heating (eg, at 37° C.) and the resulting patient-derived micro-organoid spheroids can form. In some variations, this method can be used to self-organize (e.g., biopsy material) to generate more than 10,000 (e.g., more than 20,000, more than 30,000, more than 40,000, more than 50,000, more than 60,000, more than 70,000, More than 80,000, more than 90,000, more than 100,000) uniform droplets (patient-derived micro organoid spheroids). These patient-derived micro-organoid spheroids are compatible with traditional 3D cell culture techniques. Figure 10 illustrates a plurality of patient-derived micro-organoid spheroids 1005 suspended in an immiscible material 1008 (eg, oil) as described above.

在上文說明的例示性微流體晶片中,接合處展示為T或X接合,其中微流體之流動聚焦形成可控制大小的微型類器官球體。在一些變化形式中,可藉由機器微移液例如至不混溶的流體中及/或固體或凝膠基底上形成小滴而非微流體晶片。可替代地在一些變化形式中,可藉由微毛細管生成以必需的尺寸及可再生性形成未聚合材料之小滴。可替代地用於自未聚合材料以指定大小範圍及可再生性形成微型類器官球體的技術之其他實例可包括膠體操控,例如經由外部力,諸如聲學、磁性、慣性、電濕潤或重力。In the exemplary microfluidic chips described above, the junctions are shown as T or X junctions, where the flow of microfluidics is focused to form micro organoid spheroids of controllable size. In some variations, droplets rather than microfluidic chips may be formed by robotic micropipetting, eg, into immiscible fluids and/or onto solid or gel substrates. Alternatively, in some variations, droplets of unpolymerized material can be formed with the requisite size and reproducibility by microcapillary generation. Other examples of alternative techniques for forming micro-organoid spheroids in a specified size range and reproducibility from unpolymerized materials may include colloidal manipulation, for example, via external forces such as acoustics, magnetism, inertia, electrowetting, or gravity.

圖11A及圖11B展示如上文所描述形成的於油中的患者衍生之微型類器官球體之實例。衍生自單個生檢樣本的此等患者衍生之微型類器官球體內的細胞具有活力,如由關鍵染料染色所見,如圖15A至15B及圖16A至圖16B中所展示。舉例而言,圖12A至圖12B說明具有腫瘤細胞(與圖11A至圖11B中所展示的彼等者類似)之微型類器官球體,其可經洗滌以移除不混溶的材料(例如,油)。在形成微型類器官球體之後可相對快速地移除此不混溶的材料,以便防止傷害微型類器官球體內的細胞。11A and 11B show examples of patient-derived mini-organoid spheroids in oil formed as described above. Cells within these patient-derived mini-organoid spheroids derived from a single biopsy sample were viable, as seen by key dye staining, as demonstrated in Figures 15A-15B and Figures 16A-16B. For example, FIGS. 12A-12B illustrate miniature organoid spheroids with tumor cells (similar to those shown in FIGS. 11A-11B ) that can be washed to remove immiscible material (e.g., Oil). This immiscible material can be removed relatively quickly after formation of the micro-organoid spheroids in order to prevent damage to the cells within the micro-organoid spheroids.

在此等實例中,凝膠小滴自油相回收且經由PFO (全氟辛醇)及離心再懸浮例如至PBS中。其可自微型類器官球體分離不混溶的流體。因此,此等微型類器官球體(包括基於腫瘤之微型類器官球體)可成功地生長,如上文圖1A至圖1C、圖2A至圖2C、圖3A至圖3C及圖4A至圖4E中及圖13中所展示。此為重要改良,因為必須對有活力的及生長的初代腫瘤細胞執行藥物篩選以預測患者結果,該初代腫瘤細胞保留其來自患者腫瘤之特性。此等微型類器官球體之較高數目及均勻性使得篩選有可能且可靠,如下文將描述。In these examples, gel droplets were recovered from the oil phase and resuspended via PFO (perfluorooctyl alcohol) and centrifugation eg into PBS. It can separate immiscible fluids from miniature organoid spheroids. Accordingly, these mini-organoid spheroids, including tumor-based mini-organoid spheroids, can be grown successfully, as in Figures 1A-1C, 2A-2C, 3A-3C, and 4A-4E above and shown in Figure 13. This is an important improvement because drug screening must be performed on viable and growing primary tumor cells, which retain their properties from the patient's tumor, to predict patient outcome. The higher number and uniformity of these mini-organoid spheroids made screening possible and reliable, as will be described below.

在本文中所描述之微流體晶片或裝置中之任一者中,通道可經塗佈。舉例而言,微流體裝置之通道可塗佈有疏水性材料。In any of the microfluidic chips or devices described herein, the channels can be coated. For example, channels of microfluidic devices can be coated with hydrophobic materials.

一般而言,本文中所描述之微型類器官球體的直徑高度均勻,且可具有極小的尺寸,例如直徑、差異。此例如在圖14中說明,其展示小滴直徑大小的一個實例的分佈。In general, the micro-organoid spheroids described herein are highly uniform in diameter and can have extremely small dimensions, eg, diameter, variance. This is illustrated, for example, in Figure 14, which shows the distribution of one example of droplet diameter sizes.

如所提及,圖15A至15B展示如本文中所描述形成的微型類器官球體;在圖16A至16B中,此等微型類器官球體已用展示其等存活的錐蟲藍(箭頭)染色。以此方式形成為小滴的微型類器官球體可含有生長因子及基質以模擬組織出現的生物學環境。患者樣本(例如,生檢樣本)可在獲取組織數小時內形成為微型類器官球體(包括數百、數千或數萬之微型類器官球體)。微型類器官球體可具有少至每個微型類器官球體有1個或4至6個細胞(例如,當對腫瘤取樣時為癌細胞)或多達數百個細胞。此等方法已表明對目前為止測試的(n=20)幾乎所有類型之癌組織及非癌症組織起作用,包括結腸、食道、黑色素瘤、子宮、肉瘤、腎臟、肝臟、卵巢、肺、隔膜、腸網膜、縱隔肺及乳癌組織。微型類器官球體可培養任何所需時間段,且通常展示少至3至4天的增殖及生長。其等可維持及繼代數月。如下文將更詳細地描述,其等可用於在自獲取組織(例如,生檢)的少至4至6天內篩選數千種之藥物組合物。As mentioned, Figures 15A-15B show mini-organoid spheroids formed as described herein; in Figures 16A-16B, these mini-organoid spheroids have been stained with trypan blue (arrows) showing their survival. Microorganoid spheroids formed into droplets in this way can contain growth factors and a matrix to mimic the biological environment in which the tissue emerges. Patient samples (eg, biopsy samples) can be formed into micro-organoid spheroids (including hundreds, thousands, or tens of thousands of micro-organoid spheroids) within hours of tissue harvesting. Micro-organoid spheroids can have as few as 1 or 4 to 6 cells (eg, cancer cells when sampling a tumor) or as many as several hundred cells per micro-organoid spheroid. These methods have been shown to work on nearly all types of cancerous and non-cancerous tissue tested so far (n=20), including colon, esophagus, melanoma, uterus, sarcoma, kidney, liver, ovary, lung, diaphragm, Omentum, mediastinal lung and breast cancer tissues. Miniature organoid spheroids can be cultured for any desired period of time and typically exhibit proliferation and growth in as little as 3 to 4 days. They can be maintained and subcultured for several months. As will be described in more detail below, they can be used to screen thousands of pharmaceutical compositions in as little as 4 to 6 days from harvesting tissue (eg, biopsy).

可在本文中所描述之微型類器官球體形成之後的任何時刻例如藉由將其等低溫保存來儲備。可自許多不同患者收集腫瘤微型類器官球體且可個別地或共同地使用其等以篩選多種藥物調配物以測定毒性及/或功效。可並行地生檢、儲備及/或篩選非腫瘤細胞(健康組織)。因此,此等方法及設備可允許高通量篩選。在一些變化形式中,可形成微型類器官球體且使其繼代兩次(例如,兩次倍增)及低溫保存。如所提及,可使用正常的健康組織以形成此等相同微型類器官球體以產生數百、數千或數萬之可用於分析藥物效果、藥物反應、生物標記、蛋白質體信號、基因體信號等的微型類器官球體。The micro-organoid spheroids described herein can be stored at any time after formation, for example by cryopreserving them. Tumor mini-organoid spheroids can be collected from many different patients and used individually or collectively to screen various drug formulations for toxicity and/or efficacy. Non-tumor cells (healthy tissue) can be biopsied, stockpiled and/or screened in parallel. Accordingly, such methods and devices may allow high throughput screening. In some variations, miniature organoid spheroids can be formed and passaged twice (eg, doubled twice) and cryopreserved. As mentioned, normal healthy tissue can be used to form these same miniature organoid spheroids to generate hundreds, thousands or tens of thousands of spheroids that can be used to analyze drug effects, drug responses, biomarkers, proteosome signatures, gene body signatures. and other miniature organoid spheroids.

尤其重要的是,此等微型類器官球體以生物學顯著方式存活,使得其等提供臨床上及生理學上相關資料,尤其在藥物反應方面,如圖22A至22D及圖23A至圖23D中將描述。特定而言,本文中所描述之微型類器官球體准許組織提取/生檢來源的細胞格外充分地生長且提供較多代表性資料,尤其相較於類器官或球體。在不受特定理論束縛之情況下,此可能係因為細胞可能在微型類器官球體中具有較多受限制的細胞密度,由此准許細胞在無抑制之情況下彼此通信同時共用信號。微型類器官球體亦具有極大表面積與體積之比,從而更易於准許傳輸生長因子及其他信號以滲入至微型類器官球體中(例如,微型類器官球體受到較少的擴散限制)。 分析 Importantly, these miniature organoid spheroids survived in a biologically significant manner, making them clinically and physiologically relevant, especially in terms of drug response, as shown in Figures 22A to 22D and Figures 23A to 23D describe. In particular, the miniature organoid spheroids described herein allow tissue extraction/biopsy-derived cells to grow exceptionally well and provide more representative data, especially compared to organoids or spheroids. Without being bound by a particular theory, this may be because the cells may have a more restricted cell density in the miniature organoid spheroids, thereby allowing the cells to communicate with each other while sharing signals without inhibition. Micro-organoid spheroids also have an extremely large surface area-to-volume ratio, which more easily permits the transmission of growth factors and other signals for infiltration into the micro-organoid spheroids (eg, the micro-organoid spheroids are less limited by diffusion). analyze

本文中所描述之患者衍生之微型類器官球體可用於各種不同分析中,且尤其可用於在正常及/或異常(例如,癌)組織上判定藥物調配物作用(包括毒性)。舉例而言,藥物篩選可包括將微型類器官球體塗覆至多孔(例如,96孔)盤之所有或一些孔中。可替代地,可使用定製盤(例如,可由100×100個孔形成的10,000微孔陣列)。微型類器官球體(例如,凝膠小滴)可塗覆至多個微孔陣列中或在一些變化形式中塗覆至多個微孔陣列上,且用培養基培育。微型類器官球體可培養3至5天的時程。在一些變化形式中,在第5天,該等孔(例如,微型反應器)可接著用藥物化合物(例如基於一組FDA批准的抗癌藥物)給藥,以檢查藥物集合之作用。舉例而言,測試的藥物可基於國家癌症研究所(癌症治療及診斷分部(Division of Cancer Treatment and Diagnosis) (11))篩選,由意欲實現癌症研究、藥物發現及組合藥物研究的147種藥劑組成。在第7天,微型類器官球體可經由標準螢光顯微法成像且基於藥物反應評級。The patient-derived mini-organoid spheroids described herein can be used in a variety of different assays, and are particularly useful for determining drug formulation effects, including toxicity, on normal and/or abnormal (eg, cancer) tissue. For example, drug screening can include coating mini-organoid spheroids into all or some wells of a multi-well (eg, 96-well) plate. Alternatively, a custom plate can be used (eg, a 10,000 microwell array that can be formed from 100 x 100 wells). Miniature organoid spheroids (eg, gel droplets) can be coated into or, in some variations, onto multiple microwell arrays and incubated with culture medium. Miniature organoid spheroids can be cultured for a time course of 3 to 5 days. In some variations, at day 5, the wells (eg, microreactors) can then be dosed with drug compounds (eg, based on a panel of FDA-approved anticancer drugs) to examine the effect of drug pools. For example, the drugs tested can be based on the National Cancer Institute (Division of Cancer Treatment and Diagnosis (11)) screen of 147 agents intended to enable cancer research, drug discovery, and combination drug research composition. At day 7, the mini-organoid spheroids can be imaged via standard fluorescence microscopy and graded based on drug response.

此分析技術之實例展示在圖17A至圖17E中。An example of this analytical technique is shown in Figures 17A-17E.

在此實例中, 篩選分析可為自動的。此可實現可重複及自動的工作流程,其可將篩選的藥物數目自少許增加至數百種。圖17A至圖17E說明此工作流程的一個實例。在圖17A中,獲取腫瘤生檢且如上文所描述形成複數個(例如,>10,000)微型類器官球體(在圖17A中,說明形成微型類器官球體的接合區域)。之後,微型類器官球體可回收及洗滌(例如,以移除於其中形成其等的不混溶的(例如,油)材料)。微型類器官球體可接著塗鋪至一或多個微孔盤中。如圖17C中所展示,微型類器官球體可培養一或多個世代(例如,一或多個繼代)。顯示其在第0天至第3天、第4天或第5天出現。之後,可例如藉由將藥物塗覆至一子組的複製品孔而篩選微型類器官球體,如圖17D中所展示。之後,如圖17E中所展示,在第7天,微型類器官球體中的細胞可成像及/或自動地或手動對其記分以鑑別藥物效果(例如,藥物篩選及生長剖析)。 In this example, screening analysis can be automated. This enables a reproducible and automated workflow that can increase the number of drugs screened from a few to hundreds. Figures 17A-17E illustrate an example of this workflow. In FIG. 17A , tumor biopsies were taken and a plurality (eg, >10,000) of mini-organoid spheroids formed as described above (in FIG. 17A , junctional regions forming mini-organoid spheroids are illustrated). Thereafter, the micro-organoid spheroids can be recovered and washed (eg, to remove immiscible (eg, oil) material forming them therein). The micro-organoid spheroids can then be plated into one or more microwell plates. As shown in Figure 17C, mini-organoid spheroids can be cultured for one or more passages (eg, one or more passages). It is shown to appear on day 0 to day 3, day 4 or day 5. The mini-organoid spheroids can then be screened, for example, by coating a drug to a subset of replica wells, as shown in Figure 17D. Thereafter, as shown in FIG. 17E , at day 7, the cells in the mini-organoid spheroids can be imaged and/or scored automatically or manually to identify drug effects (eg, drug screening and growth profiling).

圖17A至圖17E中展示的工作流程可使得整合式裝置能夠用於生長、給藥及/或檢查微型類器官球體。在一個例示性裝置中,可用試劑將新鮮生檢或切除的患者腫瘤樣本分離及接種至凝膠中以形成微型類器官球體(如上文所描述)。形成的微型類器官球體之一部分可低溫保存。其餘可回收及培育直至接種至微孔盤中,以用於如剛剛所描述的藥物測試或篩選。可對可成像及追蹤的微型類器官球體執行生長及存活率分析。可量測其等對藥物治療的反應,諸如IC-50、細胞毒性及生長曲線,以鑑別針對患者的腫瘤的有效療法。The workflow shown in Figures 17A-17E can enable an integrated device for growing, dosing, and/or examining micro-organoid spheroids. In one exemplary setup, fresh biopsied or resected patient tumor samples can be isolated and seeded into gels with reagents to form miniature organoid spheroids (as described above). Parts of the resulting miniature organoid spheroids can be cryopreserved. The remainder can be recovered and grown until inoculated into microwell plates for drug testing or screening as just described. Growth and viability assays can be performed on imageable and trackable miniature organoid spheroids. Their responses to drug treatments, such as IC-50, cytotoxicity, and growth curves, can be measured to identify effective therapies for a patient's tumor.

本文中所描述之方法及設備具有多種優勢,包括可再生性。樣本製造製程可藉由微流體樣本分割而自動化,其可減少對診斷測試及手動移液的專業人員的需要。此可尤其對臨床配置有幫助。此外,其可實現信號小滴之間的均勻性,從而增加分析敏感性。另外,此等分析可將產生微型類器官球體所需要的時間減至最少。基於初步資料,此等方法可能夠在少於約15分鐘內產生超過100,000個基質膠腫瘤小滴(微型類器官球體)之庫。此等方法亦為高度可延展的,且可經多工以並行地運行多個患者生檢。The methods and apparatus described herein have various advantages, including reproducibility. The sample fabrication process can be automated through microfluidic sample segmentation, which can reduce the need for specialists in diagnostic testing and manual pipetting. This may be especially helpful in clinical settings. In addition, it enables uniformity of signal droplet to increase assay sensitivity. In addition, these assays minimize the time required to generate miniature organoid spheroids. Based on preliminary data, these methods may be capable of generating a library of over 100,000 matrigel tumor droplets (miniature organoid spheroids) in less than about 15 minutes. These methods are also highly scalable and can be multiplexed to run multiple patient biopsies in parallel.

最終,此等方法為靈活的且與其他技術相容。作為研究工具,小滴類微流體通常與廣泛範圍的水凝膠材料(諸如瓊脂糖、海藻酸鹽、PEG及玻尿酸)相容。因此,起始凝膠組合物可易於經修飾以伴隨及促進微型類器官球體生長。此外,可藉由修改此等微流體裝置之大小來調節小滴大小。同時,此等允許凝膠材料組合物及微反應器大小之大量選擇。Ultimately, these methods are flexible and compatible with other techniques. As a research tool, droplet-like microfluidics are generally compatible with a wide range of hydrogel materials such as agarose, alginate, PEG, and hyaluronic acid. Thus, the starting gel composition can be readily modified to accompany and facilitate the growth of miniature organoid spheroids. Furthermore, droplet size can be tuned by modifying the size of these microfluidic devices. At the same time, these allow a large choice of gel material composition and microreactor size.

本文所描述之微型化分析(例如使用微型類器官球體)可使患者腫瘤生檢達至最大,使得能夠篩選較多藥物化合物。舉例而言,600 uL腫瘤樣本可分割至體積為約4 nL的約143,000個個別微型反應器中。藉由將組織樣本最大化,可檢驗多個實驗複製物,從而提高統計能力。此等技術可允許檢測腫瘤內部異質性、藥物微擾及鑑別稀有細胞事件,諸如藥物抗性。微型類器官球體可通常與包括單個細胞RNA轉錄組分析及表觀遺傳剖析的後續分析相容。另外,藉由將由微型類器官球體提供的組織(例如,生檢)樣本效率最大化,可儲備(例如,藉由低溫保存進行生物儲備)微型類器官球體之一部分以用於未來新穎藥物分析及/或用於證實分析,包括基因篩選。The miniaturized assays described herein (eg, using miniature organoid spheroids) can maximize patient tumor biopsy, enabling screening of more drug compounds. For example, a 600 uL tumor sample can be divided into about 143,000 individual microreactors with a volume of about 4 nL. By maximizing the tissue sample, multiple experimental replicates can be examined, increasing statistical power. These techniques may allow detection of intra-tumor heterogeneity, drug perturbation, and identification of rare cellular events such as drug resistance. Miniature organoid spheroids can generally be compatible with subsequent analyzes including single cell RNA transcriptome analysis and epigenetic profiling. Additionally, by maximizing the efficiency of tissue (e.g., biopsy) samples provided by the micro-organoid spheroids, a portion of the micro-organoid spheroids can be stockpiled (e.g., biobanked by cryopreservation) for future novel drug analysis and and/or for confirmatory analysis, including genetic screening.

舉例而言,圖18及圖19說明使用本文中所描述之方法及設備(包括微型類器官球體)的治療方法之實例。對於精準及個人化醫療,此等方法及設備可用作適當的藥物選擇之臨床指標,以改良臨床結果及藥物反應。作為一個實施例,經診斷患有轉移癌的患者將採取病理組織學的生檢,且用於篩選由如本文中所描述的生檢形成的複數個微型類器官球體。在7至10天內,可自生檢執行篩選以鑑別最有效的護理標準療法,因此患者可在大約14天開始治療。For example, Figures 18 and 19 illustrate examples of therapeutic methods using the methods and devices described herein, including miniature organoid spheroids. For precision and personalized medicine, these methods and devices can be used as clinical indicators for appropriate drug selection to improve clinical outcomes and drug responses. As an example, a histopathological biopsy of a patient diagnosed with metastatic cancer will be taken and used to screen a plurality of mini-organoid spheroids formed from the biopsy as described herein. Within 7 to 10 days, screening can be performed from the biopsy to identify the most effective standard-of-care therapy, so patients can begin treatment in approximately 14 days.

在圖18中說明此實例。在此實例中,可在第0天(例如,藉由CT掃描)鑑別腫瘤1801,且在第5天獲取生檢1805,且在同一天,可形成數百、數千或數萬個微型類器官球體且培養1至5天並篩選1805以鑑別可使用的一或多種藥物組合物。此相同步驟(形成微型類器官球體及篩選)可用於在整個疾病進程中的多個臨床決定點導引精準藥品。在此實例中,可在第14天開始使用經鑑別的一或多種藥物組合物的療法1809,且稍後可在治療過程期間監測患者(例如,在約第90天追蹤CT掃描)以確認腫瘤對治療有反應1811。若如此,可繼續療法1813且監測進行中的進展1815。This example is illustrated in FIG. 18 . In this example, a tumor 1801 can be identified on day 0 (e.g., by CT scan), and a biopsy 1805 can be taken on day 5, and on the same day, hundreds, thousands, or tens of thousands of miniature species can be formed Organ spheroids were cultured for 1 to 5 days and screened 1805 to identify one or more pharmaceutical compositions that could be used. This same step (formation of miniature organoid spheroids and screening) can be used to guide precision medicine at multiple clinical decision points throughout the disease process. In this example, therapy 1809 with the identified one or more pharmaceutical compositions can be initiated on day 14, and the patient can be monitored later during the course of treatment (e.g., follow-up CT scans at about day 90) to confirm the tumor 1811 responded to treatment. If so, therapy can be continued 1813 and ongoing progress monitored 1815.

如所提及,使用微型類器官球體分析可在治療過程期間的整個治療中的多個時刻重複。在圖19中說明此技術。舉例而言,當患者首先經診斷1907患有可切除的原發性腫瘤時,此技術(例如,微型類器官球體之產生及篩選1905)可用於判定最有效的新輔助療法1921。因此,可獲取生檢且可形成數百、數千或數萬個微型類器官球體且用一組潛在藥物組合物篩選。一旦原發性腫瘤經切除1923,則此技術1905'可指示是否應選擇輔助療法及應選擇何種輔助療法1925。若在手術移除原發性腫瘤之後發生復發或轉移1927,則可使用相同技術(例如,自新鮮生檢產生及篩選微型類器官球體1905''、1905'''、195'''')以導引護理標準療法,包括1線療法1929、2線療法1931及3線療法1933。若患者最終變得對所有護理標準療法耐受或具有耐藥性,則可執行此技術1905'''''以鑑別治療耐藥性腫瘤的仿單核準適應症外藥物1935。此技術亦可用作伴隨診斷以鑑別特定治療的患者。最後,該技術可用於衍生及保存患者衍生之微型類器官球體,以確立基於類器官球體的活癌症庫以用於篩選、基因體剖析、新穎藥物發現、藥物測試及臨床試驗設計。As mentioned, the analysis using mini-organoid spheroids can be repeated at multiple points throughout the treatment course. This technique is illustrated in FIG. 19 . For example, this technique (eg, generation and screening 1905 of mini-organoid spheroids) can be used to determine the most effective neoadjuvant therapy 1921 when a patient is first diagnosed 1907 with a resectable primary tumor. Thus, biopsies can be obtained and hundreds, thousands or tens of thousands of miniature organoid spheroids can be formed and screened with a panel of potential drug compositions. Once the primary tumor has been resected 1923, this technique 1905' can indicate whether and what adjuvant therapy should be chosen 1925. If recurrence or metastasis occurs after surgical removal of the primary tumor 1927, the same technique can be used (e.g., miniature organoid spheroids generated and screened from fresh biopsies 1905'', 1905''', 195'''') To guide the standard of care therapy, including 1-line therapy 1929, 2-line therapy 1931 and 3-line therapy 1933. If the patient eventually becomes tolerant or resistant to all standard-of-care therapies, this technique can be performed 1905''''' to identify off-label drugs 1935 for treatment of drug-resistant tumors. This technology can also be used as a companion diagnostic to identify patients for specific treatments. Ultimately, this technique can be used to derive and preserve patient-derived micro-organoid spheroids to establish living organoid-spheroid-based cancer libraries for screening, genome profiling, novel drug discovery, drug testing, and clinical trial design.

因為可以相對較低侵入性方式(例如,藉由切除或生檢)完成此等技術及產生大量微型類器官球體,以自篩選提供合理地快速結果,所以此等方法可易於調適用於護理標準。舉例而言,來自組織(例如,生檢)輸入的細胞材料之體積非常小,且可解離為例如10 µL至5 ml體積。Because these techniques can be accomplished in a relatively less invasive manner (e.g., by excision or biopsy) and generate large numbers of miniature organoid spheroids to provide reasonably rapid results for self-screening, these methods are readily adaptable to standard of care . For example, cellular material from tissue (eg, biopsy) input is very small in volume and can be dissociated into volumes of, eg, 10 µL to 5 ml.

一般而言,可為自動或手動執行使用本文中所描述之微型類器官球體進行篩選。可使用實際上任何篩選技術,包括藉由以下一或多者成像:共焦顯微鏡、螢光顯微法、液體透鏡、全像術、聲納、明視野及暗視野成像、雷射、平面雷射片,包括基於影像之分析方法(例如,使用電腦視覺及/或受監督或不受監督模式,例如CNN)之高通量實施例。後續篩選可包括對培養介質取樣及/或對來自微型類器官球體的細胞執行基因或蛋白質篩選(例如,scRNA-序列、ATAC-序列、蛋白質體學等)。 實例 In general, screening using the miniature organoid spheroids described herein can be performed automatically or manually. Virtually any screening technique can be used, including imaging by one or more of the following: confocal microscopy, fluorescence microscopy, liquid lensing, holography, sonar, brightfield and darkfield imaging, laser, planar laser Shots, including high-throughput embodiments of image-based analysis methods (eg, using computer vision and/or supervised or unsupervised modalities, such as CNNs). Subsequent screening may include sampling the culture medium and/or performing genetic or protein screening (eg, scRNA-seq, ATAC-seq, proteomics, etc.) on the cells from the mini-organoid spheroids. example

圖20及圖21說明用於形成如本文中所描述的複數個微型類器官球體的設備的另一實例。在圖20中,設備可包括複數個微型類器官球體形成接合處,其中不混溶的材料(例如,油) 2002可添加至裝置中的儲集器及/或孔口2004中。類似地,未聚合材料2006 (在此實例中,包括解離之生檢細胞及流體基質材料)可添加至設備中的儲集器或孔口2008中。在一些變化形式中,可經由第三組孔口2010添加第二或額外材料(例如,生物活性劑)。此等組分可在接合處2012(與上文所描述之彼等者類似)合併,從而在可聚合至微型類器官球體中的不混溶的材料中形成小滴。在圖20中,展示具有對應輸入端2004、2008、2010及輸出端2014的三個(或更多)平行的接合處。20 and 21 illustrate another example of an apparatus for forming a plurality of miniature organoid spheroids as described herein. In FIG. 20, the device can include a plurality of micro-organoid spheroids forming a junction where an immiscible material (eg, oil) 2002 can be added to a reservoir and/or orifice 2004 in the device. Similarly, unpolymerized material 2006 (including, in this example, dissociated biopsy cells and fluid matrix material) can be added to a reservoir or orifice 2008 in the device. In some variations, a second or additional material (eg, a bioactive agent) can be added via the third set of orifices 2010 . These components can combine at junctions 2012 (similar to those described above), forming droplets in immiscible materials that can be polymerized into miniature organoid spheroids. In FIG. 20, three (or more) parallel junctions with corresponding inputs 2004, 2008, 2010 and output 2014 are shown.

圖21說明使用如圖20中所展示的設備形成微型類器官球體的方法。在此變化形式中,所得微型類器官球體包括合併以形成微型類器官球體的目標(例如,腫瘤)生檢細胞以及一或多種額外生物活性劑兩者。舉例而言,第一通道2103可包括未聚合材料(包括解離之生檢細胞及基質材料),第二通道2107包括額外活性生物材料,且攜載不混溶的材料(例如油)的一對交叉通道2109、2109'在接合處會聚以形成大小受控制的經聚合以形成微型類器官球體2107的小滴。FIG. 21 illustrates a method of forming miniature organoid spheroids using the apparatus as shown in FIG. 20 . In this variation, the resulting mini-organoid spheroids include both target (eg, tumor) biopsy cells combined to form the mini-organoid spheroids and one or more additional bioactive agents. For example, the first channel 2103 may include unpolymerized material (including dissociated biopsy cells and matrix material), the second channel 2107 includes additional active biological material, and carries a pair of immiscible materials (such as oil). Intersecting channels 2109 , 2109 ′ converge at the junction to form size-controlled droplets that aggregate to form micro-organoid spheroids 2107 .

在此實例中,額外活性生物材料可為例如冷凍介質(例如,以輔助儲備微型類器官球體)及/或具有額外細胞(例如,免疫細胞、基質細胞、內皮細胞等)、額外支援性網狀分子(例如,ECM、膠原蛋白、酶、糖蛋白、仿生骨架等)、額外生長因子及/或藥物化合物的共培養物。 實例 2 :篩選結果 In this example, the additional active biomaterial can be, for example, a freezing medium (e.g., to aid in the storage of micro-organoid spheroids) and/or have additional cells (e.g., immune cells, stromal cells, endothelial cells, etc.), additional supportive mesh Co-cultures of molecules (eg, ECM, collagen, enzymes, glycoproteins, biomimetic scaffolds, etc.), additional growth factors, and/or pharmaceutical compounds. Example 2 : Filter results

如上文所提及,患者衍生之微型類器官球體及使用其等篩選藥物組合物的方法可用於精確地預測患者腫瘤對一或多種藥物療法的反應。在一些情況下,微型類器官球體的使用可提供精確結果,其中傳統經培養的藥物篩選並不精確地預測藥物反應。舉例而言,在圖22A至圖22D中,微型類器官球體而非細胞株能夠與患者反應相關。在圖22A中,檢驗用藥物(例如,奧沙利鉑)給藥的傳統細胞株;藥物線路展示無效果,預測腫瘤將對檢驗的所有劑量範圍下的藥物具有耐藥性。As mentioned above, patient-derived micro-organoid spheroids and methods of screening pharmaceutical compositions using the same can be used to accurately predict the response of a patient's tumor to one or more drug therapies. In some cases, the use of miniature organoid spheroids can provide precise results where traditional cultured drug screens do not precisely predict drug response. For example, in FIGS. 22A-22D , miniature organoid spheroids rather than cell lines can be correlated with patient response. In Figure 22A, conventional cell lines dosed with drug (eg, oxaliplatin) were examined; the drug line showed no effect, and tumors were predicted to be resistant to the drug at all dose ranges tested.

為進行比較,自患者生檢產生複數個微型類器官球體,如圖22B中所展示。在此實例中,患者衍生之微型類器官球體展示腫瘤微型類器官球體的細胞存活率顯著降低,從而預測藥物敏感性。實際上,當用藥物治療時,腫瘤對治療有反應,如圖22C (治療前)及圖22D (治療後)中所展示。 實例 3 微型類器官球體與患者反應之間的相關性 For comparison, a plurality of miniature organoid spheroids were generated from patient biopsies, as shown in Figure 22B. In this example, patient-derived micro-organoid spheroids exhibited significantly reduced cell survival in tumor micro-organoid spheroids, predictive of drug sensitivity. Indeed, when treated with the drug, the tumors responded to the treatment, as shown in Figure 22C (before treatment) and Figure 22D (after treatment). Example 3 : Correlation Between Miniature Organoid Spheroids and Patient Response

在類似的實驗組中,自生檢材料產生微型類器官球體(圖23A),且使用所得微型類器官球體執行藥物效果篩選。圖23B展示第一藥物(奧沙利鉑)對此等微型類器官球體的作用,顯示在藥物存在之情況下微型類器官球體之存活百分比無變化,從而預測耐藥性。類似地,用第二藥物(伊立替康)治療展示對微型類器官球體不具有作用,從而預測耐藥性,如圖23C中所展示。用奧沙利鉑及伊立替康兩者治療患者,且在治療6個月之後顯示無反應。因此,微型類器官球體與患者對護理標準藥物的反應在很大程度上相關。在此情況下,承受六個月副作用及毒性的患者可已藉由所預測的微型類器官球體的反應避免,從而指示(在自生檢的7至10天內)腫瘤將不對此等藥物有反應。 實例 4 :多種藥物篩選 In a similar set of experiments, mini-organoid spheroids were generated from biopsy material ( FIG. 23A ), and drug effect screening was performed using the resulting mini-organoid spheroids. Figure 23B shows the effect of the first drug (oxaliplatin) on these mini-organoid spheroids, showing no change in the percent survival of the mini-organoid spheroids in the presence of the drug, predicting drug resistance. Similarly, treatment with a second drug (irinotecan) showed no effect on mini-organoid spheroids, predicting drug resistance, as shown in Figure 23C. The patient was treated with both oxaliplatin and irinotecan and showed no response after 6 months of treatment. Thus, miniature organoid spheroids were largely correlated with patient response to standard-of-care medications. In this case, a patient suffering six months of side effects and toxicity could have been avoided by the predicted response of the mini-organoid spheroids, indicating (within 7 to 10 days of self-biological examination) that the tumor would not respond to the drugs . Example 4 : Multiple Drug Screening

圖24展示可使用如本文中所描述的患者衍生之複數個微型類器官球體產生的一組藥物(例如,化學治療劑)之實例。在此實例中,藉由投與複數種(27)藥物中之每一者的複數個複製物進行使用患者衍生之微型類器官球體的藥物篩選。單個腫瘤生檢用於極其快速地(例如,在少於兩週內)產生大量複數個微型類器官球體,且針對該組藥物調配物(例如,展示27種調配物)測試此等微型類器官球體。此測試並行地進行且可自動定量(例如,藉由光學偵測及定量)。在此實例中,對此特定腫瘤展示最大毒性的藥物為帕佐泮尼(Pazopanib)。Figure 24 shows an example of a panel of drugs (eg, chemotherapeutic agents) that can be produced using patient-derived plurality of mini-organoid spheroids as described herein. In this example, drug screening using patient-derived mini-organoid spheroids was performed by administering multiple replicates of each of multiple (27) drugs. A single tumor biopsy was used to generate large numbers of multiple mini-organoid spheroids extremely rapidly (eg, in less than two weeks) and test these mini-organoids against the panel of drug formulations (eg, 27 formulations demonstrated) sphere. This test is performed in parallel and can be quantified automatically (eg, by optical detection and quantification). In this example, the drug exhibiting the greatest toxicity for this particular tumor was Pazopanib.

可並行地檢驗藥物之組合以及不同藥物濃度。由於可自相同腫瘤生檢產生數百、數千或數萬個微型類器官球體,因此此分選之陣列測試藉由本文中所描述之方法及設備變得切實可行。 實例 5 :生檢樣本製備 Combinations of drugs and different drug concentrations can be tested in parallel. Since hundreds, thousands, or tens of thousands of mini-organoid spheroids can be generated from the same tumor biopsy, array testing of this sort is made feasible by the methods and devices described herein. Example 5 : Biopsy sample preparation

材料:一種用於形成微型類器官球體的設備,如上文所描述,包括小滴微流體晶片(200 µm);EvaGreen的拜耳雷德小滴生成油(Bio-rad Droplet Generation Oil)(目錄號186-4006),每次運行3至5 mL,全氟辛醇(PFO)、Sigma、含10%全氟辛醇(PFO)之Novec HFE 7500、PBS、細胞培養基(亦即,RPMI w/10% FBS及1%青黴素鏈黴素),70 µm或100 µm過濾器,50 mL錐形皮氏培養皿。Materials: An apparatus for forming miniature organoid spheroids, as described above, including droplet microfluidic wafers (200 µm); Bayer-rad Droplet Generation Oil from EvaGreen (Catalog #186 -4006), 3 to 5 mL per run, perfluorooctyl alcohol (PFO), Sigma, Novec HFE 7500 with 10% perfluorooctyl alcohol (PFO), PBS, cell culture medium (i.e., RPMI w/10% FBS with 1% Penicillin-Streptomycin), 70 µm or 100 µm filter, 50 mL conical Petri dish.

生檢樣本解離:使用生檢樣本(人類/動物)以產生來自患者之解離樣本(亦即,單個細胞組織)。塗佈微流體晶片,且組組裝微流體晶片及固持器。連接微流體導管且裝配至微型類器官球體及廢油的輸出端(例如,多孔盤,15 mL埃彭道夫(Eppendorf)等)。Biopsy Dissociation: A biopsy (human/animal) is used to generate a dissociated sample (ie, single cell tissue) from a patient. The microfluidic wafer is coated, and the microfluidic wafer and holder are assembled. Connect microfluidic tubing and fit to micro-organoid spheroids and output of waste oil (eg, multi-well disc, 15 mL Eppendorf, etc.).

運行裝置以形成微型類器官球體。自培育箱移除含有小滴的輸出端(例如,盤、埃彭道夫管等)(在至少15分鐘之後)。自輸出端移除任何過量油。小滴應為漂浮的,因此油應在瓶底部。小心不要自管移除小滴。向輸出端添加100 μL之10% (v/v) PFO。謹慎地渦旋且等待約1 min。不要移液或干擾樣本。在300 g下離心60秒。移除上清液(過量油/PFO)。不要移液或干擾樣本。移除儘可能多的PFO,此係由於此化學品可在培養期間降低細胞存活率。添加1 mL之細胞培養基。不要移液或干擾樣本。在300 g下離心60秒。移除上清液及任何過量的油/PFO。添加1 mL之細胞培養基。小心地用1 mL移液管尖端將樣本進行上下移液(約30次)。小心不要超過移液管或干擾小滴樣本。使用1 mL移液管尖端,使小滴介質溶液通過70 µm或100 µm過濾器(連接至50 mL錐形管)。一些小滴將黏著在輸出端(例如,15 mL埃彭道夫)的內部。用2至3 mL之PBS沖洗各管且上下移液。使經沖洗的PBS及小滴通過過濾器。重複此步驟兩次,或直至管看起來透明,且小滴已轉移至過濾器。使用1 mL移液管尖端,用約5 mL之PBS小心地洗滌含有小滴的過濾器。試圖覆蓋過濾器之整個表面區域。此洗滌步驟自樣本移除任何過量的油及PFO,且使得凝膠小滴之最終回收至細胞培養基中。Run the device to form miniature organoid spheroids. The output (eg, tray, Eppendorf tube, etc.) containing the droplets is removed from the incubator (after at least 15 minutes). Remove any excess oil from the output. The droplets should be floating, so the oil should be at the bottom of the bottle. Be careful not to remove the droplet from the tube. Add 100 µL of 10% (v/v) PFO to the output. Vortex carefully and wait about 1 min. Do not pipette or disturb the sample. Centrifuge at 300 g for 60 seconds. The supernatant (excess oil/PFO) was removed. Do not pipette or disturb the sample. Remove as much PFO as possible, as this chemical can reduce cell viability during culture. Add 1 mL of cell culture medium. Do not pipette or disturb the sample. Centrifuge at 300 g for 60 seconds. Remove the supernatant and any excess oil/PFO. Add 1 mL of cell culture medium. Carefully pipette the sample up and down (approximately 30 times) with a 1 mL pipette tip. Be careful not to overshoot the pipette or disturb the droplet sample. Using a 1 mL pipette tip, pass a small drop of media solution through a 70 µm or 100 µm filter (attached to a 50 mL conical tube). Some droplets will stick to the inside of the output (eg, 15 mL Eppendorf). Rinse each tube with 2 to 3 mL of PBS and pipette up and down. Pass the rinsed PBS and droplet through the filter. Repeat this step twice, or until the tube appears clear and the droplets have transferred to the filter. Using a 1 mL pipette tip, carefully wash the filter containing the droplet with approximately 5 mL of PBS. Attempt to cover the entire surface area of the filter. This washing step removes any excess oil and PFO from the sample and allows for eventual recovery of the gel droplets into the cell culture medium.

一旦恰當地排出(約1至2分鐘),則小心地自50 mL錐形管移除過濾器。上下翻轉過濾器且用新鮮細胞培養基洗滌後側,且將溶液截留於新鮮皮氏培養皿中。此操作將小滴與過濾器分離,且將其等置放在細胞培養基中。建議使用1 mL移液管尖端,且用約5 mL之培養基洗滌。Once properly drained (approximately 1 to 2 minutes), the filter was carefully removed from the 50 mL conical tube. The filter was turned upside down and the backside was washed with fresh cell culture medium, and the solution was retained in a fresh petri dish. This operation separates the droplets from the filter and places them in the cell culture medium. It is recommended to use a 1 mL pipette tip and wash with about 5 mL of medium.

在顯微鏡下檢查小滴之品質。應移除大部分/所有油。若回收不佳,則可重新過濾樣本。可藉由血球計檢查經回收的微型類器官球體之密度。 實例 6 腎臟組織微型類器官球體 The quality of the droplets was checked under a microscope. Most/all oil should be removed. If recovery is poor, the sample can be refiltered. The density of the recovered micro-organoid spheroids can be checked by a hemocytometer. Example 6 : Kidney Tissue Miniature Organoid Spheroids

在另一實例中,可自生檢腎臟組織形成微型類器官球體。舉例而言,使用的工具可包括:管旋轉器或100 μm及70 μm細胞過濾器、15 mL錐形管、50 mL錐形管、剃刀片、鑷子及手術剪刀、皮氏培養皿(100×15 mm)或組織培養皿。試劑可包括:EBM-2培養基、膠原蛋白酶(5 mg/mL儲備液)、漢克氏平衡鹽溶液(Hank's Balanced Salt Solution;HBSS)、氯化鈣(10 mM儲備溶液)、磷酸鹽緩衝溶液(1×PBS)、基質膠、0.4%錐蟲藍溶液及胰蛋白酶。In another example, miniature organoid spheroids can be formed from biopsy kidney tissue. Examples of tools used may include: tube rotators or 100 μm and 70 μm cell strainers, 15 mL conical tubes, 50 mL conical tubes, razor blades, forceps and surgical scissors, petri dishes (100× 15 mm) or tissue culture dishes. Reagents may include: EBM-2 medium, collagenase (5 mg/mL stock solution), Hank's Balanced Salt Solution (HBSS), calcium chloride (10 mM stock solution), phosphate buffered saline ( 1×PBS), matrigel, 0.4% trypan blue solution and trypsin.

腎組織儲存在低溫傳遞培養基中且始終處於冰上。2 mL之酶消化溶液可置放於15 mL錐形管中。添加600 μL之氯化鈣(最終濃度:3 mM)及添加200 μL之膠原蛋白酶(最終:0.5 mg/mL)。將腎臟樣本轉移至皮氏/培養皿中。用無菌組織或剃刀片移除所有過量或非腫瘤組織。向組織中添加1 mL之酶促溶液。用無菌剃刀片(<2 mm 2)將樣品切碎成小塊。用鑷子或用手保持該盤。將切碎的組織及酶促溶液轉移回至具有酶促溶液的15 mL管中。將該管置放在管旋轉器中或在37℃培育箱中30至60分鐘之間的15 mL管旋轉器中。自培育箱移除管。用至少6 mL EBM-2 (酶消化溶液之量至少3倍)淬滅酶消化溶液。移液以混合。將100 μm或70 μm細胞過濾器置放至50 mL錐形管上。經由過濾器轉移樣本。將溶液轉移至新的15 mL錐形管。將樣本在1500 rpm下離心5分鐘。丟棄上清液,留下細胞沈澱物。將沈澱物再懸浮在1 mL EBM-2培養基中。向封口膜片上的10 μL之錐蟲藍中添加10 μL細胞混合物,且轉移至細胞計數盤或血細胞計數器。計算細胞濃度(#/mL)。在1500 RPM下離心5分鐘,且丟棄上清液,留下沈澱物。將細胞沈澱物再懸浮在每1.25×105個細胞50 uL基質膠中。在冰上執行。將50 μL具有基質膠細胞懸浮液之圓頂平鋪在經預熱的24孔平底盤中的各孔中心中。將該盤轉移至37℃細胞培育箱且培育至少20分鐘。確認圓頂經聚合。沿著孔之壁平緩地添加500 μL經預熱的EBM-2培養基。在37℃培育箱中培育。每2天進行完全培養基更換以擴增微型類器官球體。 實例 7 肝臟微型類器官球體 Kidney tissue was stored in cryogenic transfer medium and kept on ice at all times. 2 mL of enzymatic digestion solution can be placed in a 15 mL conical tube. Add 600 μL of calcium chloride (final concentration: 3 mM) and add 200 μL of collagenase (final: 0.5 mg/mL). Transfer kidney samples to petri/petri dishes. Remove all excess or non-neoplastic tissue with sterile tissue or a razor blade. Add 1 mL of Enzyme Solution to the tissue. Samples were minced into small pieces with a sterile razor blade (<2 mm 2 ). Hold the disc with tweezers or by hand. Transfer the minced tissue and enzymatic solution back to the 15 mL tube with enzymatic solution. Place the tube in a tube rotator or a 15 mL tube rotator in a 37 °C incubator between 30 and 60 min. Remove the tube from the incubator. Quench the enzyme digestion solution with at least 6 mL of EBM-2 (at least 3 times the volume of the enzyme digestion solution). Pipette to mix. Place a 100 µm or 70 µm cell strainer onto a 50 mL conical tube. Transfer the sample through the filter. Transfer the solution to a new 15 mL conical tube. The samples were centrifuged at 1500 rpm for 5 minutes. Discard the supernatant, leaving the cell pellet. Resuspend the pellet in 1 mL of EBM-2 medium. Add 10 μL of the cell mixture to 10 μL of trypan blue on the parafilm and transfer to a cytometer or hemocytometer. Calculate the cell concentration (#/mL). Centrifuge at 1500 RPM for 5 minutes and discard the supernatant, leaving the pellet. Resuspend the cell pellet in 50 uL Matrigel per 1.25 x 105 cells. Perform on ice. Plate 50 μL of the dome with Matrigel cell suspension in the center of each well in a pre-warmed 24-well flat bottom plate. The plate was transferred to a 37°C cell incubator and incubated for at least 20 minutes. Confirm that the dome is polymerized. Gently add 500 μL of pre-warmed EBM-2 medium along the wall of the well. Cultivate in a 37°C incubator. Complete media changes were performed every 2 days to expand the micro-organoid spheroids. Example 7 : Liver Micro-Organoid Spheroids

如所提及,可自正常(例如,非癌)及/或異常組織形成微型類器官球體。舉例而言,圖25A至圖25B及圖26A至圖26B說明自已離位的小鼠肝臟組織形成的微型類器官球體的一個實例,該小鼠肝臟組織與流體基質材料合併以形成未聚合之混合物,接著聚合未聚合之混合物小滴以形成該微型類器官球體。在此實例中,微型類器官球體之直徑為約300 µm。在圖25A至圖25B中,用每個小滴單個細胞形成微型類器官球體經。在圖26A至圖26B中,用每個小滴25個細胞形成微型類器官球體。在圖25A中,展示在形成之後一天的微型類器官球體;圖25B展示培養十天之後的微型類器官球體。一些微型類器官球體中之細胞已分裂,形成呈現結構的群集;其他微型類器官球體包括分裂較慢或並未分裂的細胞。類似地,在圖26A至圖26B中,微型類器官球體最初在各微型類器官球體中包括約25個細胞。在培養十天之後,一些微型類器官球體展示大量細胞生長,從而形成結構,而其他微型類器官球體僅展示少量的生長。在兩種情況下,已發現微型類器官球體內的細胞呈現其等來源的原始組織(例如肝細胞)之特性特徵。As mentioned, miniature organoid spheroids can be formed from normal (eg, non-cancerous) and/or abnormal tissue. For example, Figures 25A-25B and Figures 26A-26B illustrate an example of a miniature organoid spheroid formed from self-exposed mouse liver tissue that was combined with a fluid matrix material to form an unpolymerized mixture , followed by polymerizing the unpolymerized mixture droplets to form the miniature organoid spheroids. In this example, the micro-organoid spheroids were approximately 300 µm in diameter. In FIGS. 25A-25B , a single cell per droplet was used to form miniature organoid spheroids. In FIGS. 26A-26B , 25 cells per droplet were used to form miniature organoid spheroids. In FIG. 25A , miniature organoid spheroids are shown one day after formation; FIG. 25B shows miniature organoid spheroids after ten days of culture. The cells in some micro-organoid spheroids had divided, forming clusters that exhibited structures; other micro-organoid spheroids included cells that divided slowly or did not divide. Similarly, in FIGS. 26A-26B , the mini-organoid spheroids initially included about 25 cells in each mini-organoid spheroid. After ten days in culture, some micro-organoid spheroids exhibited substantial cell growth forming structures, while others exhibited only minimal growth. In both cases, cells within micro-organoid spheroids have been found to exhibit characteristics characteristic of their original tissue of origin (eg, hepatocytes).

對人類肝臟組織成功地執行相同程序,如圖27A至圖27C中所展示。在此實例中,最初用約五十個細胞形成微型類器官球體,如圖27A中所展示。截至培養的第18天,一些微型類器官球體展示具有群集及形成結構的細胞,而其他具有較小結構或細胞並未分裂。 實例 8 經培養之細胞微型類器官球體 The same procedure was successfully performed on human liver tissue, as shown in Figures 27A-27C. In this example, approximately fifty cells were initially used to form miniature organoid spheroids, as shown in Figure 27A. By day 18 in culture, some micro-organoid spheroids exhibited cells that clustered and formed structures, while others had smaller structures or cells that did not divide. Example 8 : Cultured cell micro-organoid spheroids

除例如在形成微型類器官球體之前立即或不久自患者移除的初代組織之外,微型類器官球體可由一或多個經培養之細胞(包括2D培養之細胞或3D培養之細胞)形成。Micro-organoid spheroids can be formed from one or more cultured cells, including 2D-cultured cells or 3D-cultured cells, in addition to, for example, primary tissue removed from a patient immediately or shortly prior to formation of the micro-organoid spheroids.

在一些變化形式中,微型類器官球體可由生長為患者衍生之異種移植物(PDX)之部分的細胞株形成。舉例而言,圖28A至圖28D說明由經培養之PDX240細胞形成的微型類器官球體。PDX240細胞為患者衍生之異種移植物(PDX)腫瘤細胞株(基於患者來源的編號240),其為在免疫缺乏小鼠(PDX)中生長以形成活體內腫瘤的人類腫瘤。異種移植物組織經提取、解離,且用於形成如上文所描述之微型類器官球體。在此實例中,單個細胞包括於形成的各微型類器官球體中。圖28A展示在培養一天之後的微型類器官球體,而圖28B展示在培養三天之後的微型類器官球體,且圖28C及圖28D分別展示在培養五天及七天之後的微型類器官球體。隨著培養的時間進展,至少一些微型類器官球體展示細胞分裂及形成結構。In some variations, mini-organoid spheroids can be formed from cell lines grown as part of patient-derived xenografts (PDX). For example, Figures 28A-28D illustrate miniature organoid spheroids formed from cultured PDX240 cells. PDX240 cells are a patient-derived xenograft (PDX) tumor cell line (number 240 based on patient origin), which are human tumors grown in immunodeficient mice (PDX) to form tumors in vivo. Xenograft tissue was extracted, dissociated, and used to form miniature organoid spheroids as described above. In this example, a single cell was included in each miniature organoid spheroid formed. Figure 28A shows mini-organoid spheroids after one day of culture, while Figure 28B shows mini-organoid spheroids after three days of culture, and Figure 28C and Figure 28D show mini-organoid spheroids after five and seven days of culture, respectively. Over time in culture, at least some miniature organoid spheroids exhibit cell division and formation of structures.

圖29A至圖29D展示類似實驗,其中五個PDX240細胞最初包括於形成各微型類器官球體的各小滴中。隨著培養的時間(例如,第1天、第3天、第5天及第7天,如圖29A至圖29中分別所示),細胞可分裂及形成結構。 實例 9 微型類器官球體與傳統類器官之比較 Figures 29A-29D show similar experiments in which five PDX240 cells were initially included in each droplet forming each mini-organoid spheroid. Over time in culture (eg, Day 1, Day 3, Day 5, and Day 7, as shown in Figures 29A-29, respectively), cells can divide and form structures. Example 9 : Comparison of Miniature Organoid Spheroids and Traditional Organoids

類器官由患者衍生之異種移植物細胞(包括上文所描述之PDX240細胞及第二PDX細胞株PDX19187)形成且與使用相同細胞形成的微型類器官球體進行比較。使用習知技術形成類器官,其中孔或培養皿中的大量基質膠接種有細胞且經培養直至確認生長。由傳統類器官產生微型類器官球體。Organoids were formed from patient-derived xenograft cells, including the PDX240 cells described above and a second PDX cell line, PDX19187, and compared to miniature organoid spheroids formed using the same cells. Organoids are formed using well-known techniques, in which masses of Matrigel in wells or dishes are seeded with cells and cultured until growth is confirmed. Generate miniature organoid spheroids from conventional organoids.

接著用相同藥物(例如,奧沙利鉑或SN38)處理傳統(「塊狀」)類器官及微型類器官球體兩者且在處理3天之後量測細胞存活率。產生圖30及圖31中所示之藥物反應曲線,且展示類似反應曲線。舉例而言,在圖30中,PDO19187塊狀類器官及微型類器官球體之藥物反應曲線展示與奧沙利鉑濃度類似的反應曲線,PDX240塊狀類器官及微型類器官球體亦如此。在圖31中,PDX19187及PDX240兩者的藥物反應曲線亦展示塊狀類器官及微型類器官球體兩者針對SN38的類似結果。圖32展示另一抗癌藥物5-FU (氟尿嘧啶)的反應曲線,再次展示PDZ-19187及PDX-240傳統類器官及微型類器官球體的類似藥物反應曲線。Both traditional ("bulky") organoids and mini-organoid spheroids were then treated with the same drug (eg, oxaliplatin or SN38) and cell viability was measured after 3 days of treatment. The drug response curves shown in Figure 30 and Figure 31 were generated and exhibit similar response curves. For example, in Figure 30, the drug response curves of PDO19187 bulk organoids and mini-organoid spheroids showed similar response curves to oxaliplatin concentrations, as did PDX240 bulk organoids and mini-organoid spheroids. In FIG. 31 , the drug response curves of both PDX19187 and PDX240 also showed similar results against SN38 for both bulk organoids and mini-organoid spheroids. Figure 32 shows the response curve of another anticancer drug 5-FU (fluorouracil), again showing the similar drug response curves of PDZ-19187 and PDX-240 traditional organoids and micro-organoid spheroids.

因此,可較快速及可靠地形成且相較於傳統類器官可具有較高總存活速率的本文中所描述之微型類器官球體可提供與使用相同細胞形成的塊狀類器官的彼等藥物反應相當的藥物反應。然而,如本文中所描述,微型類器官球體可較快速地使用且可以顯著較大數目形成。 實例 10 微型類器官球體的藥物作用 Thus, the miniature organoid spheroids described herein, which can be formed more quickly and reliably and can have a higher overall survival rate compared to traditional organoids, can provide drug responses comparable to those of bulk organoids formed using the same cells. considerable drug reaction. However, as described herein, miniature organoid spheroids can be used relatively quickly and can be formed in significantly larger numbers. Example 10 : Drug Effects of Miniature Organoid Spheroids

一般而言,本文中所描述之微型類器官球體可用於執行一或多種分析,包括毒性分析。可執行任何適當的分析,如藉由懸浮在微型類器官球體內之組織(例如,細胞、組織結構)的分析所判定的結果。可分析或以光學方式、以化學方式、以電氣方式、以基因方式或以此項技術中已知的任何其他方式分析本文中所描述之微型類器官球體。In general, the micro-organoid spheroids described herein can be used to perform one or more assays, including toxicity assays. Any suitable analysis can be performed, as determined by analysis of tissue (eg, cells, tissue structures) suspended within the micro-organoid spheroids. The micro-organoid spheroids described herein can be analyzed or analyzed optically, chemically, electrically, genetically, or in any other manner known in the art.

光學(手動或自動)偵測可尤其適用且可包括以光學方式分析一或多種藥物調配物對微型類器官球體內的組織(包括細胞、細胞群集、細胞結構等)的作用。在一些變化形式中,如上文所提及,可針對細胞死亡(例如,經測試的微型類器官球體內的組織之數目及/或大小)分析藥物調配物。在其他變化形式中,可針對細胞生長分析微型類器官球體,包括大小、類型及/或生長速率的減少。在一些變化形式中,可針對形成的組織結構中的變化分析微型類器官球體。Optical (manual or automated) detection may be particularly useful and may include optically analyzing the effect of one or more drug formulations on tissue (including cells, cell clusters, cellular structures, etc.) within the micro-organoid spheroid. In some variations, as mentioned above, the drug formulation can be assayed for cell death (eg, the number and/or size of tissue within the tested mini-organoid spheroids). In other variations, miniature organoid spheroids can be assayed for cell growth, including reduction in size, type, and/or growth rate. In some variations, micro-organoid spheroids can be analyzed for changes in the formed tissue architecture.

舉例而言,圖33A至圖33B說明一種藥物調配物(在此實例中)乙醯胺苯酚(10 mM)對小鼠肝臟微型類器官球體的作用。圖33A為對照組,其中微型類器官球體未經處理,展示培養時生長的微型類器官球體(箭頭)內的組織。圖33B展示自用10 mM乙醯胺苯酚代替處理的小鼠肝臟形成的微型類器官的類似組。在對照組中,微型類器官球體內的組織結構與處理組相比相對較大。乙醯胺苯酚之大部分微型類器官球體中的組織較小且含有許多死亡細胞。For example, Figures 33A-33B illustrate the effect of a drug formulation (in this example) acetaminophen (10 mM) on mouse liver mini-organoid spheroids. Figure 33A is a control group in which the micro-organoid spheroids were untreated, showing the organization within the micro-organoid spheroids (arrows) grown in culture. Figure 33B shows a similar set of mini-organoids formed from mouse livers treated with 10 mM acetaminophen replacement. In the control group, the organized structures within the micro-organoid spheroids were relatively larger compared with the treated group. The tissue in most micro-organoid spheroids of acetaminophen was small and contained many dead cells.

類似地,圖34A至圖34B亦展示使用人類肝臟微型類器官球體的毒性分析。圖34A展示在包括其中形成的組織結構(由箭頭指示)的對照組中觀測到的典型人類肝臟微型類器官球體。圖34B展示處理組,其中用乙醯胺苯酚(10 mM)處理人類肝臟微型類器官球體。相較於對照組,經處理的微型類器官球體中的組織展示顯著增加的非典型組織結構(箭頭)及碎片。Similarly, Figures 34A-34B also show toxicity assays using human liver micro-organoid spheroids. Figure 34A shows typical human liver micro-organoid spheroids observed in a control group including tissue structures formed therein (indicated by arrows). Figure 34B shows the treatment group in which human liver micro-organoid spheroids were treated with acetaminophen (10 mM). Tissue in treated micro-organoid spheroids exhibited significantly increased atypical tissue architecture (arrows) and fragmentation compared to controls.

此等檢查中之任一者(包括光學檢查)可記分、分級、評級或以其他方式定量。舉例而言,在圖33A至圖33B及圖34A至圖34B中,此等兩種分析之結果可定量以指示存活/死亡細胞/組織及其類似者的大小差異、數目。在一些變化形式中,記分可為自動的。Any of these inspections, including optical inspections, can be scored, graded, rated or otherwise quantified. For example, in Figures 33A-33B and 34A-34B, the results of these two assays can be quantified to indicate size differences, numbers of live/dead cells/tissues and the like. In some variations, scoring may be automatic.

本文中所描述之方法(包括使用者介面)中之任一者可實施為軟體、硬體或韌體,且可描述為儲存能夠藉由處理器(例如,電腦、平板電腦、智慧型手機等)執行的一組指令的非暫時性電腦可讀儲存媒體,該指令在由處理器執行時使得處理器控制執行步驟中之任一者,包括但不限於:顯示、與使用者通信、分析、修改參數(包括時序、頻率、強度等)、判定、警示或其類似者。Any of the methods described herein (including user interfaces) can be implemented as software, hardware, or firmware, and can be described as storing data that can be read by a processor (e.g., computer, tablet, smartphone, etc.) ) a non-transitory computer-readable storage medium for a set of instructions that, when executed by a processor, cause the processor to control the execution of any of the steps, including but not limited to: displaying, communicating with a user, analyzing, Modify parameters (including timing, frequency, intensity, etc.), decisions, alerts, or the like.

當特徵或元件本文中被稱作「在」另一特徵或元件「上」時,其可直接在其他特徵或元件正上方或亦可存在中間特徵及/或元件。相比之下,當特徵或元件被稱作「直接在」另一特徵或元件「正上方」時,則不存在中間特徵或元件。亦將理解,當特徵或元件被稱作「連接」、「附接」或「耦接」至另一特徵或元件時,其可直接地連接、附接或耦接至另一特徵或元件或可存在中間特徵或元件。相比之下,當特徵或元件被稱作「直接連接」、「直接附接」或「直接耦接」至另一特徵或元件時,則不存在中間特徵或元件。儘管相對於一個實施例描述或展示,但如此描述或展示之特徵及元件可應用於其他實施例。熟習此項技術者亦將瞭解,對「鄰接」另一特徵安置的結構或特徵之參考可具有與鄰接特徵重疊或位於其之下的部分。When a feature or element is referred to herein as being "on" another feature or element, it can be directly on the other feature or element or intervening features and/or elements may also be present. In contrast, when a feature or element is referred to as being "directly on" another feature or element, there are no intervening features or elements present. It will also be understood that when a feature or element is referred to as being "connected", "attached" or "coupled" to another feature or element, it can be directly connected, attached or coupled to the other feature or element or Intermediate features or elements may be present. In contrast, when a feature or element is referred to as being "directly connected," "directly attached" or "directly coupled" to another feature or element, there are no intervening features or elements present. Although described or shown with respect to one embodiment, features and elements so described or shown may apply to other embodiments. Those skilled in the art will also appreciate that a reference to a structure or feature that is disposed "adjacent" to another feature may have portions that overlap or underlie the adjacent feature.

本文中所使用之術語僅出於描述特定實施例之目的且並不意欲限制本發明。舉例而言,如本文中所使用,除非上下文另外明確指示,否則單數形式「一(a/an)」及「該」意欲亦包括複數形式。應進一步理解,術語「包含(comprises及/或comprising)」在用於本說明書中時,指定所陳述之特徵、步驟、操作、元件及/或組件之存在,但不排除一或多個其他特徵、步驟、操作、元件、組件及/或其群組之存在或添加。如本文中所使用,術語「及/或」包括相關聯所列項目中的一或多者的任何及所有組合,且可被縮寫為「/」。The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. For example, as used herein, the singular forms "a/an" and "the" are intended to include the plural forms as well, unless the context clearly dictates otherwise. It should be further understood that the term "comprises and/or comprising" when used in this specification specifies the presence of stated features, steps, operations, elements and/or components, but does not exclude one or more other features , the existence or addition of steps, operations, elements, components and/or groups thereof. As used herein, the term "and/or" includes any and all combinations of one or more of the associated listed items and may be abbreviated as "/".

為易於描述,本文中可使用空間相對術語,諸如「在……之下」、「在……下方」、「下部」、「在……之上」、「上部」及其類似者來描述如圖式中所說明之一個元件或特徵與另一(些)元件或特徵之關係。應理解,除圖式中所描繪的定向之外,空間相對術語意欲涵蓋裝置在使用或操作中的不同定向。舉例而言,若將圖式中之裝置翻轉,則隨後將描述為「在」其他元件或特徵「之下」或「底下」之元件定向於其他元件或特徵「上方」。因此,例示性術語「在……之下」可涵蓋在……之上及在……之下的定向兩者。裝置可以其他方式定向(旋轉90度或處於其他定向),且本文中所使用的空間相對描述詞可相應地進行解釋。類似地,除非另外特別指示,否則本文中所使用的術語「向上」、「向下」、「豎直」、「水平」及其類似者僅用於出於解釋之目的。For ease of description, spatially relative terms such as "under", "below", "lower", "above", "upper" and the like may be used herein to describe The relationship between one element or feature and another element or feature illustrated in the drawings. It will be understood that spatially relative terms are intended to encompass different orientations of the device in use or operation in addition to the orientation depicted in the figures. For example, if the device in the figures is turned over, elements described as "below" or "beneath" other elements or features would then be oriented "above" the other elements or features. Thus, the exemplary term "beneath" can encompass both an orientation of above and below. The device may be otherwise oriented (rotated 90 degrees or at other orientations) and the spatially relative descriptors used herein interpreted accordingly. Similarly, the terms "upward," "downward," "vertical," "horizontal," and the like are used herein for purposes of explanation only, unless specifically indicated otherwise.

儘管本文中可使用術語「第一」及「第二」描述不同特徵/元件(包括步驟),但除非上下文另外指示,否則此等特徵/元件不應受此等術語限制。此等術語可用於區分一個特徵/元件與另一個特徵/元件。因此,在不脫離本發明之教示的情況下,下文論述之第一特徵/元件可稱為第二特徵/元件,且類似地,下文論述之第二特徵/元件可稱為第一特徵/元件。Although the terms "first" and "second" may be used herein to describe various features/elements (including steps), these features/elements should not be limited by these terms unless the context dictates otherwise. These terms are used to distinguish one feature/element from another feature/element. Thus, a first feature/element discussed hereinafter could be termed a second feature/element and, similarly, a second feature/element discussed hereinafter could be termed a first feature/element without departing from the teachings of the present invention. .

在整個說明書及後接申請專利範圍中,除非上下文另外要求,否則字詞「包含(comprise)」及其變化形式,諸如「包含(comprises/comprising)」意謂各個組件可共同地應用於方法及製品(例如,包括裝置之組合物及設備以及方法)中。舉例而言,術語「包含」應理解為暗示包括任何所陳述之元件或步驟,而非排除任何其他元件或步驟。Throughout this specification and claims that follow, unless the context requires otherwise, the word "comprise" and variations thereof, such as "comprises/comprising", mean that various elements can be collectively applied to methods and In articles of manufacture (eg, compositions and devices including devices and methods). For example, the term "comprising" should be understood as implying the inclusion of any stated elements or steps, not the exclusion of any other elements or steps.

一般而言,本文中所描述設備及方法中之任一者應理解為包括性的,但組件及/或步驟的所有或子集可替代地為排它性的,且可表述為「由」各個組件、步驟、子組件或子步驟「組成」或可替代地「基本上由」各個組件、步驟、子組件或子步驟「組成」。In general, any of the apparatus and methods described herein should be understood to be inclusive, but all or a subset of the components and/or steps may alternatively be exclusive and may be expressed as "by" Each component, step, sub-component or sub-step "consists of" or alternatively "consists essentially of" each component, step, sub-component or sub-step.

如本文說明書及申請專利範圍中所使用(包括如實例中所使用),除非另外明確規定,否則所有數字可讀作如同以字詞「約」或「大致」開頭,即使該術語並不明確地呈現。當描述程度及/或位置時,可使用片語「約」或「大致」以指示所描述值及/或位置在值及/或位置之合理預期範圍內。舉例而言,數值可具有為+/-0.1%之規定值(或值範圍)、+/-1%之規定值(或值範圍)、+/-2%之規定值(或值範圍)、+/-5%之規定值(或值範圍)、+/-10%之規定值(或值範圍)等之值。除非上下文另外指示,否則本文中所給出之任何數值亦應理解為包括約或近似該值。舉例而言,若揭示值「10」,則亦揭示「約10」。本文中所列舉之任何數值範圍意欲包括其中包含之所有子範圍。亦應理解,當揭示一值時,亦揭示「小於或等於」該值、「大於或等於該值」及值之間的可能範圍,如熟習此項技術者所適當地理解。舉例而言,若揭示值「X」,則亦揭示「小於或等於X」以及「大於或等於X」(例如,其中X為數值)。亦應理解,在整個申請案中,資料係以多個不同格式提供,且此資料表示端點及起點,以及資料點之任何組合之範圍。舉例而言,若揭示特定資料點「10」及特定資料點「15」,則應理解,視為揭示大於、大於或等於、小於、小於或等於及等於10及15以及在10與15之間。亦應理解,亦揭示兩個特定單元之間的每一單元。舉例而言,若揭示10及15,則亦揭示11、12、13及14。As used in the specification and claims herein (including as used in the examples), unless expressly stated otherwise, all numbers can be read as if preceded by the word "about" or "approximately," even if that term is not expressly stated presented. When describing degrees and/or locations, the phrases "about" or "approximately" may be used to indicate that the described value and/or location is within a reasonable expectation of the value and/or location. For example, a value may have a stated value (or range of values) of +/-0.1%, a stated value (or range of values) of +/-1%, a stated value (or range of values) of +/-2%, +/-5% of the specified value (or value range), +/-10% of the specified value (or value range), etc. Unless the context dictates otherwise, any numerical value given herein should also be understood to include about or approximating that value. For example, if the value "10" is revealed, "about 10" is also revealed. Any numerical range recited herein is intended to include all subranges subsumed therein. It is also understood that when a value is disclosed, "less than or equal to" the value, "greater than or equal to the value" and possible ranges between values are also disclosed, as properly understood by those skilled in the art. For example, if the value "X" is disclosed, then "less than or equal to X" and "greater than or equal to X" are also disclosed (eg, where X is a numerical value). It should also be understood that throughout this application, data is provided in a number of different formats, and that this data represents endpoints and starting points, as well as ranges for any combination of data points. For example, if a specific data point "10" and a specific data point "15" are disclosed, it is understood to be deemed to disclose greater than, greater than or equal to, less than, less than or equal to, and equal to 10 and 15, and between 10 and 15 . It is also understood that every element between the two specified elements is also disclosed. For example, if 10 and 15 are disclosed, then 11, 12, 13 and 14 are also disclosed.

儘管上文描述不同說明性實施例,但在不脫離如申請專利範圍所述之本發明之範疇的情況下,可對各種實施例進行多種改變中之任一者。舉例而言,在替代實施例中,可通常改變執行不同所描述之方法步驟之次序,且在其他替代實施例中,可完全跳過一或多個方法步驟。在一些實施例中且並不在其他實施例中,可包括不同裝置及系統實施例之視情況存在之特徵。因此,主要出於例示性目的提供前述描述且不應解釋為限制如申請專利範圍中所闡述的本發明之範疇。While various illustrative embodiments are described above, any of a number of changes may be made to the various embodiments without departing from the scope of the invention as described in the claims. For example, in alternative embodiments, the order of performing differently described method steps may generally be varied, and in other alternative embodiments, one or more method steps may be skipped entirely. In some embodiments, and not in others, optional features of different device and system embodiments may be included. Accordingly, the foregoing description is provided primarily for illustrative purposes and should not be construed as limiting the scope of the invention as set forth in the claims.

舉例而言且並不限制,本文中包括之實例及說明展示可實踐標的物之特定實施例。如所提及,可利用其他實施例且自本文中導出其他實施例,使得可在不脫離本發明之範疇的情況下作出結構及邏輯的替代及改變。本發明標的物之此類實施例在本文中可個別地或共同地由術語「發明」提及,此僅為方便起見且不意欲將此申請案之範疇自願地限於任何單一發明或發明性概念(在實際上揭示多於一個概念之情況下)。因此,儘管本文中已說明且描述特定實施例,但經計算以達成相同目的之任何配置可替代所展示之特定實施例。本發明意欲涵蓋各種實施例之任何及所有調適或變化。熟習此項技術者在審閱上述描述之後,將對上述實施例的組合以及本文中未具體描述之其他實施例顯而易見。By way of example and not limitation, the examples and descriptions included herein show specific embodiments of the subject matter that can be practiced. As mentioned, other embodiments may be utilized and derived therefrom, such that structural and logical substitutions and changes may be made without departing from the scope of the present invention. Such embodiments of the present subject matter may be referred to herein individually or collectively by the term "invention," which is for convenience only and is not intended to limit the scope of this application to any single invention or inventive concept. Concepts (where more than one concept is actually disclosed). Thus, while specific embodiments have been illustrated and described herein, any configuration calculated to achieve the same purpose may be substituted for the specific embodiments shown. This disclosure is intended to cover any and all adaptations or variations of various embodiments. Combinations of the above-described embodiments, and other embodiments not specifically described herein, will be apparent to those of skill in the art upon reviewing the above description.

275:入口 503:聚合基質材料 507:肝細胞 505:結構 601:步驟 603:步驟 605:步驟 607:步驟 609:步驟 611:步驟 613:步驟 615:步驟 617:步驟 700:設備 702:殼體 704:設備的一部分 706:容納腔室 708:腔室 712:分配器噴嘴 716:多孔盤 718:熱/溫度調節器 720:小滴形成組合件 722:分配器 724:控制器 726:泵 728:感測器 730:微流體晶片 731:出口 733:入口 735:第二入口孔口/入口孔口 737:接合區域/交叉區域 739:元件 741:通道出口/遞送路徑 743:不混溶的流體出口/連接路徑 743':不混溶的流體出口/連接路徑 803:微型類器官球體 805:細胞 839:通道區域 903:小滴 909:通道/交叉通道 909':交叉通道 911:第一通道/通道 937:接合區域 939:出口通道 1005:患者衍生之微型類器官球體 1008:不混溶的材料 1801:步驟 1803:步驟 1805:步驟 1807:步驟 1809:步驟 1811:步驟 1813:步驟 1815:步驟 1905:技術 1905' :技術 1905'':技術 1905''':技術 1905'''':技術 1905''''':技術 1907:技術 1921:技術 1923:技術 1925:技術 1927:技術 1929:技術 1931:技術 1933:技術 1935:技術 2002:不混溶的材料 2004:儲集器及/或孔口;輸入端 2006:未聚合材料 2008:儲集器或孔口;輸入端 2010:第三組孔口;輸入端 2012:接合處 2014:輸出端 2103:第一通道 2107:第二通道/微型類器官球體 2109:交叉通道 2109':交叉通道 275: Entrance 503: polymer matrix material 507: Liver cells 505: structure 601: Step 603: Step 605: Step 607: Step 609: Step 611: Step 613: Step 615: Step 617: Step 700: Equipment 702: Shell 704: part of the device 706: holding chamber 708: chamber 712: Distributor nozzle 716: porous disc 718: Heat/Temperature Regulator 720: Droplet forming assembly 722: Allocator 724: Controller 726: pump 728: sensor 730: microfluidic chip 731: export 733:Entrance 735: Second entrance orifice / entrance orifice 737:joint area/intersection area 739: Element 741: Channel exit/delivery path 743: Immiscible fluid outlet/connection path 743': Immiscible fluid outlet/connection path 803: Micro Organoid Spheroids 805: cells 839: Channel area 903: droplet 909: Channel/Cross Channel 909': cross aisle 911: first channel/channel 937: joint area 939: exit channel 1005:Patient-derived micro-organoid spheroids 1008: immiscible materials 1801: step 1803: step 1805: step 1807: step 1809: step 1811: step 1813: step 1815: step 1905: Technology 1905' : Technology 1905'': Technology 1905''': Technology 1905'''': Technology 1905''''': Technology 1907: Technology 1921: Technology 1923: Technology 1925: Technology 1927: Technology 1929: Technology 1931: Technology 1933: Technology 1935: Technology 2002: Immiscible materials 2004: Reservoirs and/or orifices; inputs 2006: Unpolymerized materials 2008: Reservoir or orifice; input 2010: Third set of orifices; input 2012: Junction 2014: Output 2103: first channel 2107: Second Passage/Miniature Organoid Spheroids 2109:Cross passage 2109': Cross Passage

本發明之新穎特徵在隨後的申請專利範圍中細緻闡述。將參考闡述利用本發明原理之說明性實施例及其附圖的以下詳細描述來獲得對本發明之特徵及優勢的較佳理解:The novel features of the invention are set forth in detail in the claims that follow. A better understanding of the features and advantages of the invention will be gained by reference to the following detailed description and accompanying drawings which illustrate illustrative embodiments utilizing the principles of the invention:

圖1A至圖1C說明如本文中所描述形成的患者衍生之微型類器官球體,每個微型類器官球體包括一個解離之初代組織細胞,該微型類器官球體在形成之後培養一天(圖1A)、在形成之後培養三天(圖1B)及在形成之後培養七天(圖1C)。細胞來源於大腸直腸癌(CRC)組織。Figures 1A-1C illustrate patient-derived micro-organoid spheroids formed as described herein, each micro-organoid spheroid comprising a dissociated primary tissue cell, cultured for one day after formation (Figure 1A), Cultured for three days after formation (Fig. IB) and seven days after formation (Fig. 1C). Cells were derived from colorectal cancer (CRC) tissue.

圖2A至圖2C說明如本文中所描述形成的患者衍生之微型類器官球體,每個微型類器官球體包括五個解離之初代組織細胞,該微型類器官球體在形成之後培養一天(圖2A)、在形成之後培養三天(圖2B)及在形成之後培養七天(圖2C)。細胞來源於大腸直腸癌(CRC)組織。Figures 2A-2C illustrate patient-derived micro-organoid spheroids formed as described herein, each micro-organoid spheroid comprising five dissociated primary tissue cells, cultured for one day after formation (Figure 2A) , three days after formation (Fig. 2B) and seven days after formation (Fig. 2C). Cells were derived from colorectal cancer (CRC) tissue.

圖3A至圖3C說明如本文中所描述形成的患者衍生之微型類器官球體,每個微型類器官球體包括二十個解離之初代組織細胞,該微型類器官球體在形成之後培養一天(圖3A)、在形成之後培養三天(圖3B)及在形成之後培養七天(圖3C)。如圖1A至圖1C及圖2A至圖2C中,細胞來源於大腸直腸癌(CRC)組織。Figures 3A-3C illustrate patient-derived micro-organoid spheroids formed as described herein, each micro-organoid spheroid comprising twenty dissociated primary tissue cells, cultured for one day after formation (Figure 3A ), three days after formation (Fig. 3B) and seven days after formation (Fig. 3C). As shown in Fig. 1A to Fig. 1C and Fig. 2A to Fig. 2C, the cells are derived from colorectal cancer (CRC) tissue.

圖4A至圖4E說明如本文中所描述形成的患者衍生之微型類器官球體之實例,每個微型類器官球體包括十個解離之初代組織細胞。圖4A展示形成之後不久的微型類器官球體(在低放大率下)。圖4B展示在培養兩天之後取得的圖4A之微型類器官球體中之一些之較高放大率視圖。圖4C展示培養三天之後的微型類器官球體。圖4D展示培養四天之後的微型類器官球體。圖4E展示培養五天之後的微型類器官球體。4A-4E illustrate examples of patient-derived micro-organoid spheroids formed as described herein, each micro-organoid spheroid comprising ten dissociated primary tissue cells. Figure 4A shows miniature organoid spheroids (at low magnification) shortly after formation. Figure 4B shows a higher magnification view of some of the miniature organoid spheroids of Figure 4A taken after two days in culture. Figure 4C shows miniature organoid spheroids after three days in culture. Figure 4D shows miniature organoid spheroids after four days in culture. Figure 4E shows miniature organoid spheroids after five days in culture.

圖5A至圖5B說明來自正常小鼠肝臟肝細胞的如本文中所描述形成的微型類器官球體之實例,該微型類器官球體在形成之後培養一天(圖5A)、在形成之後培養十天(圖5B)。小鼠肝細胞取自正常(例如,非病變)小鼠肝臟。Figures 5A-5B illustrate examples of miniature organoid spheroids formed as described herein from normal mouse liver hepatocytes cultured one day after formation (Figure 5A) and ten days after formation ( Figure 5B). Mouse hepatocytes are obtained from normal (eg, non-diseased) mouse livers.

圖6說明如本文中所描述的自初代組織(例如,生檢)樣本形成患者衍生之微型類器官球體之方法。6 illustrates a method of forming patient-derived mini-organoid spheroids from primary tissue (eg, biopsy) samples as described herein.

圖7A示意性說明用於形成如本文中所描述的患者衍生之微型類器官球體的設備之一個實例,包括作為組合件之部分的微流體晶片。圖7B為諸如展示於圖7A中的設備的一個實例微流體晶片部分的透視圖。圖7C示意性說明用於形成患者衍生之微型類器官球體的設備的微流體組合件之一部分,諸如展示於圖7A中的一者。Figure 7A schematically illustrates one example of an apparatus for forming patient-derived micro-organoid spheroids as described herein, including a microfluidic chip as part of the assembly. 7B is a perspective view of an example microfluidic wafer portion of a device such as that shown in FIG. 7A. Figure 7C schematically illustrates a portion of a microfluidic assembly of an apparatus for forming patient-derived micro-organoid spheroids, such as the one shown in Figure 7A.

圖8展示繪示使用諸如展示於圖7A中的設備形成的複數個患者衍生之微型類器官球體的影像之一個實例,其展示聚合之後不久的患者衍生之微型類器官球體,在由設備等分之前懸浮於含有不混溶之流體(例如,油)的通道內。8 shows an example of an image depicting a plurality of patient-derived micro-organoid spheroids formed using an apparatus such as that shown in FIG. Previously suspended in a channel containing an immiscible fluid (eg, oil).

圖9為一種用於形成患者衍生之微型類器官球體的設備的原型微流體組合件之一部分的影像,與展示於圖7C中彼等者類似,說明患者衍生之微型類器官球體之形成。Figure 9 is an image of a portion of a prototype microfluidic assembly of an apparatus for forming patient-derived micro-organoid spheroids, similar to those shown in Figure 7C, illustrating the formation of patient-derived micro-organoid spheroids.

圖10說明聚合之後不久的如本文中所描述的複數個患者衍生之微型類器官球體;患者衍生之微型類器官球體懸浮於不混溶之流體中。Figure 10 illustrates a plurality of patient-derived micro-organoid spheroids as described herein shortly after polymerization; the patient-derived micro-organoid spheroids are suspended in an immiscible fluid.

圖11A至圖11B說明在低放大率(圖11A)及較高放大率(圖11B)下,形成之後不久及懸浮於不混溶的流體(例如,油)中的複數個患者衍生之微類器官球體之另一實例。11A-11B illustrate a plurality of patient-derived microclasses shortly after formation and suspended in an immiscible fluid (e.g., oil) at low magnification (FIG. 11A) and higher magnification (FIG. 11B). Another example of an organ spheroid.

圖12A至圖12B展示在低放大率(圖12A)及較高放大率(圖12B)下,在形成患者衍生之微型類器官球體的數小時內自不混溶的流體分離之後的複數個患者衍生之微型類器官球體。Figures 12A-12B show multiple patients after isolation from immiscible fluids within hours of formation of patient-derived miniature organoid spheroids at low magnification (Figure 12A) and higher magnification (Figure 12B) Derived miniature organoid spheroids.

圖13為展示如本文中所描述形成的複數個患者衍生之微型類器官球體的影像之另一實例。13 is another example of an image showing a plurality of patient-derived micro-organoid spheroids formed as described herein.

圖14為說明自例示性生檢樣本形成的複數個患者衍生之微型類器官球體的直徑之大小分佈的圖表。14 is a graph illustrating the size distribution of diameters of a plurality of patient-derived micro-organoid spheroids formed from exemplary biopsy samples.

圖15A至圖15B分別說明聚合之後自解離之組織生檢樣本及流體基質材料形成的複數個患者衍生之微型類器官球體之一個實例的低及較高放大率視圖。圖15A為未染色影像,而在圖15B中類器官球體已用錐蟲藍染色以展示微型類器官球體中的解離細胞為存活的。15A-15B illustrate low and higher magnification views, respectively, of an example of a plurality of patient-derived micro-organoid spheroids formed from dissociated tissue biopsy samples and fluid matrix material after polymerization. Figure 15A is an unstained image, while in Figure 15B the organoid spheroids have been stained with trypan blue to show that the dissociated cells in the miniature organoid spheroids are viable.

圖16A至圖16B為另一實例,與展示於圖15A至圖15B中的彼等實例類似,分別展示複數個患者衍生之微型類器官球體之一個實例的低及較高放大率視圖。圖16A為未染色影像,而在圖16B中類器官球體已用錐蟲藍(箭頭)染色以展示微型類器官球體中的解離細胞,其指示細胞在微型類器官球體內保持活力(例如,活的)。FIGS. 16A-16B are another example, similar to those shown in FIGS. 15A-15B , showing low and higher magnification views, respectively, of one example of a plurality of patient-derived micro-organoid spheroids. Figure 16A is an unstained image, while in Figure 16B the organoid spheroids have been stained with trypan blue (arrow) to demonstrate dissociated cells in the mini-organoid spheroids, which indicates that the cells remain viable (e.g., viable) within the mini-organoid spheroids. of).

圖17A至圖17E說明在自患者腫瘤生檢形成的此實例中分析複數個患者衍生之微型類器官球體以判定對多種藥物調配物之藥物反應概況之方法的一個實例。所說明的程序自生檢至結果花費少於兩週(例如,大致一週)。Figures 17A-17E illustrate one example of a method for analyzing a plurality of patient-derived mini-organoid spheroids to determine drug response profiles to multiple drug formulations in this example formed from patient tumor biopsies. The illustrated procedure takes less than two weeks (eg, approximately one week) from biopsy to result.

圖18示意性地說明用於治療患者之方法的一實例,包括形成及使用複數個患者衍生之微型類器官球體作為治療程序之部分。Figure 18 schematically illustrates an example of a method for treating a patient, including forming and using a plurality of patient-derived micro-organoid spheroids as part of a treatment procedure.

圖19示意性地說明用於治療患者之方法的一實例,包括多次重複快速形成及分析複數個患者衍生之微型類器官球體作為治療程序之部分。Figure 19 schematically illustrates an example of a method for treating a patient, including multiple iterations of rapid formation and analysis of a plurality of patient-derived micro-organoid spheroids as part of a treatment procedure.

圖20示意性地說明用於形成如本文中所描述的複數個患者衍生之微型類器官球體的設備之一部分的一個變化形式。Figure 20 schematically illustrates one variation of a portion of an apparatus for forming a plurality of patient-derived micro-organoid spheroids as described herein.

圖21示意性地說明操作與展示於圖20中的彼等者類似的用於形成複數個患者衍生之微型類器官球體的設備之方法。FIG. 21 schematically illustrates a method of operating an apparatus similar to those shown in FIG. 20 for forming a plurality of patient-derived micro-organoid spheroids.

圖22A至圖22D說明使用如本文中所描述的複數個患者衍生之微型類器官球體鑑別藥物抗性之方法的驗證的一個實例。圖22A說明傳統(「2D」)腫瘤細胞分析方法預測藥物抗性之用途。圖22B說明如本文中所描述的患者衍生之微型類器官球體方法之一個實例分析藥物抗性進而預測藥物敏感性的用途。圖22C及圖22D展示與傳統培養細胞不同的基於患者衍生之微型類器官球體的方法精確地預測腫瘤之實際反應(藥物反應性)。Figures 22A-22D illustrate one example of validation of a method for identifying drug resistance using a plurality of patient-derived mini-organoid spheroids as described herein. Figure 22A illustrates the use of traditional ("2D") tumor cell analysis methods to predict drug resistance. Figure 22B illustrates the use of one example of the patient-derived micro-organoid spheroid method as described herein to analyze drug resistance and thereby predict drug sensitivity. Figures 22C and 22D demonstrate that the patient-derived micro-organoid spheroid-based approach accurately predicts the actual tumor response (drug responsiveness) as opposed to traditional cultured cells.

圖23A至圖23D說明驗證如本文中所描述的患者衍生之微型類器官球體鑑別藥物抗性之用途的另一實例,其展示對奧沙利鉑(Oxaliplatin)及伊立替康(Irinotecan)兩者所預測的藥物反應與用此等藥物治療後的實際腫瘤反應一致。Figures 23A-23D illustrate another example of validating the use of patient-derived mini-organoid spheroids as described herein to identify drug resistance, showing resistance to both oxaliplatin and irinotecan Predicted drug responses were consistent with actual tumor responses following treatment with these drugs.

圖24說明使用如本文中所描述的患者衍生之微型類器官球體的藥物篩選之一個實例,其中單個腫瘤生檢可極其快速地(例如,在少於兩週內)產生大量複數個幾乎相同的微型類器官球體及並行地針對大量藥物調配物(例如,展示27種)進行快速測試。Figure 24 illustrates an example of drug screening using patient-derived mini-organoid spheroids as described herein, where a single tumor biopsy can very rapidly (e.g., in less than two weeks) generate large numbers of nearly identical Rapid testing of miniature organoid spheroids and in parallel against a large number of drug formulations (eg, 27 displayed).

圖25A至圖25B說明自小鼠肝臟組織形成的小鼠肝臟微型類器官球體之實例,其具有300 µm之直徑及每個類器官球體有1個細胞。圖25A展示在第1天的微型類器官球體,且圖25B展示在第10天的微型類器官球體。Figures 25A-25B illustrate examples of mouse liver micro-organoid spheroids formed from mouse liver tissue with a diameter of 300 µm and 1 cell per organoid spheroid. Figure 25A shows mini-organoid spheroids at day 1, and Figure 25B shows mini-organoid spheroids at day 10.

圖26A至圖26B說明自部分肝切除小鼠肝臟組織形成的小鼠肝臟微型類器官球體之實例,其與圖25A至圖25B中所展示的彼等者類似,具有300 µm之直徑,及每個類器官球體有25個細胞。圖26A展示在第1天的微型類器官球體,且圖26B展示在第10天的微型類器官球體。Figures 26A-26B illustrate examples of mouse liver micro-organoid spheroids formed from partially hepatectomized mouse liver tissue, similar to those shown in Figures 25A-25B , having a diameter of 300 µm, and each Each organoid spheroid has 25 cells. Figure 26A shows mini-organoid spheroids at day 1, and Figure 26B shows mini-organoid spheroids at day 10.

圖27A至圖27C說明自人類肝臟組織形成的人類肝臟微型類器官球體之實例。圖27A展示在第1天的微型類器官球體,接種有40個細胞/小滴。圖27B及圖27C展示在第18天的微型類器官球體。在圖27B中,微器官球體為肝細胞樣結構,而圖27C展示膽管上皮樣微型類器官球體。27A-27C illustrate examples of human liver micro-organoid spheroids formed from human liver tissue. Figure 27A shows miniature organoid spheroids at day 1, seeded with 40 cells/droplet. Figure 27B and Figure 27C show miniature organoid spheroids at day 18. In Figure 27B, the micro-organ spheroids are hepatocyte-like structures, while Figure 27C shows biliary epithelial-like micro-organoid spheroids.

圖28A至圖28D展示自患者衍生之異種移植物腫瘤株產生的微型類器官球體之實例,其具有300 µm之直徑,及每個微型類器官球體有1個細胞,圖28A展示在第1天的微型類器官球體,圖28B展示在第3天的微型類器官球體,圖28C展示在第5天的微型類器官球體,且圖28D展示在第7天的微型類器官球體。Figures 28A-28D show examples of mini-organoid spheroids generated from patient-derived xenograft tumor lines with a diameter of 300 µm and 1 cell per mini-organoid spheroid, shown in Figure 28A at day 1 Figure 28B shows mini-organoid spheroids at day 3, Figure 28C shows mini-organoid spheroids at day 5, and Figure 28D shows mini-organoid spheroids at day 7.

圖29A至圖29D展示自患者衍生之異種移植物模型產生的微型類器官球體之實例,其具有300 µm之直徑,及每個類器官球體有5個細胞。圖29A展示在第1天的微型類器官球體,圖29B展示在第3天的微型類器官球體,圖29C展示在第5天的微型類器官球體,且圖29D展示在第7天的微型類器官球體。Figures 29A-29D show examples of miniature organoid spheroids generated from a patient-derived xenograft model with a diameter of 300 µm and 5 cells per organoid spheroid. Figure 29A shows mini-organoid spheroids on day 1, Figure 29B shows mini-organoid spheroids on day 3, Figure 29C shows mini-organoid spheroids on day 5, and Figure 29D shows mini-organoid spheroids on day 7 organ sphere.

圖30為將傳統類器官及自大腸直腸癌患者衍生之類器官形成的微型類器官球體對奧沙利鉑的反應進行比較之圖,其展示傳統類器官及微型類器官球體之相當的反應。Figure 30 is a graph comparing the response to oxaliplatin of conventional organoids and mini-organoid spheroids formed from organoids derived from colorectal cancer patients, showing comparable responses of traditional organoids and mini-organoid spheroids.

圖31為將傳統類器官及自兩個大腸直腸癌患者衍生之異種移植物模型形成的微型類器官球體對SN38 (7-乙基-10-羥基-喜樹鹼)的反應進行比較之圖,其展示相當的反應。Figure 31 is a graph comparing the response to SN38 (7-ethyl-10-hydroxy-camptothecin) of conventional organoids and mini-organoid spheroids formed from xenograft models derived from two colorectal cancer patients, It exhibits considerable response.

圖32為將傳統類器官及自大腸直腸癌患者衍生之異種移植物模型形成的微型類器官球體對5-FU (氟尿嘧啶)的反應進行比較之圖,其展示相當的反應。Figure 32 is a graph comparing the responses to 5-FU (fluorouracil) of conventional organoids and miniature organoid spheroids formed from colorectal cancer patient-derived xenograft models showing comparable responses.

圖33A及圖33B展示使用小鼠肝臟微型類器官球體的毒性分析之實例。圖33A展示對照組中小鼠肝臟微型類器官球體中的組織之大小相對較大(如箭頭所指示)。相比之下,在展示乙醯胺苯酚(10 mM)治療組的圖33A中,大部分微型類器官球體中之的組織較小且含有許多死亡細胞。Figures 33A and 33B show examples of toxicity assays using mouse liver mini-organoid spheroids. Figure 33A shows that the size of the tissue in the mouse liver mini-organoid spheroids was relatively larger (as indicated by the arrows) in the control group. In contrast, in Figure 33A showing the acetaminophen (10 mM) treated group, most of the tissue within the mini-organoid spheroids was smaller and contained many dead cells.

圖34A及圖34B展示使用人類肝臟微型類器官球體的毒性分析之實例。圖34A展示在包括組織結構的對照組中觀測到的典型人類肝臟微型類器官球體(由箭頭指示)。圖34B展示乙醯胺苯酚(10 mM)治療組中的微型類器官球體,其展示非典型組織結構(箭頭)及碎片。Figures 34A and 34B show examples of toxicity assays using human liver mini-organoid spheroids. Figure 34A shows typical human liver micro-organoid spheroids (indicated by arrows) observed in controls including tissue structures. Figure 34B shows miniature organoid spheroids in the acetaminophen (10 mM) treatment group showing atypical tissue architecture (arrows) and debris.

601:步驟 601: Step

603:步驟 603: Step

605:步驟 605: Step

607:步驟 607: Step

609:步驟 609: Step

611:步驟 611: Step

613:步驟 613: Step

615:步驟 615: Step

617:步驟 617: Step

Claims (28)

一種用於個人化癌症療法之精準藥物篩選的方法,該方法包含: 接收來自患者腫瘤之組織樣本; 使該組織樣本解離以形成解離之組織樣本; 藉由以下操作自該解離之組織樣本形成患者衍生之微型類器官球體庫: 驅動該解離之組織樣本及未聚合之流體基質材料穿過微流體設備之一或多個通道,其中該微流體設備控制該一或多個通道內的壓力、流動速率或壓力及流動速率且將該微流體設備之所有或一部分的溫度維持在20℃或更低,以使得該解離之組織樣本及該未聚合之流體基質穿過層流中的該一或多個通道, 將該微流體設備內的該解離之組織樣本及該未聚合之流體基質材料合併以形成複數個未聚合之混合物小滴; 將該複數個未聚合之混合物小滴暴露於高於25℃之溫度以聚合該流體基質材料且形成該患者衍生之微型類器官球體,其中該患者衍生之微型類器官球體各自具有在50 µm與500 µm之間的直徑,其中分佈有1至200個解離之細胞; 將該患者衍生之微型類器官球體庫培養2至14天以形成成熟的患者衍生之微型類器官球體,該成熟的患者衍生之微型類器官球體具有複製其等取樣的該患者腫瘤之結構的結構化細胞群集;及 使用該患者衍生之微型類器官球體庫分析一或多種藥物療法。 A method for precision drug screening for personalized cancer therapy, the method comprising: Receive tissue samples from patient tumors; dissociating the tissue sample to form a dissociated tissue sample; A library of patient-derived mini-organoid spheroids was formed from the dissociated tissue sample by: Driving the dissociated tissue sample and unpolymerized fluid matrix material through one or more channels of a microfluidic device, wherein the microfluidic device controls pressure, flow rate, or pressure and flow rate within the one or more channels and will The temperature of all or a portion of the microfluidic device is maintained at 20° C. or lower such that the dissociated tissue sample and the unpolymerized fluid matrix pass through the one or more channels in laminar flow, combining the dissociated tissue sample and the unpolymerized fluid matrix material within the microfluidic device to form a plurality of unpolymerized mixture droplets; exposing the plurality of unpolymerized mixture droplets to a temperature greater than 25° C. to polymerize the fluid matrix material and form the patient-derived micro-organoid spheroids, wherein each of the patient-derived micro-organoid spheroids has a diameter between 50 μm and Between 500 µm in diameter, with 1 to 200 dissociated cells distributed in it; culturing the bank of patient-derived mini-organoid spheroids for 2 to 14 days to form mature patient-derived mini-organoid spheroids having a structure that replicates the structure of the patient's tumor from which they were sampled Cell clusters; and One or more drug therapies are analyzed using the patient-derived mini-organoid spheroid library. 如請求項1之方法,其中分析包含藉由將該成熟的患者衍生之微型類器官球體中之一或多者暴露於各藥物療法來並行地分析複數種藥物療法。The method of claim 1, wherein analyzing comprises analyzing a plurality of drug therapies in parallel by exposing one or more of the mature patient-derived micro-organoid spheroids to each drug therapy. 如請求項1之方法,其進一步包含基於該患者衍生之微型類器官球體對暴露於該一或多種藥物療法之反應表徵該一或多種藥物療法對該患者腫瘤之反應。The method of claim 1, further comprising characterizing the response of the one or more drug therapies to the patient's tumor based on the response of the patient-derived micro-organoid spheroids to exposure to the one or more drug therapies. 如請求項3之方法,其中接收該組織樣本與表徵該反應之間的時間為少於21天。The method of claim 3, wherein the time between receiving the tissue sample and characterizing the response is less than 21 days. 如請求項1之方法,其中形成該患者衍生之微型類器官球體庫進一步包含基於細胞數目及/或小滴尺寸分選該患者衍生之微型類器官球體。The method of claim 1, wherein forming the patient-derived micro-organoid spheroid library further comprises sorting the patient-derived micro-organoid spheroids based on cell number and/or droplet size. 如請求項1之方法,其中形成該患者衍生之微型類器官球體庫包含以光學方式分選該患者衍生之微型類器官球體或基於細胞數目及/或小滴大小分選該患者衍生之微型類器官球體。The method of claim 1, wherein forming the patient-derived micro-organoid spheroid library comprises optically sorting the patient-derived micro-organoid spheroids or sorting the patient-derived micro-organoid spheroids based on cell number and/or droplet size organ sphere. 如請求項1之方法,其中該分析包含分析多於50種不同藥物療法。The method of claim 1, wherein the analyzing comprises analyzing more than 50 different drug therapies. 如請求項7之方法,其中該一或多種藥物療法包括一或多種藥物之不同濃度、三種或更多種藥物之不同組合、兩種或更多種藥物之不同比率、一或多種藥物的不同載劑及/或一或多種藥物的不同給藥時間。The method of claim 7, wherein the one or more drug therapies include different concentrations of one or more drugs, different combinations of three or more drugs, different ratios of two or more drugs, different doses of one or more drugs Different timing of administration of the vehicle and/or one or more drugs. 如請求項1之方法,其中形成該患者衍生之微型類器官球體庫包含形成多於100至600個患者衍生之微型類器官球體。The method of claim 1, wherein forming the patient-derived micro-organoid spheroid library comprises forming more than 100 to 600 patient-derived micro-organoid spheroids. 如請求項1之方法,其中形成該患者衍生之微型類器官球體庫包含形成多於1,000個患者衍生之微型類器官球體。The method of claim 1, wherein forming the patient-derived micro-organoid spheroid library comprises forming more than 1,000 patient-derived micro-organoid spheroids. 如請求項1之方法,其中形成該患者衍生之微型類器官球體庫包含形成多於10,000個患者衍生之微型類器官球體。The method of claim 1, wherein forming the patient-derived micro-organoid spheroid library comprises forming more than 10,000 patient-derived micro-organoid spheroids. 如請求項1之方法,其中該微流體設備在形成該複數個未聚合之混合物小滴之前維持該解離之組織樣本及該未聚合之流體基質材料之黏度。The method of claim 1, wherein the microfluidic device maintains the viscosity of the dissociated tissue sample and the unpolymerized fluid matrix material before forming the plurality of unpolymerized mixture droplets. 如請求項1之方法,其中該微流體設備經結構設計以避免該解離之組織樣本在該一或多個通道內的堵塞。The method of claim 1, wherein the microfluidic device is structurally designed to avoid clogging of the dissociated tissue sample in the one or more channels. 如請求項13之方法,其中該微流體設備經結構設計以藉由具有100 µm或更大之通道直徑來避免堵塞。The method of claim 13, wherein the microfluidic device is structurally designed to avoid clogging by having a channel diameter of 100 μm or greater. 如請求項1之方法,其中該微流體設備經結構設計維持該一或多個通道內之近似恆定的壓力。The method of claim 1, wherein the microfluidic device is structurally designed to maintain an approximately constant pressure in the one or more channels. 如請求項1之方法,其中將該複數個未聚合之混合物小滴暴露於高於25℃之溫度包含將該複數個未聚合之混合物小滴暴露於30℃或更高之溫度。The method of claim 1, wherein exposing the plurality of unpolymerized mixture droplets to a temperature higher than 25° C. comprises exposing the plurality of unpolymerized mixture droplets to a temperature of 30° C. or higher. 如請求項1之方法,其中將該複數個未聚合之混合物小滴暴露於高於25℃之溫度包含使該複數個未聚合之混合物小滴流動至維持在該微流體設備之高於25℃的區域。The method of claim 1, wherein exposing the plurality of unpolymerized mixture droplets to a temperature higher than 25° C. comprises flowing the plurality of unpolymerized mixture droplets to a temperature maintained above 25° C. in the microfluidic device Area. 如請求項1之方法,其中該微流體設備維持該一或多個通道內之恆定流動速率。The method of claim 1, wherein the microfluidic device maintains a constant flow rate in the one or more channels. 如請求項1之方法,其中該解離之組織樣本在與該微流體設備內的該未聚合之流體基質材料合併之前所採取的路徑總長少於10 cm。The method of claim 1, wherein the total length of the path taken by the dissociated tissue sample before being combined with the unpolymerized fluid matrix material in the microfluidic device is less than 10 cm. 如請求項1之方法,其進一步包含量測該一或多種藥物療法對該患者衍生之微型類器官球體內之細胞的作用。The method of claim 1, further comprising measuring the effect of the one or more drug therapies on the cells in the patient-derived mini-organoid spheroid. 如請求項1之方法,其進一步包含藉由以下操作判定在一或多次投與該一或多種藥物療法中之一種藥物療法之後,該患者腫瘤仍對該藥物療法有反應:在該患者已用該藥物療法治療之後接收該第二患者腫瘤組織及自該第二患者腫瘤組織形成第二複數個患者衍生之微型類器官球體,將該第二複數個患者衍生之微型類器官球體中之至少一些暴露於該藥物療法,及量測該藥物療法對該第二複數個患者衍生之微型類器官球體中之該至少一些內的細胞的作用。The method of claim 1, further comprising determining that after one or more administrations of one of the one or more drug therapies, the patient's tumor still responds to the drug therapy by: receiving the second patient's tumor tissue following treatment with the drug therapy and forming a second plurality of patient-derived micro-organoid spheroids from the second patient's tumor tissue, at least one of the second plurality of patient-derived micro-organoid spheroids Some are exposed to the drug therapy, and the effect of the drug therapy is measured on cells within the at least some of the second plurality of patient-derived mini-organoid spheroids. 如請求項1之方法,其進一步包含用該一或多種藥物療法中之一種藥物療法治療該患者。The method of claim 1, further comprising treating the patient with one of the one or more drug therapies. 如請求項1之方法,其中患者組織樣本包含來自轉移性腫瘤之生檢樣本。The method of claim 1, wherein the patient tissue sample comprises a biopsy sample from a metastatic tumor. 如請求項1之方法,其中該患者衍生之微型類器官球體庫中之該患者衍生之微型類器官球體具有小於25%的大小變化。The method of claim 1, wherein the patient-derived micro-organoid spheroids in the patient-derived micro-organoid spheroid bank have a size variation of less than 25%. 如請求項1之方法,其中該患者衍生之微型類器官球體形成細胞之出芽群集及/或細胞之中空結構,該等細胞複製其等取樣的該患者腫瘤之結構。The method of claim 1, wherein the patient-derived micro-organoid spheroids form budding clusters of cells and/or hollow structures of cells that replicate the structure of the patient's tumor from which they were sampled. 如請求項1之方法,其中分析該一或多種藥物療法包含分析具有類似的大小及細胞數目的患者衍生之微型類器官球體。The method of claim 1, wherein analyzing the one or more drug therapies comprises analyzing patient-derived micro-organoid spheroids with similar size and cell number. 一種用於個人化癌症療法之精準藥物篩選的方法,該方法包含: 接收來自患者腫瘤的生檢或經切除組織樣本; 使該組織樣本解離以形成解離之組織樣本; 藉由以下操作由該解離之組織樣本形成多於100個患者衍生之微型類器官球體之庫: 驅動該解離之組織樣本及未聚合之流體基質材料穿過微流體設備之一或多個通道,其中該微流體設備控制該一或多個通道內的壓力、流動速率或壓力及流動速率且維持20℃或更低之溫度,以使得該解離之組織樣本及該未聚合之流體基質穿過層流中的該一或多個通道, 將該微流體設備內的該解離之組織樣本及該未聚合之流體基質材料合併以形成複數個未聚合之混合物小滴; 將該複數個未聚合之混合物小滴暴露於高於25℃之溫度以聚合該流體基質材料且形成該患者衍生之微型類器官球體,其中該患者衍生之微型類器官球體各自具有50 µm與500 µm之間的直徑,其中分佈有1至200個解離之細胞, 其中藉由大小、細胞之數目或大小及細胞之數目兩者分選該患者衍生之微型類器官球體; 將該患者衍生之微型類器官球體庫培養2至14天以形成成熟的患者衍生之微型類器官球體,該成熟的患者衍生之微型類器官球體具有複製其等生檢或經切除的該患者腫瘤之結構的結構化細胞群集;及 使用該患者衍生之微型類器官球體庫分析複數種藥物療法。 A method for precision drug screening for personalized cancer therapy, the method comprising: Receive a biopsy or resected tissue sample from the patient's tumor; dissociating the tissue sample to form a dissociated tissue sample; A bank of more than 100 patient-derived micro-organoid spheroids was formed from the dissociated tissue sample by: Driving the dissociated tissue sample and unpolymerized fluid matrix material through one or more channels of the microfluidic device, wherein the microfluidic device controls the pressure, flow rate, or pressure and flow rate in the one or more channels and maintains a temperature of 20° C. or lower, so that the dissociated tissue sample and the unpolymerized fluid matrix pass through the one or more channels in laminar flow, combining the dissociated tissue sample and the unpolymerized fluid matrix material within the microfluidic device to form a plurality of unpolymerized mixture droplets; exposing the plurality of unpolymerized mixture droplets to a temperature above 25° C. to polymerize the fluid matrix material and form the patient-derived micro-organoid spheres, wherein the patient-derived micro-organoid spheres each have a diameter of 50 μm and 500 μm. diameter between µm, in which 1 to 200 dissociated cells are distributed, wherein the patient-derived miniature organoid spheroids are sorted by size, number of cells or both; The patient-derived mini-organoid bank is cultured for 2 to 14 days to form mature patient-derived mini-organoids with reproducible biopsy or resected tumors of the patient Structured cell clusters of the structure; and Multiple drug therapies were analyzed using the patient-derived mini-organoid spheroid library. 一種用於個人化癌症療法之精準藥物篩選的方法,該方法包含: 接收來自患者腫瘤之組織樣本; 使該組織樣本解離以形成解離之組織樣本; 藉由以下操作自該解離之組織樣本形成多於1000個患者衍生之微型類器官球體之庫: 驅動該解離之組織樣本及未聚合之流體基質材料穿過微流體設備之一或多個通道,其中該微流體設備控制該一或多個通道內的壓力、流動速率或壓力及流動速率且維持20℃或更低之溫度,以使得該解離之組織樣本及該未聚合之流體基質穿過層流中的該一或多個通道, 將該微流體設備內的該解離之組織樣本及該未聚合之流體基質材料合併以形成複數個未聚合之混合物小滴; 將該複數個未聚合之混合物小滴暴露於高於25℃之溫度以聚合該流體基質材料且形成該患者衍生之微型類器官球體,其中該患者衍生之微型類器官球體各自具有50 µm與500 µm之間的直徑,其中分佈有1至200個解離之細胞, 其中藉由大小、細胞之數目或大小及細胞之數目兩者分選該患者衍生之微型類器官球體; 將該患者衍生之微型類器官球體培養2至14天以形成成熟的患者衍生之微型類器官球體,該成熟的患者衍生之微型類器官球體具有複製其等取樣的該患者腫瘤之結構的結構化細胞群集;及 藉由將複數種藥物療法中之各藥物療法暴露於該成熟的患者衍生之微型類器官球體中之一或多者來並行地測試該複數種藥物療法。 A method for precision drug screening for personalized cancer therapy, the method comprising: Receive tissue samples from patient tumors; dissociating the tissue sample to form a dissociated tissue sample; A bank of more than 1000 patient-derived micro-organoid spheroids was formed from the dissociated tissue sample by: Driving the dissociated tissue sample and unpolymerized fluid matrix material through one or more channels of the microfluidic device, wherein the microfluidic device controls the pressure, flow rate, or pressure and flow rate in the one or more channels and maintains a temperature of 20° C. or lower, so that the dissociated tissue sample and the unpolymerized fluid matrix pass through the one or more channels in laminar flow, combining the dissociated tissue sample and the unpolymerized fluid matrix material within the microfluidic device to form a plurality of unpolymerized mixture droplets; exposing the plurality of unpolymerized mixture droplets to a temperature above 25° C. to polymerize the fluid matrix material and form the patient-derived micro-organoid spheres, wherein the patient-derived micro-organoid spheres each have a diameter of 50 μm and 500 μm. diameter between µm, in which 1 to 200 dissociated cells are distributed, wherein the patient-derived miniature organoid spheroids are sorted by size, number of cells or both; culturing the patient-derived micro-organoid spheroids for 2 to 14 days to form mature patient-derived micro-organoid spheroids having a structure that replicates the structure of the patient's tumor from which they were sampled cell clusters; and The plurality of drug therapies are tested in parallel by exposing each of the drug therapies to one or more of the mature patient-derived mini-organoid spheroids.
TW110144981A 2020-12-02 2021-12-02 Precision drug screening for personalized cancer therapy TW202237858A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202063120719P 2020-12-02 2020-12-02
US63/120,719 2020-12-02
US202017118586A 2020-12-10 2020-12-10
US17/118,586 2020-12-10

Publications (1)

Publication Number Publication Date
TW202237858A true TW202237858A (en) 2022-10-01

Family

ID=81260464

Family Applications (1)

Application Number Title Priority Date Filing Date
TW110144981A TW202237858A (en) 2020-12-02 2021-12-02 Precision drug screening for personalized cancer therapy

Country Status (5)

Country Link
JP (1) JP2023554171A (en)
KR (1) KR20230143134A (en)
CN (2) CN218755788U (en)
TW (1) TW202237858A (en)
WO (1) WO2022119966A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024081586A1 (en) * 2022-10-12 2024-04-18 University Of Florida Research Foundation, Incorporated Collagen droplet-based 3d cell spheroids as 3d printable bio-ink

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2545482A1 (en) * 2003-11-10 2005-05-26 Platypus Technologies, Llc Substrates, devices, and methods for cellular assays
AU2006206426B2 (en) * 2005-01-20 2012-03-15 The Regents Of The University Of California Cellular microarrays for screening differentiation factors
WO2014153651A1 (en) * 2013-03-28 2014-10-02 The University Of British Columbia Microfluidic devices and methods for use thereof in multicellular assays of secretion
EP3161052A4 (en) * 2014-06-26 2018-03-21 Northeastern University Microfluidic device and method for analysis of tumor cell microenvironments
WO2016145242A1 (en) * 2015-03-11 2016-09-15 Agenus Inc. Methods and compositions for high throughput screening of biomolecules using gel microdrops

Also Published As

Publication number Publication date
WO2022119966A1 (en) 2022-06-09
KR20230143134A (en) 2023-10-11
CN218755788U (en) 2023-03-28
JP2023554171A (en) 2023-12-26
CN114410466A (en) 2022-04-29

Similar Documents

Publication Publication Date Title
US11555180B2 (en) Methods and apparatuses for patient-derived micro-organospheres
Dolega et al. Controlled 3D culture in Matrigel microbeads to analyze clonal acinar development
WO2019152767A1 (en) Organoids related to immunotherapy and methods of preparing and using the same
US20220062791A1 (en) Methods and apparatuses for purification of gel droplets supporting biological tissue
Wu et al. Rapid microfluidic formation of uniform patient-derived breast tumor spheroids
Zhou et al. Microfluidic device for primary tumor spheroid isolation
US20160281061A1 (en) Tissue array for cell spheroids and methods of use
US20210285054A1 (en) Precision drug screening for personalized cancer therapy
Tang et al. On-Chip cell–cell interaction monitoring at single-cell level by efficient immobilization of multiple cells in adjustable quantities
TW202237858A (en) Precision drug screening for personalized cancer therapy
US20230003716A1 (en) Methods and apparatuses for patient-derived microorganospheres
JP2023553838A (en) Organic microspheres used in personalized medicine and drug development
WO2023042156A1 (en) Methods and apparatus for testing immuno-oncology drugs and biologics using microorganospheres
JP2021004900A (en) Method for dealing with micro droplets including sample
WO2023212732A2 (en) Methods and apparatuses for testing hepatocyte toxicity using microorganospheres
WO2024054564A2 (en) Multiple myeloma microorganospheres
WO2024040145A1 (en) Methods of delivering components to microorganospheres
Dolega Developement of microtechnologies for 3D cell culture to study prostate acini formation and carcinogenesis