TWI806907B - Treating diabetes with genetically modified beta cells - Google Patents

Treating diabetes with genetically modified beta cells Download PDF

Info

Publication number
TWI806907B
TWI806907B TW107134978A TW107134978A TWI806907B TW I806907 B TWI806907 B TW I806907B TW 107134978 A TW107134978 A TW 107134978A TW 107134978 A TW107134978 A TW 107134978A TW I806907 B TWI806907 B TW I806907B
Authority
TW
Taiwan
Prior art keywords
cells
cell
human
cxcl12
beta
Prior art date
Application number
TW107134978A
Other languages
Chinese (zh)
Other versions
TW201923070A (en
Inventor
傑拉德F 史威斯
大衛 邱立奇
Original Assignee
美商Sdf生物製藥公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 美商Sdf生物製藥公司 filed Critical 美商Sdf生物製藥公司
Publication of TW201923070A publication Critical patent/TW201923070A/en
Application granted granted Critical
Publication of TWI806907B publication Critical patent/TWI806907B/en

Links

Landscapes

  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

Described herein are human transgenic beta cells expressing fugetactic levels of CXCL12 to a subject in need thereof. Also described herein are beta cells comprising a transgene comprising a nucleic acid sequence encoding CXCL12.

Description

以基因改造β細胞治療糖尿病Treating diabetes with genetically modified beta cells

本發明涉及基因改造人類β細胞以及使用此類細胞之方法。基因改造(轉殖基因)人類β細胞表現趨除(fugetactic)量之趨除劑,藉此賦予對人類單核免疫細胞之保護。在一個實施例中,趨除劑為例如CXCL12或CXCL13。在一個實施例中,轉殖基因β細胞包含載體,其中該載體包含編碼趨除劑,且較佳人類趨除劑之核酸序列。在一個實施例中,轉殖基因β細胞經進一步改造呈衰老細胞。本發明方法包括使用此等細胞在高血糖環境中表現胰島素,包括見於糖尿病患者,尤其第I型糖尿病患者中之胰島素。The present invention relates to genetically engineered human beta cells and methods of using such cells. Genetically engineered (transgenic) human beta cells express fugetactic amounts of the scavenger, thereby conferring protection against human mononuclear immune cells. In one embodiment, the chemoattractant is, for example, CXCL12 or CXCL13. In one embodiment, the transgenic β-cell comprises a vector, wherein the vector comprises a nucleic acid sequence encoding a keloid, and preferably a human keloid. In one embodiment, the transgenic beta cells are further engineered to be senescent cells. The methods of the invention include the use of such cells to express insulin in hyperglycemic environments, including insulin found in diabetic patients, especially type 1 diabetic patients.

β細胞負責在胰臟中產生胰島素。在患有第1型糖尿病(T1D)之個體中,β細胞會受到免疫系統攻擊及破壞,且因此患有T1D之個體無法有效產生其自身胰島素。Cloning technique used to create pancreatic cells for type 1 diabetes , Diabetes.Co.Uk, www.diabetes.co.uk/news/2014/apr/cloning-technique-used-to-create-pancreatic-cells-for-type-1-diabetes-94233303.html (2014年4月29日)。Beta cells are responsible for producing insulin in the pancreas. In individuals with type 1 diabetes (T1D), the beta cells are attacked and destroyed by the immune system, and thus individuals with T1D cannot effectively produce their own insulin. Cloning technique used to create pancreatic cells for type 1 diabetes , Diabetes.Co.Uk, www.diabetes.co.uk/news/2014/apr/cloning-technique-used-to-create-pancreatic-cells-for-type-1-diabetes-94233303.html (April 29, 2014) .

當個體之高血糖持續地超過個體之β細胞的能力時,會發生第2型糖尿病(T2D)且該T2D會導致β細胞功能失常、去分化及死亡。Felicia W Pagliuca及Douglas A. Melton, 「How to make a functional β-cell」,Development 2013, 140(12), 2472-2483。Type 2 diabetes (T2D) occurs when an individual's hyperglycemia persistently exceeds the capacity of the individual's beta cells and leads to beta cell dysfunction, dedifferentiation and death. Felicia W Pagliuca and Douglas A. Melton, "How to make a functional β-cell", Development 2013, 140(12), 2472-2483.

自正常供體向糖尿病接受者進行同種異體β細胞移植(亦稱為胰島細胞移植)已視為一種治療糖尿病之方法。然而,單核免疫細胞(T細胞、B細胞及NK細胞)之浸潤引起β細胞移植失敗。Cloning technique used to create pancreatic cells for type 1 diabetes , Diabetes.Co.Uk, http://www.diabetes.co.uk/news/2014/apr/cloning-technique-used-to-create-pancreatic-cells-for-type-1-diabetes-94233303.html (2014年4月29日);Alan H. Cruickshank及Emyr W Benbow, 「Recurrence of Diabetes」, Pathology of the Pancreas (第2版 1995);Felicia W Pagliuca及Douglas A. Melton, 「How to make a functional β-cell」,Development 2013, 140(12), 2472-2483。Allogeneic β-cell transplantation (also known as islet cell transplantation) from a normal donor to a diabetic recipient has been considered as a treatment for diabetes. However, the infiltration of mononuclear immune cells (T cells, B cells and NK cells) caused the failure of β cell transplantation. Cloning Technique used to create Pancreatic Cells for Type 1 Diabetes , Diabetes.co.uk, http://www.diabetes.co.uk/news/2014/Cloning-technique- USED-Create-Pancreative-Cells-FORL-TYPE-DIABETES-94233303.html (April 29, 2014); Alan H. Cruickshank and EMYR W Benbow, "Recurrence of Diabetes", Patholo Gy of the Pancreas (2nd Edition 1995); Felicia W Pagliuca and Douglas A. Melton, "How To Make A Functional β-Cell", Development 2013, 140 (12), 2472-2483.

當前臨床實踐為經由門靜脈將含有β細胞之胰島移植於肝臟中,其中基本原理為將自胰臟釋放之大多數胰島素用於肝臟中且藉由微創手術即可易於到達該部位。然而,移植之後不久便有二分之一的β細胞死亡,且認為此係由於肝臟中之低氧張力、活性免疫反應及高含量之毒素及藥物。此外,經血液介導的瞬時發炎反應(IBMIR)將經移植之胰島包封於纖維蛋白凝塊中且增強對移植的免疫反應。因此,已測試用於移植的若干替代部位,包括腸道、腎囊、腸網膜及皮下,其出於患者安全性可為最佳的,但尚未針對胰島素之全身性釋放進行充分評估。Current clinical practice is to transplant islets containing β cells into the liver via the portal vein, where the rationale is that most of the insulin released from the pancreas is used in the liver and this site is easily accessible by minimally invasive procedures. However, one-half of the beta cells die shortly after transplantation, and this is thought to be due to low oxygen tension, active immune responses, and high levels of toxins and drugs in the liver. Furthermore, the blood-mediated transient inflammatory response (IBMIR) encapsulates transplanted islets in a fibrin clot and enhances the immune response to transplantation. Thus, several alternative sites for transplantation have been tested, including the bowel, renal capsule, omentum, and subcutaneously, which may be optimal for patient safety, but have not been adequately evaluated for systemic release of insulin.

為了防止其暴露於單核免疫細胞,已將β細胞包封於據報導會提供以下雙功能之裝置中:使此等細胞與免疫破壞分離且保護宿主不受移植物影響。當使用非自體性細胞(例如同種異體或異種細胞)來移植時或若將自體性細胞移植於自體免疫環境,諸如第1型糖尿病患者中,則需要免疫保護。此可藉由經由使用半透膜或骨架物理分離細胞來阻斷細胞反應來達成。此方法降低對免疫抑制之需求且近年來已在其他處加以詳細綜述(Sakata 等人,World J Gastrointest Pathophysiol. 2012;3:19-26;Qi, Adv Med. 2014;2014:429710)。包封亦防止細胞逃離移植物之位置,且(若需要)允許移除。此與不受控制之分化及生長特定相關,例如在移植有幹細胞衍生之胰臟前驅體的小鼠中通常觀測到畸胎瘤(Kroon等人,Nat Biotechnol. 2008;26:443-452;Kelly等人,Nat Biotechnol. 2011;29:750-756.)。畸胎瘤形成可由藉由使用細胞純化(Kelly等人,2011)或藉由改良分化方法產生由純成熟β細胞群組成之移植物來預防。若胰島素瘤/畸胎瘤係由移植細胞所形成,則包封亦預防可能的癌轉移。To prevent their exposure to mononuclear immune cells, beta cells have been encapsulated in devices that reportedly serve the dual function of isolating these cells from immune destruction and protecting the host from the graft. Immune protection is required when non-autologous cells (eg, allogeneic or xenogeneic cells) are used for transplantation or if autologous cells are transplanted in an autoimmune environment, such as in type 1 diabetic patients. This can be achieved by blocking the cellular response by physically separating the cells using a semi-permeable membrane or scaffold. This approach reduces the need for immunosuppression and has been reviewed in detail elsewhere in recent years (Sakata et al., World J Gastrointest Pathophysiol. 2012;3:19-26; Qi, Adv Med. 2014;2014:429710). Encapsulation also prevents cells from escaping the site of the graft and, if desired, allows removal. This is specifically associated with uncontrolled differentiation and growth, such as teratomas commonly observed in mice transplanted with stem cell-derived pancreatic precursors (Kroon et al., Nat Biotechnol. 2008;26:443-452; Kelly et al., Nat Biotechnol. 2011;29:750-756.). Teratoma formation can be prevented by using cell purification (Kelly et al., 2011) or by modified differentiation methods to generate grafts consisting of pure mature β-cell populations. Encapsulation also prevents possible metastasis if the insulinoma/teratoma is formed by transplanted cells.

此方法需要高效及可重複的微包封方案,且在不存在清除或移除之可能性的情況下,一些細胞可能在包封材料中死亡。一些報告已顯示,經微包封之β細胞在植入之後可繼續存活至多6個月(Orlando等人,Diabetes. 2014;63:1433- 1444)。此意謂當裝置不再起作用時,需要進行重複手術來替代微包封裝置。This method requires an efficient and reproducible microencapsulation protocol, and without the possibility of clearance or removal, some cells may die within the encapsulation material. Several reports have shown that microencapsulated β cells can continue to survive up to 6 months after implantation (Orlando et al., Diabetes. 2014;63:1433-1444). This means that when the device no longer functions, repeat surgery is required to replace the microencapsulated device.

鑒於上文,對研發有效地治療糖尿病,且尤其治療第1型糖尿病的技術的需要遠未得到滿足。In view of the above, there is a far unmet need to develop technologies to effectively treat diabetes, and especially type 1 diabetes.

本發明涉及轉殖基因人類β細胞,以及轉殖基因衰老人類β細胞,其表現有效量之趨除劑,以使得此等細胞對人類免疫細胞具有抗性。趨除劑為此項技術中所熟知的,包括「CXCL12」。亦稱為SDF-1的此細胞介素係藉由胸腺及骨髓基質產生(參見例如,Honjo等人,1998年5月26日頒佈的標題為:「人類基質衍生因子1α.及1β.」之美國專利第5,756,084號)。已報導CXCL12會排斥效應T細胞,同時將免疫抑制性調節T細胞募集至解剖部位。參見例如Poznansky等人,Nature Medicine 2000, 6:543-8。亦報導CXCL12及其受體CXCR4為血管生成之組成部分。The present invention relates to transgenic human beta cells, as well as transgenic senescent human beta cells, which express an effective amount of a scavenger to render these cells resistant to human immune cells. Removal agents are well known in the art and include "CXCL12". Also known as SDF-1, this cytokine is produced by the thymus and bone marrow stroma (see, eg, Honjo et al., US Patent No. 5,756,084, issued May 26, 1998, entitled: "Human Stromal-Derived Factor 1α. and 1β."). CXCL12 has been reported to repel effector T cells while recruiting immunosuppressive regulatory T cells to anatomical sites. See eg Poznansky et al., Nature Medicine 2000, 6:543-8. CXCL12 and its receptor CXCR4 have also been reported as components of angiogenesis.

亦揭示除CXCL12外之排斥免疫細胞之劑,其包括(但不限於) gp120、其他CXCR4配位體、IL-8、CXCR4結合抗體、CXCL13、CXCR5配位體、CXCR5結合抗體及其類似者。Also disclosed are agents that repel immune cells other than CXCL12, including, but not limited to, gp120, other CXCR4 ligands, IL-8, CXCR4-binding antibodies, CXCL13, CXCR5 ligands, CXCR5-binding antibodies, and the like.

本發明之一實施例為轉殖基因人類β細胞,其表現有效量之趨除劑,較佳CXCL12或CXCL13以使得該細胞對人類免疫細胞具有抗性。在一個實施例中,此類趨除有效量之趨除劑係由將針對該劑(例如CXCL12、CXCL13)之人類轉殖基因引入β細胞或β細胞之前驅體(例如多能幹細胞)中產生。此等人類轉殖基因β細胞之進一步特徵在於表現高血糖環境中之胰島素。因此,此等細胞可用於治療個體中之糖尿病之方法中。用於本文所述之方法中的轉殖基因人類β細胞可為自體性或非自體性,例如同種異體β細胞。在一個實施例中,患者罹患T1D。在另一實施例中,轉殖基因人類β細胞可改造為衰老細胞(不能分裂),以使得此等細胞任何的進一步分化成癌細胞得到消除且由於不當細胞分裂引起的凋亡誘導無效。One embodiment of the present invention is transgenic human beta cells expressing an effective amount of a culprit, preferably CXCL12 or CXCL13, to render the cells resistant to human immune cells. In one embodiment, such a phagocytic effective amount of a phagocytic agent is produced by introducing a human transgene directed against the agent (eg, CXCL12, CXCL13) into β cells or β cell precursors (eg, pluripotent stem cells). These human transgenic beta cells were further characterized by the expression of insulin in a hyperglycemic environment. Accordingly, these cells can be used in methods of treating diabetes in an individual. Transgenic human beta cells used in the methods described herein may be autologous or non-autologous, eg, allogeneic beta cells. In one embodiment, the patient suffers from T1D. In another example, transgenic human beta cells can be engineered into senescent cells (unable to divide) such that any further differentiation of these cells into cancer cells is abolished and induction of apoptosis due to inappropriate cell division is ineffective.

本發明之一實施例使用β細胞,例如自體性或同種異體β細胞,其由非糖尿病人類個體或患有糖尿病之人類個體獲得或衍生自其。此等β細胞包括功能性趨除劑(例如CXCL12、CXCL13)表現載體。此類載體經設計以在轉殖基因β細胞中以足以在β細胞周圍產生趨除緩衝劑之量表現該劑。在不希望受理論束縛之情況下,預期此緩衝劑使得β細胞可抵抗免疫細胞攻擊,但仍表現維持糖尿病個體中之適當血糖量將所需的胰島素。由患有糖尿病之患者產生自體性β細胞為此項技術中已知的。參見例如Egli等人,EMBO J. 2015年4月1日; 34(7): 841-855,其以全文引用之方式併入本文中。衍生自幹細胞之同種異體β細胞可在市面上購得。One embodiment of the invention uses beta cells, such as autologous or allogeneic beta cells, obtained from or derived from a non-diabetic human individual or a human individual with diabetes. These β cells include vectors expressing functional chemoattractants (eg, CXCL12, CXCL13). Such vectors are designed to express the agent in the transgenic β-cell in an amount sufficient to generate an efflux buffer around the β-cell. Without wishing to be bound by theory, it is expected that this buffer renders the beta cells resistant to immune cell attack, yet expresses the insulin that would be required to maintain proper blood glucose levels in diabetic individuals. The generation of autologous beta cells from patients with diabetes is known in the art. See, eg, Egli et al., EMBO J. 2015 Apr 1; 34(7): 841-855, which is incorporated herein by reference in its entirety. Allogeneic beta cells derived from stem cells are commercially available.

本發明之一態樣為向有需要之個體投與轉殖基因人類β細胞,以調節個體中之胰島素量且治療糖尿病,該等細胞包含編碼趨除劑(例如CXCL12、CXCL13)之轉殖基因。此外,足夠量之趨除劑的表現會防止轉殖基因β細胞因單核免疫細胞浸潤受到破壞之風險發生。轉殖基因人類β細胞可為自體性或同種異體的。在本發明之一實施例中,轉殖基因β細胞為源自患有糖尿病之患者的自體性β細胞。在另一實施例中,轉殖基因β細胞為同種異體人類β細胞。在另一實施例中,患者患有第1型糖尿病。One aspect of the invention is the administration of transgenic human beta cells comprising a transgene encoding a scavenger (eg, CXCL12, CXCL13) to an individual in need thereof to regulate insulin levels in the individual and treat diabetes. In addition, the expression of a sufficient amount of the scavenger prevents the risk of destruction of the transgenic β-cells due to the infiltration of mononuclear immune cells. Transgenic human beta cells can be autologous or allogeneic. In one embodiment of the present invention, the transgenic β-cells are autologous β-cells derived from patients with diabetes. In another embodiment, the transgenic beta cells are allogeneic human beta cells. In another embodiment, the patient has type 1 diabetes.

本發明之另一態樣係關於能夠表現趨除有效量之趨除劑(例如CXCL12、CXCL13)以對免疫破壞具有抗性的轉殖基因人類β細胞。趨除劑(例如CXCL12、CXCL13)可為內源性劑,亦即由待治療之個體表現之劑;或外源性劑,例如來自非自體性來源之劑或經改造之趨除劑。在一個實施例中,與未經改造之基因相比,編碼β細胞中之趨除劑的基因經改造為過度表現的。用於改造基因表現之方法為此項技術中已知的,舉例而言,進行定點基因編輯以用不同啟動子(例如組成型啟動子、誘導性啟動子等)替代內源性啟動子。在一個實施例中,將編碼趨除劑之重組聚核苷酸插入至β細胞中,使得趨除劑由重組聚核苷酸表現。用於將重組基因插入至細胞中之方法(轉導、轉染等)為此項技術中所熟知的,用用於插入至細胞中的重組聚核苷酸製造載體之方法亦為此項技術中所熟知的。Another aspect of the invention pertains to transgenic human beta cells capable of expressing an effective amount of a culprit (eg, CXCL12, CXCL13) to be resistant to immune disruption. A scavenger (eg, CXCL12, CXCL13) can be an endogenous agent, ie, one expressed by the individual to be treated; or an exogenous agent, such as an agent from a non-autologous source or an engineered scavenger. In one embodiment, a gene encoding a scavenger in beta cells is engineered to be overexpressed compared to an unengineered gene. Methods for engineering gene expression are known in the art, for example, performing site-directed gene editing to replace endogenous promoters with different promoters (eg, constitutive promoters, inducible promoters, etc.). In one embodiment, a recombinant polynucleotide encoding an agonist is inserted into a beta cell such that the agonist is expressed by the recombinant polynucleotide. Methods for inserting recombinant genes into cells (transduction, transfection, etc.) are well known in the art, as are methods for making vectors from recombinant polynucleotides for insertion into cells.

在一些實施例中,趨除劑為經改造之趨除劑。舉例而言,趨除劑之多肽序列可經改造以增加循環半衰期、併入保守胺基酸變化、增強結合至胞外基質、改良該劑活性等。因此,編碼經改造之趨除劑(例如CXCL12或CXCL13)之基因可經改造以使得基因與天然基因具有至少95%之序列一致性,且較佳與天然基因具有99%之序列一致性。同樣,經改造之趨除劑(例如經改造之CXCL12或CXCL13)之胺基酸序列與天然趨除劑具有至少95%且較佳99%之序列一致性。In some embodiments, the repellent is an engineered repellent. For example, the polypeptide sequence of a scavenger can be engineered to increase circulating half-life, incorporate conservative amino acid changes, enhance binding to the extracellular matrix, improve the activity of the agent, and the like. Thus, a gene encoding an engineered chemokine (eg, CXCL12 or CXCL13) can be engineered such that the gene has at least 95% sequence identity with the native gene, and preferably has 99% sequence identity with the native gene. Likewise, the amino acid sequence of the engineered keloid (eg, engineered CXCL12 or CXCL13) has at least 95% and preferably 99% sequence identity with the natural keloid.

在一個實施例中,提供一種包含載體之人類β細胞,載體本身包含編碼人類CXCL12或經改造之CXCL12的核酸序列,其中使該β細胞對人類免疫細胞具有抗性。In one embodiment, there is provided a human β-cell comprising a vector which itself comprises a nucleic acid sequence encoding human CXCL12 or engineered CXCL12, wherein the β-cell is rendered resistant to human immune cells.

在一個實施例中,人類轉殖基因β細胞為由患有第1型糖尿病之個體獲得的自體性β細胞。In one embodiment, the human transgenic beta cells are autologous beta cells obtained from an individual with type 1 diabetes.

在一個實施例中,人類β細胞為同種異體β細胞。In one embodiment, the human beta cells are allogeneic beta cells.

在一個實施例中,人類單核免疫細胞包含NK細胞、T細胞及B細胞。在一個實施例中,T細胞包含細胞毒性T細胞。In one embodiment, the human mononuclear immune cells comprise NK cells, T cells and B cells. In one embodiment, the T cells comprise cytotoxic T cells.

在一個實施例中,轉殖基因人類β細胞表現呈趨除量之人類CXCL12。In one embodiment, the transgenic human beta cells express repellent amounts of human CXCL12.

在一個實施例中,人類CXCL12係選自由CXCL12α及CXCL12β組成之群。In one embodiment, the human CXCL12 is selected from the group consisting of CXCL12α and CXCL12β.

在一個實施例中,人類轉殖基因β細胞包含在內源性CXCL12編碼區上游的轉殖基因調節區,其中該β細胞對人類免疫細胞具有抗性。較佳地,內源性CXCL12編碼區調節區包含組成型啟動子。在一些實施例中,內源性CXCL12編碼區調節區包含誘導性啟動子。In one embodiment, the human transgenic beta cell comprises a transgenic regulatory region upstream of an endogenous CXCL12 coding region, wherein the beta cell is resistant to a human immune cell. Preferably, the regulatory region of the endogenous CXCL12 coding region comprises a constitutive promoter. In some embodiments, the regulatory region of the endogenous CXCL12 coding region comprises an inducible promoter.

在一個實施例中,包含在內源性CXCL12編碼區上游的轉殖基因調節區,其中該β細胞對人類免疫細胞具有抗性的人類轉殖基因β細胞為自體性β細胞,且較佳為由患有糖尿病之患者獲得的β細胞。In one embodiment, the human transgenic β-cell comprising a transgenic regulatory region upstream of the endogenous CXCL12 coding region, wherein the β-cell is resistant to human immune cells, is an autologous β-cell, and is preferably a β-cell obtained from a patient with diabetes.

在一個實施例中,包含在內源性CXCL12編碼區上游的轉殖基因調節區,其中該β細胞對人類免疫細胞具有抗性的人類轉殖基因β細胞為同種異體β細胞。In one embodiment, the human transgenic beta cell comprising a transgenic regulatory region upstream of an endogenous CXCL12 coding region, wherein the beta cell is resistant to a human immune cell, is an allogeneic beta cell.

在一個實施例中,提供一種包含可表現之人類CXCL12或CXCL13基因之人類轉殖基因β細胞,其中該細胞表現趨除有效量之CXCL12或CXCl13以對人類免疫細胞具有抗性。In one embodiment, a human transgenic beta cell comprising an expressible human CXCL12 or CXCL13 gene is provided, wherein the cell expresses an effective amount of CXCL12 or CXCL13 to be resistant to human immune cells.

在一個實施例中,人類轉殖基因β細胞包含針對CXCL12之人類基因。In one embodiment, the human transgenic beta cells comprise the human gene for CXCL12.

在一個實施例中,人類轉殖基因β細胞包含選自由CXCL12α及CXCL12β組成之群的人類基因。In one embodiment, the human transgenic beta cell comprises a human gene selected from the group consisting of CXCL12α and CXCL12β.

在一個實施例中,人類轉殖基因β細胞包含針對CXCL12β之人類基因。In one embodiment, the human transgenic β-cell comprises the human gene for CXCL12β.

在一個實施例中,提供一種人類轉殖基因衰老β細胞,其包含可表現之人類CXCL12或CXCL13基因,其中該細胞表現趨除有效量之CXCL12或CXCl13以對人類免疫細胞具有抗性,且另外地,其中該細胞為衰老的。In one embodiment, there is provided a human transgenic senescent beta cell comprising an expressible human CXCL12 or CXCL13 gene, wherein the cell expresses an effective amount of CXCL12 or CXCL13 to be resistant to human immune cells, and additionally, wherein the cell is senescent.

在一個實施例中,人類轉殖基因衰老β細胞包含可表現之針對CXCL12之人類基因,且其能夠在高血糖介質存在下表現胰島素。In one embodiment, human transgenic senescent beta cells comprise an expressible human gene for CXCL12 and are capable of expressing insulin in the presence of hyperglycemic mediators.

在一個實施例中,人類轉殖基因衰老β細胞包含可表現之人類基因,其選自由CXCL12α及CXCL12β組成之群。In one embodiment, the human transgenic senescent beta cells comprise an expressible human gene selected from the group consisting of CXCL12α and CXCL12β.

在一個實施例中,人類轉殖基因β細胞包含可表現之人類基因,其為CXCL12β。In one embodiment, the human transgenic β-cell comprises an expressible human gene which is CXCL12β.

在一個實施例中,提供一種用於回應於高血糖環境產生胰島素之方法,該方法包含使該環境與如上文所述的人類轉殖基因β細胞群接觸。In one embodiment, there is provided a method for producing insulin in response to a hyperglycemic environment, the method comprising contacting the environment with a population of human transgenic beta cells as described above.

在一個實施例中,人類轉殖基因β細胞對選自由以下組成之群的人類免疫細胞具有抗性:T細胞、B細胞、NK細胞及其混合物。In one embodiment, the human transgenic beta cells are resistant to human immune cells selected from the group consisting of T cells, B cells, NK cells, and mixtures thereof.

在一個實施例中,本文所述之β細胞係由以下獲得: (a) 由人類個體獲得人類祖細胞或人類多能幹細胞群; (b) 將個體之祖細胞或多能幹細胞分化於β細胞中;及 (c) 將編碼趨除劑之核酸分子引入β細胞中。In one embodiment, the β cell lines described herein are obtained by: (a) obtaining a population of human progenitor cells or human pluripotent stem cells from a human individual; (b) differentiating the individual's progenitor cells or pluripotent stem cells into β cells; and (c) introducing a nucleic acid molecule encoding an attractant into the β cells.

在一個實施例中,趨除劑為細胞介素、趨化細胞素、CXCR4結合抗體、CXCR4配位體、CXCR5結合抗體或CXCR5配位體。In one embodiment, the chemoattractant is an interleukin, a chemokine, a CXCR4-binding antibody, a CXCR4 ligand, a CXCR5-binding antibody, or a CXCR5 ligand.

在一個實施例中,趨除劑為CXCL12、CXCL13、gp120或IL-8。In one embodiment, the chemoattractant is CXCL12, CXCL13, gp120 or IL-8.

在一態樣中,提供一種用於提高β細胞在包含免疫細胞之生物樣本中之存活期的方法,其係藉由改造β細胞以表現足以抑制或阻斷免疫細胞殺滅該β細胞之量的趨除劑。In one aspect, a method is provided for increasing the survival of beta cells in a biological sample comprising immune cells by engineering the beta cells to express a scavenger in an amount sufficient to inhibit or block killing of the beta cells by immune cells.

相關申請之交叉引用Cross-References to Related Applications

本申請案主張2017年10月3日申請的美國臨時申請案第62/567,604號;2017年10月4日申請的美國臨時申請案第62/568,117號;2018年3月2日申請的美國臨時申請案第62/637,913號;2018年4月25日申請的美國臨時申請案第62/662,651號;2018年7月6日申請的美國臨時申請案第62/694,634號;2018年7月11日申請的美國臨時申請案第62/696,603號;2018年8月10日申請的美國臨時申請案第62/717,587號;2018年8月20日申請的美國臨時申請案第62/719,975號;及2018年9月21日申請的美國臨時申請案第62/734,910號之優先權;其中之每一者以全文引用之方式併入本文中。序列表 This application asserts U.S. Provisional Application No. 62/567,604, filed October 3, 2017; U.S. Provisional Application No. 62/568,117, filed October 4, 2017; U.S. Provisional Application No. 62/637,913, filed March 2, 2018; U.S. Provisional Application No. 62/662,651, filed April 25, 2018; U.S. Provisional Application No. 62/694,634, filed July 6, 8; U.S. Provisional Application No. 62/696,603, filed July 11, 2018; U.S. Provisional Application No. 62/717,587, filed August 10, 2018; U.S. Provisional Application No. 62/719,975, filed August 20, 2018; Priority of U.S. Provisional Application No. 62/734,910 filed; each of which is incorporated herein by reference in its entirety. sequence listing

本申請案含有序列表,該序列表已以ASCII格式以電子方式提交且以全文引用之方式併入本文中。2018年9月25日創建的該ASCII複本命名為054610-501001WO_SL.txt且位元組大小為8,668。This application contains a Sequence Listing, which was filed electronically in ASCII format and is hereby incorporated by reference in its entirety. Created on September 25, 2018, this ASCII copy is named 054610-501001WO_SL.txt and has a byte size of 8,668.

本發明提供自體性/同種異體人類β細胞,其為轉殖基因的且包含編碼人類趨除劑(例如CXCL12、CXCL13)之轉殖基因,或經基因改造以表現或過度表現呈趨除量之內源性(人類)趨除劑(例如CXCL12、CXCL13)。在一較佳實施例中,本文所述之轉殖基因β細胞經進一步改造呈衰老細胞。在其方法態樣之另一者中,β細胞經改造或處理以表現有效量之趨除劑(例如CXCL12、CXCL13),以抑制轉殖基因人類β細胞之免疫破壞且回應於高血糖環境產生胰島素。The invention provides autologous/allogeneic human beta cells that are transgenic and comprise a transgene encoding a human scavenger (e.g., CXCL12, CXCL13), or are genetically engineered to express or overexpress an endogenous (human) scavenger (e.g., CXCL12, CXCL13) in invasive amounts. In a preferred embodiment, the transgenic β cells described herein are further engineered to be senescent cells. In another aspect of the method, β cells are engineered or treated to express an effective amount of an attractant (eg, CXCL12, CXCL13) to inhibit immune destruction of transgenic human β cells and produce insulin in response to a hyperglycemic environment.

進一步詳細揭示本發明之前,將首先界定以下術語。若術語未經界定,則其具有如此項技術中所理解的一般公認科學含義。Before disclosing the present invention in further detail, the following terms will first be defined. Where a term is not defined, it has its generally accepted scientific meaning as understood in the art.

術語「CXCL13」係指其所有已知同功異型物。由於已知CXCL13會介導某些癌細胞增殖,因此其並非較佳的。參見例如https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3839818/The term "CXCL13" refers to all known isoforms thereof. Since CXCL13 is known to mediate the proliferation of certain cancer cells, it is not preferred. See e.g. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3839818/

術語「趨除(fugetaxis)」或「趨除(fugetactic)」係指排斥(或化學排斥)具有遷移能力之真核細胞的藥劑之能力。由細胞表現的趨除量之CXCL12或CXCL13(或其他趨除劑)為足以阻斷或抑制免疫細胞朝細胞之遷移,或在一些態樣中,使細胞排斥免疫細胞之量。The term "fugetaxis" or "fugetactic" refers to the ability of an agent to repel (or chemically repel) eukaryotic cells that have the ability to migrate. The attractant amount of CXCL12 or CXCL13 (or other attractant) exhibited by the cell is an amount sufficient to block or inhibit the migration of immune cells towards the cell, or in some aspects, cause the cell to repel the immune cell.

術語「人類免疫細胞」可與術語「人類單核免疫細胞」互換使用,且包括NK細胞、T細胞及B細胞。The term "human immune cell" is used interchangeably with the term "human mononuclear immune cell" and includes NK cells, T cells and B cells.

術語「抗免疫細胞」或「對免疫系統隱形」指示β細胞表現足以阻斷或抑制免疫細胞朝細胞之遷移,或在一些態樣中,使β細胞排斥免疫細胞的趨除劑之量。在一較佳實施例中,此類阻斷或抑制係藉由將本發明之β細胞暴露於人類單核免疫細胞(例如PBMC)之後的細胞死亡之程度量測。細胞死亡可藉由自已經歷裂解的細胞釋放的乳酸脫氫酶(LDH)來評定。較佳地,本發明之抗免疫細胞β細胞可藉由在兩天培育期內,在約30:1免疫細胞與本發明之β細胞之比下,顯示LDH量相對於對照低50%的細胞來評定。更佳地,抗免疫β細胞顯示LDH量相對於對照低60%;且甚至更佳地,LDH量相對於對照低75%;且最佳地,LDH量相對於對照低95%。用於評定LDH量之程序闡述於本文實例2中。The terms "anti-immune cells" or "stealth to the immune system" indicate that the β-cells exhibit an amount of a scavenger sufficient to block or inhibit the migration of immune cells towards the cell, or in some aspects, cause the β-cells to repel the immune cells. In a preferred embodiment, such blocking or inhibition is measured by the degree of cell death following exposure of the β-cells of the invention to human mononuclear immune cells (eg, PBMC). Cell death can be assessed by lactate dehydrogenase (LDH) released from cells that have undergone lysis. Preferably, the anti-immune cell β-cells of the present invention can be assessed by cells showing a 50% lower LDH amount relative to a control at a ratio of about 30:1 immune cells to β-cells of the present invention during a two-day incubation period. More preferably, the anti-immune beta cells exhibit 60% lower amounts of LDH relative to controls; and even more preferably, 75% lower amounts of LDH relative to controls; and optimally, 95% lower amounts of LDH relative to controls. The procedure used to assess the amount of LDH is described in Example 2 herein.

驅除劑為具有驅除活性之藥劑。驅除劑可包括(但不限於) CXCL12、CXCL13、gp120、IL-8、CXCR4結合抗體、CXCR4配位體、CXCR5結合抗體或CXCR5配位體。Repellents are agents with repellent activity. Repellants may include, but are not limited to, CXCL12, CXCL13, gp120, IL-8, CXCR4-binding antibodies, CXCR4 ligands, CXCR5-binding antibodies, or CXCR5 ligands.

術語「效應T細胞」係指能夠藉由釋放細胞介素引起特異性免疫反應的分化T細胞。The term "effector T cells" refers to differentiated T cells capable of eliciting a specific immune response by releasing cytokines.

術語「調節T細胞」係指降低或抑制B細胞或其他T細胞對抗原之免疫反應的T細胞。The term "regulatory T cell" refers to a T cell that reduces or suppresses the immune response of a B cell or other T cell to an antigen.

術語「CXCL12」或「SDF-1多肽」係指此項技術中熟知的細胞介素(參見例如表1)。在一實施例中,術語係指結合CXCL12特異性抗體且具有趨化性或趨除活性之蛋白質或其片段。趨化性或趨除活性係藉由分析T細胞遷移方向(例如朝向相關藥劑或離開相關藥劑)來測定。參見例如,Poznansky等人,Nature Medicine 2000, 6:543-8;N. Papeta等人,「Long-term survival of transplanted allogeneic cells engineered to express a T Cell chemorepellent」,Transplantation 2007, 83(2), 174-183。「趨除」或「趨除遷移」為遷移性細胞離開藥劑源(亦即,朝向較低濃度之藥劑)之移動。應理解,術語「CXCL12」係指其所有已知同功異型物,包括α、β、γ、δ、ε、φ及θ同功異型物。較佳CXCL12同功異型物為α及β。已知CXCL12會誘導血管生成。The term "CXCL12" or "SDF-1 polypeptide" refers to cytokines well known in the art (see eg Table 1). In one embodiment, the term refers to a protein or fragment thereof that binds a CXCL12-specific antibody and has chemotactic or cytotoxic activity. Chemotactic or cytotoxic activity is determined by analyzing the direction of T cell migration (eg, toward or away from the relevant agent). See, eg, Poznansky et al., Nature Medicine 2000, 6:543-8; N. Papeta et al., "Long-term survival of transplanted allogeneic cells engineered to express a T Cell chemorepellent", Transplantation 2007, 83(2), 174-183. "Dress" or "dress migration" is the movement of migratory cells away from the source of the agent (ie, towards lower concentrations of the agent). It should be understood that the term "CXCL12" refers to all known isoforms thereof, including alpha, beta, gamma, delta, epsilon, phi and theta isoforms. Preferred CXCL12 isoforms are α and β. CXCL12 is known to induce angiogenesis.

術語「第1型糖尿病」及「第2型糖尿病」係指與增加之血糖相關的兩種嚴重病理生理學。第1型糖尿病之特徵在於自體免疫攻擊製造胰臟胰島素之β細胞,而第2型糖尿病與β細胞功能不良及外周胰島素抗性增加相關。與第1型相似,亦在第2型糖尿病中觀測到β細胞死亡。第1型及通常,第2型糖尿病需要個人注射胰島素。第1型糖尿病之典型特徵在於胰臟中之胰島的產胰島素β細胞有所損失,引起胰島素缺乏。此類型之糖尿病可進一步歸類為免疫介導性或特發性。大多數第1型糖尿病屬於免疫介導性,其中β細胞損失係由於T細胞介導之自體免疫攻擊。第2型糖尿病之特徵在於β細胞功能障礙以及胰島素抗性。咸信身體組織對胰島素之缺陷反應性涉及胰島素受體及下游細胞信號傳遞。與第1型糖尿病相似,β細胞群不足亦為多種第2型糖尿病患者中之病原性因素。在第2型糖尿病之早期,可藉由改善胰島素分泌且降低肝臟產生之葡萄糖的多種措施及藥物來逆轉高血糖症。隨著疾病進展,胰島素分泌出現減弱,且胰島素之治療替代物有時在某些患者中可能變得必要。The terms "type 1 diabetes" and "type 2 diabetes" refer to two serious pathophysiologies associated with increased blood glucose. Type 1 diabetes is characterized by an autoimmune attack on the insulin-producing beta cells of the pancreas, while type 2 diabetes is associated with beta cell dysfunction and increased peripheral insulin resistance. Similar to type 1, beta cell death is also observed in type 2 diabetes. Type 1 and often, type 2 diabetes require individuals to inject insulin. Type 1 diabetes is typically characterized by loss of insulin-producing beta cells in the islets of the pancreas, resulting in insulin deficiency. This type of diabetes can be further classified as immune-mediated or idiopathic. Most type 1 diabetes is immune mediated, in which beta cell loss is due to T cell mediated autoimmune attack. Type 2 diabetes is characterized by beta cell dysfunction and insulin resistance. It is believed that defective responsiveness of body tissues to insulin involves the insulin receptor and downstream cell signaling. Similar to type 1 diabetes, deficiencies in β-cell populations are also a pathogenic factor in many types of type 2 diabetes. In the early stages of type 2 diabetes, hyperglycemia can be reversed by a variety of measures and drugs that improve insulin secretion and lower glucose production by the liver. As the disease progresses, insulin secretion decreases, and therapeutic replacement of insulin may sometimes become necessary in some patients.

「個體」或「患者」係指哺乳動物,較佳人類個體。"Individual" or "patient" refers to a mammal, preferably a human individual.

「有需要之個體」或「有需要之患者」為患有第1型或第2型糖尿病之個體。A "subject in need" or "patient in need" is a subject with Type 1 or Type 2 diabetes.

如本文所用,術語「治療(treat)」、「治療(treating)」、「治療(treatment)」及其類似術語係指減少或改善病症及/或與其相關的症狀。應瞭解,雖然不排除,但治療病症或病狀並不需要完全消除該病症、病狀或與其相關之症狀。As used herein, the terms "treat", "treating", "treatment" and similar terms mean to reduce or ameliorate a disorder and/or symptoms associated therewith. It is to be understood that, although not exclusive, treating a disorder or condition does not require complete elimination of the disorder, condition or symptoms associated therewith.

在本發明中,「包含(comprises)」、「包含(comprising)」、「含有」及「具有」及其類似術語可具有美國專利法中歸屬於其之含義且可意謂「包括(includes)」、「包括(including)」及其類似術語;「主要由……組成(consisting essentially of/consists essentially)」同樣具有美國專利法中所歸屬之含義且該術語為開放的,允許存在多於所列舉者,只要所列舉者之基本或新穎特徵不因存在多於所列舉者而改變即可,但不包括先前技術實施例。In the present invention, "comprises", "comprising", "containing" and "having" and similar terms may have the meanings assigned to them in the US Patent Law and may mean "includes", "including (including)" and similar terms; Recited, but excluding prior art embodiments, so long as the essential or novel features of the recited would not be altered by the presence of more than recited.

術語「約」當用於例如溫度、時間、量、濃度及此類其他名稱之數值名稱(包括範圍)前時,指示可變化(+)或(-) 10%、5%、1%或在其之間的任何子範圍或子值的近似值。在整個本發明之上下文中亦呈現其他定義。The term "about" when used in front of a numerical designation such as temperature, time, amount, concentration and such other designations (including ranges) indicates that it may vary (+) or (-) 10%, 5%, 1%, or any subrange or approximation of a subvalue therebetween. Other definitions also appear throughout the context of the present invention.

本發明之一態樣為轉殖基因β細胞,例如人類自體性β細胞或非自體性β細胞,例如同種異體β細胞,其包含可操作地連接於啟動子的編碼趨除劑(例如CXCL12、CXCL13)之核酸,以使得趨除劑(例如CXCL12、CXCL13)在β細胞微環境中以趨除量表現。啟動子可為β細胞之內源性啟動子或β細胞中之異源性但具有功能性之啟動子。較佳地,編碼趨除劑(例如CXCL12、CXCL13)之核酸對用轉殖基因β細胞治療的個體為內源性的。在一個實施例中,同種異體β細胞源自非T1D供體。One aspect of the invention is transgenic β cells, such as human autologous β cells or non-autologous β cells, such as allogeneic β cells, comprising a nucleic acid encoding a chemoattractant (e.g., CXCL12, CXCL13) operably linked to a promoter such that the chemoattractant (e.g., CXCL12, CXCL13) is expressed in a chemoattractant amount in the microenvironment of the β cell. The promoter may be endogenous to the beta cell or a heterologous but functional promoter in the beta cell. Preferably, the nucleic acid encoding the chemoattractant (eg, CXCL12, CXCL13) is endogenous to the individual treated with the transgenic beta cells. In one embodiment, the allogeneic beta cells are derived from a non-T1D donor.

本發明之一態樣為人類β細胞,其包含編碼趨除劑(例如CXCL12、CXCL13)的基因改造之內源性人類基因,其中該基因經改造而包含可操作地連接於編碼趨除劑之序列的異源啟動子,以使得趨除劑在β細胞微環境中以趨除量由內源性基因表現。可使用此項技術中已知的基因組編輯技術將啟動子引入β細胞中以可操作地連接於編碼趨除劑之序列。熟知CXCL12具有若干同功異型物,其包括α、β、γ及θ。在一較佳實施例中,所用同功異型物為CXCL12β。本文所述之轉殖基因人類β細胞回應於高血糖環境產生胰島素。術語「胰島素」意謂涵蓋胰島素原及胰島素兩者。One aspect of the invention is a human beta cell comprising a genetically engineered endogenous human gene encoding a chemoattractant (e.g., CXCL12, CXCL13), wherein the gene has been engineered to include a heterologous promoter operably linked to a sequence encoding the chemoattractant, such that the chemoattractant is expressed by the endogenous gene in a chemoattractant amount in the microenvironment of the beta cell. The promoter can be introduced into the beta cell to be operably linked to the sequence encoding the dispersant using genome editing techniques known in the art. CXCL12 is well known to have several isoforms, including alpha, beta, gamma and theta. In a preferred embodiment, the isoform used is CXCL12β. The transgenic human beta cells described herein produce insulin in response to a hyperglycemic environment. The term "insulin" is meant to encompass both proinsulin and insulin.

一般而言,本發明提供β細胞,且較佳人類β細胞,其表現呈足以阻斷或抑制免疫細胞(例如人類免疫細胞)向β細胞之遷移或足以排斥免疫細胞之量的趨除劑(例如CXCL12、CXCL13)。術語免疫細胞及單核細胞(T細胞、B細胞及NK細胞)可互換使用。趨除劑(例如CXCL12、CXCL13)多肽排斥免疫細胞(例如效應T細胞)之能力可使用博伊登室分析(boyden chamber assay)來活體外評定。參見例如,如Poznansky等人,Journal of Clinical Investigation, 109, 1101 (2002) 中先前所述。可替代地,藉由將此類細胞與人類PBMC組合來評定轉殖基因人類β細胞之存活率。可藉由隨時間推移量測一或多種細胞死亡標記物來評估細胞死亡之比率。一種常用的此類標記物為細胞壞死期間所釋放的乳酸脫氫酶(LDH)。In general, the invention provides β cells, and preferably human β cells, that exhibit an attractant (e.g., CXCL12, CXCL13) in an amount sufficient to block or inhibit migration of immune cells (e.g., human immune cells) to β cells, or to repel immune cells. The terms immune cells and monocytes (T cells, B cells and NK cells) are used interchangeably. The ability of an attractant (eg, CXCL12, CXCL13) polypeptide to repel immune cells (eg, effector T cells) can be assessed in vitro using a boyden chamber assay. See eg, as previously described in Poznansky et al., Journal of Clinical Investigation, 109, 1101 (2002). Alternatively, the survival of transgenic human beta cells is assessed by combining such cells with human PBMCs. The rate of cell death can be assessed by measuring one or more cell death markers over time. One commonly used such marker is lactate dehydrogenase (LDH), which is released during necrosis.

在不希望受任何理論束縛之情況下,申請者預期在本發明之一態樣中,由轉殖基因β細胞產生的趨除劑(例如CXCL12、CXCL13)之量足以在β細胞微環境中提供趨除作用,但並未產生呈足以提高藥劑之全身量且打破藥劑在一個過程中之有益作用,而在另一過程中產生不利後果之間的平衡的量。此外,已知當CXCL12與其受體CXCR4結合時,其會誘導血管生成。此外,在不受任何理論束縛之情況下,預期表現CXCL12的所植入之轉殖基因β細胞之微環境將誘導提高所植入之細胞的存活率的血管生成反應。Without wishing to be bound by any theory, applicants contemplate that, in one aspect of the invention, the amount of occult (e.g., CXCL12, CXCL13) produced by the transgenic β-cells is sufficient to provide phagocytosis in the β-cell microenvironment, but not in an amount sufficient to increase the systemic amount of the agent and upset the balance between beneficial effects of the agent in one process and adverse consequences in another. Furthermore, CXCL12 is known to induce angiogenesis when it binds to its receptor CXCR4. Furthermore, without being bound by any theory, it is expected that the microenvironment of the implanted transgenic beta cells expressing CXCL12 will induce an angiogenic response that increases the survival of the implanted cells.

趨除劑(例如CXCL12、CXCL13)之趨除有效量為足以阻斷免疫細胞殺滅轉殖基因β細胞之任何量。例如,轉殖基因β細胞微環境中的趨除劑(例如CXCL12、CXCL13)之趨除有效量可為至少約100 ng/mL且較佳至少100 nM。在一些實施例中,轉殖基因β細胞微環境中的趨除劑(例如CXCL12、CXCL13)之量為至少約1000 ng/mL。舉例而言,以下特定範圍適用於本發明:約100 nM至約200 nM、約100 nM至約300 nM、約100 nM至約400 nM、約100 nM至約500 nM、約100 nM至約600 nM、約100 nM至約700 nM、約100 nM至約800 nM、約100 nM至約900 nM或約100 nM至約1 µM。A cytotoxic effective amount of a chemoattractant (eg, CXCL12, CXCL13) is any amount sufficient to block killing of transgenic β cells by immune cells. For example, an effective amount of a dispersant (eg, CXCL12, CXCL13) in the microenvironment of transgenic β cells may be at least about 100 ng/mL and preferably at least 100 nM. In some embodiments, the amount of the scavenger (eg, CXCL12, CXCL13) in the microenvironment of the transgenic β-cell is at least about 1000 ng/mL. For example, the following specific ranges are suitable for use in the present invention: about 100 nM to about 200 nM, about 100 nM to about 300 nM, about 100 nM to about 400 nM, about 100 nM to about 500 nM, about 100 nM to about 600 nM, about 100 nM to about 700 nM, about 100 nM to about 800 nM, about 100 nM to about 900 nM M or about 100 nM to about 1 µM.

在各實施例中,轉殖基因β細胞微環境中的趨除劑(例如CXCL12、CXCL13)之趨除有效量在20 ng/mL至約5 µg/mL範圍內。在各實施例中,趨除有效量在20 ng/mL至約1 µg/mL範圍內。在各實施例中,β細胞微環境中的趨除劑(例如CXCL12、CXCL13)之量為在約100 ng/mL至約500 ng/mL、約500 ng/mL至5 µg/mL,約800 ng/mL至約5 µg/mL或約1000 ng/mL至約5000 ng/mL範圍內的充足趨除量。在不希望受理論束縛之情況下,預期當在一起使用轉殖基因及非轉殖基因β細胞時,轉殖基因β細胞可表現足量之趨除劑,以使得微環境產生延伸至鄰近非轉殖基因β細胞的趨除作用。轉殖基因β細胞微環境中的趨除劑(例如CXCL12、CXCL13)之趨除有效量可為所述範圍內的任何值或子範圍(包括端值)。In various embodiments, the effective amount of the extinction agent (eg, CXCL12, CXCL13) in the microenvironment of the transgenic β cells is in the range of 20 ng/mL to about 5 μg/mL. In various embodiments, the repelling effective amount is in the range of 20 ng/mL to about 1 µg/mL. In various embodiments, the amount of the expelling agent (e.g., CXCL12, CXCL13) in the microenvironment of the β-cell is sufficient for expelling in the range of about 100 ng/mL to about 500 ng/mL, about 500 ng/mL to 5 µg/mL, about 800 ng/mL to about 5 µg/mL, or about 1000 ng/mL to about 5000 ng/mL. Without wishing to be bound by theory, it is expected that when transgenic and non-transgenic β-cells are used together, the transgenic β-cells may express sufficient amounts of culverts such that the microenvironment produces an invasive effect that extends to adjacent non-transgenic β-cells. The effective amount of the dispersing agent (eg, CXCL12, CXCL13) in the microenvironment of the transgenic β-cell can be any value or sub-range (inclusive) within the stated range.

儘管免疫學家普遍使用小鼠及小鼠DNA以進一步理解人類免疫系統之運轉方式,但人類與小鼠之間存在顯著差異。因此,由載體編碼的趨除劑(例如CXCL12、CXCL13)較佳為人類藥劑。Javier Mestas及Christopher C. W. Hughes, 「Of Mice and Not Men: Differences between Mouse and Human Immunology」,Journal of Immunology 2004, 172(5), 2731-2738;O. Cabrera等人,「The unique cytoarchitecture of human pancreatic islets has implications for islet cell function」,PNAS 2006, 103(7), 2334-2339;M. Votey, 「Of mice and men: how the nPOD program is changing the way researchers study type 1 diabetes」, diaTribe, Network for Pancreatic Organ Donors with Diabetes (2015年8月21日)。Although immunologists commonly use mice and mouse DNA to further understand how the human immune system works, there are significant differences between humans and mice. Therefore, the vector-encoded chemoattractant (eg, CXCL12, CXCL13) is preferably a human agent. Javier Mestas and Christopher CW Hughes, "Of Mice and Not Men: Differences between Mouse and Human Immunology", Journal of Immunology 2004, 172(5), 2731-2738; O. Cabrera et al., "The unique cytoarchitecture of human pancreatic islets has implications for islet cell function”, PNAS 2006, 103(7), 2334-2339; M. Votey, “Of mice and men: how the nPOD program is changing the way researchers study type 1 diabetes”, diaTribe, Network for Pancreatic Organ Donors with Diabetes (2015 August 21).

CXCL12多肽為此項技術中已知的。參見例如,Poznansky等人,Nature Medicine 2000, 6:543-8及 美國專利公開案第20170246250號,兩者均以全文引用之方式併入本文中。術語CXCL12及SDF-1可互換使用。例示性CXCL12/SDF-1同功異型物提供於美國公開案20170246250之表I中。例示性CXCL12/SDF1同功異型物亦提供於(下)表1中: 表1:人類CXCL12/SDF1同功異型物 CXCL12 polypeptides are known in the art. See, eg, Poznansky et al., Nature Medicine 2000, 6:543-8 and US Patent Publication No. 20170246250, both of which are incorporated herein by reference in their entirety. The terms CXCL12 and SDF-1 are used interchangeably. Exemplary CXCL12/SDF-1 isoforms are provided in Table I of US Publication 20170246250. Exemplary CXCL12/SDF1 isoforms are also provided in Table 1 (below): Table 1: Human CXCL12/SDF1 isoforms

在一個實施例中,CXCL12多肽與NP 001029058具有至少約85%、90%、92%、95%、96%、97%、98%、99%或100%之胺基酸序列一致性且具有趨化細胞素或趨除活性。在一個實施例中,CXCL12多肽與SEQ ID NO: 1、SEQ ID NO: 2、SEQ ID NO: 3、SEQ ID NO: 4、SEQ ID NO: 5或SEQ ID NO: 6具有至少約85%、90%、92%、95%、96%、97%、98%、99%或100%之胺基酸序列一致性且具有趨化細胞素或趨除活性。此類序列一致性係基於用已知保守第二胺基酸替代第一胺基酸。此類保守替代在此項技術中為公認的,且針對所得經改造之CXCL12多肽的趨除特性的測試亦為此項技術中所熟知的。參見例如,Poznansky,見上文。In one embodiment, the CXCL12 polypeptide has at least about 85%, 90%, 92%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to NP 001029058 and has chemoattractant or chemoattractant activity. In one embodiment, CXCL12 polypeptide and SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, or SEQ ID NO: 6 have at least 85%, 90%, 95%, 96%, 98%, 99%, or 100%of aminic acid. Sequence consistency and have cytopenin or eliminate activity. Such sequence identity is based on the substitution of a first amino acid by a known conserved second amino acid. Such conservative substitutions are well-recognized in the art, and testing for the cytotoxic properties of the resulting engineered CXCL12 polypeptides is also well known in the art. See eg, Poznansky, supra.

CXCL13肽為此項技術中已知的。CXCL13亦稱為B淋巴細胞化學引誘劑(BLC)或B細胞引誘趨化細胞素1 (BCA-1),且此等術語可互換使用。舉例而言,人類CXCL13可見於寄存編號Q53X90。在一個實施例中,CXCL13多肽具有包含以下之胺基酸序列:MKFISTSLLLMLLVSSLSPVQGVLEVYYTSLRCRCVQESSVFIPRRFIDRIQILPRGNGCPRKEIIVWKKNKSIVCVDPQAEWIQRMMEVLRKRSSSTLPVPVFKRKIP (SEQ ID NO: 7)。在一個實施例中,CXCL13多肽與Q53X90具有至少約85%、90%、92%、95%、96%、97%、98%、99%或100%之胺基酸序列一致性且具有趨化細胞素或趨除活性。在一個實施例中,CXCL13多肽與SEQ ID NO: 7具有至少約85%、90%、92%、95%、96%、97%、98%、99%或100%之胺基酸序列一致性且具有趨化細胞素或趨除活性。CXCL13 peptides are known in the art. CXCL13 is also known as B-lymphocyte chemoattractant (BLC) or B-cell-attracting chemoattractant 1 (BCA-1), and these terms are used interchangeably. For example, human CXCL13 can be found in accession number Q53X90. In one embodiment, the CXCL13 polypeptide has an amino acid sequence comprising: MKFISTSLLLMLLVSSSLSPVQGVLEVYYTSLRCRCVQESSVFIPRRFIDRIQILPRGNGCPRKEIIVWKKNKSIVCVDPQAEWIQRMMEVLRKRSSSTLPVPVFKRKIP (SEQ ID NO: 7). In one embodiment, the CXCL13 polypeptide has at least about 85%, 90%, 92%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to Q53X90 and has chemoattractant or cytotoxic activity. In one embodiment, the CXCL13 polypeptide has at least about 85%, 90%, 92%, 95%, 96%, 97%, 98%, 99%, or 100% amino acid sequence identity to SEQ ID NO: 7 and has chemoattractant or cytotoxic activity.

用於本文所述之方法中的轉殖基因β細胞可為自體性或非自體性,例如同種異體β細胞。「自體性細胞」為來自同一個體之細胞。「同種異體」細胞為來自相同物種的遺傳學類似但不相同的供體之細胞。適用於本發明方法中的同種異體細胞較佳來自人類個體。適用於本發明方法中的同種異體細胞可來自親屬,例如同胞、表親、父母或子女;或非親屬。用於選擇同種異體供體之標準為此項技術中所熟知的,參見例如,Tatum等人,Diabetes Metab Syndr Obes 2017: 10 73-78。人類同種異體β細胞可在市面上購得,且自體性β細胞係藉由由Egli 等人所述的方法產生,見上文。Transgenic beta cells used in the methods described herein may be autologous or non-autologous, eg, allogeneic beta cells. "Autologous cells" are cells from the same individual. "Allogeneic" cells are cells from a genetically similar but not identical donor of the same species. Allogeneic cells suitable for use in the methods of the invention are preferably from human individuals. Allogeneic cells suitable for use in the methods of the invention may be from a relative, such as a sibling, cousin, parent or child; or a non-relative. Criteria for selecting an allogeneic donor are well known in the art, see, eg, Tatum et al., Diabetes Metab Syndr Obes 2017: 10 73-78. Human allogeneic β-cells are commercially available, and autologous β-cell lines are generated by the method described by Egli et al., supra.

在一實施例中,本發明方法中所用之轉殖基因人類β細胞為自體性轉殖基因β細胞,可藉由利用此項技術中已知的方法自由患者獲得的多能祖細胞或多能幹細胞衍生β細胞而製備。此等衍生之β細胞可包含以下(例如用以下轉染或感染等):包含編碼趨除劑(例如CXCL12、CXCL13)之核酸序列的表現載體。In one embodiment, the transgenic human beta cells used in the methods of the invention are autologous transgenic beta cells, which can be prepared by pluripotent progenitor or pluripotent stem cell-derived beta cells obtained from a patient using methods known in the art. These derived β-cells may comprise (eg, be transfected or infected with, etc.): an expression vector comprising a nucleic acid sequence encoding an attractant (eg, CXCL12, CXCL13).

可替代地,本發明方法中所用之轉殖基因β細胞可藉由將胰島β細胞與有需要之個體分離來製備。此等經分離之胰島β細胞可包含以下(例如用以下轉染或感染等):包含編碼趨除劑(例如CXCL12、CXCL13)之核酸序列的表現載體。可替代地,β細胞可經基因改造以表現內源性趨除劑(例如CXCL12、CXCL13)基因,以使得其組成性產生趨除有效量之趨除劑(例如CXCL12、CXCL13)。Alternatively, transgenic beta cells used in the methods of the present invention can be prepared by isolating pancreatic islet beta cells from an individual in need thereof. These isolated islet β-cells may comprise (eg transfected or infected with, etc.): an expression vector comprising a nucleic acid sequence encoding an attractant (eg CXCL12, CXCL13). Alternatively, beta cells can be genetically engineered to express an endogenous scavenger (eg, CXCL12, CXCL13) gene such that they constitutively produce a scavenger (eg, CXCL12, CXCL13) in an effective amount.

在本發明之一實施例中,β細胞包含以下(例如用以下轉染或感染等):包含編碼趨除劑(例如CXCL12、CXCL13)之核酸分子的表現載體,該核酸分子可操作地連接於適用於β細胞中之表現的啟動子。載體可整合至β細胞之基因組中或其可以游離方式存在且不整合至基因組中。In one embodiment of the invention, the β-cell comprises (e.g., transfected or infected with, etc.): an expression vector comprising a nucleic acid molecule encoding an attractant (e.g., CXCL12, CXCL13) operably linked to a promoter suitable for expression in the β-cell. The vector may integrate into the genome of the beta cell or it may exist episomally and not integrate into the genome.

本發明之轉殖基因β細胞亦可由成體幹細胞藉由以下製備:將成體幹細胞與個體分離,在合適條件下培養幹細胞以擴展細胞群且誘導分化成β細胞。細胞可藉由以下經改造以表現趨除有效量之趨除劑(例如CXCL12、CXCL13):將編碼趨除有效量之趨除劑(例如CXCL12、CXCL13)之表現載體引入細胞中;或編輯基因組以表現趨除有效量之趨除劑(例如CXCL12、CXCL13)。載體可在分化成β細胞之前引入幹細胞中或幹細胞之基因組可經編輯以含有異源啟動子。可替代地,載體可引入所得β細胞中或所得β細胞之基因組可經編輯以含有異源啟動子。The transgenic β-cells of the present invention can also be prepared from adult stem cells by isolating the adult stem cells from the individual, culturing the stem cells under appropriate conditions to expand the cell population and induce differentiation into β-cells. Cells can be engineered to express an effective amount of a chemoattractant (eg, CXCL12, CXCL13) by introducing an expression vector encoding an effective amount of a chemoattractant (eg, CXCL12, CXCL13) into the cell; or editing the genome to express an effective amount of a chemoattractant (eg, CXCL12, CXCL13). The vector can be introduced into the stem cell prior to differentiation into beta cells or the genome of the stem cell can be edited to contain a heterologous promoter. Alternatively, the vector can be introduced into the resulting beta cells or the genome of the resulting beta cells can be edited to contain a heterologous promoter.

本發明之轉殖基因β細胞亦可藉由以下製備:由個體之體細胞,例如β細胞、纖維母細胞或角質細胞產生誘導多能幹(iPS)細胞;處理iPS細胞以誘導分化成β細胞;且將包含編碼趨除劑(例如CXCL12、CXCL13)之核酸序列的表現載體引入經分化之β細胞中。The transgenic β-cells of the present invention can also be prepared by: generating induced pluripotent stem (iPS) cells from individual somatic cells, such as β-cells, fibroblasts or keratinocytes; treating the iPS cells to induce differentiation into β-cells; and introducing an expression vector comprising a nucleic acid sequence encoding a chemoattractant (such as CXCL12, CXCL13) into the differentiated β-cells.

本發明之轉殖基因β細胞亦可藉由以下製備:製備由個體體細胞產生之誘導多能幹(iPS)細胞;將包含編碼趨除劑(例如CXCL12、CXCL13)之核酸序列的表現載體引入iPS細胞中;且在引入轉殖基因之前或之後處理iPS細胞,以誘導分化成β細胞。The transgenic β cells of the present invention can also be prepared by: preparing induced pluripotent stem (iPS) cells generated from individual somatic cells; introducing an expression vector comprising a nucleic acid sequence encoding a chemoattractant (such as CXCL12, CXCL13) into the iPS cells; and treating the iPS cells before or after introducing the transgene to induce differentiation into β cells.

本發明之轉殖基因β細胞亦可利用此項技術中已知的方法藉由以下產生:獲得祖細胞或類祖細胞之細胞,例如胰臟β-細胞祖細胞;將包含編碼趨除劑(例如CXCL12、CXCL13)之核酸序列的載體引入細胞中;且在引入載體之前或之後處理細胞以誘導分化成β細胞,或對體內葡萄糖量起反應之胰島素釋放細胞;參見例如,Millman等人Nature Communications (2016年5月10日) 第1-8頁;Baek等人 Curr Stem Cell Rep (2016) 2:52-61;Russ等人,EMBO. J. 34, 1759-1772 (2015);及 Qadir等人,Cell Reports 22,2408-2420 (2018年2月27日)。祖細胞及類祖細胞之細胞可為用轉殖基因細胞治療的個體之自體性或非自體性,例如同種異體細胞。對體內葡萄糖量起反應的產胰島素細胞(參見例如Qadir等人,見上文)可如本文所述經基因改造以表現趨除量之趨除劑(例如CXCL12、CXCL13),且亦為本發明之一實施例。此類基因改造之產胰島素細胞亦可用於本發明方法中來治療如本文所述之糖尿病。The transgenic β-cells of the present invention can also be produced using methods known in the art by obtaining progenitor cells or progenitor-like cells, such as pancreatic β-cell progenitor cells; introducing a vector comprising a nucleic acid sequence encoding an attractant (e.g., CXCL12, CXCL13) into the cells; and treating the cells before or after introducing the vector to induce differentiation into β-cells, or insulin-releasing cells that respond to the amount of glucose in vivo; see, for example, Millman et al. Nature Communications (May 2016 10) pp. 1-8; Baek et al. Curr Stem Cell Rep (2016) 2:52-61; Russ et al., EMBO. J. 34, 1759-1772 (2015); and Qadir et al., Cell Reports 22, 2408-2420 (27 February 2018). Progenitor cells and progenitor-like cells may be autologous or non-autologous to the individual being treated with the transgenic cells, eg, allogeneic cells. Insulin producing cells that respond to the amount of glucose in vivo (see eg Qadir et al., supra) can be genetically engineered to express a decimator (eg CXCL12, CXCL13) as described herein and is also an embodiment of the invention. Such genetically engineered insulin-producing cells may also be used in the methods of the invention to treat diabetes as described herein.

可藉由此項技術中已知的方法將來自個體的任何適合體細胞再程式化為iPS細胞,參見例如,Pagliuca及Melton (2013) How to make a functional β-cell, Development (3013) 140(12); 2472-2483;Yu等人(2007). Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917-1920;Takahashi及Yamanaka, 2006, Cell 126(4):663-676;Wernig等人,2007, Nature 448:7151;Okita等人,2007 Nature 448:7151;Maherali等人,2007 Cell Stem Cell 1:55-70;Lowry等人,2008 PNAS 105:2883-2888;Park等人,2008 Nature 451:141-146.;Takahashi等人,2007 Cell 131, 861-872;美國專利第8,546,140號;美國專利第7,033,831號及美國專利第8,268,620號。可使用此項技術中已知的方法將iPS細胞分化成β細胞,參見例如美國專利公開案第20170081641號及美國專利公開案第20130164787號,以及Millette及Georgia, 「Gene Editing and Human Pluripotent Stem Cells: Tools for advancing Diabetes Disease Modeling and Beta Cell Development」, Current Diabetes Reports 2017年11月, 17: 116;美國專利申請案第20130273651號;Shi, Y.,等人 Stem Cells., 25: 656-662 (2005);或Tateishi, K.等人,J Biol Chem., 283: 31601-31607 (2008)。Any suitable somatic cells from an individual can be reprogrammed into iPS cells by methods known in the art, see for example, Pagliuca and Melton (2013) How to make a functional β-cell, Development (3013) 140(12); 2472-2483; Yu et al. (2007). Induced pluripotent stem cell lines der ived from human somatic cells. Science 318, 1917-1920; Takahashi and Yamanaka, 2006, Cell 126(4):663-676; Wernig et al., 2007, Nature 448:7151; Okita et al., 2007 Nature 448:7151; Maherali et al., 2007 C ell Stem Cell 1:55-70; Lowry et al., 2008 PNAS 105:2883-2888; Park et al., 2008 Nature 451:141-146.; Takahashi et al., 2007 Cell 131, 861-872; U.S. Patent No. 8,546,140; U.S. Patent No. 7,033,831 and U.S. Patent No. 8,268,620. iPS cells can be differentiated into beta cells using methods known in the art, see, e.g., US Patent Publication No. 20170081641 and US Patent Publication No. 20130164787, and Millette and Georgia, "Gene Editing and Human Pluripotent Stem Cells: Tools for advancing Diabetes Disease Modeling and Beta Cell Development", Current Diabetes betes Reports 2017 Nov, 17: 116; US Patent Application No. 20130273651; Shi, Y., et al. Stem Cells., 25: 656-662 (2005); or Tateishi, K. et al., J Biol Chem., 283: 31601-31607 (2008).

較佳地,編碼趨除劑之序列可操作地連接於適用於β細胞中之表現的調節區。適合調節區為此項技術中已知的,且包括啟動子,諸如哺乳動物啟動子,其包括例如次黃嘌呤磷酸核糖基轉移酶(HPTR)、腺苷去胺酶、丙酮酸激酶、β-肌動蛋白啟動子、肌肉肌酸激酶啟動子及人類延長因子啟動子(EF1α)、GAPDH啟動子、肌動蛋白啟動子及泛素啟動子以及病毒啟動子(包括SV40早期啟動子、SV40晚期啟動子、金屬硫蛋白啟動子、鼠乳腺腫瘤病毒啟動子、勞斯肉瘤病毒啟動子(Rous sarcoma virus promoter)、多角體蛋白啟動子、人類免疫缺乏病毒(HIV)啟動子、細胞巨大病毒(CMV)啟動子、腺病毒啟動子、腺相關病毒啟動子、或單純疱疹病毒之胸苷激酶啟動子)。其他相關啟動子,例如病毒及真核啟動子亦為此項技術中熟知的(參見例如,Sambrook及Russell (Molecular Cloning: a laboratory manual, Cold Spring Harbor Laboratory Press))。可操作地連接於編碼趨除劑之序列的調節區可為適用於個體細胞中之表現的任何組成型啟動子。Preferably, the sequence encoding the chemoattractant is operably linked to a regulatory region suitable for expression in beta cells. Suitable regulatory regions are known in the art and include promoters such as mammalian promoters including, for example, hypoxanthine phosphoribosyltransferase (HPTR), adenosine deaminase, pyruvate kinase, β-actin promoter, muscle creatine kinase promoter and human elongation factor promoter (EF1α), GAPDH promoter, actin promoter and ubiquitin promoter as well as viral promoters (including SV40 early promoter, SV40 late promoter, metallothionein promoter, mouse milk Adenotumor virus promoter, Rous sarcoma virus promoter (Rous sarcoma virus promoter), polyhedrin promoter, human immunodeficiency virus (HIV) promoter, cytomegalovirus (CMV) promoter, adenovirus promoter, adeno-associated virus promoter, or herpes simplex virus thymidine kinase promoter). Other related promoters, such as viral and eukaryotic promoters, are also well known in the art (see, eg, Sambrook and Russell (Molecular Cloning: a laboratory manual, Cold Spring Harbor Laboratory Press)). The regulatory region operably linked to the sequence encoding the keloid may be any constitutive promoter suitable for expression in the individual cell.

本發明之表現趨除劑(例如CXCL12、CXCL13)之轉殖基因細胞,無論自體性或非自體性,例如同種異體,均可藉由此項技術中已知的用於投與β細胞的任何手段向有需要之個體投與。可以足夠提供能夠緩解與低胰島素量相關的症狀中之至少一些的胰島素量的量投與本發明之轉殖基因細胞。Transgenic cells of the present invention expressing a phenotypic agent (eg, CXCL12, CXCL13), whether autologous or non-autologous, eg, allogeneic, can be administered to an individual in need by any means known in the art for administering β cells. Transgenic cells of the invention can be administered in an amount sufficient to provide an amount of insulin capable of alleviating at least some of the symptoms associated with low insulin amounts.

本發明之另一態樣為一種治療有需要之個體中的糖尿病之方法,其包含以下步驟:(a)由個體獲得或衍生β細胞或產胰島素類β細胞;(b)將編碼趨除劑(例如CXCL12、CXCL13)之適合表現載體引入細胞中以形成表現所引入之趨除劑(例如CXCL12、CXCL13)的自體性轉殖基因細胞;及(c)將自體性轉殖基因細胞移植於個體中。Another aspect of the present invention is a method of treating diabetes in an individual in need thereof, comprising the steps of: (a) obtaining or deriving beta cells or insulin-producing beta cells from the individual; (b) introducing a suitable expression vector encoding a chemoattractant (e.g., CXCL12, CXCL13) into the cells to form autologous transgenic cells expressing the introduced chemoattractant (e.g., CXCL12, CXCL13); and (c) transplanting the autologous transgenic cells into the individual.

可利用適用於將外源性基因轉移於哺乳動物細胞,例如β細胞中之多種載體,包括整合至基因組中之載體及不整合至基因組中但以游離基因體之形式存在的載體;及用於將此類載體引入細胞中之方法,且各載體及方法為此項技術中已知的。舉例而言,可使用反轉錄病毒載體、慢病毒載體、腺病毒載體、腺相關(AAV)類載體及EBV類載體。參見例如,US 20110280842;Narayanavari及Izsvák, Cell Gene Therapy Insights 2017;3(2),131-158;Hardee等人,Genes 2107, 8, 65;Tipanee等人,Bioscience Reports (2017) 37;及Chira等人 Oncotarget, 第6卷, 第31期, 第30675-30703頁。Various vectors suitable for transferring exogenous genes into mammalian cells, such as β cells, including vectors integrated into the genome and vectors not integrated into the genome but present in the form of episomes; and methods for introducing such vectors into cells are available, and each vector and method are known in the art. For example, retroviral vectors, lentiviral vectors, adenoviral vectors, adeno-associated (AAV) type vectors and EBV type vectors can be used. See, eg, US 20110280842; Narayanavari and Izsvák, Cell Gene Therapy Insights 2017;3(2), 131-158; Hardee et al., Genes 2107, 8, 65; Tipanee et al., Bioscience Reports (2017) 37; and Chira et al. Oncotarget, Vol. 6, No. 3 Issue 1, pp. 30675-30703.

本發明之另一態樣為一種用於提高β細胞在包含免疫細胞之生物樣本中之存活期的方法,其包含將編碼趨除劑(例如CXCL12、CXCL13)之表現載體引入β細胞中;或藉由編輯β細胞之基因組以使得β細胞表現趨除量之趨除劑(例如CXCL12、CXCL13)。在本發明之一態樣中,趨除劑(例如CXCL12、CXCL13)係由β細胞以足以阻斷或抑制免疫細胞(例如,T細胞、B細胞及/或NK細胞)向β細胞遷移之量表現。在本發明之一態樣中,趨除劑(例如CXCL12、CXCL13)係由β細胞以足以使β細胞排斥免疫細胞之量表現。在本發明之一態樣中,基因改造β細胞處於個體,例如患有第1型或第2型糖尿病之人類個體中。在一個實施例中,β細胞為個體之自體性β細胞。Another aspect of the present invention is a method for increasing the survival period of β-cells in a biological sample containing immune cells, which comprises introducing an expression vector encoding a chemoattractant (such as CXCL12, CXCL13) into the β-cell; or editing the genome of the β-cell so that the β-cell expresses the chemoattractant (such as CXCL12, CXCL13). In one aspect of the invention, the chemoattractant (eg, CXCL12, CXCL13) is expressed by β cells in an amount sufficient to block or inhibit migration of immune cells (eg, T cells, B cells and/or NK cells) to β cells. In one aspect of the invention, an attractant (eg, CXCL12, CXCL13) is expressed by a beta cell in an amount sufficient for the beta cell to repel the immune cell. In one aspect of the invention, the genetically engineered beta cells are in an individual, such as a human individual with type 1 or type 2 diabetes. In one embodiment, the beta cells are autologous beta cells of the individual.

用於向哺乳動物細胞遞送病毒載體及非病毒載體之方法為此項技術中所熟知的,且包括例如脂質體轉染、顯微注射、彈道法(ballistics)、病毒體(virosome)、脂質體、免疫脂質體、聚陽離子或脂質-核酸結合物、裸DNA、人工病毒粒子及DNA之試劑增強性攝取。脂質體轉染試劑為市售的(例如TransfectamTM 及LipofectinTM )。適用於聚核苷酸之高效受體識別脂質體轉染的陽離子型及中性脂質為已知的。核酸可遞送至細胞(離體投與)或目標組織(活體內投與)。脂質:核酸複合物,包括靶脂質體,諸如免疫脂質之複合物的製備為熟習此項技術者熟知的。可使用經重組介導之系統來將載體引入細胞中。此類重組方法包括例如使用位點特異性重組酶,如Cre、Flp或PHIC31 (參見例如,Oumard等人,Cytotechnology (2006) 50: 93-108) ,其可介導轉殖基因之定向插入。Methods for delivering viral and non-viral vectors to mammalian cells are well known in the art and include, for example, lipofection, microinjection, ballistics, virosomes, liposomes, immunoliposomes, polycations or lipid-nucleic acid conjugates, naked DNA, artificial virions, and agent-enhanced uptake of DNA. Lipofectamine reagents are commercially available (eg Transfectam and Lipofectin ). Cationic and neutral lipids suitable for efficient receptor-recognizing liposome transfection of polynucleotides are known. Nucleic acids can be delivered to cells (administration ex vivo) or tissues of interest (administration in vivo). The preparation of lipid:nucleic acid complexes, including targeted liposomes, such as immunolipid complexes is well known to those skilled in the art. Recombination-mediated systems can be used to introduce vectors into cells. Such recombination methods include, for example, the use of site-specific recombinases, such as Cre, Flp, or PHIC31 (see, e.g., Oumard et al., Cytotechnology (2006) 50: 93-108), which can mediate directed insertion of transgenes.

適用於本發明中之載體包括包含編碼趨除劑(例如CXCL12、CXCL13)之核酸的表現載體,該核酸可操作地連接於啟動子以指導轉錄。適合啟動子為此項技術中所熟知的,且描述於例如Sambrook及Russell (Molecular Cloning: a laboratory manual, Cold Spring Harbor Laboratory Press)中。用於指導趨除劑(例如CXCL12、CXCL13)之表現的啟動子可為例如SV40早期啟動子、SV40晚期啟動子、金屬硫蛋白啟動子、鼠乳腺腫瘤病毒啟動子、勞斯肉瘤病毒啟動子或顯示對在哺乳動物細胞中之表現有效的其他啟動子。Vectors suitable for use in the present invention include expression vectors comprising a nucleic acid encoding an attractant (eg, CXCL12, CXCL13) operably linked to a promoter to direct transcription. Suitable promoters are well known in the art and are described, for example, in Sambrook and Russell (Molecular Cloning: a laboratory manual, Cold Spring Harbor Laboratory Press). The promoter used to direct the expression of a chemoattractant (e.g., CXCL12, CXCL13) can be, for example, the SV40 early promoter, the SV40 late promoter, the metallothionein promoter, the murine mammary tumor virus promoter, the Rous sarcoma virus promoter, or other promoters shown to be effective for expression in mammalian cells.

適用於本發明方法中之載體包括例如SV40載體、乳頭狀瘤病毒載體、埃-巴二氏病毒載體(Epstein-Barr virus)、反轉錄病毒載體及慢病毒載體。Vectors suitable for use in the methods of the present invention include, for example, SV40 vectors, papilloma virus vectors, Epstein-Barr virus vectors, retroviral vectors, and lentiviral vectors.

本發明中所用之載體可包含來自真核病毒,例如SV40、乳頭狀瘤病毒及埃-巴二氏病毒之調節元件,包括例如針對轉錄物之高效聚腺苷酸化、轉錄終止、核糖體結合及/或轉譯終止之信號。載體之額外元件可包括例如強化子及異源剪接內含子信號。Vectors used in the present invention may contain regulatory elements from eukaryotic viruses, such as SV40, papillomavirus, and Epstein-Barr virus, including, for example, signals for efficient polyadenylation of transcripts, transcription termination, ribosome binding and/or translation termination. Additional elements of the vector may include, for example, enhancers and heterologous splicing intron signals.

在本發明之一實施例中,β細胞之基因組可經基因改造以增加內源性趨除劑(例如CXCL12、CXCL13)基因之表現量。此類增加之表現可藉由以下來達成:引入可操作地連接於內源性趨除劑(例如CXCL12、CXCL13)基因之異源啟動子;或改變內源性趨除劑(例如CXCL12、CXCL13)啟動子,以使得β細胞表現趨除量之趨除劑(例如CXCL12、CXCL13)。此類增加之表現可藉由來達成:將啟動子引入β細胞之基因組中以使得其可操作地連接於編碼內源性趨除劑之序列,且由此表現或過度表現呈趨除量之趨除劑。In one embodiment of the present invention, the genome of β cells can be genetically modified to increase the expression of endogenous chemokine (eg CXCL12, CXCL13) genes. Such increased expression can be achieved by: introducing a heterologous promoter operably linked to the endogenous chemoattractant (e.g., CXCL12, CXCL13) gene; or altering the endogenous chemoattractant (e.g., CXCL12, CXCL13) promoter such that the beta cells express a chemoattractant (e.g., CXCL12, CXCL13). Such increased expression can be achieved by introducing a promoter into the genome of the beta cell such that it is operably linked to a sequence encoding an endogenous qualifier, and thereby expresses or overexpresses the qualifier in invasive amounts.

用於改造基因組之基因編輯技術為此項技術中所熟知的,且包括例如CRISPR/CAS 9、Piggybac、睡美人(Sleeping Beauty)基因組編輯系統(參見例如,Zhang等人 Molecular Therapy Nucleic Acids, 第9卷, 2017年12月, 第230-241頁);系統(參見例如,Cong等人,Science. 2013; 339(6121): 819-23;Mali等人,Science. 2013; 339(6121): 823-6;González等人,Cell Stem Cell. 2014; 15(2): 215-26;He等人,Nucleic Acids Res. 2016; 44(9);Hsu等人,Cell. 2014; 157(6): 1262-78.);鋅指核酸酶類系統(參見例如,Porteus及Carroll, Nat Biotechnol. 2005; 23(8): 967-73;Urnov等人,Nat Rev Genet. 2010; 11(9): 636-46);TALEN類系統(類轉錄活化因子效應核酸酶) (參見例如,Cermak等人,Nucleic Acids Res. 2011; 39(12);Hockemeyer等人,Nat Biotechnol. 2011; 29(8): 731-4;Joung及Sander JD, Nat Rev Mol Cell Biol. 2013; 14(1): 49-55;Miller等人,Nat Biotechnol. 2011; 29(2): 143-8;及Reyon等人,Nat Biotechnol. 2012; 30(5): 460-5)。Gene editing technologies for modifying genomes are well known in the art and include, for example, CRISPR/CAS 9, Piggybac, Sleeping Beauty genome editing systems (see, e.g., Zhang et al. Molecular Therapy Nucleic Acids, Vol. 9, Dec. 2017, pp. 230-241); systems (see, e.g., Cong et al., Science. 2013; 339(6121): 819-23; Mali et al., Science. 2013; 339(6121): 823-6; González et al., Cell Stem Cell. 2014; 15(2): 215-26; He et al., Nucleic Acids Res. 2016; 44(9); 2014; 157(6): 1262-78.); zinc finger nuclease-like systems (see for example, Porteus and Carroll, Nat Biotechnol. 2005; 23(8): 967-73; Urnov et al., Nat Rev Genet. 2010; 11(9): 636-46); TALEN-like systems (transcription activator-like effector nucleases) (See eg, Cermak et al., Nucleic Acids Res. 2011; 39(12); Hockemeyer et al., Nat Biotechnol. 2011; 29(8): 731-4; Joung and Sander JD, Nat Rev Mol Cell Biol. 2013; 14(1): 49-55; Miller et al., Nat Biotechnol. . 2011; 29(2): 143-8; and Reyon et al., Nat Biotechnol. 2012; 30(5): 460-5).

在一個實施例中,用使得細胞可存活且能夠控制患者之血糖但無法複製(亦即,誘導細胞衰老)之試劑處理本文所述之轉殖基因β細胞。一種此類試劑為絲裂黴素C,其為已知的DNA交聯劑。處理後,此等細胞中之DNA交聯,藉此使得不可能形成複製所需的單鏈DNA。此類處理防止細胞,尤其由幹細胞產生之彼等細胞分裂,以使得若細胞變成癌細胞,其不可分裂。能夠誘導細胞衰老之其他已知試劑包括由Petrova等人,「Small Molecule Compounds that Induce Cellular Senescence」 Aging Cell, 15(6):999-1017 (2016)敍述之彼等試劑,,其以全文引用之方式併入本文中。僅舉例而言,此類試劑包括引起端粒由於複製相關端粒縮短,次細胞毒性應激,諸如暴露於UV、γ照射、過氧化氫及低氧發生功能障礙之試劑。使本發明之β細胞呈現非複製之特定手段並非至關重要,但其限制條件為此等細胞可在不存在細胞分裂之風險下植入。研究顯示,成人胰臟具有極少的β細胞轉換,其表明限制細胞分裂之能力將幾乎不會對植入細胞產生胰島素造成影響。. 參見例如,Perl等人,The Journal of Clinical Endocrinology & Metabolism , 第95卷, 第10期, 2010年10月1日, 第 E234-E239頁。In one embodiment, the transgenic beta cells described herein are treated with an agent that renders the cells viable and capable of controlling the patient's blood glucose but unable to replicate (ie, induce cellular senescence). One such reagent is mitomycin C, a known DNA cross-linking agent. After treatment, the DNA in these cells is cross-linked, thereby making it impossible to form the single-stranded DNA required for replication. Such treatments prevent cells, especially those arising from stem cells, from dividing, so that if a cell becomes a cancer cell, it cannot divide. Other known agents capable of inducing cellular senescence include those described by Petrova et al., "Small Molecule Compounds that Induce Cellular Senescence" Aging Cell, 15(6):999-1017 (2016), which is incorporated herein by reference in its entirety. By way of example only, such agents include agents that cause telomere dysfunction due to replication-associated telomere shortening, secondary cytotoxic stress, such as exposure to UV, gamma irradiation, hydrogen peroxide, and hypoxia. The particular means by which the β-cells of the invention are rendered non-replicating is not critical, but the proviso is that these cells can be implanted without the risk of cell division. The adult pancreas has very little beta-cell turnover, the study showed, suggesting that limiting the ability of the cells to divide would have little effect on insulin production by the implanted cells. . See eg, Perl et al., The Journal of Clinical Endocrinology & Metabolism , Vol. 95, No. 10, Oct. 1, 2010, pp. E234-E239.

本發明之另一態樣為一種調節個體中之胰島素量的方法,其包含向有需要之個體投與本發明之β細胞,其中β細胞表現胰島素且產生呈趨除量之趨除劑(例如CXCL12、CXCL13)。β細胞可為自體性β細胞或非自體性β細胞,例如同種異體β細胞,且可具有表現趨除劑之載體,該載體可整合至β細胞基因組中或以游離方式存在。在本發明之一實施例中,轉殖基因β細胞可經基因改造以過度表現呈趨除量之內源性趨除劑(例如CXCL12、CXCL13)。Another aspect of the invention is a method of modulating the amount of insulin in an individual comprising administering to an individual in need thereof a β-cell of the invention, wherein the β-cell expresses insulin and produces a scavenger (eg, CXCL12, CXCL13) in a depressant amount. The beta cell may be an autologous beta cell or a non-autologous beta cell, such as an allogeneic beta cell, and may have a vector expressing a tactic agent, either integrated into the beta cell genome or present episomally. In one embodiment of the invention, the transgenic β-cells can be genetically engineered to overexpress endogenous scavenger agents (eg, CXCL12, CXCL13) in scavenging amounts.

將本文所述之轉殖基因β細胞引入個體中的方法為熟習此項技術者熟知的,且包括(但不限於)注射、靜脈內、門靜脈內或非經腸投與。可進行單次、多次、連續或間歇投與。參見例如, Schuetz及Markmann, Curr Transplant Rep. 2016年9月; 3(3): 254-263。Methods of introducing transgenic beta cells described herein into an individual are well known to those skilled in the art and include, but are not limited to, injection, intravenous, portal vein or parenteral administration. Single, multiple, continuous or intermittent administration can be performed. See, eg, Schuetz and Markmann, Curr Transplant Rep. 2016 Sep; 3(3): 254-263.

醫藥學上可接受之載劑包括無菌水溶液或分散液及用於臨時製備無菌可注射溶液或分散液之無菌散劑。用於醫藥學活性物質,包括細胞的培養基及試劑的使用為此項技術中熟知的。用於靜脈內輸注β細胞之典型醫藥組合物可經製備以含有250 ml無菌林格氏溶液(Ringer's solution)及100 mg組合。用於製備可非經腸投與之化合物的實際方法將為熟習此項技術者已知或顯而易知的且較詳細地描述在例如Remington's Pharmaceutical Science, 第17版, Mack Publishing Company, Easton, Pa. (1985)及其第18版及第19版中,,其以引用之方式併入本文中。Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. The use of media and reagents for pharmaceutically active substances, including cells, is well known in the art. A typical pharmaceutical composition for intravenous infusion of beta cells may be prepared to contain 250 ml of sterile Ringer's solution and 100 mg of combination. Actual methods for preparing parenterally administrable compounds will be known or apparent to those skilled in the art and are described in some detail, for example, in Remington's Pharmaceutical Science, 17th ed., Mack Publishing Company, Easton, Pa. (1985) and 18th and 19th ed., which are incorporated herein by reference.

本發明之轉殖基因及基因改造β細胞可引入若干不同的此項技術中熟知的個體之部位中之任一者中,包括(但不限於)胰臟、腹腔、腎臟、肝臟、門靜脈或脾臟。The transgenic and genetically modified beta cells of the present invention can be introduced into any of a number of different sites in an individual well known in the art, including but not limited to the pancreas, abdominal cavity, kidney, liver, portal vein, or spleen.

此外,為了避免發生可引起患者罹患腫瘤之可能性的任何可能的轉殖基因β細胞轉變成癌細胞,可藉由與已知試劑,諸如絲裂黴素C接觸或暴露於來自電離輻射、低氧、過氧化氫等之次毒性應激而使轉殖基因β細胞為衰老細胞。衍生自多能幹細胞之細胞典型地在不當細胞分裂期間或由於免疫細胞清除而經歷細胞凋亡。本文所述之衰老的轉殖基因β細胞不能分裂,藉此消除在細胞分裂期間出現的細胞凋亡觸發事件。此外,本文所述之衰老的轉殖基因β細胞具有抗免疫細胞性,藉此提供防止由於免疫細胞清除導致細胞凋亡誘導。因此,預期本文所述之轉殖基因β細胞將具有比非衰老轉殖基因β細胞要長至顯著較長的壽命。Furthermore, in order to avoid any possible transformation of transgenic beta cells into cancerous cells that would raise the possibility of the patient developing a tumor, the transgenic beta cells can be rendered senescent by exposure to known agents such as mitomycin C or exposure to secondary toxic stress from ionizing radiation, hypoxia, hydrogen peroxide, etc. Cells derived from pluripotent stem cells typically undergo apoptosis during inappropriate cell division or as a result of immune cell clearance. The senescent transgenic beta cells described herein are unable to divide, thereby eliminating the apoptosis triggering event that occurs during cell division. In addition, the senescent transgenic beta cells described herein are immune cell resistant, thereby providing protection against the induction of apoptosis due to immune cell clearance. Accordingly, it is expected that the transgenic beta cells described herein will have an up to significantly longer lifespan than non-senescent transgenic beta cells.

轉殖基因,且較佳衰老的改造β細胞可經由移植物移植於個體中。理想的β細胞移植部位將為支持移植細胞在個體中之植入、長期功能及存活期且出於最大患者安全性而可輕易獲得的部位。植入部位包括肝臟、腸道、皮下及胰臟部位。Transgenic, and preferably senescent, engineered beta cells can be transplanted into an individual via a graft. The ideal site for beta cell transplantation will be one that supports engraftment, long-term function and survival of the transplanted cells in an individual and is readily accessible for maximum patient safety. Implantation sites include liver, intestinal tract, subcutaneous and pancreatic sites.

本文中使用之以下縮寫具有以下含義,且若縮寫未加以界定,則其具有其一般公認科學含義。在本文中使用已獲確認的單字母縮寫來敍述胺基酸。 FLAG = DYKDDDDK蛋白質標籤(SEQ ID NO: 10) g/L = 每公升之公克數 HRP = 辣根過氧化酶 LDH = 乳酸去氫酶 iBLOT = 半乾燥蛋白質傳送裝置(Invitrogen) MES = 2-(N-嗎啉基)乙磺酸 mL = 毫升 N/A = 不適用 nM = 奈莫耳 PBMC = 外周血單核細胞 PBS = 磷酸鹽緩衝鹽水 TMB = 3,3',5,5'-四甲基聯苯胺 mL = 微升 mg = 微克 × g = 倍重力 實例實例 1 :用於評定 CXCL12-a CXCL12-b 同功異型物之表現量的模型細胞 The following abbreviations used herein have the following meanings and, if not defined, have their generally accepted scientific meanings. Amino acids are described herein using established one-letter abbreviations. FLAG = DYKDDDDK protein tag (SEQ ID NO: 10) g/L = grams per liter HRP = horseradish peroxidase LDH = lactate dehydrogenase iBLOT = semi-dry protein delivery device (Invitrogen) MES = 2-(N-morpholino)ethanesulfonic acid mL = milliliters N/A = not applicable nM = nanomoles PBMC = peripheral blood mononuclear cells PBS = phosphate buffered salineTM B = 3,3',5,5'-tetramethylbenzidine mL = microliter mg = microgram × g = double gravity Example Example 1 : Model cells used to evaluate the expression of CXCL12-a and CXCL12-b isoforms

用2種不同的CXCL12 (α及β)之同功異型物使用可在市面上購得的針對各同功異型物之質體(購自GenScript之質體)轉染HEK293細胞。用250 μg/mL G418(可購自ThermoFisher)選定經轉染之細胞且產生針對各同功異型物之穩定池。使細胞處於適合培養基中3天。用分析稀釋緩衝液1:1稀釋來自表現CXCL12α及CXCL12β的經轉染HEK293細胞之改良性培養基。針對各同功異型物確立兩個獨立池,且隨後藉由使用標準化濃度曲線之吸收度來獲得各同功異型物在溶液中之濃度。重複此實驗兩次且結果如下: CXCL12α CXCL12β 1. 310 nM 1410 nM 2. 274 nM 1330 nMHEK293 cells were transfected with 2 different isoforms of CXCL12 (α and β) using commercially available plastids for each isoform (plastids purchased from GenScript). Transfected cells were selected with 250 μg/mL G418 (available from ThermoFisher) and a stable pool was generated for each isoform. Cells were kept in appropriate medium for 3 days. Modified media from transfected HEK293 cells expressing CXCL12α and CXCL12β were diluted 1:1 with Assay Dilution Buffer. Two independent cells were established for each isoform, and the concentration of each isoform in solution was then obtained by using the absorbance of the normalized concentration curve. This experiment was repeated twice and the results were as follows: CXCL12α CXCL12β 1. 310 nM 1410 nM 2. 274 nM 1330 nM

以上結果顯示,與表現CXCL12α之轉殖基因模型細胞相比,轉殖基因模型細胞表現呈明顯較高量之CXCL12β。 實例2 -用於評定 CXCL12 之其他同功異型物之表現量的模型細胞 用5種不同的CXCL12 (α及β)之同功異型物使用可在市面上購得的針對各同功異型物之質體(購自GenScript之質體)轉染HEK293細胞。用250 μg/mL G418(可購自ThermoFisher)選定經轉染之細胞且產生針對各同功異型物之穩定池。使細胞處於適合培養基中3天。用MES緩衝液在4%至8% NuPage凝膠(可購自ThermoFisher)中分離改良性培養基且轉移至硝化纖維素(iBLOT)。The above results show that, compared with the transgenic model cells expressing CXCL12α, the transgenic model cells express a significantly higher amount of CXCL12β. Example 2 - Model Cells for Assessing the Expression of Other Isoforms of CXCL12 HEK293 cells were transfected with 5 different isoforms of CXCL12 (α and β) using commercially available plastids for each isoform (plastids purchased from GenScript). Transfected cells were selected with 250 μg/mL G418 (available from ThermoFisher) and a stable pool was generated for each isoform. Cells were kept in appropriate medium for 3 days. Modified medium was separated on 4% to 8% NuPage gels (available from ThermoFisher) with MES buffer and transferred to nitrocellulose (iBLOT).

用經HRP標記的抗FLAG標籤抗體/TMB色素原(購自GenScript)在西方墨點法上偵測表現量,如圖1中所示。結果證明,CXCL12之γ、δ及θ同功異型物具有比α或β同功異型物要高的濃度。實例 3 :轉殖基因 β 細胞之製備 The expression level was detected by Western blot method with HRP-labeled anti-FLAG tag antibody/TMB chromogen (purchased from GenScript), as shown in FIG. 1 . The results demonstrate that the gamma, delta and theta isoforms of CXCL12 have higher concentrations than the alpha or beta isoforms. Example 3 : Preparation of transgenic β cells

衍生自人類誘導之多能幹細胞的胰臟β細胞係購自Takara Bio USA公司(加利福尼亞州山景城(Mountain View, CA))且根據所提供之說明書進行培養。Pancreatic β-cell lines derived from human induced pluripotent stem cells were purchased from Takara Bio USA (Mountain View, CA) and cultured according to the provided instructions.

用含有人類CXCL12同型(CXCL12a/SDF-1α或CXCL12b/SDF-1β)或對照之慢病毒載體(pLenti-C-Myc-DDK,馬里蘭州羅克維爾(Rockville, MD)之OriGene Technologies)轉導細胞。以每個β細胞約10:1之比率使用慢病毒載體。包括標籤(帶下劃線)之序列提供於下文中。藉由ELISA (佐治亞州諾克羅斯(Norcross, GA)之RayBioTech)測定CXCL12同型之濃度(表1)。 CXCL12a (亦稱為SDF1a) 寄存編號NM_199168 CXCL12b (亦稱為SDF1b) 寄存編號NM_000609 實例 4 :轉殖基因 β 細胞排斥 PBMC Cells were transduced with a lentiviral vector (pLenti-C-Myc-DDK, OriGene Technologies, Rockville, MD) containing a human CXCL12 isotype (CXCL12a/SDF-1α or CXCL12b/SDF-1β) or a control. Lentiviral vectors were used at a ratio of approximately 10:1 per beta cell. Sequences including tags (underlined) are provided below. Concentrations of CXCL12 isoforms were determined by ELISA (RayBioTech, Norcross, GA) (Table 1). CXCL12a (also known as SDF1a) deposit number NM_199168 CXCL12b (also known as SDF1b) deposit number NM_000609 Example 4 : Transgenic β Cells Reject PBMCs

使來自實例3之轉殖基因β細胞與人類外周血單核細胞(PBMC,密歇根州諾維(Novi, MI)之Innovative Research)以30:1 (PBMC與β細胞)之比率接觸。簡言之,將PBMC再懸浮於全β培養基中,計數且添加100 μL PBMC (以將經表現之CXCL12的稀釋度降至最低)加以調整以實現30:1 PBMC:β細胞比率。最終體積為1.1 mL。亦產生不存在PBMC下之β細胞及不存在β細胞下之PBMC的背景對照。緊接著,自各樣本移除150 μL培養基且以1200 × g離心10分鐘。移除上清液且儲存於4℃下(零時)。使細胞返回至培育箱且以與零時樣本相似之方式在之後24及48小時時取樣。Transgenic β cells from Example 3 were contacted with human peripheral blood mononuclear cells (PBMCs, Innovative Research, Novi, MI) at a ratio of 30:1 (PBMCs to β cells). Briefly, PBMCs were resuspended in complete beta medium, counted and adjusted by adding 100 μL of PBMCs (to minimize dilution of expressed CXCL12) to achieve a 30:1 PBMC:β cell ratio. The final volume was 1.1 mL. Background controls for β cells in the absence of PBMCs and PBMCs in the absence of β cells were also generated. Next, 150 μL of medium was removed from each sample and centrifuged at 1200×g for 10 minutes. The supernatant was removed and stored at 4°C (hour zero). Cells were returned to the incubator and sampled 24 and 48 hours later in a similar manner to the time zero sample.

在根據製造商說明書使用Pierce LDH細胞毒性分析套組(Thermo Scientific)接觸之後24及48小時時測試LDH之釋放量。增加之LDH為細胞毒性(細胞溶解)之指標。LDH release was tested at 24 and 48 hours after exposure using the Pierce LDH Cytotoxicity Assay Kit (Thermo Scientific) according to the manufacturer's instructions. Increased LDH is an indicator of cytotoxicity (cytolysis).

來自代表性實驗之資料(減除背景)提供於表1及圖2A中。來自第二代表性實驗之資料提供於圖2B中。 表1. LDH及CXCL12量 Data (background subtracted) from representative experiments are provided in Table 1 and Figure 2A. Data from a second representative experiment are presented in Figure 2B. Table 1. LDH and CXCL12 levels

此等資料表明,藉由β胰島細胞表現之CXCL12保護β胰島細胞不受免疫細胞攻擊,藉此使得其具有抗性。在此實驗中表現呈比SDF1a/CXCL12a要高之量的表現SDF1b/CXCL12b的β細胞顯示,在PBMC存在下基本上沒有細胞毒性。實例 5 :轉殖基因 β 細胞之替代性製備 These data suggest that CXCL12 expressed by β-islet cells protects β-islet cells from attack by immune cells, thereby rendering them resistant. Beta cells expressing SDF1b/CXCL12b in higher amounts than SDF1a/CXCL12a in this experiment showed essentially no cytotoxicity in the presence of PBMCs. Example 5 : Alternative Preparation of Transgenic Beta Cells

用編碼CXCL12之反轉錄病毒表現載體或不編碼CXCL12之對照反轉錄病毒載體活體外轉染或感染自患有第1型糖尿病之受試者分離的β細胞。使用博伊登室分析如Poznansky等人,Journal of Clinical Investigation, 109, 1101 (2002)中先前所述,針對趨除量之CXCL12的表現來分析攜帶編碼CXCL12之反轉錄病毒載體的轉殖基因β細胞。預期在此分析中,表現至少100 nM CXCL12之轉殖基因β細胞將排斥免疫細胞。實例 6 :轉殖基因 β 細胞之強迫衰老對轉殖基因細胞介素表現之影響 β cells isolated from subjects with type 1 diabetes were transfected or infected in vitro with a retroviral expression vector encoding CXCL12 or a control retroviral vector not encoding CXCL12. Transgenic β cells carrying a retroviral vector encoding CXCL12 were analyzed for the expression of excitatory amounts of CXCL12 using Boyden chamber assay as previously described in Poznansky et al., Journal of Clinical Investigation, 109, 1101 (2002). It was expected that transgenic β cells expressing at least 100 nM CXCL12 would reject immune cells in this assay. Example 6 : Effect of forced senescence of transgenic β cells on expression of transgenic cytokines

如實例3中所述製備β細胞。藉由ELISA分析在絲裂黴素C (購自Santa Cruz Biotechnology)處理之前的SDF1a/CXCL12a及SDF1b/CXCL12b之表現量以測定基線表現(「之前」)。用含有10 μg/mL絲裂黴素C (一種已知會誘導衰老的試劑)之新製培養基替代培養基。使細胞返回至培育箱持續2小時。藉由平緩移液移除含有絲裂黴素C之培養基。用PBS洗滌細胞兩次。第二次洗滌之後,將細胞補充新製完全培養基。藉由ELISA分析測定SDF1a/CXCL12a或SDF1b/CXCL12b表現。Beta cells were prepared as described in Example 3. Baseline expression ("before") was determined by ELISA analysis of the expression levels of SDF1a/CXCL12a and SDF1b/CXCL12b prior to mitomycin C (purchased from Santa Cruz Biotechnology) treatment. The medium was replaced with fresh medium containing 10 μg/mL mitomycin C, an agent known to induce senescence. The cells were returned to the incubator for 2 hours. The medium containing mitomycin C was removed by gentle pipetting. Cells were washed twice with PBS. After the second wash, the cells were supplemented with fresh complete medium. SDF1a/CXCL12a or SDF1b/CXCL12b expression was determined by ELISA analysis.

來自兩個代表性實驗之資料顯示在表2及圖3A中。SDF1a/CXCL12a及SDF1b/CXCL12b表現不受轉殖基因β細胞之強迫衰老之影響。Data from two representative experiments are shown in Table 2 and Figure 3A. SDF1a/CXCL12a and SDF1b/CXCL12b expression was not affected by forced senescence of transgenic β cells.

表2:絲裂黴素C處理之前及之後的CXCL12a及CXCL12b量 實例 7 :轉殖基因 β 細胞之強迫衰老對 PBMC 攻擊之影響 Table 2: CXCL12a and CXCL12b levels before and after mitomycin C treatment Example 7 : Effect of Forced Senescence of Transgenic β Cells on PBMC Challenge

如實例3中所述製備β細胞。用絲裂黴素C或對照如實例4中所述處理細胞。如實例2中所述,使細胞與PBMC接觸。Beta cells were prepared as described in Example 3. Cells were treated with mitomycin C or control as described in Example 4. Cells were contacted with PBMCs as described in Example 2.

來自兩個代表性實驗之資料顯示在圖4A及圖4B中。LDH量不受轉殖基因β細胞之強迫衰老之影響。實例 8 :轉殖基因 β 細胞之強迫衰老對胰島素產生之影響 Data from two representative experiments are shown in Figure 4A and Figure 4B. LDH levels were not affected by forced senescence of transgenic β cells. Example 8 : Effect of Forced Senescence of Transgenic β Cells on Insulin Production

如實例3中所述製備β細胞。用絲裂黴素C或對照如實例4中所述處理細胞。Beta cells were prepared as described in Example 3. Cells were treated with mitomycin C or control as described in Example 4.

用1 mL第2培養基替代完全生長培養基且每24小時用培養基替代物在第2培養基上維持2天。在第3天,用高血糖培養基(4.5g/L葡萄糖)攻擊β細胞。高血糖攻擊之後24小時取用經調節之培養基的樣本且藉由夾心ELISA量測胰島素表現。Complete growth medium was replaced with 1 mL of 2nd medium and maintained on 2nd medium with medium replacement every 24 hours for 2 days. On day 3, β cells were challenged with high blood glucose medium (4.5 g/L glucose). Samples of conditioned medium were taken 24 hours after hyperglycemic challenge and insulin expression was measured by sandwich ELISA.

來自兩個代表性實驗之資料顯示在表3及圖5中。結果顯示,轉殖基因β細胞及轉殖基因衰老β細胞產生與回應於高血糖攻擊之對照β細胞實質上相等的量之胰島素。 表3:回應於高血糖攻擊之胰島素表現 實例 9 :轉殖基因 β 細胞之活體內評估 Data from two representative experiments are shown in Table 3 and Figure 5. The results showed that transgenic β cells and transgenic senescent β cells produced substantially equal amounts of insulin as control β cells in response to a hyperglycemic challenge. Table 3: Insulin performance in response to hyperglycemic challenge Example 9 : In Vivo Evaluation of Transgenic β Cells

向具有人類化免疫系統之人類化小鼠,參見例如,N. Walsh, 「Humanized mouse models of clinical disease」, Annu Rev Pathol 2017, 12, 187-215;E. Yoshihara等人,投與表現趨除量之CXCL12的轉殖基因人類β細胞或對照轉殖基因人類β細胞,且在初始投與之後的不同時間點分析胰島素產量及轉殖基因β細胞在小鼠中之存活期。預期表現趨除量之CXCL12的轉殖基因人類β細胞將比對照轉殖基因人類β細胞存活更長時段。亦預期,接受表現趨除量之CXCL12的轉殖基因人類β細胞的小鼠亦將比接受對照轉殖基因人類β細胞的小鼠具有較高量之人類胰島素,且相較於投與對照轉殖基因人類β細胞的小鼠中之量,較高量之人類胰島素將存留較長時段。Humanized mice with a humanized immune system, see e.g., N. Walsh, "Humanized mouse models of clinical disease", Annu Rev Pathol 2017, 12, 187-215; E. Yoshihara et al., were administered transgenic human beta cells expressing a depletion of CXCL12 or control transgenic human beta cells and analyzed insulin production and migration at various time points after initial administration Genetic β-cell survival in mice. It is expected that transgenic human beta cells expressing a depleted amount of CXCL12 will survive for a longer period of time than control transgenic human beta cells. It is also expected that mice receiving transgenic human β cells expressing depletion amounts of CXCL12 will also have higher amounts of human insulin than mice receiving control transgenic human β cells, and that higher amounts of human insulin will persist for longer periods of time compared to the amount in mice administered control transgenic human β cells.

向具有人類化免疫系統之人類化小鼠,參見例如,N. Walsh, 「Humanized mouse models of clinical disease」, Annu Rev Pathol 2017, 12, 187-215;E. Yoshihara等人,投與過度表現CXCL12的由內源性CXCL12基因基因改造之人類β細胞或對照人類β細胞,且在初始投藥之後的不同時間點分析人類胰島素之產量及β細胞在小鼠中之存活期。預期,過度表現CXCL12之基因改造人類β細胞將比未經基因改造以過度表現CXCL12的對照人類β細胞存活更長時段。亦預期,接受過度表現CXCL12的基因改造人類β細胞的小鼠亦將比接受對照人類β細胞的小鼠具有較高量之人類胰島素,且相較於投與對照人類β細胞的小鼠中之量,較高量之人類胰島素將存留較長時段。Humanized mice with humanized immune systems, see e.g., N. Walsh, "Humanized mouse models of clinical disease", Annu Rev Pathol 2017, 12, 187-215; E. Yoshihara et al., were administered CXCL12-overexpressing human beta cells genetically engineered with the endogenous CXCL12 gene or control human beta cells, and humans were analyzed at various time points after initial administration Insulin production and β-cell survival in mice. It is expected that genetically engineered human beta cells that overexpress CXCL12 will survive for a longer period of time than control human beta cells that are not genetically engineered to overexpress CXCL12. It is also expected that mice receiving genetically modified human β cells overexpressing CXCL12 will also have higher amounts of human insulin than mice receiving control human β cells, and that higher amounts of human insulin will persist for a longer period of time compared to the amount in mice administered control human β cells.

視情況地,用使細胞內之DNA交聯的試劑(例如絲裂黴素C)處理細胞以防止細胞分裂。Optionally, cells are treated with an agent that cross-links DNA within the cell (eg, mitomycin C) to prevent cell division.

僅為說明本發明闡述前述描述且並不意謂為限制性的。由於本領域熟習此項技術者可進行併入本發明之精神及主旨的所述之實施例之修改,因此本發明應視為廣義上包括在申請專利範圍之範疇內的所有變化及其等效物。The foregoing description has been set forth in illustration of the invention only and is not meant to be limiting. Since those skilled in the art can make modifications to the described embodiments that incorporate the spirit and gist of the present invention, the present invention should be regarded as broadly including all changes and equivalents within the scope of the patent application.

圖1為西方墨點法之像片,其顯示當各過度表現於β細胞中時,CXCL12α、CXCL12β、CXCL12γ、CXCL12θ及CXCL12δ及CXCL14之相對量。Figure 1 is a Western blot photograph showing the relative amounts of CXCL12α, CXCL12β, CXCL12γ, CXCL12θ, and CXCL12δ and CXCL14 when each is overexpressed in β cells.

圖2A及圖2B為顯示自與PBMC以30:1 PBMC:β細胞比一起培育24及48小時的表現CXCL12α或表現CXCL12β之β細胞釋放的乳酸去氫酶(LDH,細胞溶解之標記物)的相對量的條形圖。2A and 2B are bar graphs showing the relative amount of lactate dehydrogenase (LDH, a marker of cell lysis) released from CXCL12α- or CXCL12β-expressing β-cells incubated with PBMCs at a 30:1 PBMC:β-cell ratio for 24 and 48 hours.

圖3顯示表現各細胞介素之兩個β細胞組中CXCL12α或CXCL12β之表現量。Fig. 3 shows the expression levels of CXCL12α or CXCL12β in two β cell groups expressing each cytokine.

圖4A及圖4B為顯示在存在或不存在β細胞之經誘導之衰老(藉由絲裂黴素C處理)下,自與PBMC以30:1 PBMC:β細胞比一起培育24及48小時的表現CXCL12α或表現CXCL12β之β細胞釋放的LDH之相對量的條形圖。4A and 4B are bar graphs showing the relative amount of LDH released from CXCL12α- or CXCL12β-expressing β-cells incubated with PBMCs at a 30:1 PBMC:β-cell ratio for 24 and 48 hours in the presence or absence of induced senescence of β-cells by mitomycin C treatment.

圖5顯示在存在或不存在用絲裂黴素C處理下,由表現CXCL12α或CXCL12β之β細胞的高血糖攻擊引起的胰島素誘導。Figure 5 shows insulin induction by hyperglycemic challenge of beta cells expressing CXCL12α or CXCL12β in the presence or absence of treatment with mitomycin C.

Figure 12_A0101_SEQ_0001
Figure 12_A0101_SEQ_0002
Figure 12_A0101_SEQ_0003
Figure 12_A0101_SEQ_0004
Figure 12_A0101_SEQ_0005
Figure 12_A0101_SEQ_0001
Figure 12_A0101_SEQ_0002
Figure 12_A0101_SEQ_0003
Figure 12_A0101_SEQ_0004
Figure 12_A0101_SEQ_0005

Figure 12_A0101_SEQ_0001
Figure 12_A0101_SEQ_0002
Figure 12_A0101_SEQ_0003
Figure 12_A0101_SEQ_0004
Figure 12_A0101_SEQ_0005
Figure 12_A0101_SEQ_0001
Figure 12_A0101_SEQ_0002
Figure 12_A0101_SEQ_0003
Figure 12_A0101_SEQ_0004
Figure 12_A0101_SEQ_0005

Claims (21)

一種人類β細胞,其表現或過度表現足以使得該β細胞對人類免疫細胞具有抗性的量的人類趨除(fugetactic)劑,其中該β細胞能夠表現胰島素且不能進行細胞分裂。 A human beta cell expressing or overexpressing a human fugetactic agent in an amount sufficient to render the beta cell resistant to human immune cells, wherein the beta cell is capable of expressing insulin and unable to undergo cell division. 一種人類β細胞,其包含載體,該載體本身包含編碼人類CXCL12之核酸序列,其中該β細胞對人類免疫細胞具有抗性。 A human β cell comprising a vector which itself comprises a nucleic acid sequence encoding human CXCL12, wherein the β cell is resistant to human immune cells. 如請求項1或2之人類β細胞,其中該細胞為:a)自患有第1型糖尿病之個體獲得的自體性β細胞,或b)同種異體β細胞。 The human β cell according to claim 1 or 2, wherein the cell is: a) an autologous β cell obtained from an individual suffering from type 1 diabetes, or b) an allogeneic β cell. 如請求項1或2之人類β細胞,其中該細胞表現呈趨除量之人類CXCL12。 The human β-cell according to claim 1 or 2, wherein the cell expresses a suppressed amount of human CXCL12. 如請求項4之人類β細胞,其中該CXCL12係選自由CXCL12α及CXCL12β組成之群。 The human β cell according to claim 4, wherein the CXCL12 is selected from the group consisting of CXCL12α and CXCL12β. 一種人類β細胞,其包含在內源性CXCL12編碼區上游的轉殖基因調節區,其中該β細胞對人類免疫細胞具有抗性。 A human beta cell comprising a transgenic regulatory region upstream of an endogenous CXCL12 coding region, wherein the beta cell is resistant to human immune cells. 如請求項6之人類β細胞,其中該細胞為自體性β細胞。 The human β-cell according to claim 6, wherein the cell is an autologous β-cell. 如請求項6之人類β細胞,其中該細胞為同種異體β細胞。 The human β-cell according to claim 6, wherein the cell is an allogeneic β-cell. 如請求項7之人類β細胞,其中該細胞為自患有第1型糖尿病之個體獲得或衍生的自體性β細胞。 The human β cell according to claim 7, wherein the cell is an autologous β cell obtained or derived from an individual suffering from type 1 diabetes. 如請求項8之人類β細胞,其中該細胞為自沒有第1型糖尿病之個體獲得或衍生的同種異體β細胞。 The human beta cell according to claim 8, wherein the cell is an allogeneic beta cell obtained or derived from an individual without type 1 diabetes. 如請求項6至10中任一項之人類β細胞,其中該轉殖基因調節區為外源性組成型或誘導性啟動子。 The human β cell according to any one of claims 6 to 10, wherein the transgene regulatory region is an exogenous constitutive or inducible promoter. 如請求項6至10中任一項之人類β細胞,其中該細胞表現呈趨除量之CXCL12。 The human beta cell according to any one of claims 6 to 10, wherein the cell expresses CXCL12 in a depressive amount. 如請求項12之人類β細胞,其中該CXCL12係選自由CXCL12α及CXCL12β組成之群。 The human β cell according to claim 12, wherein the CXCL12 is selected from the group consisting of CXCL12α and CXCL12β. 如請求項2及6至10中任一項之人類β細胞,其中該β細胞不能進行細胞分裂。 The human β cell according to any one of claims 2 and 6 to 10, wherein the β cell cannot perform cell division. 如請求項1、2及6至10中任一項之人類β細胞,其中該人類CXCL12係選自由以下組成之群:CXCL12α、CXCL12β、CXCL12δ及 CXCL12γ。 The human β cell according to any one of claims 1, 2 and 6 to 10, wherein the human CXCL12 is selected from the group consisting of CXCL12α, CXCL12β, CXCL12δ and CXCL12γ. 如請求項1、2及6至10中任一項之人類β細胞,其中該等人類免疫細胞包含NK細胞、細胞毒性T細胞及B細胞。 The human β cell according to any one of claims 1, 2 and 6 to 10, wherein the human immune cells comprise NK cells, cytotoxic T cells and B cells. 一種產生用於回應於高血糖環境之胰島素之活體外方法,其包含使該環境與如請求項1至16中任一項之人類β細胞群接觸。 An in vitro method of producing insulin in response to a hyperglycemic environment comprising contacting the environment with a population of human beta cells according to any one of claims 1 to 16. 如請求項17之方法,其中該高血糖環境包含人類免疫細胞。 The method according to claim 17, wherein the hyperglycemic environment comprises human immune cells. 如請求項18之方法,其中該等人類免疫細胞包含NK細胞、T細胞及B細胞。 The method according to claim 18, wherein the human immune cells comprise NK cells, T cells and B cells. 一種組合物,其供用於治療糖尿病,該組合物包含如請求項1至16中任一項之人類β細胞。 A composition for treating diabetes, the composition comprising the human β-cell according to any one of claims 1-16. 一種如請求項1至16中任一項之人類β細胞之用途,其係用於製備用於治療糖尿病之藥物。 A use of the human beta cell according to any one of claims 1 to 16, which is used for preparing a drug for treating diabetes.
TW107134978A 2017-10-03 2018-10-03 Treating diabetes with genetically modified beta cells TWI806907B (en)

Applications Claiming Priority (20)

Application Number Priority Date Filing Date Title
US201762567604P 2017-10-03 2017-10-03
US62/567,604 2017-10-03
US201762568117P 2017-10-04 2017-10-04
US62/568,117 2017-10-04
US201862637913P 2018-03-02 2018-03-02
US62/637,913 2018-03-02
US201862662651P 2018-04-25 2018-04-25
US62/662,651 2018-04-25
US201862694634P 2018-07-06 2018-07-06
US62/694,634 2018-07-06
US201862696603P 2018-07-11 2018-07-11
US62/696,603 2018-07-11
US201862717587P 2018-08-10 2018-08-10
US62/717,587 2018-08-10
US201862719975P 2018-08-20 2018-08-20
US62/719,975 2018-08-20
US201862723415P 2018-08-27 2018-08-27
US62/723,415 2018-08-27
US201862734910P 2018-09-21 2018-09-21
US62/734,910 2018-09-21

Publications (2)

Publication Number Publication Date
TW201923070A TW201923070A (en) 2019-06-16
TWI806907B true TWI806907B (en) 2023-07-01

Family

ID=67702074

Family Applications (1)

Application Number Title Priority Date Filing Date
TW107134978A TWI806907B (en) 2017-10-03 2018-10-03 Treating diabetes with genetically modified beta cells

Country Status (1)

Country Link
TW (1) TWI806907B (en)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
期刊 Papeta, N., Chen, T., Vianello, F., Gererty, L., Malik, A., Mok, Y. T., ... & Poznansky, M. C. Long-term survival of transplanted allogeneic cells engineered to express a T cell chemorepellent. Transplantation 83(2) 2007 174-183

Also Published As

Publication number Publication date
TW201923070A (en) 2019-06-16

Similar Documents

Publication Publication Date Title
US11186827B1 (en) Genetically modified stem cells
JP2023510916A (en) Safety switch for control of gene expression
CN110678190A (en) Method for protecting transplanted tissue from rejection
EP1928504B1 (en) Liver-directed gene therapy
EP1401268A1 (en) Treatment or replacement therapy using transgenic stem cells delivered to the gut
TWI806907B (en) Treating diabetes with genetically modified beta cells
US20180066253A1 (en) Methods and compositions for modifying endothelial cells
US20240024371A1 (en) Treating diabetes
EP3938497A1 (en) Optimised rag1 deficient gene therapy
EP4132545A1 (en) Treating autoimmune diseases with genetically modified cells
EP4310188A1 (en) Gene coding for chimeric receptor for anti-acetylcholine receptor autoantibody
JP7308750B2 (en) Engineered cells to induce tolerance
KR101699567B1 (en) Composition for Preventing or Treating Immune Disease Comprising inhibitor
DAVIES ENGINEERING A SURROGATE (-CELL
Delivered Genetic Diseases II
MIYAZAKI et al. Establishment of a Pancreatic 6 Cell Line That Retains Glucose-Inducible Insulin Secretion: Special Reference to Expression of Glucose Transporter Isoforms
AU2002344233A1 (en) Treatment or replacement therapy using transgenic stem cells delivered to the gut
AU2008200852A1 (en) Treatment or replacement therapy using transgenic stem cells delivered to the gut