scholarly journals Innate Immune Activation by Tissue Injury and Cell Death in the Setting of Hematopoietic Stem Cell Transplantation

2015 ◽  
Vol 6 ◽  
Author(s):  
Todd V. Brennan ◽  
Victoria R. Rendell ◽  
Yiping Yang
Blood ◽  
2022 ◽  
Author(s):  
Asim Saha ◽  
Sharon Hyzy ◽  
Tahirih Laforest Lamothe ◽  
Katelyn J. Hammond ◽  
Nicholas M Clark ◽  
...  

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative treatment for patients with non-malignant or malignant blood disorders. Its success has been limited by graft-versus-host disease (GVHD). Current genotoxic conditioning regimens mediate tissue injury and potentially incite and amplify GVHD, limiting use of this potentially curative treatment beyond malignant disorders. Minimizing genotoxic conditioning while achieving alloengraftment without global immune suppression is highly desirable. Antibody-drug-conjugates (ADCs) targeting hematopoietic cells can specifically deplete host stem and immune cells and enable alloengraftment. Here we report an anti-mouse CD45-targeted-ADC (CD45-ADC) that facilitates stable murine multi-lineage donor cell engraftment. Conditioning with CD45-ADC (3mg/kg) was effective as a single agent in both congenic and minor-mismatch transplant models resulting in full donor chimerism comparable to lethal total body irradiation (TBI). In an MHC-disparate allo-HSCT model, pre-transplant CD45-ADC (3mg/kg) combined with low-dose TBI (150cGy) and a short course of costimulatory blockade with anti-CD40 ligand antibody enabled 89% of recipients to achieve stable alloengraftment (mean value: 72%). When CD45-ADC was combined with pre-transplant TBI (50cGy) and post-transplant Rapamycin, Cytoxan or a JAK inhibitor, 90-100% of recipients achieved stable chimerism (mean: 77%, 59%, 78%, respectively). At a higher dose (5mg/kg), CD45-ADC as a single agent was sufficient for rapid, high level multi-lineage chimerism sustained through the 22 weeks observation period. Therefore, CD45-ADC has potential utility to confer the benefit of fully myeloablative conditioning but with substantially reduced toxicity when given as a single agent or at lower doses in conjunction with reduced intensity conditioning.


Blood ◽  
2015 ◽  
Vol 126 (4) ◽  
pp. 437-444 ◽  
Author(s):  
Martin Chopra ◽  
Andreas Brandl ◽  
Daniela Siegmund ◽  
Anja Mottok ◽  
Viktoria Schäfer ◽  
...  

Key Points Fn14 activation is involved in intestinal apoptosis after allo-HCT and contributes to gastrointestinal GVHD. Fn14 blockade with an ADCC-defective human immunoglobulin G1 antibody reduces GVHD severity without modulating GVL responses.


Author(s):  
Simona Jurkovic Mlakar ◽  
Chakradhara Rao Uppugunduri Satyanarayana ◽  
Tiago Nava ◽  
Vid Mlakar ◽  
Hadrien Golay ◽  
...  

Abstract Purpose This study aimed to retrospectively evaluate the genetic association of null variants of glutathione S-transferases GSTM1 and GSTT1 with relapse incidence in children with hematological malignancies (HMs) undergoing busulfan (BU)- containing allogeneic hematopoietic stem cell transplantation (HSCT) and to assess the impact of these variants on BU-induced cytotoxicity on the immortalized lymphoblastoid cell lines (LCLs) and tumor THP1 GST gene-edited cell models. Methods GSTM1- and GSTT1-null alleles were genotyped using germline DNA from whole blood prior to a conditioning BU-based regimen. Association of GSTM1- and GSTT1-null variants with relapse incidence was analyzed using multivariable competing risk analysis. BU-induced cell death studies were conducted in GSTs- null and non-null LCLs and CRISPR–Cas9 gene-edited THP1 leukemia cell lines. Results Carrying GSTM1/GSTT1 double null genotype was found to be an independent risk factor for post-HSCT relapse in 86 children (adjusted HR: 6.52 [95% Cl, 2.76–15.42; p = 1.9 × 10–5]). BU-induced cell death preferentially in THP1GSTM1(non−null) and LCLsGSTM1(non−null) as shown by decreased viability, increased necrosis and levels of the oxidized form of glutathione compared to null cells, while GSTT1 non-null cells showed increased baseline proliferation. Conclusion The clinical association suggests that GSTM1/GSTT1 double null genotype could serve as genetic stratification biomarker for the high risk of post-HSCT relapse. Functional studies have indicated that GSTM1 status modulates BU-induced cell death. On the other hand, GSTT1 is proposed to be involved in baseline cell proliferation.


2018 ◽  
Vol 24 (2) ◽  
pp. 112-121 ◽  
Author(s):  
Sarah L Caddy ◽  
Meng Wang ◽  
Pramila Krishnamurthy ◽  
Benjamin Uttenthal ◽  
Anita Chandra ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document