WO2016193836A1 - Procédé de préparation de cellules stromales mésenchymateuses pour une indication spécifique et composition correspondante - Google Patents

Procédé de préparation de cellules stromales mésenchymateuses pour une indication spécifique et composition correspondante Download PDF

Info

Publication number
WO2016193836A1
WO2016193836A1 PCT/IB2016/052636 IB2016052636W WO2016193836A1 WO 2016193836 A1 WO2016193836 A1 WO 2016193836A1 IB 2016052636 W IB2016052636 W IB 2016052636W WO 2016193836 A1 WO2016193836 A1 WO 2016193836A1
Authority
WO
WIPO (PCT)
Prior art keywords
mesenchymal stromal
pooled
cells
analysis
composition
Prior art date
Application number
PCT/IB2016/052636
Other languages
English (en)
Inventor
Udaykumar KOLKUNDKAR
Suresh Kannan
Sudha BALASUBRAMANIAN
Parvathy VENUGOPAL
Priyanka SWAMYNATHAN
Anish Sen Majumdar
Original Assignee
Stempeutics Research Private Limited
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 Stempeutics Research Private Limited filed Critical Stempeutics Research Private Limited
Publication of WO2016193836A1 publication Critical patent/WO2016193836A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0605Cells from extra-embryonic tissues, e.g. placenta, amnion, yolk sac, Wharton's jelly
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0668Mesenchymal stem cells from other natural sources

Definitions

  • the present disclosure relates to stem cell technology.
  • a method of preparing stem cell based compositions is provided.
  • the said compositions comprise pooled cells which are arrived by selective pooling of individual cells from characterized Donor cell banks (DCB).
  • DCB Donor cell banks
  • Specific/selective pooling of DCBs is based on the analysis of characters such as but not limiting to growth kinetics, gender, immunosuppressive studies, gene profiling and secretome profiling.
  • Such composition is used to target specific indication or disease condition.
  • MSCs Mesenchymal stem cells
  • BM bone marrow
  • BM-MSCs Mesenchymal stem cells
  • the origin of MSCs may determine their fate and functional characteristics. Being from a fetal-derived cord, that is discarded after birth, wharton's jelly (WJ)-derived MSCs have higher proliferation and ex vivo expansion capabilities than adult MSCs. Although, MSCs from different origins share important biological properties, till date it is not clear which type or source of MSC should be selected for the different therapeutic approaches.
  • the umbilical cord can provide an inexhaustible source of stem cells for therapy without involving any invasive procedures or ethical concerns.
  • WJ Wharton's jelly
  • the present disclosure relates to a method of preparing pooled mesenchymal stromal cell based composition, said method comprising acts of: a) culturing mesenchymal stromal cells of individual donors to a first predetermined passage to obtain individual donor cell banks (DCBs), b) characterizing each donor cell bank (DCB) at the first predetermined passage, c) culturing the mesenchymal stromal cells of DCBs up to a second predetermined passage and pooling the mesenchymal stromal cells from plurality of donor cell banks (DCBs) based on the characterization of step (b) to obtain working cell bank (WCB), and d) culturing the mesenchymal stromal cells of the working cell bank (WCB) up to a third predetermined passage to obtain the pooled mesenchymal stromal cell based composition; a mesenchymal stromal cell based composition comprising mesenchymal stromal cells and cryopreservation medium
  • Figure 1 shows a flow diagram for establishing and characterizing donor cell bank and further preparing indication specific working cell bank.
  • Figure 2 shows the morphology of individual and pooled cells (N, R, S and NRS; O, P, Q and OPQ and Q, R, S and QRS) at Passage 5. Magnification 10X.
  • Figure 3 shows the proliferation rate/ growth kinetics and CFU-F assay of passage 2 to passage 5 cells.
  • Fig 3(A) shows the proliferation rate of cells from individual donors and pooled cells (N, R, S and NRS; O, P, Q and OPQ and Q, R, S and QRS) from passage 2 to passage 5; their total cell number; cumulative population doubling and population doubling time.
  • Figure 3(B) shows characterization based on the CFU-F assay of pooled cells (NRS, OPQ and QRS) from passage 2 to passage 5 indicating that pooled and upscaled cells in Mesencult medium retained the clonal formation efficacy till passage 5.
  • Figure 4 shows the surface marker characterization of cells from individual donors (having phenotypes N, R, S;) versus pooled cells (NRS); OPQ;QRS) at passages 5. , indicating that pooling does not alter surface marker expression.
  • Figure 5 shows the trilineage differentiation potential of pooled cells of NRS, OPQ and QRS at passage -5 indicating that pooling does not affect trilineage differentiation.
  • Figure 6 shows Karyotype analysis of pooled cells by Giemsa -Banding (G-Banding) and Flourescent in situ hybridization (FISH) for X & Y specific probes.
  • Figure 6(A) shows the Karyotype analysis (G-Banding) of pooled cells (NRS; OPQ and QRS) at passage 6, indicating that at the end of passage 6, the cells from all the three pools (NRS, OPQ and QRS) exhibit normal karyotype.
  • Mosaic karyoview is observed for the QRS mixed pool in the ratio of (XY 75 : XX 25).
  • Figure 6(B) shows the FISH results of QRS mixed pool. Evaluation of percentage of two clones using X/Y centromeric probe by FISH technique on interphase nuclei for better delineation of the two clones. Hybridization shows the presence of one signal for X chromosome (Green) and one signal for Y chromosome (Red) in 75% [XY] and two signals for X (Green) in the remaining 25% of cells analyzed [XX]. The FISH results corroborate with the karyotype data of mixed pool QRS.
  • Figure 7 shows the immunosuppression of cells from individual donors N, R, S at passages P4 and P6.
  • Figure 8 shows the gene expression profiles of individual and pooled cells.
  • FIG. 8(A) shows the gene expression profile of individual (N, R, S) and pooled cells (NRS) at P6 for expression of Pro angiogenic cytokine, (CXCL5) anti- inflammatory gene (TGF b) and pro inflammatory markers (TNF -alpha & IL lb) and tumor suppressor marker P53 at passage 6.
  • CXCL5 Pro angiogenic cytokine,
  • TGF b anti- inflammatory gene
  • TNF -alpha & IL lb pro inflammatory markers
  • Figure 8 (B) shows the gene expression profile of individual and pooled cells (N, R, S and NRS) at passage 6.
  • Figure 8(E) shows a summary of gene expression profile of pooled cells (NRS, OPQ and QRS) [summary of figure 8B, 8C and 8D].
  • Figure 8(F) shows a comparison of gene expression profile at passage 6 for three pooled donors (NRS, OPQ and QRS).
  • Figure 8(G) shows the comparison of gene expression profile of individual and pooled cells (N, R, S and NRS; O, P, Q and OPQ and Q, R, S and QRS) at passage 6.
  • Figure 9 shows the secretome analysis of individual and pooled cells (N, R, S and NRS; O, P, Q and OPQ and Q, R, S and QRS) conditioned media collected at passage 6.
  • Figure 10 shows the STR analysis of individual and pooled cells (N, R, S and NRS; O, P, Q and OPQ and Q, R, S and QRS) at passage 6.
  • the present disclosure relates to a method of preparing pooled mesenchymal stromal cell based composition, said method comprising acts of:
  • DCBs donor cell banks
  • DCB donor cell bank
  • the mesenchymal stromal cells are Wharton jelly derived mesenchymal stromal cells.
  • the mesenchymal stromal cells are cultured in a xeno-free culture medium comprising Basal medium, 20% Proliferation Supplement, L- Glutamine, penicillin and streptomycin.
  • the first predetermined passage is passage 2.
  • the characterization in step (b) is carried out by performing analyses selected from a group comprising growth kinetics, gender analysis, gene expression, secretome analysis and immunosuppression analysis, or any combination thereof.
  • the pooling in step (c) is performed by combining the mesenchymal stromal cells of characterized donor cell banks (DCBs) showing similar properties.
  • DCBs characterized donor cell banks
  • the second predetermined passage is passage 4.
  • the pooled mesenchymal stromal cell of the composition obtained in step (d) is characterized by performing analyses selected from a group comprising gender analysis, karyotype analysis, growth kinetics, gene expression analysis, secretome analysis, immunosuppressive analysis, trilineage potential analysis, surface marker characterization, tumorigenicity, STR analysis and clonal expression, or any combination thereof.
  • the third predetermined passage is passage 6.
  • the pooled mesenchymal stromal cell based composition is specific to an indication selected from group comprising but not limiting to Rheumatoid arthritis, Diabetes mellitus, graft-versus-host disease, diabetic foot ulcer, acute myocardial infarction, liver cirrhosis, stroke, multiple sclerosis and critical limb ischemia, or any combination thereof.
  • the present disclosure relates to a mesenchymal stromal cell based composition comprising mesenchymal stromal cells and cryopreservation medium wherein said composition is prepared by the method comprising steps of:
  • DCBs donor cell banks
  • DCB donor cell bank
  • the cryopreservation medium is animal protein- free defined cryopreservation medium or a cryopreservation medium comprising ionic buffer, cryoprotectant, protein, energy substrate and anti-aging agent.
  • the composition is cryopreserved, and wherein said cryopreserved composition comprises mesenchymal stromal cells at a concentration ranging from about 10 million cells per ml to 25 million cells per ml of the medium.
  • the present disclosure relates to a method of managing a disease, said method comprising act of administering the composition comprising mesenchymal stromal cells and cryopreservation medium to a subject in need thereof.
  • the disease is selected from a group comprising Rheumatoid arthritis, Diabetes mellitus, graft-versus-host disease, diabetic foot ulcer, acute myocardial infarction, liver cirrhosis, stroke, multiple sclerosis and critical limb ischemia, or any combination thereof.
  • Mesenchymal Stem Cells are known for their use in treating various auto immune diseases. They have the characteristics of modulating immune response by secreting various cytokines and growth factors. But the properties of MSCs largely depend on the donor characteristics and the clinical capabilities of the MSCs vary across the donors. For example, MSCs from some donors might have higher proliferation rate, high immunosuppressive capacities than compared to other donors.
  • MSCs from different donors are pooled.
  • pooled MSCs show reduced variability with regards to properties such as proliferation rate, gene expression, immunosuppressive capacities etc., which has been shown by comparing individual samples vs. pooled samples.
  • WJ-derived MSCs share some properties unique to fetal derived MSCs like having faster proliferation and greater ex-vivo expansion capabilities than adult MSCs.
  • MSC transplantation requires harvesting a large number of cells. MSCs are present in very low percentage in their niches and a vast number of cells are required for their subsequent use in therapeutic applications. Hence, it is important to define optimal culture conditions which would maximize the yield of MSCs from WJ of umbilical cord within a reasonable time frame.
  • the terms "mesenchymal stromal cell", “mesenchymal stem cell” and “MSC” are employed interchangeably within the instant disclosure.
  • cell composition As used herein, the terms “cell composition”, “cell product”, “Investigational Medicinal product (IMP)”, “final MSC composition” are used interchangeably within the instant disclosure and means the cell product comprising pooled MSCs obtained by the method of the instant disclosure.
  • management refers to preventing a disease or disorder from occurring in a subject, decreasing the risk of death due to a disease or disorder, delaying the onset of a disease or disorder, inhibiting the progression of a disease or disorder, partial or complete cure of a disease or disorder and/or adverse effect attributable to the said disease or disorder, obtaining a desired pharmacologic and/or physiologic effect (the effect may be prophylactic in terms of completely or partially preventing a disorder or disease or condition, or a symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease or disorder and/or adverse effect attributable to the disease or disorder), relieving a disease or disorder (i.e.
  • the present disclosure also envisages managing/treating said disorder by administering therapeutically effective dosage of the composition/MSCs prepared by the method of the invention.
  • the present disclosure relates to a general method of establishing improved stem cell compositions targeting specific disease conditions wherein the method uses the concept of indication specific pooling of stem cells based on stem cell characteristics.
  • the said method involves establishing well characterized disease specific cell banks referred to as working cells banks (WCBs).
  • WBs working cells banks
  • the disease specific working cell bank is used in developing the disease specific therapeutic cell products/compositions.
  • 'indication specific composition', 'indication specific pooled mesenchymal stromal cell based composition', 'indication specific MSC composition' or 'indication specific MSCs/mesenchymal stromal cells' refers to composition/MSCs which has been prepared for management/treatment of a specific disease(s) or disorder(s) or medical condition(s)/indication(s).
  • said disease or disorder or condition/indication is selected from a group comprising but not limiting to Rheumatoid arthritis, Diabetes Mellitus (DM), graft-versus-host disease (GVHD), Diabetic Foot Ulcer (DFU), Critical limb Ischemia (CLI), Acute myocardial infarction (AMI), Liver cirrhosis (LC), stroke and Multiple sclerosis (MS).
  • DM Diabetes Mellitus
  • GVHD graft-versus-host disease
  • DFU Diabetic Foot Ulcer
  • CLI Critical limb Ischemia
  • AMI Acute myocardial infarction
  • LC Liver cirrhosis
  • MS Multiple sclerosis
  • the method involves establishing a well characterized Donor cell Bank (DCB) or Master cell bank (MCB) of each stem cell donor and thereafter indication specific Working Cell Bank (WCB) is established by pooling DCBs.
  • DCB Donor cell Bank
  • MBB Master cell bank
  • WCB Working Cell Bank
  • the WCB is optionally cryopreserved and later the WCBs are further used in developing indication specific cell product/composition for clinical/therapeutic application.
  • Such an indication specific cell product system ensures reduced variability in the products manufactured from the single established WCB.
  • the steps involved in obtaining and characterizing DCBs and subsequent establishment of WCB is important which determines the quality and specificity of the final composition/product towards indications/diseases.
  • the method involves preparing MSC based compositions, wherein said MSCs are derived from source selected from a group comprising Wharton's jelly, bone marrow, adipose tissue and dental pulp, or any combination thereof.
  • the MSCs are derived from Wharton's jelly (WJ).
  • the selection of DCBs for pooling and preparing the WCB passes through specific stages and this involves extensive characterization of the DCB and identification of DCBs with similar characteristics. Depending on the similarity of the selected characteristics, DCBs are pooled to generate WCB.
  • One of the important steps involved in the establishment of indication-specific WCBs and subsequent final composition is the initial characterization of DCB at specific time points of culturing. The characterization results are the main criteria for selection of DCBs for pooling.
  • the indication specific WCBs which are established after pooling the characterized DCBs are further expanded/passaged till specific time-point.
  • the cell products/compositions prepared at such time-point of expansion will be indication specific and are potent for the said indication/disease.
  • the pooled MSCs forming the final composition have high expression levels of bioactive factors such as cytokines and growth factors which are beneficial for the relevant indications thus making the compositions more efficacious for a specific indication/disease condition.
  • the method of preparing WJ-MSC based compositions broadly comprises acts of:
  • the MSCs are derived from human donors selected from male, female, or a combination thereof.
  • the number of donors is at least two.
  • the number of donors can be three, four, five, six, seven, eight, nine or ten, and so on, as suitable.
  • the MSCs of final composition (at P6) is further analysed/characterized for clinical potency.
  • the MSCs of DCB and the final composition are analyzed/ characterized for characteristics selected from a group comprising proliferation rate/ growth kinetics (population doubling and population doubling time), gender (male, female or mixed), immunomodulatory analysis (immunogenicity and immunosuppression analysis), gene expression (expression related to chondrogenesis, immunomodulation /angiogenesis etc.), secretome analysis (secretion of cytokines and growth factors), tumorigenicity assay, trilineage potential, surface marker characterization, STR analysis, clonal expression and karyotype analysis, or any combination of characteristics thereof.
  • the MSCs of DCB is characterized at passage 2 for characteristics- growth kinetics, gender, gene expression, secretome analysis and immunosuppression.
  • the MSCs of final composition is characterized for characteristics- growth kinetics, gene expression, secretome analysis, immunosuppression, trilineage potential, surface marker characteristics, clonal expansion, karyotype analysis and STR analysis, or any combination thereof.
  • the WCB comprises 'pooled' WJ-MSCs wherein the WCB pool is prepared based on the analysis/characterization of each DCB as described above. Further, said WCB is selected from a female pool (i.e.
  • the pooled WJ-MSCs are from female DCBs), male pool (i.e. the pooled WJ-MSCs are from male DCBs) or mixed pool (i.e. the pooled WJ-MSCs are a combination male and female DCBs).
  • the present disclosure further provides for stem cell based pooled compositions targeting specific indications/diseases. More particularly, the disclosure provides potent indication specific pooled WJ-MSC compositions.
  • the compositions are prepared by the present method for indication/disease selected from a group including but not limited to Rheumatoid arthritis, Diabetes Mellitus (DM), graft-versus-host disease (GVHD), Diabetic Foot Ulcer (DFU), Critical limb Ischemia (CLI), Acute myocardial infarction (AMI), Liver cirrhosis (LC), stroke and Multiple sclerosis (MS) or any combination thereof.
  • preparation of Rheumatoid arthritis Working Cell Bank (WCB-RA), Diabetes Mellitus (WCB-DM) and Critical limb Ischemia Working Cell Bank (WCB-CLI) have been disclosed in the present disclosure.
  • the final composition is in the form of a cell suspension.
  • the pooled cell population of WCB is cryopreserved in known commercial or in-house developed cryopreservation solution.
  • the in-house cryo-formulation comprises of ionic buffer, cryoprotectant, protein, energy substrate and anti-aging agent.
  • the ionic buffer is selected from a group comprising PlasmaLyte A, ringer lactate solution, sucrose, and combinations thereof;
  • the cryoprotectant is selected from a group comprising dimethylsulfoxide (DMSO), glycerol, triglycerol, poly-L-lysine, and combinations thereof;
  • the protein is selected from a group comprising human serum albumin, recombinant plant derived xenofree human serum albumin, and a combination thereof;
  • the energy substrate is selected from a group comprising trehalose, hydroethyl starch, and a combination thereof;
  • the anti-aging agent is selected from a group comprising L-Glutamine, poly-L-lysine, ectoine, and combinations thereof.
  • the pooled cell population of WCB is cryopreserved in commercial available cryopreservation/cryo-formulation solution selected from but not limited to animal protein-free, defined cryopreservation medium CryoStorTM CS2, CryoStorTM CS5, CryoStorTM CS10, from Biolife solution and other known cryopreservation solutions used in the art can be used without limitation.
  • cryopreservation/cryo-formulation solution selected from but not limited to animal protein-free, defined cryopreservation medium CryoStorTM CS2, CryoStorTM CS5, CryoStorTM CS10, from Biolife solution and other known cryopreservation solutions used in the art can be used without limitation.
  • the P6 expanded indication - specific pooled cell population of 10 million to 25 million is cryopreserved in 1ml of the either in- house or commercially available cryopreservation solution.
  • isolating or obtaining mesenchymal stem cells from a donor does not involve operation or surgery or any invasive means/methods performed by doctors or medical practitioners.
  • a person having average skill in the field of stem cell technology can obtain or isolate mesenchymal stem cells from discarded umbilical cords using means which does not involve or require invasive steps (such as surgery) or the intervention of a doctor or a medical practitioner.
  • wharton's jelly derived mesenchymal stem cells WJ-MSCs are isolated from discarded umbilical cords (obtained after child birth) by the inventors without performing any step of surgery or any means practiced by doctors or medical practitioners.
  • mesenchymal stem cells can be obtained from sources including but non-limiting to stem cell banks- a repository of stem cells, as gifts from collaborating laboratories or a scientific person in this field of technology, and so on, but does not include any surgical/invasive step for obtaining said stem cells. Further, all the aforementioned approaches or sources of isolating/obtaining stem cells are within the scope of the present disclosure.
  • the WJ-MSC is obtained from a cell bank.
  • the term "Bank" in the present disclosure means source for obtaining the WJ-MSC. In an embodiment, such source is pre-processed and/or cryopreserved WJ-MSC or umbilical cords, stored for instant or future use.
  • the source is a Mesenchymal Stem Cell Bank.
  • any depository that stores umbilical cord or WJ-MSC for a pre-determined period of time is a bank.
  • any medium such as apparatus or device or vessel or container that stores WJ-MSC for a pre-determined period of time is considered to be a bank.
  • information about the bank is procured from Cord blood registry.
  • the WJ MSCs are isolated from the umbilical cords by enzymatic method.
  • the WJ MSCs are isolated from the umbilical cords by explant culture method.
  • the isolation by the enzymatic method is carried out after dissection and removal of the umbilical cord arteries, vein and amniotic epithelium.
  • the cords are cut into 2-3 cm segments after disinfecting the cords.
  • MSCs isolation from WJ cord is achieved by enzymatic dissociation with collagenase- 1 and hyaluronidase (Sigma-Aldrich, St. Louis, MO, USA) at about 37°C for about 16 hours followed by trypsin digestion at about 37°C for about lOmins for generation of WJ cell pellets.
  • Suspended cell pellets are thereafter seeded onto tissue cultured pre-coated flask with MesenCultTM-XF Attachment Substrate, in MesenCultTM-XF.
  • Complete medium consisting of MesenCultTM- XF Basal medium supplemented with about 20% MesenCultTM-XF Proliferation Supplement (all from STEMCELL Technologies TM Inc., Vancouver BC, Canada), L-Glutamine to a final concentration of about 2mM, about 100 U/ml penicillin and about 100 ⁇ g/ml streptomycin.
  • Cultures of WJ tissue suspended cell pellets are maintained at about 37 °C with about 5% C0 2 and about 21% 0 2 in a humidified atmosphere. For optimal cell growth, medium is changed at about 50% confluency and harvested at about 80% confluence.
  • Fresh and healthy human umbilical cords are obtained with informed consent using the guidelines approved by the Institutional Committee for Stem Cell Research and Therapy (ICSCRT) and Institutional Ethics Committee (IEC), at the Manipal Hospital, Bangalore, India.
  • Six umbilical cords named as N, O, P, Q, R, S are used for the present process. Further, this process can use any number of umbilical cords without limitation.
  • the WJ-MSCs are isolated from each of these cords by non-surgical/non-invasive method as per the below protocol to establish 6 Donor Cell Banks (DCB). Thereafter, each individual DCB is characterized for population doubling time, gender, immunogenicity, immunosuppressive properties, gene expression and cytokines secretion (secretome analysis).
  • the Wharton's Jelly derived MSC from umbilical cords are isolated by explant culture method and/ or enzymatic digestion method (i.e. non-surgical methods).
  • WJ-MSCs can also be obtained non-surgically) from Cell Banks or Depositories that store WJ-MSCs for predetermined period of time.
  • explant culture method is employed for isolation of WJ MSCs from umbilical cord.
  • Explants culture is based on the mesenchymal migratory capabilities and is also referred to as "plate and wait” method wherein the cord segments are simply plated in the culture medium and the MSCs are obtained by waiting for them to migrate out.
  • the explant culture method is preferred as it is simple, reliable and less laborious, and yielding pure and viable cultures of human umbilical cord derived WJ-MSCs.
  • the individual cords measuring about 5-10 cms are used.
  • the cords collected are subjected to multiple wash using normal saline (about 0.9% w/v Sodium Chloride) to clean the cord and remove all the blood clots.
  • the cords are then cut into multiple bits and placed in normal saline with anti-anti and kept in the refrigerator for about half an hour.
  • the cords are given a quick rinse with about 70% ethanol for not more than about 30 seconds and followed by rinsing with normal saline couple of times.
  • the cords are then dissected longitudinally and the inner part of the cord is exposed. After removing the veins and arteries manually, the cords are cut into small pieces of size of about 0.4 to 0.7 cms. Approximately 40 to 50 such pieces are placed in 100 mm tissue culture dish containing serum free culture media. The explants are left undisturbed in 5% C0 2 incubator at about 37°C for about 3-4 days, after which fresh culture medium is added to the culture dishes. By this time, the MSCs start to migrate out and grow in the dishes. The MSCs isolated at passage 0 (P0) are further passaged to P2 to eliminate any other tissue and to obtain pure cell population. The cells at P2 are cryopreserved and stored. The WJ-MSCs from each individual cord are cryopreserved at P2 and are referred to as Donor Cell Banks (DCBs).
  • DCBs Donor Cell Banks
  • the isolation of the WJ-MSCs used in the present disclosure does not involve invasive or surgical procedures as the explant culture method and the enzymatic digestion method do not require the intervention/performance of operation or surgery by a doctor or a medical practitioner.
  • the WJ-MSCs used in the present method can also be sourced from depositories or cell banks.
  • DCBs After the establishment of DCBs at P2, each of the DCBs is extensively characterized based on their proliferation kinetics/ growth kinetics, gender, immunogenicity, immunosuppressive properties, gene expression and secretome analysis. Depending on the disease/indication which needs to be treated, the DCBs showing similar properties are selected to be pooled together for establishing indication specific WCB at P4. Depending on requirements, WCB is expanded up to passage 6 to prepare final product- IMP.
  • the individual DCBs which have higher expression of anti-inflammatory gene (IL-10, TGFP) and low expression of pro-inflammatory genes (IL- ⁇ , TNFa ) as assayed at gene level and on secretomes level, are chosen along with population doubling and similar gender to be pooled.
  • IL-10 anti-inflammatory gene
  • TGFP anti-inflammatory gene
  • IL- ⁇ pro-inflammatory gene
  • TNFa pro-inflammatory gene
  • WJ-MSCs from the prepared WCBs are expanded to P6 and are further characterized to analyse their proliferation kinetics/growth kinetics, trilineage potential, expression of MSC markers, immunogenicity, immunosuppressive properties, gene expression (especially anti and pro inflammatory markers), secretome analysis, and tumorigenicity assay.
  • the characterization of the cells at passage 6 is an optional step which is performed to check the potency of the cells for clinical use.
  • Figure 1 gives the illustration for the concept of pooling DCBs based on selected characteristics for preparing and establishing indication specific WCB for various indications and manufacturing indication specific cell product for clinical application. The illustration in figure 1 shows that the cells from 6 different cords are isolated to prepare DCB.
  • the DCB cells are characterized at passage 2 for their population doubling (PD), gender, gene expression, secretome analysis etc. Based on the PD and gender, the first level screening for selection is performed. This is followed by gene expression analysis to select DCB for pooling to prepare an indication specific WCB at P4. Thereafter, the pooled WCB cells are expanded up to passage P6 (based on requirement) to prepare F P and P6 cells are extensively characterized to establish the potency for clinical application.
  • the population doubling time, number of population doublings in a passage and the cumulative population doubling over the passages are calculated. Thereafter, at least 2 DCBs having similar values for the above said parameters are pooled together.
  • the 6 cords are cultured in serum free and xeno free culture media (MesenCultTM-XF Basal medium, 20% MesenCultTM-XF Proliferation Supplement, L-Glutamine, penicillin and streptomycin).
  • Figure 3A shows the proliferation rate of pooled and individual samples.
  • Figure 3A and table 1 shows the growth/ proliferation kinetics of the WJ-MSC from each DCB.
  • Their mean population doubling time (PDT) and cumulative population doubling (CPD) is 28.8 ⁇ 4.09 and 18.07 ⁇ 1.21 respectively.
  • Table 1 Population kinetics of WJ-MSCs from each DCB
  • the gender details of the baby are collected during the birth.
  • the cords of the same gender are pooled and expanded.
  • Table 2 gives the details of the gender of the 6 DCB.
  • RNA isolated from MSC of each individual DCBs are analyzed for their gene expression related to pro and anti inflammatory properties/angiogenesis / disease specificity, using Real time PCR.
  • Figures 8a to 8g show the gene expression profile of individual and pooled samples.
  • the conditioned media collected from MSCs of each DCB samples are analyzed for the presence of cytokines and growth factors by ELISA method.
  • DCB showing similar secretomes are selected for pooling. For example, to treat CLI, the individuals that show high level of VEGF secretion are pooled together.
  • Figure 9 shows the secretome analysis of individual and pooled samples.
  • MLR Mixed Lymphocyte Reaction
  • PBMC peripheral blood mononuclear cells
  • Responder PBMC are then added to each well, at a ratio of about 1 :2.5 (MSCs: Responder) and the cultures are maintained for about 5 days, at which point, they are pulsed with 5-bromo-2-deoxyuridine (BrdU) for the final 24 h.
  • Cell proliferation is measured using a fluorimetric immunoassay kit (Calbiochem) for the quantification of BrdU incorporation, according to the manufacturer's instructions.
  • MLR mixed lymphocyte reactions
  • responder ratio are used as positive controls for PBMC proliferation in response to alloantigen.
  • Mitomycin C-treated MSCs are seeded in 96-well plates and allowed to attach overnight.
  • a one-way MLR at a ratio of about 1 :2.5 is then added to each well. Lymphocyte proliferation is measured at the end of 5 days, as described above. A oneway MLR cultured in the absence of MSCs is considered as the 100% proliferation control.
  • Four individual donors are analyzed in each group. All treatments are performed in triplicate wells in RPMI 1640 medium (Invitrogen) supplemented with about 10% FBS (Hyclone), about 2 mM glutamine (Invitrogen) and about 0.05 mM ⁇ -mercaptoethanol (Sigma-Aldrich).
  • WJ-MSCs exhibit hypo immunogenicity and arrest the growth of T cell proliferation in dose dependent manner when co cultured with peripheral blood mononuclear cells (PBMCs). Also WJ-MSCs suppress mixed lymphocyte reaction (MLR) where PBMCs from mismatched donors are co cultured with the MSCs.
  • MLR mixed lymphocyte reaction
  • Table 3 gives the list of gene expression and secretome analysis which is considered for each indication/ disease condition to establish the pooled WCB.
  • Table 3 List of gene expression and secretome analysis considered for various indications/ disease conditions
  • the DCBs at passage 2 are selected and segregated for pooling.
  • the selected DCBs are further expanded till passage 4 (P).
  • the WJ-MSCs from selected DCBs are then pooled in equal proportion to prepare indication specific working cell banks at passage 4.
  • the three WCB pools prepared based on the characterization of the MSC from DCB are RS, OPQ and QRS.
  • QRS is a mixed pool, where the individual DCB from male and female donors having low and high population doublings are pooled. Apart from population doubling time, other characteristics as described in Example 2 are also considered for pooling.
  • pooled cells are plated and expanded till P6 to prepare final product.
  • pooled samples are assayed for certain selected characteristics as mentioned in the DCB characterization at P2 along with a few additional assays.
  • MSC of final composition is further analysed/characterized for clinical potency. The said characterization of final product at P6 is described below: (i) Growth kinetics of pooled MSCs
  • the average Cumulative Population Doubling (CPD) of all the three pools is 16.05 ⁇ 0.384.
  • the average PD and PDT at passage 6 is 4.22 ⁇ 0.496622 and 34.43667 ⁇ 3.79961 respectively.
  • Average total cell number at the end of Passage 5 for the pooled and upscaled cells is 1.81X10 11 ⁇ 0.49X10 11 .
  • Figure 3(a) shows the proliferation rate of individual and pooled samples.
  • Tables 4a and 4b gives the result of growth kinetics of the pooled samples.
  • RNA extracted from individual and pooled WJ-MSCs are reverse transcribed and the gene expression is analyzed using Real time PCR.
  • Figure 8a to 8g shows the gene expression profiles of individual and pooled samples.
  • Figure 8a shows that the expression pattern of pooled MSCs averages out the individual values, but in some the pooling is advantageous - as like for IL-lb, the pooled samples have lesser expression of this pro inflammatory gene.
  • figures 8(e) and 8(f) summarizes the gene expression profile of pooled samples (NRS, OPQ and QRS) wherein it is shown that pooling overall brings down the expression of all genes in female (NRS) and male (OPQ) pools whereas the mixed pool (QRS) have mixed results and mostly averages out the individuals. From figure 8(g), it is seen that on comparison with the individual donor cells, pooled samples have normalized expression profiles, and are thus better in clinical applications vis-a-vis individual donor cells.
  • FIG. 9 shows the secretome analysis of individual and pooled samples. It is found that pooling increases the HGF secretion and in case of TGF- ⁇ , the pooled sample averages out the individual values. On a whole, it is observed from the comparison of the individual versus pooled cells that the characteristics of the pooled samples are normalized and therefore can be accordingly employed for further applications.
  • the immunosuppressive properties of WJ-MSCs are maintained even after pooling and even outweigh the individual immunosuppressive capacities. For example, at passage 6 and at about 1 :2.5 ratio of MSCs to responder (i.e. PBMCs isolated from another donor), the pooled MSCs show higher immunosuppression than the individuals. Thus, pooling affects the immunosuppression and the WCBs and final cell product prepared are highly immunosuppressive.
  • Figure 7 shows the immunosuppressive properties of individual samples at P4 and P6.
  • Figure 9 shows the secretome analysis of individual and pooled cells (N, R, S and NRS; O, P, Q and OPQ and Q, R, S and QRS) condition media collected at passage 6, with respect to a 'control' sample. This shows that the pooling of the MSCs normalizes the expression of cytokines- HGF and TGF- ⁇ at P6.
  • the pooled WJ-MSCs form adipocytes, osteocytes and chondrocytes as similar to individual MSCs and thus their differentiation capacity is not affected by the pooling as shown in the figure 5.
  • the pooled MSCs are positive for CD 90, 73 and 105 but negative for hematopoietic marker and class II MHCs.
  • Figure 4 shows surface marker characterization of pooled cells.
  • Table 5 provides the average of expression of various markers in all three pools.
  • the cells are plated at a density of 100 cells per 35mm dish. Complete media change is carried out every third day. The cells are incubated for about 8 days, at which point they are fixed and stained with 0.1% Toluidene Blue in 1% Paraformaldehyde (all reagents from Sigma-Aldrich), to visualize the colonies. Stained colonies are manually counted. The assay for each sample is carried out in triplicates.
  • STR analysis Short tandem repeat (STR) analysis is a molecular biology method used to compare specific loci on DNA from different donors.
  • a short tandem repeat is a microsatellite, consisting of a unit of two to thirteen nucleotides repeated hundreds of times in a row on the DNA strand.
  • STR analysis measures the exact number of repeating units. This method differs from restriction fragment length polymorphism analysis (RFLP) since STR analysis does not cut the DNA with restriction enzymes. Instead, probes are attached to desired regions on the DNA, and a polymerase chain reaction (PCR) is employed to discover the lengths of the short tandem repeats. Number of repeats for particular allele is unique for each donor.
  • RFLP restriction fragment length polymorphism analysis
  • PCR polymerase chain reaction
  • the method of the present disclosure aims at establishing improved stem cell compositions targeting specific disease conditions wherein the method uses the concept of pooling of stem cells from individual donors based on stem cell characteristics. Cryopreservation of the composition:
  • the pooled cell population of WCB is cryopreserved in known commercial or in-house developed cryopreservation solution.
  • the in-house cryo-formulation comprises of ionic buffer, cryoprotectant, protein, energy substrate and anti-aging agent.
  • the ionic buffer is selected from a group comprising PlasmaLyte A, ringer lactate solution, sucrose, and combinations thereof;
  • the cryoprotectant is selected from a group comprising dimethylsulfoxide (DMSO), glycerol, triglycerol, poly-L-lysine, and combinations thereof;
  • the protein is selected from a group comprising human serum albumin, recombinant plant derived xenofree human serum albumin, and a combination thereof;
  • the energy substrate is selected from a group comprising trehalose, hydroethyl starch, and a combination thereof;
  • the anti-aging agent is selected from a group comprising L-Glutamine, poly-L-lysine,
  • the pooled cell population of WCB can be cryopreserved in commercial available cryopreservation/cryo-formulation solution selected from but not limited to animal protein-free, defined cryopreservation medium CryoStorTM CS2, CryoStorTM CS5, CryoStorTM CS10, from Biolife solution and other known cryopreservation solutions used in the art can be used without limitation.
  • the P6 expanded indication- specific pooled cell population of 10 million to 25 million is cryopreserved in 1ml of the either in-house or commercially available cryopreservation solution.
  • the culture conditions for culturing of the MSCs are optimized which maximizes the yield of MSCs from WJ of umbilical cord within a reasonable time frame.
  • the method for culturing MSCs are described in the present disclosure involves establishing well characterized indication specific cell banks referred to as working cells banks (WCBs).
  • the indication specific working cell bank is used in developing the indication specific therapeutic cell products/compositions.
  • the pooled MSCs forming the final composition have high expression levels of bioactive factors such as cytokines and growth factors which are beneficial for the relevant indications thus making the compositions more efficacious for a specific disease condition.
  • pooled MSCs show reduced variability with regards to properties such as proliferation rate, gene expression, immunosuppressive capacities etc., which are substantiated in the above examples by comparing individual samples vs. pooled samples.
  • the present disclosure provides potent indication specific pooled WJ-MSC compositions, wherein the compositions are prepared by the present method for indication/disease selected from a group including but not limited to Rheumatoid arthritis, Diabetes Mellitus (DM), graft-versus-host disease (GVHD), Diabetic Foot Ulcer (DFU), Critical limb Ischemia (CLI), Acute myocardial infarction (AMI), Liver cirrhosis (LC), stroke and Multiple sclerosis (MS) or any combination thereof.
  • indication/disease selected from a group including but not limited to Rheumatoid arthritis, Diabetes Mellitus (DM), graft-versus-host disease (GVHD), Diabetic Foot Ulcer (DFU), Critical limb Ischemia (CLI), Acute myocardial infarction (AMI), Liver cirrhosis (LC), stroke and Multiple sclerosis (MS) or any combination thereof.
  • the MSC- based composition of the present disclosure is obtained by pooling of the cells, wherein cells from DCBs are pooled based on the results of characterization of the DCBs (at passage 2).
  • This method of pooling is advantageous over random pooling (i.e., which is not based on any characterization studies), since pooling minimizes variability among the cells of the DCBs, with regards to properties such as proliferation rate, gene expression, cytokine secretion, immunosuppressive capacities etc., averages out individual values and normalizes the expression profiles of the pooled cells which thus make these cells better for clinical applications.

Abstract

La présente invention concerne un procédé de préparation de compositions à base de cellules stromales. Lesdites compositions comprennent des cellules regroupées qui proviennent d'un regroupement de cellules issues de donneurs individuels. Ledit regroupement des cellules est basé sur l'analyse des caractéristiques telles que mais non limitées à la cinétique de croissance, au sexe, à l'immunosuppression, au profil génétique et au profil du sécrétome. Ainsi, le présent procédé peut être utilisé pour préparer des cellules stromales regroupées pour des indications/applications cliniques spécifiques en fonction des besoins.
PCT/IB2016/052636 2015-06-01 2016-05-09 Procédé de préparation de cellules stromales mésenchymateuses pour une indication spécifique et composition correspondante WO2016193836A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN2757CH2015 2015-06-01
IN2757/CHE/2015 2015-06-01

Publications (1)

Publication Number Publication Date
WO2016193836A1 true WO2016193836A1 (fr) 2016-12-08

Family

ID=56101760

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2016/052636 WO2016193836A1 (fr) 2015-06-01 2016-05-09 Procédé de préparation de cellules stromales mésenchymateuses pour une indication spécifique et composition correspondante

Country Status (1)

Country Link
WO (1) WO2016193836A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021028583A1 (fr) 2019-08-15 2021-02-18 Nextcell Pharma Ab Composition allogénique pour le traitement de troubles du système nerveux central

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011064733A1 (fr) * 2009-11-27 2011-06-03 Stempeutics Research Pvt. Ltd. Procédés de préparation de cellules souches mésenchymateuses, compositions et nécessaires associés
WO2012131618A1 (fr) * 2011-03-30 2012-10-04 Stempeutics Research Private Limited Composition comprenant des cellules souches mésenchymateuses dérivées de la gelée de wharton et procédés associés

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011064733A1 (fr) * 2009-11-27 2011-06-03 Stempeutics Research Pvt. Ltd. Procédés de préparation de cellules souches mésenchymateuses, compositions et nécessaires associés
WO2012131618A1 (fr) * 2011-03-30 2012-10-04 Stempeutics Research Private Limited Composition comprenant des cellules souches mésenchymateuses dérivées de la gelée de wharton et procédés associés

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
GOTTIPAMULA SANJAY ET AL: "Isolation, expansion and characterization of bone marrow-derived mesenchymal stromal cells in serum-free conditions", CELL AND TISSUE RESEARCH, SPRINGER, DE, vol. 356, no. 1, 22 January 2014 (2014-01-22), pages 123 - 135, XP035331249, ISSN: 0302-766X, [retrieved on 20140122], DOI: 10.1007/S00441-013-1783-7 *
O RINGDÉN ET AL: "Pooled MSCs for treatment of severe hemorrhage", BONE MARROW TRANSPLANTATION, vol. 46, no. 8, 1 August 2011 (2011-08-01), pages 1158 - 1160, XP055140414, ISSN: 0268-3369, DOI: 10.1038/bmt.2010.262 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021028583A1 (fr) 2019-08-15 2021-02-18 Nextcell Pharma Ab Composition allogénique pour le traitement de troubles du système nerveux central
WO2022034220A1 (fr) 2019-08-15 2022-02-17 Nextcell Pharma Ab Composition allogénique pour le traitement de troubles de la covid-19

Similar Documents

Publication Publication Date Title
Leri et al. Cardiac stem cells and mechanisms of myocardial regeneration
JP4336821B2 (ja) 哺乳動物の骨髄細胞または臍帯血由来細胞と脂肪組織を利用した心筋細胞の誘導
Deasy et al. Long-term self-renewal of postnatal muscle-derived stem cells
JP5469457B2 (ja) 胎盤幹細胞を使用する腫瘍抑制
KR101378874B1 (ko) 태반 줄기세포를 이용한 면역 조절
US10526581B2 (en) Modulation of cardiac stem-progenitor cell differentiation, assays and uses thereof
Kreke et al. Cardiospheres and cardiosphere-derived cells as therapeutic agents following myocardial infarction
WO2009027563A1 (fr) Population de cellules souches adultes issues du tissu adipeux cardiaque et son utilisation dans la régénération cardiaque
MX2011005229A (es) Celulas adherentes derivadas del amnios.
JP2013510582A (ja) 間葉幹細胞の球状集合体
KR102155623B1 (ko) 성체 심장 줄기세포 군집
CN110713984B (zh) 诱导人间充质干细胞生成功能内皮细胞的方法
JP2009531021A (ja) 骨格筋周囲血管芽細胞および心筋中胚葉性血管芽細胞、それらの単離および使用方法
US20210230537A1 (en) Angiogenesis using stimulated placental stem cells
Ennis et al. Isolation, characterization, and differentiation of human umbilical cord perivascular cells (HUCPVCs)
KR20080042761A (ko) 렙틴을 이용한 줄기세포 증식방법
CN109646458B (zh) 使用胎盘间充质干细胞制剂治疗硬化病的方法
JP6147419B2 (ja) 心臓内膜由来の成体幹細胞及びこれの製造方法
KR101834800B1 (ko) 심장 조직-유래 세포
WO2016193836A1 (fr) Procédé de préparation de cellules stromales mésenchymateuses pour une indication spécifique et composition correspondante
Ludke et al. Uterine-derived progenitor cells are immunoprivileged and effectively improve cardiac regeneration when used for cell therapy
CN102119936B (zh) 制备利用人羊膜间充质细胞治疗缺血性脑损伤注射液的方法及其注射液
US20200190475A1 (en) Methods for identifying and isolating cardiac stem cells and methods for making and using them
Ellison et al. Adult cardiac stem cells: identity, location and potential
Suyoko et al. Simple Procedure for the Isolation of Mesenchymal Stem Cells from Different Parts of the Human Umbilical Cord

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16727225

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16727225

Country of ref document: EP

Kind code of ref document: A1