1
|
Traynor R, Vignola I, Sarkar S, Prochazkova M, Cai Y, Shi R, Underwood S, Ramanujam S, Yates B, Silbert S, Jin P, Dreyzin A, Shah NN, Somerville RP, Stroncek DF, Song HW, Highfill SL. Efficient manufacturing of CAR-T cells from whole blood: a scalable approach to reduce costs and enhance accessibility in cancer therapy. Cytotherapy 2025; 27:400-409. [PMID: 39652017 PMCID: PMC11810577 DOI: 10.1016/j.jcyt.2024.11.013] [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: 08/27/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 02/12/2025]
Abstract
BACKGROUND Chimeric antigen receptor T (CAR-T) cells have significantly advanced the treatment of cancers such as leukemia and lymphoma. Traditionally, T cells are collected from patients through leukapheresis, an expensive and potentially invasive process that requires specialized equipment and trained personnel. Although whole blood collections are much more technically straightforward, whole blood starting material has not been widely utilized for clinical CAR-T cell manufacturing, in part due to lack of manufacturing processes designed for use in a good manufacturing practice (GMP) environment. Collecting cellular starting material from whole blood without leukapheresis could reduce manufacturing complexity and cost, thereby improving accessibility to CAR-T cell therapy. METHODS Whole blood samples were collected from eight healthy donors and one pediatric B-cell acute lymphoblastic leukemia (B-ALL) patient. These samples were processed using the Sepax C-Pro (Cytiva) instrument to isolate mononuclear cells (MNCs) via density gradient separation. CAR-T cells were then manufactured from the isolated MNCs using a GMP-compliant 7-day protocol, whereby T cells were activated with anti-CD3 and IL-2, transduced with GMP lentiviral vector encoding a CD19/CD22 bispecific CAR, and expanded in gas permeable cell culture bags. The resulting CAR-T cells were then evaluated for their phenotypic and functional properties using flow cytometry, cytokine release and cytotoxicity assays. RESULTS From an average 77.7 mL of whole blood from healthy donors (range = 29-96 mL), we isolated an average of 42.2 × 106 CD3⁺ T cells (range 7.3-63.0) postprocessing. CAR-T cell cultures were initiated from thaw using 1-10 × 106 starting CD3+ T cells, yielding a median T cell number of 105 × 106 cells on day 7 (range 61-188 × 106). We observed 66 ± 11% mean transduction efficiency and produced a mean of 77.4 × 106 transduced CAR-T cells (range 30.8-143.5 × 106). Similar results were obtained when using a blood sample (28mL) obtained from a patient with relapsed B-ALL who had received recent chemotherapy. CONCLUSIONS Therapeutically relevant doses of CD19/CD22 CAR-T cells can be successfully manufactured from whole blood. On average, 80 mL of whole blood yields enough CAR-T cells to create a single dose for a pediatric patient (50 kg) at a dosage of 1 × 106 CAR-T cells/kg. For larger patients, scaling up is straightforward by collecting a larger blood volume. This method also demonstrates a cost-effective approach to T cell activation and expansion which, alongside a more straightforward collection of whole blood, makes it more widely accessible especially for middle- and low-income countries. By reducing costs and labor, this strategy has the potential to significantly expand global access to CAR-T cell therapy.
Collapse
Affiliation(s)
- Roshini Traynor
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Isabella Vignola
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Sarmila Sarkar
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Michaela Prochazkova
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Yihua Cai
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Rongye Shi
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Sarah Underwood
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Supriya Ramanujam
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Bonnie Yates
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sara Silbert
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ping Jin
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Alexandra Dreyzin
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Robert P Somerville
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - David F Stroncek
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Hannah W Song
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Steven L Highfill
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
2
|
Hanson MD, Groh D, Barsoom M. Use of Therapeutic Plasma Exchange and Intravenous Immunoglobulin to Prevent Complications in a K+ Sensitized Pregnancy. Cureus 2024; 16:e72254. [PMID: 39583478 PMCID: PMC11584755 DOI: 10.7759/cureus.72254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 11/26/2024] Open
Abstract
The K antigen is a major cause of hemolytic disease of the fetus and newborn (HDFN). K-HDFN is unique in that it can result in destruction of not just mature erythrocytes but fetal erythrocyte progenitors, causing severe fetal anemia earlier in pregnancy than other antigens. This poses a danger to fetal health as intrauterine transfusion (IUT), the preferred method of managing HDFN, becomes riskier earlier in pregnancy. This report follows a K-negative mother managed with an alternative treatment, designed to delay the need for IUT. The patient is a 32-year-old K-negative female, G2P1001, sensitized against K by her previous pregnancy with a 256 anti-K antibody titer. To prevent HDFN, she opted for preventative treatment to lower her immune response. She received three rounds of therapeutic plasma exchange, which lowered her titer to 64, followed by weekly IVIG administration at a dosage of 1g/kg body weight. Fetal anemia was monitored via middle cerebral artery Doppler imaging. The fetus did require two IUTs; however, they were not required until the third trimester. A healthy baby was delivered at 36 weeks with mild anemia and a positive direct antiglobulin test. The standard of care for K-sensitized pregnancies involves watchful waiting and correction of anemia with IUT. However, K-HDFN, if untreated, can cause severe anemia in early pregnancy when IUT is less viable. This case study joins a handful of others in reported literature where a K-sensitized pregnancy was treated prophylactically with immune-modulating therapies and argues that these treatments deserve further recognition and study.
Collapse
Affiliation(s)
- Mary D Hanson
- Pathology, Creighton University School of Medicine, Omaha, USA
| | - Darren Groh
- Pathology, Creighton University School of Medicine, Omaha, USA
| | - Michael Barsoom
- Maternal/Fetal Medicine, Creighton University School of Medicine, Omaha, USA
| |
Collapse
|
3
|
Walker H, O'Reilly E, Millard Z, Cusack B, Duncan A, Clucas L, Fleming J, Hughes D, Greenway A, Metz D. Small but mighty: Case report and practical guidance for peripheral blood stem cell collection in small infants. J Clin Apher 2024; 39:e22141. [PMID: 39060110 DOI: 10.1002/jca.22141] [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: 02/20/2024] [Revised: 05/31/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024]
Abstract
Modern apheresis devices, with increased procedural precision, automation, and monitoring, have been shown to allow for safe delivery of apheresis therapies in young children. Medical advances are increasing demand for apheresis procedures like mononuclear cell collection in infants <10 kg, including stem-cell supported chemotherapy, cell collection for chimeric antigen receptor T cell development, and now ex vivo gene therapies for rare genetic diseases. Nevertheless, safe delivery in small infants involves a range of unique considerations and challenges, beyond just size, and experience will vary between centers. In this case report we describe our experience performing mononuclear cell collection in our smallest patient to date and outline a practice guideline developed following a literature review and discussion with both international experts and device representatives. This case may help to inform other clinicians aiming to provide apheresis care to very small infants in their own centers.
Collapse
Affiliation(s)
- Hannah Walker
- Children's Cancer Centre, The Royal Children's Hospital, Melbourne, Victoria, Australia
- Apheresis Service, The Royal Children's Hospital, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Division of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Erin O'Reilly
- Department of Clinical Haematology, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Zoe Millard
- Apheresis Service, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Brendan Cusack
- Apheresis Service, The Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Nephrology, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Anna Duncan
- Apheresis Service, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Luisa Clucas
- Apheresis Service, The Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Clinical Haematology, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Jacqueline Fleming
- Children's Cancer Centre, The Royal Children's Hospital, Melbourne, Victoria, Australia
- Apheresis Service, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - David Hughes
- Children's Cancer Centre, The Royal Children's Hospital, Melbourne, Victoria, Australia
- Apheresis Service, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Anthea Greenway
- Apheresis Service, The Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Clinical Haematology, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - David Metz
- Apheresis Service, The Royal Children's Hospital, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Nephrology, The Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Paediatrics, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
4
|
Zikou X, Vaia D, Vasiliki P, Panagiotis C, Stavros A. Use of Therapeutic Apheresis methods in ICU. Transfus Apher Sci 2024; 63:103853. [PMID: 38049358 DOI: 10.1016/j.transci.2023.103853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
Apheresis is a modern medical approach in which plasma or cellular components are separated from the whole blood. Apheresis can be either diagnostic or therapeutic. Diagnostic apheresis is typically applied in hematology and cancer research. Therapeutic Apheresis (TA) includes a broad spectrum of extracorporeal treatments applied in various medical specialties, including Intensive Care Unit (ICU). Considering the complexity of the pathophysiologic characteristics of various clinical entities and in particular sepsis, apheresis methods are becoming increasingly applicable. Therapeutic Plasma Exchange (TPE) is the most common used method in ICU. It is considered as first line therapy for Thrombotic Thrombocytopenic Purpura (TTP) and Guillain Barre Syndrome, while the current data for sepsis are scarce. Over the last decades, technologic evolution has led to increasing application of new and more selective methods based on adsorptive techniques. In this review we will describe the current data of characteristics of different techniques, safety and clinical impact of apheresis methods used in ICUs.
Collapse
|
5
|
Mina-Osorio P, Tran MH, Habib AA. Therapeutic Plasma Exchange Versus FcRn Inhibition in Autoimmune Disease. Transfus Med Rev 2024; 38:150767. [PMID: 37867088 DOI: 10.1016/j.tmrv.2023.150767] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 10/24/2023]
Abstract
Therapeutic plasma exchange (TPE or PLEX) is used in a broad range of autoimmune diseases, with the goal of removing autoantibodies from the circulation. A newer approach for the selective removal of immunoglobulin G (IgG) antibodies is the use of therapeutic molecules targeting the neonatal Fc receptor (FcRn). FcRn regulates IgG recycling, and its inhibition results in a marked decrease in circulating autoantibodies of the IgG subtype. The difference between FcRn inhibition and PLEX is often questioned. With anti-FcRn monoclonal antibodies (mAbs) and fragments only recently entering this space, limited data are available regarding long-term efficacy and safety. However, the biology of FcRn is well understood, and mounting evidence regarding the efficacy, safety, and potential differences among compounds in development is available, allowing us to compare against nonselective plasma protein depletion methods such as PLEX. FcRn inhibitors may have distinct advantages and disadvantages over PLEX in certain scenarios. Use of PLEX is preferred over FcRn inhibition where removal of antibodies other than IgG or when concomitant repletion of missing plasma proteins is needed for therapeutic benefit. Also, FcRn targeting has not yet been studied for use in acute flares or crisis states of IgG-mediated diseases. Compared with PLEX, FcRn inhibition is associated with less invasive access requirements, more specific removal of IgG versus other immunoglobulins without a broad impact on circulating proteins, and any impacts on other therapeutic drug levels are restricted to other mAbs. In addition, the degree of IgG reduction is similar with FcRn inhibitors compared with that afforded by PLEX. Here we describe the scientific literature regarding the use of PLEX and FcRn inhibitors in autoimmune diseases and provide an expert discussion around the potential benefits of these options in varying clinical conditions and scenarios.
Collapse
Affiliation(s)
| | - Minh-Ha Tran
- Department of Pathology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Ali A Habib
- Department of Neurology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
6
|
Singh KR, Natarajan A, Pandey SS. Bioinspired Multifunctional Silver Nanoparticles for Optical Sensing Applications: A Sustainable Approach. ACS APPLIED BIO MATERIALS 2023; 6:4549-4571. [PMID: 37852204 DOI: 10.1021/acsabm.3c00669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Silver nanoparticles developed via biosynthesis are the most fascinating nanosized particles and encompassed with excellent physicochemical properties. The bioinspired nanoparticles with different shapes and sizes have attracted huge attention due to their stability, low cost, environmental friendliness, and use of less hazardous chemicals. This is an ideal method for synthesizing a range of nanosized metal particles from plants and biomolecules. Optical biosensors are progressively being fabricated for the attainment of sustainability by using opportunities offered by nanotechnology. This review focuses mainly on tuning the optical properties of the metal nanoparticles for optical sensing to explore the importance and applications of bioinspired silver nanoparticles. Further, this review deliberates the role of bioinspired silver nanoparticles (Ag NPs) in biomedical, agricultural, environmental, and energy applications. Profound insight into the antimicrobial properties of these nanoparticles is also appreciated. Tailor-made bioinspired nanoparticles with effectuating characteristics can unsurprisingly target tumor cells and distribute enwrapped payloads intensively. Existing challenges and prospects of bioinspired Ag NPs are also summarized. This review is expected to deliver perceptions about the progress of the next generation of bioinspired Ag NPs and their outstanding performances in various fields by promoting sustainable practices for fabricating optical sensing devices.
Collapse
Affiliation(s)
- Kshitij Rb Singh
- Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, 2-4 Hibikino, Wakamatsu, Kitakyushu 808-0196, Japan
| | - Arunadevi Natarajan
- Department of Chemistry, PSGR Krishnammal College for Women, Coimbatore, Tamil Nadu 641004, India
| | - Shyam S Pandey
- Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, 2-4 Hibikino, Wakamatsu, Kitakyushu 808-0196, Japan
| |
Collapse
|
7
|
Asensi Cantó P, Sanz Caballer J, Solves Alcaína P, de la Rubia Comos J, Gómez Seguí I. Extracorporeal Photopheresis in Graft-versus-Host Disease. Transplant Cell Ther 2023; 29:556-566. [PMID: 37419324 DOI: 10.1016/j.jtct.2023.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/09/2023]
Abstract
Graft-versus-host disease (GVHD) is a major cause of mortality and morbidity following allogeneic hematopoietic stem cell transplantation. Extracorporeal photopheresis (ECP), which exposes mononuclear cells to ultraviolet A irradiation in the presence of a photosensitizing agent, has shown efficacy in the treatment of GVHD. Recent observations in molecular and cell biology have revealed the mechanisms by which ECP can reverse GVHD, including lymphocyte apoptosis, differentiation of dendritic cells from circulating monocytes, and modification of the cytokine profile and T cell subpopulations. Technical innovations have made ECP accessible to a broader range of patients; however, logistical constraints may limit its use. In this review, we scrutinize the development of ECP from its origins to recent insights into the biology underlying ECP efficacy. We also review practical aspects that may complicate successful ECP treatment. Finally, we analyze how these theoretical concepts translate into clinical practice, summarizing the published experiences of leading research groups worldwide.
Collapse
Affiliation(s)
- Pedro Asensi Cantó
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain.
| | - Jaime Sanz Caballer
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Pilar Solves Alcaína
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain; CIBERONC, Instituto Carlos III, Madrid, Spain
| | - Javier de la Rubia Comos
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain; School of Medicine and Dentistry, Catholic University of Valencia, Valencia, Spain
| | - Inés Gómez Seguí
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain; CIBERONC, Instituto Carlos III, Madrid, Spain
| |
Collapse
|
8
|
Jalal Eldin A, Lyonga A, Ohiokpehai B, Rizwan M, Musa A. COVID-19, Hypertriglyceridemia, and Acute Pancreatitis: A Case Report and Clinical Considerations. Cureus 2023; 15:e35431. [PMID: 36994303 PMCID: PMC10040487 DOI: 10.7759/cureus.35431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2023] [Indexed: 02/27/2023] Open
Abstract
Acute pancreatitis (AP) is a serious condition that can result in numerous negative outcomes including death. The underlying causes of AP are varied, with both COVID-19 and hypertriglyceridemia being documented in the medical literature. Here, we present the case of a young man with a pre-existing diagnosis of prediabetes and class 1 obesity who developed severe hypertriglyceridemia, AP, and mild diabetic ketoacidosis while concurrently infected with COVID-19. It is crucial for healthcare providers to be vigilant in recognizing the potential complications associated with COVID-19, regardless of whether the patient has received a vaccination.
Collapse
|
9
|
Aksoy BA. Apheresis in pediatric patients: Current differences and difficulties. Transfus Apher Sci 2023; 62:103679. [PMID: 36870906 DOI: 10.1016/j.transci.2023.103679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Affiliation(s)
- Başak Adaklı Aksoy
- Altinbas University, Medical Park Bahcelievler Hospital, Pediatric Hematology, Oncology and Stem Cell Transplantation Unit, Turkey.
| |
Collapse
|
10
|
Kosmadakis G. Rheopheresis: A narrative review. Int J Artif Organs 2022; 45:445-454. [PMID: 35389284 DOI: 10.1177/03913988221086597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Rheopheresis is an extracorporal selective double-filtration procedure with an initial separation of plasma from the whole blood and a further filtration of the plasma through a second filter in order to eliminate a certain number of high molecular weight proteins such LDL, Lp(a), fibrinogen, α2-macroglobulin, Factor von Willebrand, and IgM Immunoglobulin. METHODS In this narrative review we discuss the available data on the effects of Rheopheresis in various clinical conditions. RESULTS Rheopheresis is considerd to exert a rapid effect on clinical conditions associated with seriously affected microcirculation and rheologic parameters such as, the dry age-related macular degeneration (AMD), sudden sensorineural hearing loss (SSHS), peripheral artery disease (PAD), calciphylaxis, systemic sclerosis and diabetic foot. CONCLUSIONS Rheopheresis is a promising technique for conditions associated with affected microcirculatory rheologic parameters.
Collapse
|
11
|
Ahaneku H, Gupta R, Anusim N, Umeh CA, Anderson J, Jaiyesimi I. Leucocytoclastic Vasculitis, Cryoglobulinemia, or Plasma Cell Leukemia: A Diagnostic Conundrum. Cureus 2021; 13:e16832. [PMID: 34513422 PMCID: PMC8409693 DOI: 10.7759/cureus.16832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2021] [Indexed: 11/06/2022] Open
Abstract
Plasma cell leukemia is rare and could be life-threatening. Even rarer and equally life-threatening is cryoglobulinemia. Both of them occurring together paints a grim clinical picture. We present the case of a 63-year-old male with plasma cell leukemia complicated by cryoglobulinemia with skin lesions. The report briefly reviews the clinical and diagnostic characteristics of plasma cell leukemia and well as available treatment options. It also highlights the need to consider non-chemotherapy-based regimens and clinical trials in the care of plasma cell leukemia patients.
Collapse
Affiliation(s)
| | - Ruby Gupta
- Hematology and Oncology, Beaumont Health, Royal Oak, USA
| | | | | | | | | |
Collapse
|
12
|
Drexler B, Buser A, Infanti L, Stehle G, Halter J, Holbro A. Extracorporeal Photopheresis in Graft-versus-Host Disease. Transfus Med Hemother 2020; 47:214-225. [PMID: 32595426 DOI: 10.1159/000508169] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/21/2020] [Indexed: 12/16/2022] Open
Abstract
Background and Summary Extracorporeal photopheresis (ECP) is a leukapheresis-based procedure used in the therapy of acute and chronic graft-versus-host disease (aGvHD, cGvHD) and other diseases. Based on the substantial efficacy and the excellent safety profile in the absence of immunosuppression ECP has established itself as a major treatment form for steroid-refractory GvHD. Here we review the current literature on ECP as a treatment option for patients with aGvHD as well as cGvHD. Key Messages ECP is a well-established second-line therapy for cGvHD. Its role in the treatment of aGvHD is less clear but also points towards an effective second-line therapy option. In the future ECP could play a role in the prevention of GvHD. More experimental and randomized controlled trials are needed to define the best patient selection criteria, settings, and therapy regimens for GvHD.
Collapse
Affiliation(s)
- Beatrice Drexler
- Division of Hematology, University Hospital Basel, Basel, Switzerland.,Blood Transfusion Center, Swiss Red Cross, Basel, Switzerland
| | - Andreas Buser
- Division of Hematology, University Hospital Basel, Basel, Switzerland.,Blood Transfusion Center, Swiss Red Cross, Basel, Switzerland
| | - Laura Infanti
- Division of Hematology, University Hospital Basel, Basel, Switzerland.,Blood Transfusion Center, Swiss Red Cross, Basel, Switzerland
| | - Gregor Stehle
- Division of Hematology, University Hospital Basel, Basel, Switzerland.,Blood Transfusion Center, Swiss Red Cross, Basel, Switzerland
| | - Joerg Halter
- Division of Hematology, University Hospital Basel, Basel, Switzerland
| | - Andreas Holbro
- Division of Hematology, University Hospital Basel, Basel, Switzerland.,Blood Transfusion Center, Swiss Red Cross, Basel, Switzerland
| |
Collapse
|
13
|
Mansouri Taleghani B, Buser A. Therapeutic Apheresis. Transfus Med Hemother 2019; 46:391-393. [PMID: 31933568 PMCID: PMC6944895 DOI: 10.1159/000504143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 10/15/2019] [Indexed: 01/04/2023] Open
Affiliation(s)
- Behrouz Mansouri Taleghani
- University Clinic of Hematology and Central Hematology Laboratory, Division of Transfusion Medicine, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Andreas Buser
- Regional Blood Transfusion Service Swiss Red Cross Basel, and Department of Hematology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|