1
|
Iqbal M, McLennan AL, Mukhamedshin A, Dinh MTP, Liu Q, Junco JJ, Mohan A, Srivaths PR, Rabin KR, Fogarty TP, Gifford SC, Shevkoplyas SS, Lam FW. Ultra-low extracorporeal volume microfluidic leukapheresis is safe and effective in a rat model. Nat Commun 2025; 16:1930. [PMID: 39994179 PMCID: PMC11850925 DOI: 10.1038/s41467-025-57003-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 01/29/2025] [Indexed: 02/26/2025] Open
Abstract
Leukapheresis is a potentially life-saving therapy for children with symptomatic hyperleukocytosis. However, the standard centrifugation-based approach exposes pediatric patients to significant complications due to its large extracorporeal volume, high flow rates, and considerable platelet loss. Here, we tested whether performing cell separation with a high-throughput microfluidic technology could alleviate these limitations. In vitro, our microfluidic devices removed ~85% of large leukocytes and ~90% of spiked leukemic blasts from undiluted human whole blood, while minimizing platelet losses. Multiplexed devices connected in parallel allowed for faster, clinically relevant flow rates in vitro with no difference in leukocyte collection efficiency. When connected to Sprague-Dawley rats, the devices removed large leukocytes with ~80% collection efficiency, reducing the leukocyte count in recirculating blood by nearly half after a 3-hour procedure. Evaluation of plasma biomarkers and end-organ histology revealed no adverse effects compared to sham control. Overall, our study suggests that microfluidics-based leukapheresis is safe and effective at selectively removing leukocytes from circulation, with separation performance sufficiently high to ultimately enable low extracorporeal volume leukapheresis in children.
Collapse
Affiliation(s)
- Mubasher Iqbal
- Department of Biomedical Engineering; University of Houston, Houston, TX, USA
| | - Alexandra L McLennan
- Department of Pediatrics; Baylor College of Medicine, Houston, TX, USA
- Center for Translational Research on Inflammatory Diseases; Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Anton Mukhamedshin
- Department of Biomedical Engineering; University of Houston, Houston, TX, USA
| | - Mai T P Dinh
- Department of Biomedical Engineering; University of Houston, Houston, TX, USA
| | - Qisheng Liu
- Center for Translational Research on Inflammatory Diseases; Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
- Department of Medicine; Baylor College of Medicine, Houston, TX, USA
| | - Jacob J Junco
- Department of Pediatrics; Baylor College of Medicine, Houston, TX, USA
| | - Arvind Mohan
- Department of Pediatrics; Baylor College of Medicine, Houston, TX, USA
| | | | - Karen R Rabin
- Department of Pediatrics; Baylor College of Medicine, Houston, TX, USA
| | - Thomas P Fogarty
- Department of Pediatrics; Baylor College of Medicine, Houston, TX, USA
| | | | | | - Fong W Lam
- Department of Pediatrics; Baylor College of Medicine, Houston, TX, USA.
- Center for Translational Research on Inflammatory Diseases; Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.
| |
Collapse
|
2
|
Mishra A, Huang SB, Dubash T, Burr R, Edd JF, Wittner BS, Cunneely QE, Putaturo VR, Deshpande A, Antmen E, Gopinathan KA, Otani K, Miyazawa Y, Kwak JE, Guay SY, Kelly J, Walsh J, Nieman LT, Galler I, Chan P, Lawrence MS, Sullivan RJ, Bardia A, Micalizzi DS, Sequist LV, Lee RJ, Franses JW, Ting DT, Brunker PAR, Maheswaran S, Miyamoto DT, Haber DA, Toner M. Tumor cell-based liquid biopsy using high-throughput microfluidic enrichment of entire leukapheresis product. Nat Commun 2025; 16:32. [PMID: 39746954 PMCID: PMC11696112 DOI: 10.1038/s41467-024-55140-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 11/28/2024] [Indexed: 01/04/2025] Open
Abstract
Circulating Tumor Cells (CTCs) in blood encompass DNA, RNA, and protein biomarkers, but clinical utility is limited by their rarity. To enable tumor epitope-agnostic interrogation of large blood volumes, we developed a high-throughput microfluidic device, depleting hematopoietic cells through high-flow channels and force-amplifying magnetic lenses. Here, we apply this technology to analyze patient-derived leukapheresis products, interrogating a mean blood volume of 5.83 liters from seven patients with metastatic cancer. High CTC yields (mean 10,057 CTCs per patient; range 100 to 58,125) reveal considerable intra-patient heterogeneity. CTC size varies within patients, with 67% overlapping in diameter with WBCs. Paired single-cell DNA and RNA sequencing identifies subclonal patterns of aneuploidy and distinct signaling pathways within CTCs. In prostate cancers, a subpopulation of small aneuploid cells lacking epithelial markers is enriched for neuroendocrine signatures. Pooling of CNV-confirmed CTCs enables whole exome sequencing with high mutant allele fractions. High-throughput CTC enrichment thus enables cell-based liquid biopsy for comprehensive monitoring of cancer.
Collapse
Affiliation(s)
- Avanish Mishra
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Shih-Bo Huang
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Howard Hughes Medical Institute, Bethesda, MD, 20815, USA
| | - Taronish Dubash
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Risa Burr
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Jon F Edd
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Ben S Wittner
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Quinn E Cunneely
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Victor R Putaturo
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Akansha Deshpande
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Ezgi Antmen
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Kaustav A Gopinathan
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Keisuke Otani
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Yoshiyuki Miyazawa
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Ji Eun Kwak
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Sara Y Guay
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Justin Kelly
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - John Walsh
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Linda T Nieman
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Isabella Galler
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - PuiYee Chan
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Michael S Lawrence
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Ryan J Sullivan
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Aditya Bardia
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
- Hematology/Oncology, University of California, Los Angeles, USA
| | - Douglas S Micalizzi
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Lecia V Sequist
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Richard J Lee
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Joseph W Franses
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - David T Ting
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Patricia A R Brunker
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Shyamala Maheswaran
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - David T Miyamoto
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA.
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| | - Daniel A Haber
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA.
- Howard Hughes Medical Institute, Bethesda, MD, 20815, USA.
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA.
| | - Mehmet Toner
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
- Shriners Children's Boston, Boston, MA, 02114, USA.
| |
Collapse
|
3
|
Zhan L, Edd J, Mishra A, Toner M. Label-Free Microfluidic Apheresis of Circulating Tumor Cell Clusters. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405853. [PMID: 39199012 PMCID: PMC11515904 DOI: 10.1002/advs.202405853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/17/2024] [Indexed: 09/01/2024]
Abstract
Screening liters of blood (i.e., apheresis) represents a generalized approach to promote the reliable access to circulating tumor cell clusters (CTCCs), which are known to be highly metastasis-competent, yet ultrarare. However, no existing CTCC sorting technology has demonstrated high throughput, high yield, low shear stress, and minimal blood dilution simultaneously as required in apheresis. Here, a label-free method is introduced termed Precision Apheresis for Non-invasive Debulking of cell Aggregates (PANDA) to continuously isolate CTCCs from undiluted blood to clean buffer through size sorting, processing 1.4 billion cells per second. The cell focusing is optimized within whole blood leveraging secondary transverse flow and margination. The PANDA chip recovers >90% of spiked ≈24 rare HeLa cell clusters from 100 mL undiluted blood samples (equivalent to ≈500 billion blood cells) at 1 L h-1 throughput, with ≤20s device residence time, ≤15 Pa shear stress, and >99.9% return of blood components. The technology lays the groundwork for future routine isolation to increase the recovery of these ultrarare yet clinically significant tumor cell populations from large volumes of blood to advance cancer research, early detection, and treatment.
Collapse
Affiliation(s)
- Li Zhan
- Center for Engineering in Medicine and SurgeryMassachusetts General HospitalBostonMA02129USA
- Harvard Medical SchoolBostonMA02115USA
| | - Jon Edd
- Center for Engineering in Medicine and SurgeryMassachusetts General HospitalBostonMA02129USA
- Cancer CenterMassachusetts General HospitalBostonMA12129USA
| | - Avanish Mishra
- Center for Engineering in Medicine and SurgeryMassachusetts General HospitalBostonMA02129USA
- Harvard Medical SchoolBostonMA02115USA
- Cancer CenterMassachusetts General HospitalBostonMA12129USA
| | - Mehmet Toner
- Center for Engineering in Medicine and SurgeryMassachusetts General HospitalBostonMA02129USA
- Harvard Medical SchoolBostonMA02115USA
- Shriners Hospitals for ChildrenBostonMA02114USA
| |
Collapse
|
4
|
Bojanić I, Novosel G, Lukač Baričević M, Škrnjug P, Horvat E, Mazić S, Batinić J, Kinda SB, Ćepulić BG. Vascular access for autologous peripheral blood stem cells collection by large volume leukapheresis: Single center experience. J Clin Apher 2024; 39:e22095. [PMID: 37941512 DOI: 10.1002/jca.22095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 09/21/2023] [Accepted: 10/12/2023] [Indexed: 11/10/2023]
Abstract
INTRODUCTION Peripheral blood stem cell (PBSC) harvesting requires reliable and safe vascular access. In our institution, a change of practice was implemented and the central venous catheter (CVC) placement for all autologous PBSC collections was abandoned in favor of a careful evaluation of peripheral venous access (PVA) for each individual patient. The aim of this prospective study was to evaluate the rate of patients with adequate peripheral veins for autologous PBSC collection and compare patient characteristics, collection efficacy, and complication rate between patients with PVA and CVC. METHOD Peripheral veins were assessed by the apheresis nurse team in all patients referred between January 2020 and July 2021 to autologous PBSC collection. Only in case of difficult venous access, CVC was inserted. Large volume leukapheresis (LVL) procedures, which processed ≥3 total blood volumes, were performed. RESULTS In 65 (57%) patients PVA was used, while 49 (43%) patients required placement of short-term CVC. Peripheral venous access was successfully used significantly more often in males (69.8%) (P = 0.010), and patients with multiple myeloma (71.0%) than in patients with non-Hodgkin's lymphoma (35.9%) and Hodgkin's lymphoma patients (33.3%) (P < 0.001). There was a significant difference in the type of prior administered chemotherapy; in the patients who received cytostatics free chemotherapy, PVA was used more often (75.0%) (P = 0.007). In terms of the efficacy and safety of LVLs, there were no differences between procedures performed using PVA and CVCs. CONCLUSION Peripheral venous access is feasible for autologous PBSC collection in more than a half of patients, in particular in those with multiple myeloma. Changes in the treatment of multiple myeloma, using new proteasome inhibitors-based and immunomodulatory agents that do not adversely affect peripheral veins, have enabled the use of PVA even at the high blood flow rates required by LVL. Peripheral venous access is not associated with safety issues or with a lesser collection efficiency, and it is cost-effective as well. Each patient referred to autologous PBSC collection needs to be evaluated individually by the experienced apheresis team for the most appropriate venous access.
Collapse
Affiliation(s)
- Ines Bojanić
- Department of Transfusion Medicine and Transplantation Biology, University Hospital Center Zagreb, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
- University of Applied Health Sciences Zagreb, Zagreb, Croatia
| | - Greta Novosel
- Department of Transfusion Medicine and Transplantation Biology, University Hospital Center Zagreb, Zagreb, Croatia
- University of Applied Health Sciences Zagreb, Zagreb, Croatia
| | - Marijana Lukač Baričević
- Department of Transfusion Medicine and Transplantation Biology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Petar Škrnjug
- Department of Transfusion Medicine and Transplantation Biology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Elena Horvat
- Department of Transfusion Medicine and Transplantation Biology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Sanja Mazić
- Department of Transfusion Medicine and Transplantation Biology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Josip Batinić
- School of Medicine, University of Zagreb, Zagreb, Croatia
- Division of Hematology, Department of Internal Medicine, University Hospital Center Zagreb, Zagreb, Croatia
| | - Sandra Bašić Kinda
- Division of Hematology, Department of Internal Medicine, University Hospital Center Zagreb, Zagreb, Croatia
| | - Branka Golubić Ćepulić
- Department of Transfusion Medicine and Transplantation Biology, University Hospital Center Zagreb, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
- University of Applied Health Sciences Zagreb, Zagreb, Croatia
| |
Collapse
|
5
|
Mukhamedshin A, Reddington RC, Dinh MTP, Abhishek K, Iqbal M, Manheim M, Gifford SC, Shevkoplyas SS. Rapid, label-free enrichment of lymphocytes in a closed system using a flow-through microfluidic device. Bioeng Transl Med 2024; 9:e10602. [PMID: 38193116 PMCID: PMC10771558 DOI: 10.1002/btm2.10602] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 01/10/2024] Open
Abstract
The majority of adoptive cellular therapies are produced from peripheral mononuclear cells obtained via leukapheresis and further enriched for the cells of interest (e.g., T cells). Here, we present a first-of-its-kind closed system, which effectively removes ~85% of monocytes and ~88% of platelets, while recovering ~88% of concentrated T cells in a separate output stream, as the leukapheresis sample flows through a microfluidic device at 5 mL/min. The system is driven by a common peristaltic pump, enabled by a novel pressure wave dampener, and operates in a closed bag-to-bag configuration, without requiring any specialized, dedicated equipment. When compared to standard density gradient centrifugation on paired samples, the new system demonstrated a 1.5-fold increase in T cell recovery and a 2-fold reduction in inter-sample variability for this separation outcome. The T cell-to-monocyte ratio of the leukapheresis sample was increased to 20:1, whereas with density gradient processing it decreased to 2:1. As a result of superior purity and/or gentler processing, T cells enriched by the system showed a 2.7-times higher fold expansion during subsequent culture, and an overall 3.5-times higher cumulative yield. This centrifugation-free and label-free closed system for enriching lymphocytes could significantly simplify and standardize the manufacturing of life-saving cellular therapies.
Collapse
Affiliation(s)
- Anton Mukhamedshin
- Department of Biomedical EngineeringUniversity of HoustonHoustonTexasUSA
| | | | - Mai T. P. Dinh
- Department of Biomedical EngineeringUniversity of HoustonHoustonTexasUSA
| | - Kumar Abhishek
- Department of Biomedical EngineeringUniversity of HoustonHoustonTexasUSA
| | - Mubasher Iqbal
- Department of Biomedical EngineeringUniversity of HoustonHoustonTexasUSA
| | - Marc Manheim
- Halcyon Biomedical, IncorporatedFriendswoodTexasUSA
| | | | | |
Collapse
|
6
|
Pessach I, Nagler A. Leukapheresis for CAR-T cell production and therapy. Transfus Apher Sci 2023; 62:103828. [PMID: 37838564 DOI: 10.1016/j.transci.2023.103828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is an effective, individualized immunotherapy, and novel treatment for hematologic malignancies. Six commercial CAR-T cell products are currently approved for lymphatic malignancies and multiple myeloma. In addition, an increasing number of clinical centres produce CAR-T cells on-site, which enable the administration of CAR-T cells on site. The CAR-T cell products are either fresh or cryopreserved. Manufacturing CAR-T cells is a complicated process that begins with leukapheresis to obtain T cells from the patient's peripheral blood. An optimal leukapheresis product is crucial step for a successful CAR-T cell therapy; therefore, it is imperative to understand the factors that may affect the quality or T cells. The leukapheresis for CAR-T cell production is well tolerated and safe for both paediatric and adult patients and CAR-Τ cell therapy presents high clinical response rate in many studies. CAR-T cell therapy is under continuous improvement, and it has transformed into an almost standard procedure in clinical haematology and stem cell transplantation facilities that provide both autologous and allogeneic stem cell transplantations. In patients suffering from advanced haematological malignancies, CAR-T cell therapy shows incredible antitumor efficacy. Even after a single infusion of autologous CD19-targeting CAR-T cells in patients with relapsed or refractory diffuse large B cell lymphoma (DLBCL) and acute lymphoblastic leukaemia (ALL), long lasting remission is observed, and a fraction of the patients are being cured. Future novel constructs are being developed with better T cell persistence and better expansion. New next-generation CAR-T cells are currently designed to avoid toxicities such as cytokine release syndrome and neurotoxicity.
Collapse
Affiliation(s)
- Ilias Pessach
- Hematology Department, Athens Medical Center, Athens, Greece
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Israel.
| |
Collapse
|
7
|
Rico LG, Juncà J, Salvia R, Ward MD, Bradford JA, Petriz J. Efficient discrimination of functional hematopoietic stem cell progenitors for transplantation by combining alkaline phosphatase activity and CD34 + immunophenotyping. Methods Cell Biol 2023; 195:101-113. [PMID: 40180450 DOI: 10.1016/bs.mcb.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Alkaline phosphatase (ALP) is a membrane-associated hydrolase enzyme with dimeric structure that catalyzes phosphate esters, optimally at alkaline pH. ALP has a focus of interest, since this enzyme is highly expressed in primitive stem cells, such as progenitor cells, non-differentiating cells, and primordial cells. We previously adapted a fluorescent microscopy-based assay for quantifying ALPhigh and ALPlow cells by flow cytometry in combination with immunophenotyping. Our method uses a minimal sample perturbation approach, avoiding the use of erythrocyte lysing solutions and washing steps, and offering opportunities to combine live cell response and functional assessment with cell immunophenotyping, while minimizing sample preparation effects on the cell biology. Here we provide a detailed experiment protocol to determine alkaline phosphatase activity in CD34+ hematopoietic stem cells from blood and apheresis products obtained from patients involved in a stem cell mobilization process for allo- or auto-transplant. This study may provide the early detection of progenitors at different levels of differentiation and therefore, relate this information to long-term engraftment in hematopoietic stem cell transplants.
Collapse
Affiliation(s)
- Laura G Rico
- Functional Cytomics Lab, Germans Trias i Pujol Research Institute (IGTP), ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Barcelona, Spain
| | - Jordi Juncà
- Functional Cytomics Lab, Germans Trias i Pujol Research Institute (IGTP), ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Barcelona, Spain
| | - Roser Salvia
- Functional Cytomics Lab, Germans Trias i Pujol Research Institute (IGTP), ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Barcelona, Spain
| | - Michael D Ward
- Thermo Fisher Scientific, Fort Collins, CO, United States
| | | | - Jordi Petriz
- Functional Cytomics Lab, Germans Trias i Pujol Research Institute (IGTP), ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Barcelona, Spain.
| |
Collapse
|
8
|
Piñeyroa JA, Cid J, Vlagea A, Carbassé G, Henao P, Bailo N, Ortiz-Maldonado V, Martínez-Cibrian N, Español M, Delgado J, Urbano-Ispizua Á, Lozano M. Evaluation of cell collection efficiency in non-mobilized adult donors for autologous chimeric antigen receptor T-cell manufacturing. Vox Sang 2023; 118:217-222. [PMID: 36516201 DOI: 10.1111/vox.13394] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 11/08/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND OBJECTIVES Data about collection efficiency 1 (CE1), which takes into account blood cell counts before and after collection, thus providing a more accurate estimate, in the collection of autologous T lymphocytes by apheresis for chimeric antigen receptor (CAR) T-cells remain scarce. We evaluated donor- and procedure-related characteristics that might influence the CE1 of lymphocytes. MATERIALS AND METHODS We retrospectively reviewed all mononuclear cell (MNC) collections) performed for CAR T-cell manufacturing in our institution from May 2017 to June 2021 in adult patients. Age, gender, weight, total blood volume (TBV), prior haematopoietic cell transplant, diagnosis, days between last treatment and apheresis, pre-collection cell counts, duration of apheresis, TBV processed, vascular access, inlet flow and device type were analysed as potential factors affecting CE1 of lymphocytes. RESULTS A total of 127 autologous MNC collections were performed on 118 patients diagnosed with acute lymphoblastic leukaemia (n = 53, 45%), non-Hodgkin lymphoma (n = 40, 34%), multiple myeloma (n = 19, 16%), and chronic lymphocytic leukaemia (n = 6, 5%). The median CE1 of lymphocytes was 47% (interquartile range: 32%-65%). In multiple regression analysis, Amicus device was associated with higher CE1 of lymphocytes (p = 0.01) and lower CE1 of platelets (p < 0.01) when compared with Optia device. CONCLUSION The knowledge of the MNC and lymphocyte CE1 of each apheresis device used to collect cells for CAR T therapy, together with the goal of the number of cells required, is essential to define the volume to be processed and to ensure the success of the collection.
Collapse
Affiliation(s)
- Juan A Piñeyroa
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, ICMHO, Hospital Clínic, Barcelona, Spain
| | - Joan Cid
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, ICMHO, Hospital Clínic, Barcelona, Spain.,IDIBAPS, Barcelona, Spain.,University of Barcelona, Barcelona, Spain
| | - Alexandru Vlagea
- Department of Immunology, CDB, Hospital Clínic, Barcelona, Spain
| | - Gloria Carbassé
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, ICMHO, Hospital Clínic, Barcelona, Spain
| | - Paola Henao
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, ICMHO, Hospital Clínic, Barcelona, Spain
| | - Noemí Bailo
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, ICMHO, Hospital Clínic, Barcelona, Spain
| | | | | | - Marta Español
- Department of Immunology, CDB, Hospital Clínic, Barcelona, Spain
| | - Julio Delgado
- IDIBAPS, Barcelona, Spain.,University of Barcelona, Barcelona, Spain.,Department of Hematology, ICMHO, Hospital Clínic, Barcelona, Spain
| | - Álvaro Urbano-Ispizua
- IDIBAPS, Barcelona, Spain.,University of Barcelona, Barcelona, Spain.,Department of Hematology, ICMHO, Hospital Clínic, Barcelona, Spain
| | - Miquel Lozano
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, ICMHO, Hospital Clínic, Barcelona, Spain.,IDIBAPS, Barcelona, Spain.,University of Barcelona, Barcelona, Spain
| |
Collapse
|
9
|
A high-throughput microfluidic device based on controlled incremental filtration to enable centrifugation-free, low extracorporeal volume leukapheresis. Sci Rep 2022; 12:13798. [PMID: 35963876 PMCID: PMC9376077 DOI: 10.1038/s41598-022-16748-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/14/2022] [Indexed: 11/08/2022] Open
Abstract
Leukapheresis, the extracorporeal separation of white blood cells (WBCs) from red blood cells (RBCs) and platelets (PLTs), is a life-saving procedure used for treating patients with cancer and other conditions, and as the initial step in the manufacturing of cellular and gene-based therapies. Well-tolerated by adults, leukapheresis poses a significant risk to neonates and low-weight infants because the extracorporeal volume (ECV) of standard centrifugation-based machines represents a particularly large fraction of these patients' total blood volume. Here we describe a novel high-throughput microfluidic device (with a void volume of 0.4 mL) based on controlled incremental filtration (CIF) technology that could replace centrifugation for performing leukapheresis. The CIF device was tested extensively using whole blood from healthy volunteers at multiple hematocrits (5-30%) and flow rates (10-30 mL/min). In the flow-through regime, the CIF device separated WBCs with > 85% efficiency and 10-15% loss of RBCs and PLTs while processing whole blood diluted with saline to 10% hematocrit at a flow rate of 10 mL/min. In the recirculation regime, the CIF device demonstrated a similar level of separation performance, virtually depleting WBCs in the recirculating blood (~ 98% reduction) by the end of a 3.5-hour simulated leukapheresis procedure. Importantly, the device operated without clogging or decline in separation performance, with minimal activation of WBCs and PLTs and no measurable damage to RBCs. Compared to the typical parameters of centrifugation-based leukapheresis, the CIF device had a void volume at least 100-fold smaller, removed WBCs about twice as fast, and lost ~ 2-3-fold fewer PLTs, while operating at a flow rate compatible with the current practice. The hematocrit and flow rate at which the CIF device operated were significantly higher than previously published for other microfluidic cell separation methods. Finally, this study is the first to demonstrate a highly efficient separation of cells from recirculating blood using a microfluidic device. Overall, these findings suggest the feasibility of using high-throughput microfluidic cell separation technology to ultimately enable centrifugation-free, low-ECV leukapheresis. Such a capability would be particularly useful in young children, a vulnerable group of patients who are currently underserved.
Collapse
|
10
|
Considerations for immune effector cell therapy collections: a white paper from the American Society for Apheresis. Cytotherapy 2022; 24:916-922. [DOI: 10.1016/j.jcyt.2022.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/18/2022]
|
11
|
Thibodeaux SR, Aqui NA, Park YA, Schneiderman J, Su LL, Winters JL, Zubair AC, Schwartz J, Liu HD. Lack of defined apheresis collection criteria in publicly available CAR-T cell clinical trial descriptions: Comprehensive review of over 600 studies. J Clin Apher 2022; 37:223-236. [PMID: 35085413 DOI: 10.1002/jca.21964] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 11/20/2021] [Accepted: 12/16/2021] [Indexed: 01/28/2023]
Abstract
BACKGROUND Chimeric antigen receptor T (CAR-T) cell successes have encouraged continued clinical study. Apheresis collection of starting material for CAR-T cell therapy product manufacturing is critical but described approaches suggest variability and clinical guidelines are currently lacking. The goal of this study was to gather and assess variability in apheresis collection descriptions in publicly available CAR T-cell therapy clinical trials. STUDY DESIGN We searched clinicaltrials.gov (a publicly available clinical trial database) for "chimeric antigen receptor T cells" on July 01, 2020 and studies accessed July 30, 2020-August 15, 2020. Data collected included date posted, study characteristics, apheresis mentions (number, location, and context), laboratory parameters and transfusion allowances. Apheresis context was analyzed using a qualitative inductive approach of grounded theory method with open coding. Text was classified into 37 context codes, grouped into 12 categories, and then consolidated into patient, procedure, product, and miscellaneous themes. RESULTS Apheresis was mentioned 1044 times in 322 (51.9%) of 621 total studies. Laboratory parameters mentioned included white blood cells (100 studies), absolute neutrophil count (220 studies), absolute lymphocyte count (102 studies), CD3+ cell (38 studies), hemoglobin (233 studies, 54 studies specified transfusion allowance), and platelet (269 studies, 48 studies specified transfusion allowance). CONCLUSIONS Apheresis collection of CAR-T cell products is not well-defined in clinical study descriptions and the context is inconsistent. Laboratory parameters useful for apheresis collection are variably present and do not consistently align with current practices. Further exploration, and clinical guideline development will encourage alignment of apheresis collections for CAR-T cell products.
Collapse
Affiliation(s)
- Suzanne R Thibodeaux
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nicole A Aqui
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yara A Park
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jennifer Schneiderman
- Department of Pediatric Hematology/Oncology/Neuro-oncology/Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Leon L Su
- Department of Pathology and Laboratory Medicine, Phoenix Children's Hospital, Phoenix, Arizona, USA
| | - Jeffrey L Winters
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Abba C Zubair
- Laboratory Medicine and Pathology and Center for Regenerative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Joseph Schwartz
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine, New York, New York, USA
| | - Hien D Liu
- Department of Bone Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | | |
Collapse
|
12
|
Maru B, Nadeau L, McKeague M. Enhancing CAR-T Cell Therapy with Functional Nucleic Acids. ACS Pharmacol Transl Sci 2021; 4:1716-1727. [PMID: 34927006 DOI: 10.1021/acsptsci.1c00188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Indexed: 02/07/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy is a relatively new form of immunotherapy that has had success in treating patients with hematologic malignancies, leading to three recent United States Food and Drug Administration approvals. However, several challenges hinder the widespread use of CAR-T therapy. Here, we review the application of functional nucleic acids such as aptamers and ribozymes as novel tools to improve a variety of steps in CAR-T cell therapy development. We critically examine key studies that highlight the benefits of functional nucleic acids at different stages of cell-based therapy and discuss the feasibility of their practical clinical application. Finally, we offer insights into potential opportunities where chemists can significantly contribute to the innovative incorporation of functional nucleic acids to overcome challenges associated with this cutting-edge immunotherapy.
Collapse
Affiliation(s)
- Bruktawit Maru
- Pharmacology and Therapeutics, Faculty of Medicine, McGill University, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada
| | - Lea Nadeau
- Pharmacology and Therapeutics, Faculty of Medicine, McGill University, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada
| | - Maureen McKeague
- Pharmacology and Therapeutics, Faculty of Medicine, McGill University, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada.,Department of Chemistry, Faculty of Science, McGill University, 801 Sherbrooke Street West, Montreal, QC H3A 0B8, Canada
| |
Collapse
|
13
|
Livingston J, Di-Mola M, Lowry J, Ruse N, Chiang KY, Chopra Y, Schechter T, Ali M, Licht C, Wall D, Krueger J. Peripheral venous catheter collection of immune effector cells and hematopoietic stem cells is feasible and safe in older pediatric patients. Transfusion 2021; 61:3413-3419. [PMID: 34751944 DOI: 10.1111/trf.16717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND The Collection of hematopoietic stem cells (HSC) and immune effector cells (IEC) has unique challenges in children. To maintain adequate blood flow, central venous catheters (CVCs) remain the standard of care in many centers, but are associated with procedural risks and increased resource utilization. The goal of this study was to determine feasibility and safety of peripheral venous catheter (PVC) cell collection in older children. METHODS Patients and donors requiring venous access with weight >25 kg, age >8 years were screened for PVC collection via 18G PVCs. Those with poor venous access (on history/exam/pre-screening ultrasound) or unable to maintain suitable procedural position were excluded. Comparison was made to CVC collections in a matched patient cohort. RESULTS Thirty-eight individuals were screened and met age/weight criteria for PVC collection. Five did not have PVC collection attempted due to poor access (n = 4) or behavioral concerns (n = 1). Thirty-three had PVC collection attempt (HSC = 22; IEC = 11) with median age 15.3 year (range 9.7-18.0) and weight 58.5 kg (range 27.9-115.4). Thirty-two of 33 (97%) patients were collected successfully by PVC without adverse events. Comparing PVC to matched CVC collection cohort (n = 18), there was no significant difference in flow rate (48.2 mL/h vs 53.9 mL/h, p = 0.12), collection time (266 min vs 262 min, p = 0.85) or collection efficiency (IEC/CD3 60.9% vs 60.8% p = 0.99; HSC/CD34 53.6% vs 41.3% p = 0.05). CONCLUSION PVC collection of HSC and IEC is feasible and safe in older children with comparable collection efficiency to CVC collections. Ultrasound screening may reduce failure rates. PVC collections can reduce the risk of CVC insertions and associated healthcare costs.
Collapse
Affiliation(s)
- Joel Livingston
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, The University of Toronto, Toronto, Ontario, Canada
| | - Maria Di-Mola
- Dialysis and Apheresis Program, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jane Lowry
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nigel Ruse
- Vascular Access Service, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kuang-Yueh Chiang
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, The University of Toronto, Toronto, Ontario, Canada
| | - Yogi Chopra
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, The University of Toronto, Toronto, Ontario, Canada
| | - Tal Schechter
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, The University of Toronto, Toronto, Ontario, Canada
| | - Muhammad Ali
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, The University of Toronto, Toronto, Ontario, Canada
| | - Christoph Licht
- Department of Pediatrics, The University of Toronto, Toronto, Ontario, Canada.,Dialysis and Apheresis Program, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Donna Wall
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, The University of Toronto, Toronto, Ontario, Canada
| | - Joerg Krueger
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, The University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Tian M, Li X, Liu F, Xu D, Sun L, Li Q, Ling Y, Bai Y, Wu X, Liu Y, Qiao J. Efficacy of peripheral arterial access for peripheral blood stem cells collection. J Clin Apher 2021; 36:864-869. [PMID: 34558738 DOI: 10.1002/jca.21940] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND Transplantation of peripheral blood stem cells (PBSCs) mobilized by cytokines is increasingly applied to treat patients with hematologic diseases, such as lymphoma, multiple myeloma, leukemia, etc. Successful hematopoietic stem cell transplantation (HSCT) increasingly depends on the collection of hematopoietic stem cells (HSCs) from peripheral blood. Peripheral vein (PV) is the most common type of blood access. When the blood vessels are not well filled and the blood flow is insufficient, the machine will appear repeated low pressure alarm or pipeline coagulation, which seriously affects the collection efficiency. A peripheral artery (PA) is utilized for drawing blood, while a peripheral vein is used for blood return, that is a way to perform apheresis. The advantages of PA are that it ensures adequate extracorporeal circulation blood flow, stable blood flow rate, simple operation, and relatively low price. However, there are very few studies on the efficacy of peripheral arterial access for HSCs collection. Therefore, this retrospective study was conducted to assess the effectiveness of PA and PV access for PBSCs collection. METHODS We performed a retrospective analysis of 150 apheresis procedures on 26 patients and 95 healthy donors collected by PV or PA access from March 1, 2020 to March 1, 2021. We compared the CD34+ cell count, collection efficiency (CE), duration of processing a single blood volume, number of low-pressure alarms, average blood flow rate and number of punctures between the two groups. Also, we analyzed adverse events. RESULTS There was no significant difference in the quality of apheresis blood components between the PA group and the PV group. The CD34+ cells collected was 274.16 ± 216.31 × 106 in the PV group and 246.63 ± 127.94 × 106 in the PA group. The CE in the PA group was 49.50 ± 9.88%, higher than 42.39 ± 14.62% in the PV group. The duration of processing a single blood volume was 90.67 ± 15.35 min in the PV group and 79.68 ± 10.28 min in the PA group. The number of low-pressure alarms in the PA group was 0.38 ± 0.98, <2.42 ± 1.76 in the PV group, and the average blood flow rate in the PA group was 59.27 ± 2.18, higher than 54.21 ± 3.41 in the PV group. The difference was statistically significant (P < .05). The Number of punctures was 1.35 ± 0.75 in the PA group and 1.41 ± 1.01 in the PV group. There was no statistically significant difference. CONCLUSION Peripheral artery is a safe, reliable, economical, convenient, and fast vascular access, which opens a new way to the establishment of vascular access for PBSCs collection.
Collapse
Affiliation(s)
- Min Tian
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Xiangmin Li
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University &Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Feng Liu
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Dongmei Xu
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Lanlan Sun
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Qiuhuan Li
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yue Ling
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yingxue Bai
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Xiaoling Wu
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yun Liu
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Jianhong Qiao
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| |
Collapse
|
15
|
Augustine M, Murugesan M, Nayanar SK, Padmanabhan M. Adopting a vein assessment tool improves procedural outcomes in double dose platelet collections - A prospective study. Transfus Apher Sci 2021; 60:103198. [PMID: 34187772 DOI: 10.1016/j.transci.2021.103198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND This study aims to determine the phlebotomy and procedural outcomes using a vein assessment tool (VAT) in Double Dose Platelet (DDP) collections by apheresis. METHODS VAT was based on assessing vein visibility, palpation and size with maximum score of 12 and the least being 0 and the scores were graded as adequate and inadequate. A vein-viewer was used for studying cubital vein patterns (type 1-5). Phlebotomy outcome was defined based on need for re-puncture. Procedural outcomes in terms of target yield attained and RBC reinfusion completed. Chi square test and Mann- Whitney U test were used to assess the vein score and pattern against phlebotomy and procedural outcome. RESULTS Out of 200 DDP collections, the phlebotomy was successful in 88 % with good procedural outcome in 94 % donations. The cut off in VAT scores for successful phlebotomy was ≥8 (AUC: 70 %). Median vein scores of the arm selected for phlebotomy was 9 and graded adequate in 154 (77 %) donations.Odds for successful phlebotomy was 3.7 times higher when donors had an adequate VAT grades(p = 0.003). Procedural outcomes was favourable when at least one arm had adequate VAT grade when compared to both arms being inadequate (98 % vs 82 %; p < 0.001). Phlebotomy failure was more with first time apheresis donors than repeat apheresis donors (p = 0.014). CONCLUSION This study indicated that a VAT score with a cut off of ≥8 had better phlebotomy and procedural outcomes in DDP collections and that donor with at least one arm having the VAT score of ≥8 are preferred for DDP collections.
Collapse
Affiliation(s)
- Merline Augustine
- Transfusion Medicine, Malabar Cancer Centre, Thalassery, Kerala, India
| | | | | | - Maya Padmanabhan
- Clinical Research and Biostatistics, Malabar Cancer Centre, Thalassery, Kerala, India
| |
Collapse
|
16
|
Konno S, Yanagisawa R, Motoki N, Shimodaira S. Predictive factors of poor blood collecting flow during leukocyte apheresis for cellular therapy. Ther Apher Dial 2021; 25:1001-1011. [PMID: 33522707 DOI: 10.1111/1744-9987.13631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/21/2020] [Accepted: 01/21/2021] [Indexed: 11/27/2022]
Abstract
Leukocyte apheresis is necessary in various cellular therapies. However, maintenance of a stable flow rate during leukocyte apheresis is often difficult, even in patients or donors without major problems. Despite this, predictive methods and evidence regarding the reality of the situation are limited. We conducted a retrospective analysis involving adult patients who required leukocyte apheresis for the treatment of neoplasms using WT1-pulsed dendritic cell vaccine. Monocytes were separated from apheresis products to obtain dendritic cells. All the patients were pre-evaluated based on laboratory and chest X-ray findings and subjected to an identical apheresis procedure. The occurrence of poor blood collecting flow during leukocyte apheresis was monitored, and the frequency, clinical information, and associated risk factors were analyzed. Among 160 cases, poor blood collecting flow was observed in 53 cases (33.1%) in a median time of 54 min (range, 2-127 min) post-initiation of leukocyte apheresis. Owing to difficulty in obtaining higher collecting flow, a longer procedure time was required, and in some cases, the scheduled apheresis cycles could not be completed. Consequently, the number of harvested monocytes was low. Multivariable analysis indicated that female patients have an increased risk of poor inlet flow rate. Furthermore, prolonged QT dispersion (QTD) calculated using Bazett's formula was found to be a risk factor. Although the patients did not present any major problems during leukocyte apheresis, poor blood collecting flow was observed in some cases. Sex and pre-evaluated QTD might be useful predictors for these cases; however, further prospective evaluation is necessary.
Collapse
Affiliation(s)
- Saori Konno
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Japan.,Division of Blood Transfusion, Shinshu University Hospital, Matsumoto, Japan
| | - Ryu Yanagisawa
- Division of Blood Transfusion, Shinshu University Hospital, Matsumoto, Japan.,Center for Advanced Cell Therapy, Shinshu University Hospital, Matsumoto, Japan
| | - Noriko Motoki
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Shigetaka Shimodaira
- Department of Regenerative Medicine, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
17
|
Couzin C, Manceau S, Diana JS, Joseph L, Magnani A, Magrin E, Amrane H, Dupont E, Raphalen JH, Sibon D, Marcais A, Suarez F, Cavazzana M, Lefrère F, Delville M. Vascular access for optimal hematopoietic stem cell collection. J Clin Apher 2020; 36:12-19. [PMID: 32854142 DOI: 10.1002/jca.21828] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Autologous and allogeneic hematopoietic stem cell transplantation of cytokine-mobilized peripheral blood stem cells (PBSCs) is increasingly used to treat patients with hematologic disorders. Different types of vascular access have been exploited for the apheresis procedure, including peripheral veins (PV) and central venous catheter (CVC). In some cases, PV access is unavailable. There are few published data on the efficiency and quality of harvesting with different types of vascular access. This study brings out complications and morbidity of this procedure linked to these different access. METHODS We performed a comparative, retrospective, single-center study of hematopoietic stem cell collection using these two types of vascular access. We compared the efficiency and complication rate for 617 adults apheresis sessions in 401 patients and healthy donors, for PBSC collection via PV or CVC between 2010 and 2016. The quality of the HSC product was evaluated in terms of the total CD34 + count and neutrophil contamination. RESULTS The PV and CVC groups did not differ significantly in terms of the quality of the apheresis product, mean ± SD CD34 + cells collected in PV group was 383.1 ± 402.7 × 10e6 and 298.8 ± 372.7 × 10e6 and the level of neutrophil contamination was 21.0 ± 17.8% in the PV group and 20.6 ± 18.4% in the CVC group. The complication rate did not differ between the two groups. CONCLUSION The type of vascular access for apheresis hematopoietic stem cell harvesting must be determined by trained staff. Successful harvesting can be performed via PV then CVC is not needed or not available.
Collapse
Affiliation(s)
- Chloé Couzin
- Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France.,Paris Descartes University, Paris, France
| | - Sandra Manceau
- Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Sébastien Diana
- Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France.,Paris Descartes University, Paris, France
| | - Laure Joseph
- Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | - Alessandra Magnani
- Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | - Elisa Magrin
- Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | - Horiya Amrane
- Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Emilie Dupont
- Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Herlé Raphalen
- Intensive Care Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - David Sibon
- Haematology Department, Necker-Children's Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Ambroise Marcais
- Haematology Department, Necker-Children's Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Felipe Suarez
- Paris Descartes University, Paris, France.,Haematology Department, Necker-Children's Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Marina Cavazzana
- Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France.,Paris Descartes University, Paris, France
| | - François Lefrère
- Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | - Marianne Delville
- Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France.,Paris Descartes University, Paris, France
| |
Collapse
|
18
|
Caime A, Piredda A, Lucchetti B, Magarò A, Zencovich C, Clerici M, Laszlo D. Midline catheter as effective device in healthy allogeneic donors and patients without an adequate peripheral venous access for HPC collection by apheresis: Preliminary experience at IEO. Transfus Apher Sci 2020; 59:102740. [DOI: 10.1016/j.transci.2020.102740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/05/2019] [Accepted: 11/05/2019] [Indexed: 01/31/2023]
|
19
|
Egri N, Ortiz de Landazuri I, San Bartolomé C, Ortega JR, Español-Rego M, Juan M. CART manufacturing process and reasons for academy-pharma collaboration. Immunol Lett 2019; 217:39-48. [PMID: 31669547 DOI: 10.1016/j.imlet.2019.10.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/14/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023]
Abstract
The success of genetically engineered T-cells modified with a chimeric antigen receptor as an adoptive cell immunotherapy and the subsequent last regulatory approvals of products based on this therapy are leading to a crescent number of both academic and pharmaceutical industry clinical trials testing new approaches of this "living drugs". The aim of this review is to outline the latest developments and regulatory considerations in this field, with a particular emphasis to differences and similarities between academic and industry approaches and the role they should play to coexist and move forward together. To do that, the main considerations for the manufacturing process are firstly discussed, from the chimeric antigen receptor design to final production steps, passing through ex vivo T-cell handling, gene delivery methods, patient´s final product infusion observations or possible associated side effects of this treatment.
Collapse
Affiliation(s)
- Natalia Egri
- Servei d'Immunologia, Hospital Clínic de Barcelona (HCB), University of Barcelona (UB), Banc de Sang i Teixits (BST) - HCB Immunotherapy Platform Barcelona, Spain
| | - Iñaki Ortiz de Landazuri
- Servei d'Immunologia, Hospital Clínic de Barcelona (HCB), University of Barcelona (UB), Banc de Sang i Teixits (BST) - HCB Immunotherapy Platform Barcelona, Spain
| | - Clara San Bartolomé
- Servei d'Immunologia, Hospital Clínic de Barcelona (HCB), University of Barcelona (UB), Banc de Sang i Teixits (BST) - HCB Immunotherapy Platform Barcelona, Spain
| | - J Ramón Ortega
- Servei d'Immunologia, Hospital Clínic de Barcelona (HCB), University of Barcelona (UB), Banc de Sang i Teixits (BST) - HCB Immunotherapy Platform Barcelona, Spain
| | - Marta Español-Rego
- Servei d'Immunologia, Hospital Clínic de Barcelona (HCB), University of Barcelona (UB), Banc de Sang i Teixits (BST) - HCB Immunotherapy Platform Barcelona, Spain
| | - Manel Juan
- Servei d'Immunologia, Hospital Clínic de Barcelona (HCB), University of Barcelona (UB), Banc de Sang i Teixits (BST) - HCB Immunotherapy Platform Barcelona, Spain; IDIBAPS, University of Barcelona (UB), Banc de Sang i Teixits (BST) - HCB Immunotherapy Platform Barcelona, Spain; University of Barcelona (UB), Banc de Sang i Teixits (BST) - HCB Immunotherapy Platform Barcelona, Spain; Hospital Sant Joan de Déu, University of Barcelona (UB), Banc de Sang i Teixits (BST) - HCB Immunotherapy Platform Barcelona, Spain.
| |
Collapse
|
20
|
Abstract
Transplants using peripheral blood hemopoietic stem/progenitor (PBHS) cells are widely performed for the treatment of patients with hematologic disorders in routine practice and clinical trials. Although the process from mobilization to infusion of PBHS cells has been mostly established, optimal conditions for each process remain undetermined. Adverse reactions caused by PBHS cell infusions have not been systematically recorded. In transplants using PBHS cells, a number of problems still exist. In this section, the current status of and future perspectives regarding PBHS cells are described.
Collapse
Affiliation(s)
- Kazuo Muroi
- Division of Cell Transplantation and Transfusion, Jichi Medical University Hospital, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan.
| |
Collapse
|