Alloantigen Presenting Capacity, T Cell Alloreactivity and NK Function of G-CSF-Mobilized Peripheral Blood Cells

Total Page:16

File Type:pdf, Size:1020Kb

Alloantigen Presenting Capacity, T Cell Alloreactivity and NK Function of G-CSF-Mobilized Peripheral Blood Cells Bone Marrow Transplantation, (1998) 22, 631–637 1998 Stockton Press All rights reserved 0268–3369/98 $12.00 http://www.stockton-press.co.uk/bmt Alloantigen presenting capacity, T cell alloreactivity and NK function of G-CSF-mobilized peripheral blood cells D Rondelli1, D Raspadori1, C Anasetti2, G Bandini1,FRe1, M Arpinati1, M Stanzani1, A Morelli3, C Baccini3, A Zaccaria4, RM Lemoli1 and S Tura1 1Institute of Hematology and Medical Oncology ‘Sera`gnoli’, University of Bologna; 3Toxicology and Pharmacology Unit, 4Division of Hematology, ‘S Maria delle Croci’ Hospital, Ravenna, Italy; and 2Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA Summary: Hematopoietic stem cells mobilized into the peripheral blood with granulocyte colony-stimulating factor (G-CSF) In this study we addressed whether the proportion and or granulocyte–macrophage colony-stimulating factor the function of antigen presenting cells (APC), T and NK (GM-CSF) have been demonstrated to be as effective as lymphocytes are modified in the apheresis product of six marrow stem cells in reconstituting all blood lineages after healthy donors who received a stem cell mobilizing treat- allogeneic transplantation.1–3 However, leukaphereses col- ment with glycosylated G-CSF at 10 ␮g/kg/day × 5 days lected from normal donors receiving G-CSF contain large s.c. Flow cytometry analysis showed comparable percent- numbers of T cells,4 potentially increasing the risk of acute ages of HLA-DR+,CD19+, CD86+, CD80+ and CD1a+ cells graft-versus-host disease (aGVHD). Nevertheless, previous in preG-CSF-peripheral blood mononuclear cells (preG- reports demonstrated that the incidence and the severity of PBMC) and after mobilization in G-PBMC, whereas the aGVHD after allogeneic transplantation with G-CSF mobil- proportion of CD14+ monocytes significantly increased in ized peripheral blood mononuclear cells (G-PBMC) are not -G-PBMC (3 ؎ 1% vs 17 ؎ 8%, P = 0.003). Analysis of higher than after allogeneic transplantation with steady lymphocyte subsets in preG-PBMC and G-PBMC showed state marrow cells.3,5–7 similar proportions of CD3+, CD4+, CD8+ and CD28+ T Experimental studies addressed whether G-CSF may cells, but a significantly lower percentage of CD16+ modulate alloimmune responses by directly modifying T vs 4 ؎ 1%, P = 0.01), CD56+ (15 ؎ 6% vs cell function or by inducing cell-mediated suppression of 7% ؎ 11) -P = 0.008), CD57+ (16 ؎ 9% vs 5 ؎ 2%, T cell alloreactivity. In particular, it was previously demon ,2% ؎ 5 P = 0.04), CD25+ (19 ؎ 2% vs 9 ؎ 6%, p = 0.009) and strated that aGVHD is more likely due to the activation CD122+ (5 ؎ 2% vs 2 ؎ 1%, P = 0.05) cells in G-PBMC. of type-1 donor cells (secreting IL-2 and IFN-␥)8 and that Unfractionated preG-PBMC and G-PBMC were pretreatment of mice with G-CSF polarizes donor T cells irradiated and tested in primary mixed leukocyte culture toward type-2 cytokine production (IL-4 and IL-10), thus (MLC) with two HLA-incompatible responders and reducing aGVHD.9,10 Mielcarek et al11 showed that G-CSF induced efficient alloresponses in four of six cases, whereas mobilization increases the frequency of CD14+ monocytes G-PBMC stimulated poorly in the remaining two cases. in the blood of normal donors and that a large number of Also, in allo-MLC with irradiated G-PBMC we detected CD14+ cells in the leukapheresis could suppress T cells lower amounts of IFN-␥ (P = 0.04) and of IL-2 (P = 0.06) mainly in a non-contact way, suggesting that this effect than in allo-MLC with preG-PBMC. Furthermore, freshly may contribute to a lower incidence of aGVHD after G- isolated preG-PBMC and G-PBMC from each donor PBMC allogeneic transplantation.11 Also, we recently dem- exerted comparable allogeneic responses to HLA-incom- onstrated that G-PBMC induce a lower allogeneic CD4+ T patible irradiated mononuclear cells in all cases. However, cell response than purified CD34+ blood cells in primary G-PBMC showed no NK activity against K562 target cells MLC.12 at any effector:target ratio tested. These data suggest that Cytolytic cell precursors are present in G-PBMC,13 how- normal G-PBMC may prevent Th1 alloresponses, main- ever previous data showed that G-CSF could downregulate tain efficient alloreactivity to HLA mismatched antigens the NK activity by inhibiting TNF-␣ and IFN-␥ production and have impaired NK activity. by NK cells.14 Nevertheless, Neubauer et al15 reported a Keywords: peripheral blood stem cells; APC; IFN-␥;T higher proportion of CD8+, CD56+ and p75+ cells and a cell; NK activity more efficient IL-2-induced lymphokine-activated killer (LAK) activity in G-PBMC of patients undergoing an auto- logous transplantation than in normal controls. In the present study we analyzed the proportion of anti- gen presenting cells (APC), T and NK cells, as well as the immunological functions of these cell populations in Correspondence: Dr D Rondelli, Institute of Hematology and Medical Oncology ‘Sera`gnoli’, University of Bologna, Policlinico S. Orsola, via the blood of healthy donors before and after treatment Massarenti, 9, 40138 Bologna, Italy with G-CSF. Our results suggest that unfractionated G- Received 4 March 1998; accepted 19 June 1998 PBMC may prevent HLA-incompatible type-1 T cell allo- Alloimmune effects of mobilized PBMC D Rondelli et al 632 responses. In addition, although maintaining the capacity Table 1 Expression of APC markers on mononuclear cells from heal- of responding to alloantigen in MLC, they do not exert thy donors before (PreG-PBMC) and after (G-PBMC) stem cell mobilizing NK activity. treatment with G-CSF PreG-PBMC (%) G-PBMC (%) P value Materials and methods HLA-DR 21 Ϯ 726Ϯ 9NS CD14 3 Ϯ 117Ϯ 8 0.003 Cell preparation CD86 5 Ϯ 15Ϯ 3NS CD80 Ͻ1 Ͻ1 Blood and apheresis samples were obtained with informed CD1a NEG NEG consent from healthy adult peripheral blood stem cell CD19 4 Ϯ 14Ϯ 2NS donors. Blood samples were collected before initiating stem Data represent the mean Ϯ s.d. of six separate cases. cell mobilization with glycosylated G-CSF (lenograstim; = = Ͻ Rhone-Poulenc Rorer, Milan, Italy) at a dose of NS not significant; NEG 0.5%. 10 ␮g/kg/day s.c. × 5–6 days and apheresis samples were obtained from the first apheresis on day 5 of treatment. Blood samples from HLA-DR incompatible healthy vol- ounteers were also collected. All samples were separated 18 h before harvest on day 6. The stimulation index (SI) by centrifugation over Ficoll/Hypaque (Nycomed Pharma, was calculated for each individual experiment as: Oslo, Norway). Light density cells were washed twice in = + RPMI medium enriched with 10% fetal calf serum (FCS) SI c.p.m. (T cell responders stimulators)/c.p.m. (T (Seromed Biochrome, Berlin, Germany). Freshly isolated cell responders). mononuclear cells were then used for cytofluorimetric and functional assays. NK activity Flow cytometry The NK assay was performed using preG-PBMC or G- PBMC and third-party mononuclear cells as effector cells Flow cytometric analysis was carried out on preG-PBMC and the K562 cell line as the target in a 4 h 51Cr release and G-PBMC by direct immunofluorescence. Human assay. Briefly, 1 × 106 target cells were incubated for 1 h monoclonal antibodies (mAbs) used in this study are the at 37°C with 100 ␮Ci Na51Cr, washed twice, mixed in trip- following: CD3 fluorescein isthyocianate (FITC), CD4 licate with effector cells at final effector:target (E:T) ratios FITC, CD8 FITC, CD16 FITC, CD56 phycoerythrin (PE), of 25:1, 12:1 and 6:1 in round-bottomed 96-well plates for CD57 FITC, CD80 (anti-B7–1) PE, CD25 (anti-p55 IL-2 4 h at 37°C, and then centrifuged at 400 g for 10 min. An receptor) FITC, CD28 PE, CD14 FITC and CD19 FITC aliquot (100 ␮l) of the supernatant was collected and coun- (Becton Dickinson, San Jose´, CA, USA); CD1a PE and ted in a gamma scintillation counter. The percentage CD86 (anti-B7–2) PE (PharMingen, San Diego, CA, USA); of 51Cr release was expressed as: CD122 (anti-p75 IL-2 receptor) FITC (Janssen, Brussels, Belgium). Isotype controls IgG1 FITC and PE and IgG2a % lysis = (E − S)/(M − S) × 100 PE are from Becton Dickinson and IgG2b PE is from PharMingen. Data acquisition and analysis were assessed (where E = mean c.p.m. release in the presence of effector by Lysis II software (Becton Dickinson). cells; S = mean c.p.m. spontaneously released by target cells incubated with medium alone; M = mean c.p.m. of resuspended target cells). Primary MLC Freshly isolated preG-PBMC or G-PBMC were either irradiated with 3000 cGy and used as stimulators, or used Cytokine assays as responders in primary allogeneic mixed lymphocyte cul- tures (MLC). Each donor preG- or G-PBMC were tested Levels of IL-2, IFN-␥, IL-4 and IL-10 in MLC supernatants with the same two third-party mononuclear cells from two were determined by commercially available enzyme-linked HLA incompatible healthy volounteers. In selected experi- immunosorbent assay (ELISA) kits (IL-2: Immunotech, ments over 90% T cells were isolated from preG- and G- Marseille, France; IFN-␥, IL-4 and IL-10: Endogen, Bos- PBMC and from third party mononuclear cells by rosetting ton, MA, USA) and a standard curve was generated to with sheep red blood cells, as previously described.16 determine the cytokine concentration in the sample. Detec- Irradiated stimulator and responder cells were resus- tion limits of the assays were 5 pg/ml for IL-2, Ͻ2 pg/ml pended in medium containing RPMI-1640, 25 mm Hepes, for IFN-␥, Ͻ2 pg/ml for IL-4 and Ͻ3 pg/ml for IL-10.
Recommended publications
  • Factors Affecting Mobilization of Peripheral Blood Progenitor Cells in Patients with Lymphoma’
    Vol. 4, 311-316, February 1998 Clinical Cancer Research 311 Factors Affecting Mobilization of Peripheral Blood Progenitor Cells in Patients with Lymphoma’ Craig H. Moskowitz,2 Jill R. Glassman, (median, 13 versus 22 days; P 0.06). Patients who received 1l cycles of chemotherapy prior to PBPC mobilization David Wuest, Peter Maslak, Lilian Reich, tended to have delayed platelet recovery to >20,090/&l and Anthony Gucciardo, Nancy Coady-Lyons, to require more platelet transfusions than less extensively Andrew D. Zelenetz, and Stephen D. Nimer pretreated patients (median, 13.5 versus 23.5 days; P 0.15; Division of Hematologic Oncology, Department of Medicine median number of platelet transfusion episodes, 13 versus 9; [C. H. M., D. W., P. M., L. R., A. G., N. C-L., A. D. Z., S. D. N.] and P = 0.17). Department of Biostatistics [J. R. G.], Memorial Sloan-Kettering Cancer Center, New York, New York 10021 These data suggest that current strategies to mobilize PBPCs may be suboptimal in patients who have received either stem cell-toxic chemotherapy or 11 cycles of chem- ABSTRACT otherapy prior to PBPC mobilization. Alternative ap- The objective of this study was to identify factors asso- proaches, such as ex vivo expansion or the use of other ciated with poor mobilization of peripheral blood progenitor growth factors in addition to G-CSE, may improve mobili- cells (PBPCs) or delayed platelet engraftment after high- zation of progenitor cells for PBPC transplantation. dose therapy and autologous stem cell transplantation in patients with lymphoma. INTRODUCTION Fifty-eight patients with Hodgkin’s disease or non- The use of high-dose chemoradiotherapy supported by Hodgkin’s lymphoma underwent PBPC transplantation as cryopreserved autologous hematopoietic progenitor cells is ef- the “best available therapy” at Memorial Sloan-Kettering fective in treating relapsed HD3 and NHL; a high complete Cancer Center (New York, NY) between 1993 and 1995.
    [Show full text]
  • 45 Part 606—Current Good Man- Ufacturing Practice
    Food and Drug Administration, HHS Pt. 606 a presentation. The presiding officer ucts approved under § 601.91, the re- may, as a matter of discretion, permit strictions would no longer apply when questions to be submitted to the pre- FDA determines that safe use of the bi- siding officer for response by a person ological product can be ensured making a presentation. through appropriate labeling. FDA also (f) Judicial review. The Commissioner retains the discretion to remove spe- of Food and Drugs’ decision constitutes cific postapproval requirements upon final agency action from which the ap- review of a petition submitted by the plicant may petition for judicial re- sponsor in accordance with § 10.30 of view. Before requesting an order from a this chapter. court for a stay of action pending re- view, an applicant must first submit a PART 606—CURRENT GOOD MAN- petition for a stay of action under § 10.35 of this chapter. UFACTURING PRACTICE FOR BLOOD AND BLOOD COMPO- [67 FR 37996, May 31, 2002, as amended at 70 NENTS FR 14984, Mar. 24, 2005] § 601.93 Postmarketing safety report- Subpart A—General Provisions ing. Sec. Biological products approved under 606.3 Definitions. this subpart are subject to the post- marketing recordkeeping and safety Subpart B—Organization and Personnel reporting applicable to all approved bi- ological products. 606.20 Personnel. § 601.94 Promotional materials. Subpart C—Plant and Facilities For biological products being consid- 606.40 Facilities. ered for approval under this subpart, unless otherwise informed by the agen- Subpart D—Equipment cy, applicants must submit to the agency for consideration during the 606.60 Equipment.
    [Show full text]
  • 0985.03CC Non-Mobilized Donor Consent
    Fred Hutchinson Cancer Research Center Consent to take part in a research study: 985.03CC – Non-Mobilized Donor Consent to Participate as a Donor of Non-Mobilized Peripheral Blood Mononuclear Cells for Laboratory Research and Process Development Studies. Principal Investigator: Derek Stirewalt, MD, Member, FHCRC, (206 667-5386) Investigators: Michael Linenberger, M.D., FACP Professor, Division of Hematology, UW, Robert & Phyllis Henigson Endowed Chair, Program Director, Hematology/Oncology Fellowship Medical Director, Apheresis and Cellular Therapy, SCCA, Member, FHCRC, Associate Professor of Medicine, UW, (206 667-5021); Laura Connelly Smith, Assistant Member, FHCRC, Assistant Professor, UW (206 606 -6938) Research Coordinator: Aubrey McMillan, (206) 667-3539 Emergency Phone (24 hours) UW Transplant unit 8NE: (206) 598-8902 UW Nocturnist on call provider (7:00 PM -7:00 AM): (206) 598-1062 Donor recruitment and participation: 206-667-5318 Important things to know about this study. You are invited to participate in a research study. The purpose of this research is to collect blood cells by a procedure called “leukapheresis”. Leukapheresis is a procedure that allows for a greater collection of peripheral blood cells than can be obtained by standard blood donation. Our purpose is to collect both small blood draws and larger samples of peripheral blood cells for laboratory research studies. This research may involve analysis of your genetic material called DNA and RNA. The studies may also examine other components of the cell like proteins. These analyses can be used to better understand cancer and other diseases. People who agree to join the study will be asked to attend 2 appointments over up to 30 days.
    [Show full text]
  • Leukapheresis Protocol for Nonhuman Primates Weighing Less Than 10 Kg
    Journal of the American Association for Laboratory Animal Science Vol 52, No 1 Copyright 2013 January 2013 by the American Association for Laboratory Animal Science Pages 70–77 Leukapheresis Protocol for Nonhuman Primates Weighing Less than 10 Kg Vimukthi Pathiraja, Abraham J Matar, Ashley Gusha, Christene A Huang, and Raimon Duran-Struuck* Leukapheresis is a common procedure for hematopoietic cell transplantation in adults. The main challenge in applying this procedure to human infants and small monkeys is the large extracorporeal blood volume (165 mL on average) necessary for priming the apheresis machine. This volume represents greater than 50% of the total circulating blood volume of a human neonate or small monkey. In this report, we document a safe leukapheresis protocol developed for rhesus macaques (3.9 to 8.7 kg). To avoid sensitizing donor animals undergoing leukapheresis to third-party blood products, autologous blood col- lected during the weeks prior to leukapheresis was used to volume-expand the same donor while priming the machine with saline on the day of leukapheresis. During the procedures, blood pressure was controlled by monitoring the inlet volume, and critical-care support was provided by the anesthesia team. Electrolytes and hemogram parameters were monitored intermit- tently. Overall, our research subjects underwent effective 4- to 6-h leukapheresis. A total of 9 leukapheresis procedures were performed, which yielded 1 × 109 to 6 × 109 peripheral blood mononuclear cells containing 1.1 to 5.1 × 106 CD34+ cells (assessed in 4 of 9 macaques) in a volume of 30 to 85 mL. All macaques showed decreases in Hct and platelet counts.
    [Show full text]
  • Current Challenges in Providing Good Leukapheresis Products for Manufacturing of CAR-T Cells for Patients with Relapsed/Refractory NHL Or ALL
    cells Article Current Challenges in Providing Good Leukapheresis Products for Manufacturing of CAR-T Cells for Patients with Relapsed/Refractory NHL or ALL Felix Korell 1,*, Sascha Laier 2, Sandra Sauer 1, Kaya Veelken 1, Hannah Hennemann 1, Maria-Luisa Schubert 1, Tim Sauer 1, Petra Pavel 2, Carsten Mueller-Tidow 1, Peter Dreger 1, Michael Schmitt 1 and Anita Schmitt 1 1 Department of Internal Medicine V, University Hospital Heidelberg, 69120 Heidelberg, Germany; [email protected] (S.S.); [email protected] (K.V.); [email protected] (H.H.); [email protected] (M.-L.S.); [email protected] (T.S.); [email protected] (C.M.-T.); [email protected] (P.D.); [email protected] (M.S.); [email protected] (A.S.) 2 Institute of Clinical Transfusion Medicine and Cell Therapy (IKTZ), 89081 Heidelberg, Germany; [email protected] (S.L.); [email protected] (P.P.) * Correspondence: [email protected] Received: 9 April 2020; Accepted: 13 May 2020; Published: 15 May 2020 Abstract: Background: T lymphocyte collection through leukapheresis is an essential step for chimeric antigen receptor T (CAR-T) cell therapy. Timing of apheresis is challenging in heavily pretreated patients who suffer from rapid progressive disease and receive T cell impairing medication. Methods: A total of 75 unstimulated leukaphereses were analyzed including 45 aphereses in patients and 30 in healthy donors. Thereof, 41 adult patients with Non-Hodgkin’s lymphoma (85%) or acute lymphoblastic leukemia (15%) underwent leukapheresis for CAR-T cell production.
    [Show full text]
  • Transfusion Reactions 7
    HEMATOLOGY Immunohematology, transfusion medicine and bone marrow transplantation Institute of Pathological Physiology First Facultry of Medicine, Charles University in Prague http://patf.lf1.cuni.cz Questions and Comments: MUDr. Pavel Klener, Ph.D., [email protected] Presentation in points 1. Imunohematology 2. AB0 blood group system 3. Rh blood group system 4. Hemolytic disease of the newborn and neonatal alloimmune thrombocytopenia 5. Pre-transfusion examinations 6. Transfusion reactions 7. Transfusion medicine and hemapheresis 8. HLA system 9. Stem cell/ bone marrow transplantation Origins of immunohematology and transfusion medicine Imunohematology as a branch of medicine developed hand in hand with the origins of transfusion therapy. It focuses on concepts and questions associated with transfusion therapy, immunisation (as a result of transfusion therapy and pregnancy) and organ transplantation. In 1665, an English physiologist, Richard Lower, successfully performed the first animal- to-animal blood transfusion that kept ex-sanguinated dogs alive by transfusion of blood from other dogs. In 1667, Jean Bapiste Denys, transfused blood from the carotid artery of a lamb into the vein of a young man, which at first seemed successful. However, after the third transfusion of lamb’s blood the man suffered a reaction and died. Due to the many disastrous consequences resulting from blood transfusion, transfusions were prohibited from 1667 to 1818- when James Blundell of England successfully transfused human blood to women suffering from hemorrhage at childbirth. Revolution in 1900 In 1900 Karl Landsteiner discovered the AB0 blood groups. This landmark event initiated the era of scientific – based transfusion therapy and was the foundation of immunohematology as a science.
    [Show full text]
  • How Do I Perform Whole Blood Exchange?
    HOW DO I? How do I perform whole blood exchange? † † † David Ming-Hung Lin ,1, Joanne Becker,2, YanYun Wu,1 and Laura Cooling3, here are a number of clinical scenarios in which crossmatched against plasma or adsorbed plasma, if avail- simultaneous exchange of the patient’s plasma able. Because preparation of reconstituted WB is considered and red blood cells (RBCs) are indicated. Some of an open system with a 24-hour outdate, we prepared only these clinical indications for therapeutic whole one reconstituted WB unit at a time (i.e., after dispensing Tblood exchange (WBEx) include hemolytic disease of the one unit, the next unit was prepared). WBEx was performed newborn, severe autoimmune hemolytic anemia (AIHA), by physically removing WB with a syringe, followed by infu- – babesiosis, sickle cell disease, and hyperleukocytosis.1 4 sion of reconstituted WB. In adult patients, this involved WBEx has been performed with manual, semiautomated, removal of 200 mL (50 mL per draw with 60-mL syringes) and fully automated methods using replacements fluids over 10 minutes through a triple-lumen central venous (i.e., RBCs with plasma, RBCs with 5% albumin, and whole catheter, followed by infusion of WB over 1 to 1.5 hours. blood [WB]) reconstituted to a prespecified target hemato- Overall, it required 20 hours to complete a manual WBEx crit (Hct); however, methodologies vary widely and each exchange with 12 units of WB as replacement. For pediatric carry advantages and disadvantages. Here, we describe our patients, a similar process was followed using a central or combined experiences in performing WBEx, including clini- femoral venous catheter (5 mL/kg per draw).
    [Show full text]
  • Leukopak PBMC Sample Processing for Preparing Quality Control Material to Support Proficiency Testing Programs
    JIM-11874; No of Pages 8 Journal of Immunological Methods xxx (2014) xxx–xxx Contents lists available at ScienceDirect Journal of Immunological Methods journal homepage: www.elsevier.com/locate/jim Leukopak PBMC sample processing for preparing quality control material to support proficiency testing programs Ambrosia Garcia a, Sarah Keinonen a, Ana M. Sanchez a, Guido Ferrari a,d,e, Thomas N. Denny a,b,e, M. Anthony Moody a,c,⁎ a Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA b Departments of Medicine, Duke University Medical Center, Durham, NC, USA c Departments of Pediatrics, Duke University Medical Center, Durham, NC, USA d Departments of Surgery, Duke University Medical Center, Durham, NC, USA e Duke Global Health Institute, Duke University Medical Center, Durham, NC, USA article info abstract Article history: External proficiency testing programs designed to evaluate the performance of end-point Received 19 November 2013 laboratories involved in vaccine and therapeutic clinical trials form an important part of Received in revised form 14 May 2014 clinical trial quality assurance. Good clinical laboratory practice (GCLP) guidelines recommend Accepted 31 May 2014 both assay validation and proficiency testing for assays being used in clinical trials, and such Available online xxxx testing is facilitated by the availability of large numbers of well-characterized test samples. These samples can be distributed to laboratories participating in these programs and allow Keywords: monitoring of laboratory performance over time and among participating sites when results Proficiency testing are obtained with samples derived from a large master set. The leukapheresis procedure Leukocytes provides an ideal way to collect samples from participants that can meet the required number Leukapheresis of cells to support these activities.
    [Show full text]
  • Leukapheresis: an Overview
    IMPORTANCE OF IMMUNE CELLS IN RESEARCH Your immune cell donation may assist researchers to advance medical treatments, lead to a scientific breakthrough or even develop cures for certain diseases or disorders. Immunologists worldwide have a need for high quality immune cells and cellular subsets, and this is where you come in! Researchers will use donations like yours in an attempt to find novel ways to prime the immune system against cancers and other diseases such as HIV, auto-immune disorders, multiple sclerosis, Crohn’s, CONTACT US allergies and asthma. Immunotherapy treatments are especially effective against lung cancer, skin cancer and blood cancers such (267) 310-1400 LEUKAPHERESIS: as leukemia and lymphoma. [email protected] AN OVERVIEW biospecialty.com WHITE BLOOD CELL DONATION PROCESS Blood is made up of many components, specifically platelets, red blood cells, plasma and white blood cells, which can all be Biological Specialty Company individually collected. FDA-registered donor center operating under Investigational Review Board (IRB) approval 2165 N. Line Street | Colmar, PA 18915 1401 W. Green Street | Allentown, PA 18102 22 South 4th Street | Reading, PA 19602 LEUKAPHERESIS, noun leu· ka· phe· re· sis | \ ˌlü-kə-fə-ˈrē-səs\ The process of leukapheresis allows for peripheral blood Derived from the latin roots mononuclear cells (PBMCs) to be collected from the blood while returning red blood cells, platelets and plasma back to the donor. An of leuk (meaning white) and apheresis machine is used to draw blood through a vein in one arm, aphaeresis (translates to take remove the needed component and return the unneeded parts back away).
    [Show full text]
  • Peripheral Blood Stem Cell Transplantation As an Alternative to Bone Marrow Transplantation: an Overview
    Henry Ford Hospital Medical Journal Volume 39 Number 2 Article 10 6-1991 Peripheral Blood Stem Cell Transplantation as an Alternative to Bone Marrow Transplantation: An Overview Nalini Janakiraman Follow this and additional works at: https://scholarlycommons.henryford.com/hfhmedjournal Part of the Life Sciences Commons, Medical Specialties Commons, and the Public Health Commons Recommended Citation Janakiraman, Nalini (1991) "Peripheral Blood Stem Cell Transplantation as an Alternative to Bone Marrow Transplantation: An Overview," Henry Ford Hospital Medical Journal : Vol. 39 : No. 2 , 103-107. Available at: https://scholarlycommons.henryford.com/hfhmedjournal/vol39/iss2/10 This Article is brought to you for free and open access by Henry Ford Health System Scholarly Commons. It has been accepted for inclusion in Henry Ford Hospital Medical Journal by an authorized editor of Henry Ford Health System Scholarly Commons. Peripheral Blood Stem Cell Transplantation as an Alternative to Bone Marrow Transplantation: An Overview Nalini Janakiraman, MD' Slem cells capable of resloring hemalopoiesis following lethal bone marrow injury circulate in Ihc blood of many animals, including humans. When collected ihrough leukapheresis and reinfused following high-dose chenwiherapy, slem cells offer a Irealmenl optiim not currently open lo some patients who are unable lo undergo autologous hone marrow transplantation because of tumor involvement in the pelvis, prior pelvic radiation, or intolerance to general anesthesia. After stem cell infusion, hematologic and immunologic recovery are rapid in comparison lo that after autologous hone marrow reinfusion: however, in some cases platelet engraftment is slower. There is some evidence lhal tumor conlaminalion in the peripheral hlood is much less than in the marrow.
    [Show full text]
  • Blood Transfusions an Overview
    Blood Transfusions: An Overview Jassin M. Jouria, MD Dr. Jassin M. Jouria is a medical doctor, professor of academic medicine, and medical author. He graduated from Ross University School of Medicine and has completed his clinical clerkship training in various teaching hospitals throughout New York, including King’s County Hospital Center and Brookdale Medical Center, among others. Dr. Jouria has passed all USMLE medical board exams, and has served as a test prep tutor and instructor for Kaplan. He has developed several medical courses and curricula for a variety of educational institutions. Dr. Jouria has also served on multiple levels in the academic field including faculty member and Department Chair. Dr. Jouria continues to serves as a Subject Matter Expert for several continuing education organizations covering multiple basic medical sciences. He has also developed several continuing medical education courses covering various topics in clinical medicine. Recently, Dr. Jouria has been contracted by the University of Miami/Jackson Memorial Hospital’s Department of Surgery to develop an e-module training series for trauma patient management. Dr. Jouria is currently authoring an academic textbook on Human Anatomy & Physiology. Abstract Being able to replace blood and blood components that a patient has lost or that his or her body is not able to produce is a vital part of providing excellent medical care. With the aid of donor blood, or even sometimes the patient’s own blood that has been previously banked, medical professionals have a safe strategy for treatment of patients who have lost blood to illness or injury. This course will explain the different types of blood products, the requirements for performing a transfusion, and how to recognize and respond to complications of the transfusion process.
    [Show full text]
  • Case Reports
    CASE REPORTS units that were group A, D+, E−, K−, and Fy(a−). Severe delayed hemolytic transfusion reaction Although anti-Le a was microscopically positive at poly - due to anti-Fy3 in a patient with sickle cell disease ethylene glycol (PEG) IgG, anti-Lea is not routinely undergoing red cell exchange prior to matched for at the study institution nor had been hematopoietic progenitor cell collection for gene matched for with transfusions at his primary medical therapy facility. After RCE, his Hb rose from 9.3 to 11.2 g/dL, and his HbS% decreased from 78% to 22% (HbA% of 72%). To our knowledge, most patients with sickle cell di- The following day, the patient was given a single 240 sease (SCD) who have undergone hematopoietic progen - µg/kg dose of plerixafor for HPC mobilization and started itor cell (HPC) collection for gene therapy have been con - leukapheresis about 3 hours afterward. He had no ditioned with a series of monthly simple or exchange adverse events and was discharged home after his leuka - transfusions. The rationale for this practice is to improve pheresis. safety and decrease bone marrow inflammation, poten - Five days after RCE, the patient developed throbbing + tially leading to higher CD34 mobilization and low back pain, subjective fevers, dark urine, and scleral decreased risk of adverse events during HPC collection. icterus, symptoms differing from his typical vaso-occlu - + Whether transfusion actually improves CD34 mobiliza - sive crises usually manifesting as right-sided flank/upper tion is not clear however, and we previously showed that back discomfort and priapism.
    [Show full text]