The largest database of trusted experimental protocols

Clinimacs system

Manufactured by Miltenyi Biotec
Sourced in Germany

The CliniMACS system is a laboratory device designed for cell processing and isolation. It utilizes magnetic cell separation technology to selectively isolate specific cell populations from complex biological samples. The core function of the CliniMACS system is to enable precise and efficient cell separation for research and clinical applications.

Automatically generated - may contain errors

21 protocols using clinimacs system

1

Isolation and Activation of Primary T Cells

Check if the same lab product or an alternative is used in the 5 most similar protocols
Leukapheresis products were obtained from health donors (Bloodworks Northwest). Primary CD4+ and CD8+ T cells were isolated serially using the CliniMACS System (Miltenyi) and cryopreserved for later use in activation studies. Cells were cultured in RPMI 1640 (Corning) supplemented with 10% fetal bovine serum (FBS) (ThermoFisher), recombinant human IL-2 (50 U/mL), and recombinant human IL-15 (0.5 U/mL) (Miltenyi). Cells were maintained in a 37 °C and 5% CO2 humidified incubator.
+ Open protocol
+ Expand
2

Isolation and Characterization of Immune Cells

Check if the same lab product or an alternative is used in the 5 most similar protocols
Heparinized peripheral blood was obtained from the three patients, their mother, and healthy donors at a similar age as the patients. Peripheral blood mononuclear cells (PBMCs) were isolated from peripheral blood by density gradient centrifugation using Biocoll® Trennlösung (BIO&SELL, Nuremberg, Germany) according to the manufacturer’s instructions. CD3+ T cells were collected from PBMCs with the magnetic-activated cell sorting system (CliniMACS System, Miltenyi Biotec, Bergisch Gladbach, Germany) based on the manufacturer’s instructions of MACS CD3 Microbeads and LS separation columns. The purity of gathered CD3+ T cells determined by flow cytometry (BD FACS Calibur, BD Biosciences, Franklin Lakes, NJ, USA) was >90% for all experiments (data not shown). Various subsets of CD3+, CD4+, CD8+, CD14+, CD19+, CD56+, αβ, γδ, and NKT (CD3+/CD56+) cells were sorted from PBMCs using the cell sorter (MACS Quant® Tyto®, Miltenyi Biotec, Bergisch Gladbach, Germany) with corresponding anti-human antibodies (sorting purity of subpopulations > 95%). Genomic DNA (gDNA) was isolated from peripheral blood cells and the different sorted populations with NucleoSpin Tissue kit (Macherey Nagel, Düren, Germany) following the manufacturer’s instructions.
+ Open protocol
+ Expand
3

Allogeneic Stem Cell Transplant with CD34+ Selection

Check if the same lab product or an alternative is used in the 5 most similar protocols
T-cells were harvested from all donors after informed consent and shipped to Milano for transduction. Eleven patients received total body irradiation (TBI; 12 Gy) and cyclophosphamide (120 mg/kg) followed by a CD34-enriched stem cell graft from matched related donors (MRDs; Table 1). Donors received G-CSF (2 × 5 μg/kg daily) for 4–5 days and blood leukocytes were collected at the Institute for Transfusion Medicine (MHH; Borchers et al., 2011 (link)). CD34-selection was performed under GMP conditions using the CliniMACS-system (Miltenyi; Bergisch Gladbach, Germany) at the Center for Cellular Therapy (former Cytonet, Hannover, Germany). At least 3.9 × 106/kg CD34-positive cells were transplanted on day 0, and CD3+ T-cells were usually below 1 × 104 cells/kg body weight (Table 1). CD34-selection was the only GvHD prophylaxis. Missing informed consent, acute GvHD grade II or more, life threatening infections, or relapse at the time of transfusion were exclusion criteria for gene therapy. Relapse and declining donor chimerism were treated with additional, non-transduced DLI (see Table 2). One patient (UPN 1505) received a second transduced DLI to treat relapse.
+ Open protocol
+ Expand
4

Adoptive NK Cell Immunotherapy

Check if the same lab product or an alternative is used in the 5 most similar protocols
Patients with rising absolute neutrophil counts ≥ 300/μL and platelet counts ≥ 30,000/μL received the following conditioning regimen: Cyclophosphamide (60 mg/kg) was intravenously (IV) administered on Day − 7, and fludarabine (25 mg/m2 per day) was IV administered on Days − 6 through − 2. Patients received interleukin-2 (1 × 106 units/m2) subcutaneously on Days − 1, 1, 3, 5, 7, and 9. Donors underwent apheresis on Day − 1, and mononuclear cells were purified in a two-step procedure for CD56+/CD3 NK cells with the CliniMACS system (Miltenyi Biotec, Woburn, MA) as preciously described [8 (link)], allowing for a CD56/CD3+ cell dose of < 0.05 × 106 cells/kg. Purified, unmanipulated NK cells were infused on Day 0 at a desired dose of > 2 × 106 CD56+/CD3 cells/kg recipient body weight.
+ Open protocol
+ Expand
5

Allogeneic NK Cell Infusion Protocol

Check if the same lab product or an alternative is used in the 5 most similar protocols
Patients were enrolled on the St. Jude NKAML or NKHEM protocols (ClinicalTrials.gov NCT00697671 and NCT00187096) and received clofarabine 40 mg/m2, etoposide 100 mg/m2, and cyclophosphamide 400 mg/m2 on days-6 through -2 before NK cell infusion.[32 (link)] On alternate days, 1 million units/m2 of IL-2 was administered subcutaneously for 6 doses starting on day –1 to activate and expand circulating donor NK cells. The donors underwent apheresis on day-1 and the product was purified for CD3 CD56+ cells in our Human Applications Laboratory by a 2-step procedure using magnetic activated cell sorting.[33 (link)] First, the CliniMACS system (Miltenyi Biotec, Cambridge, MA) was used to deplete T cells from the mononuclear cell product obtained by leukapheresis by using anti-CD3 antibody-coated beads. Second, the CD3-depleted product was enriched for CD56+ cells by incubation with anti-CD56 antibody-coated beads. The entire cell-purification process lasted less than 12 hours and the final products were infused immediately without in vitro exposure to IL-2 or other cytokines.
+ Open protocol
+ Expand
6

Monocyte Adhesion to Endothelial Cells

Check if the same lab product or an alternative is used in the 5 most similar protocols
Leukapheresis products and discards were obtained from consented research participants (healthy donors) under protocols approved by the City of Hope Internal Review Board (IRB #09025). Peripheral blood mononuclear cells (PBMCs) were isolated by density gradient centrifugation over Ficoll-Paque (GE Healthcare) followed by multiple washes in PBS/ethylenediaminetetraacetic acid (EDTA) (Miltenyi Biotec). Cells were rested overnight at room temperature on a rotator, and subsequently washed and resuspended in complete X-VIVO. Up to 5 × 109 PBMC were incubated with anti-CD14 microbeads (Miltenyi Biotec) for 30 min at room temperature and magnetically enriched using the CliniMACS system (Miltenyi Biotec) according to the manufacturer’s protocol. The monocytes were labeled with CellTracker Green CMFDA Dye (Thermo Fisher Scientific) and incubated with monolayer HUVECs (4 × 103 cells per cm2) for 15–30 min in a cell culture incubator. The nonattached monocytes were then washed off with complete EC growth medium. The attached monocyte numbers were quantified on Cytation1 Cell Imaging Multi-Mode Reader (BioTek) using green fluorescent channel. Average numbers per condition were calculated from two randomly selected fields of technical duplicates.
+ Open protocol
+ Expand
7

Leukapheresis-Derived Monocyte Transduction

Check if the same lab product or an alternative is used in the 5 most similar protocols
After informed consent, leukapheresis was obtained from healthy adult volunteers (HLA-A*02:01) with a COBE Spectra apheresis system. CD14+ monocytes were enriched by immunomagnetic separation using a GMP-compliant CliniMACS system (Miltenyi Biotec). Quantitative and qualitative analyses of the selected CD14+ fraction and flow through were performed by flow cytometry. From the enriched CD14+ fraction, 2 × 108 cells were resuspended in 25 ml of serum-free CellGro DC medium (CellGenix) and seeded in a 100 ml bag (CellGenix). Cells were preconditioned with 25 ml of medium containing hGM-CSF and human IL-4 cytokines (50 ng ml−1 each, CellGenix) for 8 h. Cells were transduced with 1 × 109 infective particles (multiplicity of infection of 5) in 50 ml medium containing Protamine sulfate (5 μg ml−1). The bag was incubated at 37 °C and 5% CO2 for 16 h. Next day, cells were washed three times with CellGro medium. After washing, cell number and viability was determined. Transduced cells were cryopreserved in aliquots of 2 × 106 cell ml−1 per vial. Surplus, non-transduced monocytes were cryopreserved in aliquots of 2 × 106 cell ml−1 per vial and 50 × 106 cell ml−1 per vial and were used as controls for the characterization experiments. Sterility tests were performed with the ‘Bactec' system (BD Biosciences).
+ Open protocol
+ Expand
8

Monocyte-derived Dendritic Cell Protocol

Check if the same lab product or an alternative is used in the 5 most similar protocols
Leukapheresates were washed with PBS/EDTA supplemented with 2% HSA, incubated with CD14 MicroBeads (Miltenyi Biotec) for 15 minutes then washed. CD14+ cells were isolated using the CliniMACS System (Miltenyi Biotec). Positively selected cells were washed and plated in RPMI-1640 supplemented with 1% autologous plasma, GM-CSF (Genzyme) and IL-4 (R & D Systems) over 6 days. Cells were pulsed with tumor cells, matured and harvested in the same manner as Adherence DCs.
+ Open protocol
+ Expand
9

Generating CAR T Cells for B-ALL Treatment

Check if the same lab product or an alternative is used in the 5 most similar protocols
CD19-CAR T cells were generated using peripheral blood mononuclear cells (PBMC) obtained via apheresis from pediatric and young adult patients with relapsed/refractory B cell ALL, treated on our institutional CD19-CAR T cell clinical study (NCT03573700; Hines et al., 2021 (link)). The protocol was approved by the Food and Drug Administration and by the institutional review boards at St. Jude Children’s Research Hospital. CD19-CAR T cell products were generated within the St Jude Children’s Research Hospital GMP facility. Briefly, T cells are selected from the PBMC product using CD8 and CD4 electromagnetic bead reagents (CliniMACS system; Miltenyi Biotech). T cells are then activated using CD3/CD28 monoclonal antibodies, transduced with the clinical grade CD19-CAR lentiviral vector, and expanded using IL-7 and IL-15 for 7 – 10 days until desired dose is achieved. The final CAR T cell product the undergoes QA/QC testing prior to release for clinical use. Patients then receive lymphodepleting chemotherapy, followed by CD19-CAR T cell infusion at protocol defined dosing. Characteristics of the manufactured product and post-infusion expansion were performed as described by A. Talleur (unpublished data).
+ Open protocol
+ Expand
10

Expansion of Anti-CD123 CAR T Cells

Check if the same lab product or an alternative is used in the 5 most similar protocols
Healthy donor-derived human peripheral blood apheresis cells were purchased from Key Biologics. The cells were labeled with anti-CD4/CD8 microbeads, and CD4+/CD8+ T cells were purified by using the CliniMACS system (Miltenyi Biotec, Bergisch Gladbach, Germany). CD4+ and CD8+ T cells from peripheral blood mononuclear cells were suspended in X-VIVO 15 media with 5% human AB serum (Valley Biomedicals, Winchester, VA, UA) and 10 ng/mL of recombinant human IL-7 and IL-15 (Miltenyi Biotec) (X-VIVO 15–5% HSA/IL-7/IL-15, hereafter). Then, cells were activated with T Cell TransAct (Miltenyi Biotec) according to the manufacturer’s protocol in six-well plates (Thermo Fisher Scientific) at 37°C with 5% CO2 for 24 h. The activated T cells were washed and suspended in X-VIVO 15–5% HSA/IL-7/IL-15 followed by transduction with four different batches of anti-CD123 chimeric antigen receptor-expressing vectors at a MOI of 25 in a total volume of 1 mL. After 22 h of transduction, the cells were transferred to a G-Rex plate (Wilson Wolf, St. Paul, MN, USA) and incubated in 30 mL of X-VIVO 15–5% HSA/IL-7/IL-15. CD123-specific chimeric antigen receptor-expressing T cell expression, viability, and expansion were determined 5 days post transduction.
+ Open protocol
+ Expand

About PubCompare

Our mission is to provide scientists with the largest repository of trustworthy protocols and intelligent analytical tools, thereby offering them extensive information to design robust protocols aimed at minimizing the risk of failures.

We believe that the most crucial aspect is to grant scientists access to a wide range of reliable sources and new useful tools that surpass human capabilities.

However, we trust in allowing scientists to determine how to construct their own protocols based on this information, as they are the experts in their field.

Ready to get started?

Sign up for free.
Registration takes 20 seconds.
Available from any computer
No download required

Sign up now

Revolutionizing how scientists
search and build protocols!