scholarly journals Antirelapse effect of pretransplant exposure to rabbit antithymocyte globulin

2019 ◽  
Vol 3 (9) ◽  
pp. 1394-1405 ◽  
Author(s):  
Rosy Dabas ◽  
Kareem Jamani ◽  
Shahbal B. Kangarloo ◽  
Poonam Dharmani-Khan ◽  
Tyler S. Williamson ◽  
...  

AbstractIt remains unknown why rabbit antithymocyte globulin (ATG; Thymoglobulin) has not affected relapse after hematopoietic cell transplantation (HCT) in randomized studies. We hypothesized that high pre-HCT ATG area under the curve (AUC) would be associated with a low incidence of relapse, whereas high post-HCT AUC would be associated with a high incidence of relapse. We measured serum levels of ATG capable of binding to mononuclear cells (MNCs), lymphocytes, T cells, CD4 T cells, or CD33 cells. We estimated pre- and post-HCT AUCs in 152 adult recipients of myeloablative conditioning and blood stem cells. High pre-HCT AUCs of MNC- and CD33 cell–binding ATG were associated with a low incidence of relapse and high relapse-free survival (RFS). There was a trend toward an association of high post-HCT AUC of lymphocyte-binding ATG with a high incidence of relapse and low RFS. High pre-HCT AUCs were also associated with faster engraftment and had no impact on graft-versus-host disease (GVHD) or fatal infections. High post-HCT AUCs were associated with a low risk of GVHD, seemed associated with an increased risk of fatal infections, and had no impact on engraftment. In conclusion, pre-HCT AUC seems to have a positive, whereas post-HCT AUC seems to have a negative, impact on relapse.

Author(s):  
Derek J Hanson ◽  
Hu Xie ◽  
Danielle M Zerr ◽  
Wendy M Leisenring ◽  
Keith R Jerome ◽  
...  

Abstract We sought to determine whether donor-derived human herpesvirus (HHV) 6B–specific CD4+ T-cell abundance is correlated with HHV-6B detection after allogeneic hematopoietic cell transplantation. We identified 33 patients who received HLA-matched, non–T-cell–depleted, myeloablative allogeneic hematopoietic cell transplantation and underwent weekly plasma polymerase chain reaction testing for HHV-6B for 100 days thereafter. We tested donor peripheral blood mononuclear cells for HHV-6B–specific CD4+ T cells. Patients with HHV-6B detection above the median peak viral load (200 copies/mL) received approximately 10-fold fewer donor-derived total or HHV-6B–specific CD4+ T cells than those with peak HHV-6B detection at ≤200 copies/mL or with no HHV-6B detection. These data suggest the importance of donor-derived immunity for controlling HHV-6B reactivation.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1116-1116
Author(s):  
Pamala A. Jacobson ◽  
Sheiren Farag El-Massah ◽  
John Rogosheske ◽  
Amber Kerr ◽  
Janel Boyle-Long ◽  
...  

Abstract Mycophenolate mofetil (MMF) is frequently used as part of the immunosuppressive regimen after allogeneic nonmyeloablative HCT. We and others have previously shown that low mycophenolic acid (MPA) exposure, the active component of MMF, is associated with poorer rates of engraftment and increased risk of graft-vs-host disease (GVHD). As therapeutic plasma targets are difficult to achieve in adult HCT recipients with doses of MMF 2 gm/day, higher doses are required. To determine if higher MMF doses of 3 gm/day would achieve the therapeutic plasma targets we conducted a prospective pharmacokinetic study in adult recipients of nonmyeloablative HCT treated with cyclophosphamide 50 mg/kg, fludarabine 200 mg/m2 and total body irradiation 200 cGy in combination with cyclosporine and MMF as either 1.5 gm every 12 hours (n=15) or 1 gm every 8 hours (n=15). MMF was initiated on day -3 intravenously and switched to the oral form within the first week at the same dose. Pharmacokinetic sampling at steady state was performed once in each patient between days -1 and +5 while on intravenous therapy and then repeated once between days 5 and 14 posttransplant after oral administration. There were no differences in total or unbound MPA 24 hour cumulative area under the curve (AUC), concentration at steady state (Css) or troughs between the two dosing regimens. Total MPA 24 hour AUC (median, [range]) exposures were not different between intravenous 1 gm every 8 hours (53.59 [22.86–101.99] mcg*hr/mL) and intravenous 1.5 gm every 12 hours (60.90 [35.89–127.24] mcg*hr/mL) regimen (p=0.34). Unbound MPA 24 hour AUC exposures were also not different between the intravenous 1 gm every 8 hours (0.942 [0.389–1.722] mcg*hr/mL) and intravenous 1.5 gm every 12 hours (1.081 [0.610–2.194] mcg*hr/mL)(p=0.25). Following oral therapy, total and unbound MPA 24 hour AUC exposures after 1 gm every 8 hours and 1.5 gm every 12 hours were also not different (p≥0.43). Total MPA Css after intravenous 1 gm every 8 hours, oral 1 gm every 8 hours, intravenous 1.5 gm every 12 hours, oral 1.5 gm every 12 hours were 2.23 [0.95–4.25], 2.05 [1.32–3.24], 2.53 [1.46–5.24] and 2.35 [1.47–4.05] mcg/ml, respectively (p≥0.15). We previously found that an unbound 24 hour cumulative MPA AUC <0.600 mcg*hr/mL was associated with higher risk of acute GVHD. The 3 gm/day regimens achieved an unbound 24 hour cumulative AUC target >0.600 mcg*hr/mL in 87–100% of subjects. All patients with the every 8 hour dosing had neutrophil recovery (ANC >500 cells/uL x 3 consecutive days) at a median [range] of day 10 [6–38] as compared to day 12 [0–31] in those with every 12 hour dosing. Acute GVHD grades II–IV developed in 3 (20%) and 6 (40%) patients in the MMF every 8 hour and every 12 hour dosing cohorts, respectively. When compared to our historical controls, MPA exposure is higher with MMF 3 gm/day compared to 2 gm/day regardless of dosing schedule and achieves adequate exposure in most adult patients. In conclusion, MMF 3 g/day should be considered the standard starting dose for adult patients undergoing nonmyeloablative HCT.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. SCI-9-SCI-9
Author(s):  
Alexandra H. Filipovich

Abstract Abstract SCI-9 Hemophagocytic lymphohistiocytosis (HLH) is a group of immunodeficiencies characterized by clinical signs and symptoms of extreme inflammation. HLH is now more widely recognized and no longer viewed as a disorder of young children only, as more adults are being diagnosed and treated. HLH is defined by a unique pattern of clinical findings. In addition to fevers, cytopenias, hepatitis, and splenomegaly, markedly increased levels of inflammatory markers in the blood (ferritin and sCD163 reflecting activation of antigen presenting cells; sIL2Ra and neopterin reflecting activation of T cells) constitute the collection of diagnostic criteria. Activation of inflammatory cells within the central nervous system (CNS) is found in approximately 50 percent of children at diagnosis and requires targeted therapy. In many cases immune defects affecting cytotoxicity of T cells and natural killer cells underlie the susceptibility to HLH. Autosomal recessive disorders include perforin deficiency (the major cytotoxin of the immune system), or defects in proteins involved in degranulation and exocytosis of perforin and granzyme B (MUNC 13–4, MUNC18-2, STX11, Rab27a). The latter proteins are involved in degranulation generally within the hematopoietic system, thus impacting the function of neutrophils and platelets as well. A rare defect of granulogenesis, Chediak Higashi syndrome, is also associated with a high incidence of HLH. Two forms of X-linked lymphoproliferative syndrome (XLP1 – SAP deficiency, and XLP2 – XIAP deficiency), as well as the rare autosomal recessive disorder ITK (IL-2 inducible T cell kinase) deficiency, are characterized by a high incidence of Epstein-Barr virus-driven HLH and lymphoproliferation. A common pathogenic mechanism underlying these consequences has not yet been elucidated. Effective initial treatment for HLH consists of cytotoxic and anti-inflammatory agents. The most widely used over the past 20 years has been a combination of CNS-penetrating steroid (Decadron) and etoposide. Another approach has been to use anti-thymocyte globulin (ATG) as induction therapy. Both treatments have resulted in approximately 60 percent responses during the first month of therapy. Supportive care with broad-spectrum antimicrobials is a critical adjunct. More recently, a new induction protocol—hybrid immunotherapy for HLH, combining the features of early ATG followed by etoposide, with steroids—has been opened in the United States (http://clinicaltrials.gov/ct2/show/NCT01104025) and Europe. However, HLH persists or reactivates in nearly half of patients as immune suppression is reduced. While a common approach to reactivation is to reintensify previous therapy, no clear guidelines have been developed for this complication. The use of Campath, a humanized monoclonal anti-CD52 antibody, as salvage therapy prior to hematopoietic cell transplantation (HCT) is being tested, as both activated T cells and activated monocyte/macrophages (histiocytes) are targeted through CD52. Historically, the three-year survival after HCT in patients treated with HLH-94 was 60 percent. More recently, use of Campath-based reduced intensity conditioning protocols have led to improved results after HCT. Campath has the advantage of reducing graft-versus-host disease if properly timed prior to HCT. In a recent contemporaneous series of HCT from unrelated adult donors, three-year posttransplant survival improved from 43 percent to 92 percent with no early transplant-related mortality. Disclosures: No relevant conflicts of interest to declare.


Author(s):  
Isabella-Maria Giese ◽  
Simone Renner ◽  
Eckhard Wolf ◽  
Stefanie M. Hauck ◽  
Cornelia A. Deeg

AbstractPeople with diabetes mellitus have an increased risk for infections, however, there is still a critical gap in precise knowledge about altered immune mechanisms in this disease. Since diabetic INSC94Y transgenic pigs exhibit elevated blood glucose and a stable diabetic phenotype soon after birth, they provide a favourable model to explore functional alterations of immune cells in an early stage of diabetes mellitus in vivo. Hence, we investigated peripheral blood mononuclear cells (PBMC) of these diabetic pigs compared to non-transgenic wild-type littermates. We found a 5-fold decreased proliferative response of T cells in INSC94Y tg pigs to polyclonal T cell mitogen phytohaemagglutinin (PHA). Using label-free LC-MS/MS, a total of 2,704 proteins were quantified, and distinct changes in protein abundances in CD4+ T cells of early-stage diabetic pigs were detectable. Additionally, we found significant increases in mitochondrial oxygen consumption rate (OCR) and higher basal glycolytic activity in PBMC of diabetic INSC94Y tg pigs, indicating an altered metabolic immune cell phenotype in diabetics. Thus, our study provides new insights into molecular mechanisms of dysregulated immune cells triggered by permanent hyperglycaemia.


2020 ◽  
Vol 51 (1) ◽  
pp. 11-16 ◽  
Author(s):  
Jan Styczyński

AbstractThe most frequent and severe complications after chimeric antigen receptor T-cells (CAR-T cells) therapy include cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), macrophage activation syndrome/hemophagocytic lymphohistiocytosis (MAS/HLH), tumor lysis syndrome (TLS), followed by B-cell aplasia and hypogammaglobulinemia. With these immunologically related events, cytokine storm and immunosuppression, there is a high risk of sepsis and infectious complications. The objective of this review was to present current knowledge on incidence, risk factors, clinical characteristics, and outcome of infections in patients following CAR-T cells therapy, as well as to present current recommendations on prophylaxis of infections after CAR-T cells therapy. Comparable to hematopoietic cell transplantation setting, specific pre- and post-CAR-T cells infusion phases can be determined as early (from 0 to +30 days), intermediate (from +31 to +100 days), and late (beyond day +100). These phases are characterized by CAR-T cells therapy-related factors and immune system defects contributing to an increased risk of infections. It is recommended that in case of active infection, CAR-T cells infusion should be delayed until infection has been successfully treated. After CAR-T cells therapy, prophylaxis should be implemented (anti-bacterial, anti-viral, anti-fungal, anti-pneumocystis), as well as treatment of neutropenia and immunoglobulin replacement should be considered. No recommendations so far can be given on revaccinations after CAR-T cells therapy.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4555-4555
Author(s):  
Mariam Khalil ◽  
Aaron Foster ◽  
Christine Gagliardi

Abstract Introduction: Genetically modified T cells are being investigated to treat a variety of disorders and have been particularly successful in treating B cell cancers. As more effort is poured into new targets, molecular switches, and various other modifications, development of processes to quickly manufacture new products must keep up. Current manufacturing processes often require highly skilled operators and specialized equipment. Here, we demonstrate a simplified, novel method for transduction of T cells, followed by robust expansion in the G-Rex bioreactor with no need for intervention until harvest. A scaled-up, closed-version of the same process, including a closed harvest step with the GatheRex is currently under evaluation. Methods: Frozen peripheral blood mononuclear cells (PBMCs) from healthy donors were used as starting material. PBMCs were thawed, washed, and activated with soluble anti-CD3 and anti-CD28 antibodies either in cell culture bags (32-C, Saint-Gobain Cell Therapy) or in G-Rex bioreactors (Wilson Wolf Corporation). Cells were cultured in TexMACS GMP medium (Miltenyi Biotec) with IL-7 and IL-15 throughout. For the transduction step, activated PMBCs and retroviral supernatant were incubated in cell culture bags coated with Retronectin (Takara) or in G-Rex bioreactors with vectofusin-1 (Miltenyi Biotec). Viral constructs contained either a CD34 or CD19 marker detectable by antibody staining. For transduction in the G-Rex, various cell densities, volumes, constructs, and multiplicity of infection (MOIs) were tested. Where applicable, the GatheRex device (Wilson Wolf Corporation) was used for volume reduction and harvest. Transduction efficiency and T cell phenotype were measured by flow cytometry. Cell count and viability were assessed with the NC-3000 (Chemometic). Glucose and lactate concentrations were checked daily for in-processing monitoring. Results: Overall transduction efficiency ranged from 30-90% depending on the experimental conditions. Incubating 1x107 activated PBMCs in 10 ml of medium in a 10-cm2 G-Rex (1.0 ml/cm2) with retrovirus at an MOI of 1 resulted in 3% transduced cells. Addition of vectofusin-1 to the same condition yielded transduction efficiency of 44%. Increasing the MOI to 10 lead to 86% transduced cells. Decreasing the transduction volume from 1.0 ml/cm2 to 0.4 ml/cm2 increased transduction efficiency from 34% to 55%. Reducing the volume further to 0.2 ml/cm2 did not improve efficiency, and rather had a negative impact compared to the 0.4 ml/cm2 condition (38%). 16-24 hrs after transduction, the volume of medium was increased to 10.0 ml/cm2 without a wash step. The dilution in place of a wash step had no negative impact on cell viability. 10.0 ml/cm2 medium supported high viability (>90%) and expansion (30-50 fold) over an additional 9 days without operator intervention. The phenotype of cells expanded in the G-Rex contained a mixed population of CD45RO+ and CD45RA+ cells, with a similar distribution of naive and memory cell subsets in G-Rex and bag cultures. Harvest of cells with the GatheRex was efficient; a 1L volume was reduced 10-fold in 5 minutes, and 95% of cells were recovered. Summary: T cells can be transduced with retroviral vectors in the G-Rex bioreactor. Clinically relevant levels of transgene expression can be achieved by combining reagents in the G-Rex, without complicated coating steps or time-consuming spinning steps. This simplified procedure reduces the hands-on time of the T cell transduction to minutes rather than hours. Transgenic cells can be expanded 30-50-fold in the G-Rex with limited operator intervention and without specialized equipment. Disclosures Khalil: Bellicum Pharmaceuticals: Employment. Foster:Bellicum: Employment, Equity Ownership. Gagliardi:Bellicum Pharmaceuticals: Employment.


2014 ◽  
Vol 2014 ◽  
pp. 1-3 ◽  
Author(s):  
Hilana H. Hatoum ◽  
Anita Patel ◽  
K. K. Venkat

Delayed graft function (DGF) of kidney transplants increases risk of rejection. We aimed to assess the utility of weekly biopsies during DGF in the setting of currently used immunosuppression and identify variables associated with rejection during DGF. We reviewed all kidney transplants at our institution between January 2008 and December 2011. All patients received rabbit antithymocyte globulin/Thymoglobulin (ATG) or Basiliximab/Simulect induction with maintenance tacrolimus + mycophenolate + corticosteroid therapy. Patients undergoing at least one weekly biopsy during DGF comprised the study group. Eighty-three/420 (19.8%) recipients during this period experienced DGF lasting ≥1 week and underwent weekly biopsies until DGF resolved. Biopsy revealed significant rejection only in 4/83 patients (4.8%) (one Banff 1-A and two Banff 2-A cellular rejections, and one acute humoral rejection). Six other/83 patients (7.2%) had Banff-borderline rejection of uncertain clinical significance. Four variables (ATG versus Basiliximab induction, patient age, panel reactive anti-HLA antibody level at transplantation, and living versus deceased donor transplants) were statistically significantly different between patients with and without rejection, though the clinical significance of these differences is questionable given the low incidence of rejection. Conclusions. Under current immunosuppression regimens, rejection during DGF is uncommon and the utility of serial biopsies during DGF is limited.


2018 ◽  
Vol 10 (469) ◽  
pp. eaat8410 ◽  
Author(s):  
Sonia Tugues ◽  
Ana Amorim ◽  
Sabine Spath ◽  
Guillaume Martin-Blondel ◽  
Bettina Schreiner ◽  
...  

Allogeneic hematopoietic cell transplantation (allo-HCT) not only is an effective treatment for several hematologic malignancies but can also result in potentially life-threatening graft-versus-host disease (GvHD). GvHD is caused by T cells within the allograft attacking nonmalignant host tissues; however, these same T cells mediate the therapeutic graft-versus-leukemia (GvL) response. Thus, there is an urgent need to understand how to mechanistically uncouple GvL from GvHD. Using preclinical models of full and partial MHC-mismatched HCT, we here show that the granulocyte-macrophage colony-stimulating factor (GM-CSF) produced by allogeneic T cells distinguishes between the two processes. GM-CSF drives GvHD pathology by licensing donor-derived phagocytes to produce inflammatory mediators such as interleukin-1β and reactive oxygen species. In contrast, GM-CSF did not affect allogeneic T cells or their capacity to eliminate leukemic cells, retaining undiminished GvL responses. Last, tissue biopsies and peripheral blood mononuclear cells from patients with grade IV GvHD showed an elevation of GM-CSF–producing T cells, suggesting that GM-CSF neutralization has translational potential in allo-HCT.


Sign in / Sign up

Export Citation Format

Share Document