1
|
Dhir V, Prince C, Allan D, Atkins H, Bredeson C, Kekre N, Kennah M, Masurekar A, Vasudevan Nampoothiri R. Impact of Day 11 Methotrexate Dose Adjustments due to Mucositis on the Outcomes Following Allogeneic Stem Cell Transplant in the Setting of Anti Thymocyte Globulin (ATG) Based GVHD Prophylaxis. Eur J Haematol 2025. [PMID: 39757868 DOI: 10.1111/ejh.14380] [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: 11/21/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Dose adjustments of Day 11 Methotrexate (MTx) for GVHD prophylaxis after allogeneic hematopoietic stem cell transplantation (HCT) are common due to mucositis, renal injury, or other reasons. The impact of omitting or adjusting doses of MTx in the era of ATG-based GVHD prophylaxis remains unexplored. METHODS We retrospectively analyzed the outcomes of all adult patients undergoing allogeneic HCT who received ATG-based GVHD prophylaxis at The Ottawa Hospital from January 2019 to December 2022. We compared outcomes of patients having only Day 11 MTx dose reductions due to mucositis(MTxRD group) with patients receiving full dose MTx on all 4 days (MTxFD group). The impact of Day 11 MTx dose reduction on outcomes were assessed using Kaplan-Meier analyses and log rank test. RESULTS Three hundred and four patients (median age 58 [17-74] years; 64% male) underwent allogeneic HCT during the study period. Baseline characteristics were similar between the MTxRD group (n = 69) and MTxFD group (n = 199) except for an increased proportion of MAC regimens in MTxRD group. The incidence of severe aGVHD (7.2% vs. 7.5%; p = 0.96) and chronic GVHD (15.9% vs. 15.6%; p = 0.89) were not different between the two groups. The 2-year OS (59% vs. 69.8%; p = 0.11), GRFS (42.4% vs. 47.6%; p = 0.32), NRM (17.7% vs. 12.2%; p = 0.45) or relapse/progression (CIR 27.4 vs. 26.6%; p = 0.55) were also similar between the two groups. CONCLUSIONS In patients receiving ATG-based GVHD prophylaxis regimens, there were similar GVHD and survival outcomes in patients who received no or reduced D11 MTx when compared to full dose MTx. Dose adjustments of D11 MTx due to mucositis appear to be safe in the era of ATG-based GVHD prophylaxis regimens.
Collapse
Affiliation(s)
- Vinita Dhir
- Transplant and Cellular Therapy Program, Division of Hematology, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Connor Prince
- Transplant and Cellular Therapy Program, Division of Hematology, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - David Allan
- Transplant and Cellular Therapy Program, Division of Hematology, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Harold Atkins
- Transplant and Cellular Therapy Program, Division of Hematology, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Christopher Bredeson
- Transplant and Cellular Therapy Program, Division of Hematology, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Natasha Kekre
- Transplant and Cellular Therapy Program, Division of Hematology, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Michael Kennah
- Transplant and Cellular Therapy Program, Division of Hematology, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Ashish Masurekar
- Transplant and Cellular Therapy Program, Division of Hematology, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Ram Vasudevan Nampoothiri
- Transplant and Cellular Therapy Program, Division of Hematology, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
2
|
Ngwube A, Rangarajan H, Shah N. Role of abatacept in the prevention of graft- versus-host disease: current perspectives. Ther Adv Hematol 2023; 14:20406207231152644. [PMID: 36845849 PMCID: PMC9943961 DOI: 10.1177/20406207231152644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 11/28/2022] [Indexed: 02/11/2023] Open
Abstract
Administration of abatacept following transplantation has been reported to inhibit graft rejection and graft-versus-host-disease (GvHD) in mouse models associated with allogeneic hematopoietic stem cell transplant (HSCT). This strategy has recently been adopted in clinical practice for GvHD prevention in human allogeneic HSCT and offers a unique approach to optimizing GvHD prophylaxis following alternative donor HSCTs. When combined with calcineurin inhibitors and methotrexate, abatacept had shown to be safe and effective in preventing moderate to severe acute GvHD in myeloablative HSCT using human leukocyte antigen (HLA) unrelated donors. Equivalent results are being reported in recent studies using alternative donors, in reduced-intensity conditioning HSCT and nonmalignant disorders. These observations have led to hypothesizing that even in the setting of increasing donor HLA disparity, abatacept when given with traditional GvHD prophylaxis does not worsen general outcomes. In addition, in limited studies, abatacept have being protective against the development of chronic GvHD through extended dosing and in the treatment of steroid-refractory chronic GvHD. This review summarized all the limited reports of this novels approach in the HSCT setting.
Collapse
Affiliation(s)
| | - Hemalatha Rangarajan
- Division of Pediatric Hematology, Oncology,
Blood and Marrow Transplant, Nationwide Children’s Hospital, Columbus, OH,
USA
| | - Niketa Shah
- Section of Pediatric Hematology Oncology and
Bone Marrow transplant, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
3
|
Enok Bonong PR, Buteau C, Duval M, Lacroix J, Laporte L, Tucci M, Robitaille N, Spinella PC, Cuvelier GDE, Lewis V, Vercauteren S, Alfieri C, Trottier H. Risk factors for post-transplant Epstein-Barr virus events in pediatric recipients of hematopoietic stem cell transplants. Pediatr Transplant 2021; 25:e14052. [PMID: 34076939 DOI: 10.1111/petr.14052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Epstein-Barr virus (EBV) can cause severe disease following hematopoietic stem cell transplant (HSCT), including post-transplant lymphoproliferative disorder (PTLD). The objective was to analyze risk factors associated with post-transplant EBV outcomes among pediatric allogeneic HSCT recipients. METHODS We used data from 156 pediatric allogeneic HSCT recipients enrolled in the Canadian multicenter TREASuRE study. Cox and Prentice-Williams-Petersen models were used to analyze risk factors for post-transplant EBV events including occurrence and recurrence of EBV DNAemia, increase in EBV viral load (EBV-VL), and preemptive use of rituximab, an effective therapy against PTLD. RESULTS Females were at higher risk for increasing EBV-VL (adjusted hazard ratio (HR) = 2.83 [95% confidence intervals (CI): 1.33-6.03]) and rituximab use (HR = 3.08 [1.14-8.30]), but had the same EBV DNAemia occurrence (HR = 1.21 [0.74-1.99]) and recurrence risks (HR=1.05 [0.70-1.58]) compared to males. EBV DNAemia was associated with recipient pre-transplant EBV seropositivity (HR = 2.47 [1.17-5.21]) and with graft from an EBV-positive donor (HR = 3.53 [1.95-6.38]). Anti-thymocyte globulin (ATG) was strongly associated with all EBV outcomes, including the use of rituximab (HR = 5.33 [1.47-19.40]). Mycophenolate mofetil (MMF) significantly decreased the risk of all EBV events including the rituximab use (HR = 0.13 [0.03-0.63]). CONCLUSION This study in pediatric allogeneic HSCT patients reveals a reduced risk of all EBV outcomes with the use of MMF. Risk factors for EBV events such as EBV-VL occurrence and recurrence include EBV positivity in the donor and recipient, and use of ATG, whereas risk factors for the most severe forms of EBV outcome (EBV-VL and the use of rituximab) include female sex and ATG use.
Collapse
Affiliation(s)
- Pascal R Enok Bonong
- Department of Social and Preventive Medicine, CHU Sainte-Justine, Université de Montréal, Montreal, QC, Canada
| | - Chantal Buteau
- Division of Infectious Diseases, Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, QC, Canada
| | - Michel Duval
- Division of Hematology-Oncology, Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, QC, Canada
| | - Jacques Lacroix
- Division of Pediatric Intensive Care Medicine, Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, QC, Canada
| | - Louise Laporte
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Marisa Tucci
- Division of Pediatric Intensive Care Medicine, Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, QC, Canada
| | - Nancy Robitaille
- Division of Hematology-Oncology, Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Héma-Québec, Montreal, QC, Canada
| | - Philip C Spinella
- St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, MO, USA
| | - Geoffrey D E Cuvelier
- Pediatric Blood and Marrow Transplant, Department of Pediatric Hematology-Oncology-BMT, Department of Pediatrics and Child Health, CancerCare Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Victor Lewis
- Department of Pediatrics and Department of Oncology, Alberta Children's Hospital, University of Calgary, Calgary, AB, Canada
| | - Suzanne Vercauteren
- Department of Pathology and Laboratory Medicine, BC Children's Hospital, University of British Colombia, Vancouver, BC, Canada
| | - Caroline Alfieri
- Department of Microbiology, Infectiology and Immunology, Centre de recherche du CHU Sainte-Justine, Université de Montréal, Montreal, QC, Canada
| | - Helen Trottier
- Department of Social and Preventive Medicine, CHU Sainte-Justine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
4
|
Arrabi L, Jan A, Hosing C, Milton DR, Yeh J. Transitioning tacrolimus to sirolimus in allogeneic hematopoietic cell transplantation. Eur J Haematol 2021; 107:634-641. [PMID: 34431142 DOI: 10.1111/ejh.13701] [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: 03/23/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Calcineurin inhibitor (CNI) use for acute graft-versus-host disease (aGVHD) prophylaxis in allogeneic hematopoietic cell transplantation (allo-HCT) recipients has been associated with toxicities. Toxicities may be managed by converting CNI to sirolimus as often done in solid organ transplantation. This study aimed to characterize allo-HCT patients who completely transitioned from tacrolimus to sirolimus and evaluate the incidence of aGVHD within 100 days post-transition, overall survival (OS), and incidence of relapse. METHODS Safety and efficacy data were collected at baseline and at day 30 and 90 post-transition from tacrolimus to sirolimus and at one-year post-HCT. RESULTS Most patients who transitioned had acute leukemia, received a matched unrelated donor allo-HCT, and transitioned due to nephrotoxicity or neurotoxicity. The resolution rate was 83% and 48% in the nephrotoxicity group, 78% and 61% in the neurotoxicity group, 33% and 33% in the group that developed both nephrotoxicity and transplant-associated thrombotic microangiopathy at 30 and 90 days of assessments, respectively. Patients who transitioned before day 55 post-allo-HCT were more likely to develop new or worsening aGVHD. The one-year OS and relapse rates were 37% and 20%, respectively. CONCLUSIONS The conversion from tacrolimus to sirolimus demonstrates promising resolution of acute toxicities; however, overall mortality remains high.
Collapse
Affiliation(s)
- Linda Arrabi
- Department of Pharmacy, Henry Ford Health System, Detroit, MI, USA
| | - Anna Jan
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chitra Hosing
- Division of Cancer Medicine, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Denái R Milton
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason Yeh
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
5
|
AlJohani NI. Role of folinic acid in methotrexate-based prophylaxis of graft-versus-host disease following hematopoietic stem cell transplantation. ACTA ACUST UNITED AC 2021; 26:620-627. [PMID: 34411497 DOI: 10.1080/16078454.2021.1966222] [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: 10/20/2022]
Abstract
Methotrexate (MTX) is one of the main therapeutic agents currently used for the prophylaxis of graft-versus-host disease (GvHD) following hematopoietic stem cell transplantation. However, it is associated with significant toxicity and considerable side effects in many patients, which lead to either early withdrawal or dose reductions that may expose patients to the risk of GvHD and graft failure. Folinic acid (FA) can bypass the inhibitory effects of MTX on folate availability and control MTX toxicity. However, concerns that FA might inhibit the anti-GvHD effect of MTX and limited reports on its clinical usefulness have led to reluctance in its inclusion in standard GvHD prophylaxis regimens. Additionally, universal dosing and timing guidelines are lacking. I discuss the available literature and evaluate the evidence for the effect of FA on MTX toxicity and its safety regarding GvHD development and graft rejection in both adult and pediatric patients. Although FA administration appears to be safe, its efficacy for routine use in all types of transplants in adult patients is unproven and further research is required to confirm its MTX toxicity-lowering effect, identify the individual parameters that influence its usefulness in clinical practice, and evaluate its potential when developing a personalized prophylaxis regimen.
Collapse
Affiliation(s)
- Naif I AlJohani
- King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| |
Collapse
|
6
|
Immune checkpoint inhibitor treatment induces colitis with heavy infiltration of CD8 + T cells and an infiltration pattern that resembles ulcerative colitis. Virchows Arch 2021; 479:1119-1129. [PMID: 34338882 PMCID: PMC8724151 DOI: 10.1007/s00428-021-03170-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 07/14/2021] [Accepted: 07/22/2021] [Indexed: 12/17/2022]
Abstract
Colitis is a common, but poorly understood, adverse event of immune checkpoint inhibitors that are standard-of-care for an expanding range of cancer types. This explorative study aimed to describe the immune infiltrates in the colon from individuals developing checkpoint inhibitor colitis and compare them to well-known immunophenotypes of acute graft-versus-host disease, ulcerative colitis, and Crohn’s disease. Colon biopsies (n = 20 per group) of patients with checkpoint inhibitor colitis, acute graft-versus-host disease, ulcerative colitis and Crohn’s disease, all colitis treatment-naïve, and of individuals with a normal colon were analyzed using immunohistochemistry: CD8 for cytotoxic T cells, CD4 for T helper cells, and CD68 to identify cells of macrophage lineage. CD8 + T cell, CD4 + T cell, and CD68 + cell counts were performed. Cell infiltration was scored as scattered/patchy or band-like in the superficial and deep gut mucosa. Checkpoint inhibitor colitis was found to be heavily infiltrated by CD8 + T cells. Comparative analysis between groups showed that both CD8 + T cell counts (P < 0.01) and immune cell infiltration patterns in checkpoint inhibitor colitis were most similar to those observed in ulcerative colitis, with a deep band-like CD4 + T cell infiltration pattern and a superficial band-like CD68 + cell infiltration pattern in both. In conclusion, this is the first immunohistopathological study comparing infiltrate characteristics of checkpoint inhibitor colitis, acute graft-versus-host disease, ulcerative colitis, and Crohn’s disease. Checkpoint inhibitor colitis samples are heterogeneous, heavily infiltrated by CD8 + T cells, and show an immune cell infiltration pattern that is more similar to ulcerative colitis than to colonic acute graft-versus-host disease or colonic Crohn’s disease.
Collapse
|
7
|
Butera S, Cerrano M, Brunello L, Dellacasa CM, Faraci DG, Vassallo S, Mordini N, Sorasio R, Zallio F, Busca A, Bruno B, Giaccone L. Impact of anti-thymocyte globulin dose for graft-versus-host disease prophylaxis in allogeneic hematopoietic cell transplantation from matched unrelated donors: a multicenter experience. Ann Hematol 2021; 100:1837-1847. [PMID: 33948721 PMCID: PMC8195753 DOI: 10.1007/s00277-021-04521-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 04/08/2021] [Indexed: 01/07/2023]
Abstract
Despite the widespread use of rabbit anti-thymocyte globulin (ATG) to prevent acute and chronic graft-versus-host disease (aGVHD, cGVHD) after allogeneic hematopoietic cell transplantation (allo-HCT), convincing evidence about an optimal dose is lacking. We retrospectively evaluated the clinical impact of two different ATG doses (5 vs 6-7.5 mg/kg) in 395 adult patients undergoing HSCT from matched unrelated donors (MUD) at 3 Italian centers. Cumulative incidence of aGVHD and moderate-severe cGVHD did not differ in the 2 groups. We observed a trend toward prolonged overall survival (OS) and disease-free survival (DFS) with lower ATG dose (5-year OS and DFS 56.6% vs. 46.3%, p=0.052, and 46.8% vs. 38.6%, p=0.051, respectively) and no differences in relapse incidence and non-relapse mortality. However, a significantly increased infection-related mortality (IRM) was observed in patients who received a higher ATG dose (16.7% vs. 8.8% in the lower ATG group, p=0.019). Besides, graft and relapse-free survival (GRFS) was superior in the lower ATG group (5-year GRFS 43.1% vs. 32.4%, p=0.014). The negative impact of higher ATG dose on IRM and GRFS was confirmed by multivariate analysis. Our results suggest that ATG doses higher than 5 mg/kg are not required for MUD allo-HCT and seem associated with worse outcomes.
Collapse
Affiliation(s)
- Sara Butera
- Department of Oncology, SSD Trapianto Allogenico di Cellule Staminali, A.O.U. Città della Salute e della Scienza di Torino, Via Genova 3, 10126, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy
| | - Marco Cerrano
- Department of Oncology, Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
| | - Lucia Brunello
- Department of Oncology, SSD Trapianto Allogenico di Cellule Staminali, A.O.U. Città della Salute e della Scienza di Torino, Via Genova 3, 10126, Torino, Italy
- Department of Hematology, A.O. Santissimi Antonio e Biagio e C Arrigo, Alessandria, Italy
| | - Chiara Maria Dellacasa
- Department of Oncology, SSD Trapianto Allogenico di Cellule Staminali, A.O.U. Città della Salute e della Scienza di Torino, Via Genova 3, 10126, Torino, Italy
| | - Danilo Giuseppe Faraci
- Department of Oncology, SSD Trapianto Allogenico di Cellule Staminali, A.O.U. Città della Salute e della Scienza di Torino, Via Genova 3, 10126, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy
| | - Sara Vassallo
- Department of Oncology, SSD Trapianto Allogenico di Cellule Staminali, A.O.U. Città della Salute e della Scienza di Torino, Via Genova 3, 10126, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy
| | - Nicola Mordini
- Division of Hematology, A.O. Santi Croce e Carle, Cuneo, Italy
| | - Roberto Sorasio
- Division of Hematology, A.O. Santi Croce e Carle, Cuneo, Italy
| | - Francesco Zallio
- Department of Hematology, A.O. Santissimi Antonio e Biagio e C Arrigo, Alessandria, Italy
| | - Alessandro Busca
- Department of Oncology, SSD Trapianto Allogenico di Cellule Staminali, A.O.U. Città della Salute e della Scienza di Torino, Via Genova 3, 10126, Torino, Italy
| | - Benedetto Bruno
- Department of Oncology, SSD Trapianto Allogenico di Cellule Staminali, A.O.U. Città della Salute e della Scienza di Torino, Via Genova 3, 10126, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy
| | - Luisa Giaccone
- Department of Oncology, SSD Trapianto Allogenico di Cellule Staminali, A.O.U. Città della Salute e della Scienza di Torino, Via Genova 3, 10126, Torino, Italy.
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Torino, Italy.
| |
Collapse
|
8
|
Enok Bonong PR, Zahreddine M, Buteau C, Duval M, Laporte L, Lacroix J, Alfieri C, Trottier H. Factors Associated with Post-Transplant Active Epstein-Barr Virus Infection and Lymphoproliferative Disease in Hematopoietic Stem Cell Transplant Recipients: A Systematic Review and Meta-Analysis. Vaccines (Basel) 2021; 9:288. [PMID: 33808928 PMCID: PMC8003684 DOI: 10.3390/vaccines9030288] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 12/29/2022] Open
Abstract
This systematic review was undertaken to identify risk factors associated with post-transplant Epstein-Barr virus (EBV) active infection and post-transplant lymphoproliferative disease (PTLD) in pediatric and adult recipients of hematopoietic stem cell transplants (HSCT). A literature search was conducted in PubMed and EMBASE to identify studies published until 30 June 2020. Descriptive information was extracted for each individual study, and data were compiled for individual risk factors, including, when possible, relative risks with 95% confidence intervals and/or p-values. Meta-analyses were planned when possible. The methodological quality and potential for bias of included studies were also evaluated. Of the 3362 titles retrieved, 77 were included (62 for EBV infection and 22 for PTLD). The overall quality of the studies was strong. Several risk factors were explored in these studies, but few statistically significant associations were identified. The use of anti-thymocyte globulin (ATG) was identified as the most important risk factor positively associated with post-transplant active EBV infection and with PTLD. The pooled relative risks obtained using the random-effect model were 5.26 (95% CI: 2.92-9.45) and 4.17 (95% CI: 2.61-6.68) for the association between ATG and post-transplant EBV infection and PTLD, respectively. Other risk factors for EBV and PTLD were found in the included studies, such as graft-versus-host disease, type of conditioning regimen or type of donor, but results are conflicting. In conclusion, the results of this systematic review indicate that ATG increases the risk of EBV infection and PTLD, but the link with all other factors is either nonexistent or much less convincing.
Collapse
Affiliation(s)
- Pascal Roland Enok Bonong
- Department of Social and Preventive Medicine, Université de Montréal, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada; (P.R.E.B.); (M.Z.)
| | - Monica Zahreddine
- Department of Social and Preventive Medicine, Université de Montréal, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada; (P.R.E.B.); (M.Z.)
| | - Chantal Buteau
- Department of Pediatrics, Division of Infectious Diseases, CHU Sainte-Justine, Université de Montréal, Montréal, QC H3T 1C5, Canada;
| | - Michel Duval
- Department of Pediatrics, Division of Hematology-Oncology, CHU Sainte-Justine, Université de Montréal, Montréal, QC H3T 1C5, Canada;
| | - Louise Laporte
- Research Center of CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada;
| | - Jacques Lacroix
- Department of Pediatrics, Division of Pediatric Intensive Care Medicine, CHU Sainte-Justine, Université de Montréal, Montréal, QC H3T 1C5, Canada;
| | - Caroline Alfieri
- Departement of Microbiology, Infectiology and Immunology, Université de Montréal, CHU Sainte-Justine Research Center, Montréal, QC H3T 1C5, Canada;
| | - Helen Trottier
- Department of Social and Preventive Medicine, Université de Montréal, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada; (P.R.E.B.); (M.Z.)
| |
Collapse
|
9
|
Dikmetas O, Kocabeyoglu S, Mocan MC. The Association between Meibomian Gland Atrophy and Corneal Subbasal Nerve Loss in Patients with Chronic Ocular Graft-versus-host Disease. Curr Eye Res 2021; 46:796-801. [PMID: 33427504 DOI: 10.1080/02713683.2020.1846754] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Purpose: To investigate the association between meibomian gland (MG) loss and corneal subbasal nerve plexus density in patients with chronic graft-versus-host disease (GVHD) related dry eye disease (DED).Materials and Methods: This cross-sectional study included 22 adult patients with severe DED secondary to chronic GVHD. Control group comprised age- and sex-matched 28 healthy subjects with no evidence of ocular disease. All subjects underwent tear breakup time (TBUT), corneal staining, Schirmer I test without anesthesia, quantitative MG drop-out assessment using infrared meibography and corneal subbasal nerve density measurements with in vivo confocal microscopy (IVCM) (ConfoScan4, Nidek, Japan). One eye per patient was included for statistical purposes. Mann-Whitney U test and one-way multivariate ANOVA test were used for comparative analyses.Results: Compared to healthy subjects (mean age = 26.9 ± 13.5 years (range = 20-44 years)), patients with chronic GVHD (mean age = 29.6 ± 12.6 years (range = 19-45 years)) had worse meibography scores (p < .001), reduced corneal subbasal nerve plexus densities (p < .001), lower TBUT scores (p = .012), lower Schirmer I values (p = .001) and higher corneal staining scores (p = 003). Meiboscores in the GVHD and control groups were 2.9 ± 1.1 (range = 1-4) vs. 0.7 ± 0.4 (range = 0-2) for the superior (p < .001), and 3.2 ± 1.2 (range = 2-4) vs. 0.5 ± 0.3 (range = 0-2) for inferior (p < .001) eyelids, respectively. Corneal subbasal nerve densities of patients with GVHD did not reveal a correlation with meiboscores (r = 0.030; p = .709 for the inferior and r = 0.268; p = .075 for the superior eyelids) but showed a weak correlation with Schirmer I test values (r = 0.268; p = .014).Conclusions: Patients with chronic GVHD are at high risk for developing DED and MG dysfunction. In the setting of chronic GVHD-related DED, MG loss does not appear to be a significant factor for corneal subbasal nerve damage.
Collapse
Affiliation(s)
- Ozlem Dikmetas
- Department of Ophthalmology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Sibel Kocabeyoglu
- Department of Ophthalmology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Mehmet Cem Mocan
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
10
|
Kim S, Santhanam S, Lim S, Choi J. Targeting Histone Deacetylases to Modulate Graft-Versus-Host Disease and Graft-Versus-Leukemia. Int J Mol Sci 2020; 21:ijms21124281. [PMID: 32560120 PMCID: PMC7349873 DOI: 10.3390/ijms21124281] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/06/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the main therapeutic strategy for patients with both malignant and nonmalignant disorders. The therapeutic benefits of allo-HSCT in malignant disorders are primarily derived from the graft-versus-leukemia (GvL) effect, in which T cells in the donor graft recognize and eradicate residual malignant cells. However, the same donor T cells can also recognize normal host tissues as foreign, leading to the development of graft-versus-host disease (GvHD), which is difficult to separate from GvL and is the most frequent and serious complication following allo-HSCT. Inhibition of donor T cell toxicity helps in reducing GvHD but also restricts GvL activity. Therefore, developing a novel therapeutic strategy that selectively suppresses GvHD without affecting GvL is essential. Recent studies have shown that inhibition of histone deacetylases (HDACs) not only inhibits the growth of tumor cells but also regulates the cytotoxic activity of T cells. Here, we compile the known therapeutic potential of HDAC inhibitors in preventing several stages of GvHD pathogenesis. Furthermore, we will also review the current clinical features of HDAC inhibitors in preventing and treating GvHD as well as maintaining GvL.
Collapse
Affiliation(s)
- Sena Kim
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
- Correspondence: (S.K.); (J.C.)
| | | | - Sora Lim
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Jaebok Choi
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
- Correspondence: (S.K.); (J.C.)
| |
Collapse
|
11
|
Lawitschka A, Lucchini G, Strahm B, Dalle JH, Balduzzi A, Gibson B, Diaz De Heredia C, Wachowiak J, Dalissier A, Vettenranta K, Yaniv I, Bordon V, Bauer D, Bader P, Meisel R, Peters C, Corbacioglu S. Pediatric acute graft-versus-host disease prophylaxis and treatment: surveyed real-life approach reveals dissimilarities compared to published recommendations. Transpl Int 2020; 33:762-772. [PMID: 32133691 PMCID: PMC7384018 DOI: 10.1111/tri.13601] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/06/2020] [Accepted: 02/28/2020] [Indexed: 12/22/2022]
Abstract
Pediatric allogeneic hematopoietic cell transplantation (HCT) practices differ from those of adults, particularly the heterogeneity of transplantable nonmalignant diseases and the lower incidence of graft‐versus‐host disease (GVHD). Several guidelines regarding the management of acute (a) GVHD in adult HCT have been published. We aimed to capture the real‐life approaches for pediatric aGVHD prophylaxis/treatment, and data from 75/193 (response rate 39%) EBMT centers (26 countries) were included, representing half (48%) of the pediatric EBMT‐HCT activity. Results with ≥75% approval from respondents (74/75) for GVHD prophylaxis after myeloablative HCT for malignancies partially contradict published guidelines: Single‐agent cyclosporine A (CsA) was used for matched sibling donor HCT in 47%; blood CsA levels were reported lower; the relapse risk in malignant diseases influenced GVHD prophylaxis with early withdrawal of CsA; distinct longer duration of CsA was employed in nonmalignant diseases. Most centers used additional anti‐thymocyte globulin for matched unrelated and mismatched donor HCT, but not for matched siblings. Regarding prophylaxis in nonmyeloablative conditioning (mainly for nonmalignant diseases), responses showed broad heterogeneity. High conformity was found for first‐line treatment; however, results regarding steroid‐refractory aGVHD indicate an earlier diagnosis in children. Our findings highlight the need for standardized pediatric approaches toward aGVHD prophylaxis/treatment differentiated for malignant and nonmalignant underlying diseases.
Collapse
Affiliation(s)
- Anita Lawitschka
- SCT-Unit, St. Anna Children's Hospital, Medical University Vienna, Vienna, Austria
| | - Giovanna Lucchini
- Stem Cell Transplantation Department, Great Ormond Street Hospital, London, UK
| | - Brigitte Strahm
- Division of Pediatric Hematology, and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Centre, University of Freiburg, Freiburg im Breisgau, Germany
| | - Jean-Hugues Dalle
- Hemato-Immunology Department, Robert Debré Hospital, Paris, France.,Immuno-Hematology Unit, Robert Debré Hospital, Paris 7 - Paris Diderot University, Paris, France
| | - Adriana Balduzzi
- Clinica Pediátrica, Università degli Studi di Milano Bicocca, Monza, Italy
| | - Brenda Gibson
- Brenda E.S. Gibson, Royal Hospital for Sick Children, Glasgow, UK
| | | | - Jacek Wachowiak
- Department of Pediatric Oncology, Hematology and Transplantology, University of Medical Sciences, Poznan, Poland
| | - Arnaud Dalissier
- EBMT Pediatric Diseases Working Party Office, Hematology and Cell Therapy Department, Saint-Antoine Teaching Hospital, Paris, France
| | - Kim Vettenranta
- Hospital for Children & Adolescents, University of Helsinki, Helsinki, Finland
| | - Isaac Yaniv
- Department of Pediatric Haematology Oncology and BMT Unit, Sackler Faculty of Medicine, Schneider Children's Medical Center of Israel, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Victoria Bordon
- Pediatric Hematology, Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Dorothea Bauer
- SCT-Unit, St. Anna Children's Hospital, Medical University Vienna, Vienna, Austria
| | - Peter Bader
- Division for Stem Cell Transplantation, University Children's Hospital, Frankfurt/Main, Germany
| | - Roland Meisel
- Division of Pediatric Stem Cell Therapy, Clinic for Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Christina Peters
- SCT-Unit, St. Anna Children's Hospital, Medical University Vienna, Vienna, Austria
| | - Selim Corbacioglu
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Hospital Regensburg, Regensburg, Germany
| | | |
Collapse
|
12
|
Amin R, He R, Gupta D, Zheng W, Burmakin M, Mohammad DK, DePierre JW, Sadeghi B, Olauson H, Wernerson A, El-Andaloussi S, Hassan M, Abedi-Valugerdi M. The kidney injury caused by the onset of acute graft-versus-host disease is associated with down-regulation of αKlotho. Int Immunopharmacol 2020; 78:106042. [DOI: 10.1016/j.intimp.2019.106042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/24/2019] [Accepted: 11/08/2019] [Indexed: 12/11/2022]
|
13
|
Abstract
Graft‐versus‐host disease (GvHD) is an important complication that can be observed after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Acute GvHD (aGvHD) is seen after allo-HSCT and the incidence of aGvHD is around 30%-50%. aGvHD prophylaxis is essential in patients undergoing allo-HSCT. Initial therapy for aGvHD is steroids. Prognosis is poor in aGvHD patients not responding to steroids. In this article, the pathobiology, clinical findings, prophylaxis, and treatment of aGvHD will be summarized.
Collapse
Affiliation(s)
- Elifcan Aladağ
- Hacettepe University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| | - Engin Kelkitli
- Ondokuz Mayıs University Faculty of Medicine, Department of Hematology, Samsun, Turkey
| | - Hakan Göker
- Hacettepe University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| |
Collapse
|
14
|
Durand N, Russell A, Zubair AC. Effect of Comedications and Endotoxins on Mesenchymal Stem Cell Secretomes, Migratory and Immunomodulatory Capacity. J Clin Med 2019; 8:jcm8040497. [PMID: 30979082 PMCID: PMC6517980 DOI: 10.3390/jcm8040497] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are becoming an increasingly popular therapeutic option among patients with a broad range of ailments to modulate immunity and induce regeneration. The majority of patients receiving these MSC therapies are on concurrent medication or have ongoing infection. In the present study, we examined the effect of immunosuppressive drugs and lipopolysaccharides (LPS)/endotoxins on the secretory profile, migration towards site of injury, and suppression of lymphocyte proliferation of bone marrow-derived MSCs (BMSCs). Generally, LPS coculture augmented the secretory capacity of BMSCs while exposure to immunosuppressive drugs resulted primarily in no change or attenuated secretion, with some cases of increased secretion, dependent on the cytokine assayed. Among the immunosuppressants evaluated, Hydrocortisone had the most widespread inhibitory effect, while LPS from E. coli O111:B4 had the most potent stimulatory effect. In addition, we also showed that Hydrocortisone or LPS from E. coli O111:B4 affected the migratory and immunosuppressive capacity of BMSCs. Following simulation with Hydrocortisone, BMSC migration was attenuated, and immunosuppressive capacity against T cell proliferation was enhanced, however, the opposite effects were seen with LPS from E. coli O111:B4. Our data suggests that the clinical outcomes of MSC-based therapy are affected by the use of immunosuppressive medication or the presence of endotoxemia in patients.
Collapse
Affiliation(s)
- Nisha Durand
- Transfusion Medicine, Department of Laboratory Medicine and Pathology and Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL 32224, USA.
| | | | | |
Collapse
|
15
|
Thangavelu G, Blazar BR. Achievement of Tolerance Induction to Prevent Acute Graft-vs.-Host Disease. Front Immunol 2019; 10:309. [PMID: 30906290 PMCID: PMC6419712 DOI: 10.3389/fimmu.2019.00309] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/06/2019] [Indexed: 01/04/2023] Open
Abstract
Acute graft-vs.-host disease (GVHD) limits the efficacy of allogeneic hematopoietic stem cell transplantation (allo-HSCT), a main therapy to treat various hematological disorders. Despite rapid progress in understanding GVHD pathogenesis, broad immunosuppressive agents are most often used to prevent and remain the first line of therapy to treat GVHD. Strategies enhancing immune tolerance in allo-HSCT would permit reductions in immunosuppressant use and their associated undesirable side effects. In this review, we discuss the mechanisms responsible for GVHD and advancement in strategies to achieve immune balance and tolerance thereby avoiding GVHD and its complications.
Collapse
Affiliation(s)
- Govindarajan Thangavelu
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
16
|
Hematopoietic Cell Transplantation for Paroxysmal Nocturnal Hemoglobinuria in the Age of Eculizumab. Biol Blood Marrow Transplant 2019; 25:1331-1339. [PMID: 30711779 DOI: 10.1016/j.bbmt.2019.01.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/28/2019] [Indexed: 01/07/2023]
Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is a rare, acquired clonal hematopoietic cell disease characterized by the destruction of hematopoietic cells through activation of the complement system with manifestations that can be life-threatening including hemolysis, thrombosis, and marrow failure. Allogeneic hematopoietic cell transplantation (HCT) remains the sole cure for PNH, but eculizumab, a terminal complement inhibitor of C5, has been used to prevent complement-mediated hemolysis in patients with PNH since its approval by the Food and Drug Administration in 2007. We examined outcomes of HCT in patients with PNH to evaluate the effects of disease subtype, conditioning intensity, and eculizumab use either pre-HCT or post-HCT. Fifty-five patients with a diagnosis of PNH underwent at least 1 HCT, with 4 patients requiring a second HCT for graft failure. The median age at the time of first HCT was 30.0 years (range, 4.2 to 66.9 years). Seventeen patients (30.9%) had classical PNH, and the remaining 38 patients had PNH associated with another marrow disorder (aplastic anemia in 26 of the 38). Indications for HCT included pancytopenia in 47.3% of the patients, myeloid malignancy (myelodysplastic syndrome, myeloproliferative neoplasm, or acute myelogenous leukemia) in 21.8%, recurrent hemolysis in 20.0%, and thrombosis in 10.9%. Of the 55 first HCTs, 26 were performed with myeloablative conditioning, 27 were performed with reduced-intensity conditioning, and 2 sets of identical twins underwent HCT without any conditioning. Donor types included HLA-matched related in 38.2%, HLA-matched unrelated in 34.5%, single HLA-allele mismatched unrelated in 16.4%, umbilical cord blood in 5.5%, syngeneic in 3.6%, and HLA-haploidentical in 1.8%. The median duration of follow-up in surviving patients was 6.1 years (range, 2.1 to 46.1 years) after first HCT. The median time to neutrophil and platelet engraftment was 17 days and 19 days, respectively; all but 2 patients (96.3%) had sustained engraftment. Overall survival was 70% at 5 years. Neither the choice of conditioning intensity nor PNH subtype affected survival. Nineteen patients died during follow-up, including 12 patients before day +365. Six patients received treatment with eculizumab before HCT, and 2 were treated after HCT. All patients treated with eculizumab were alive at a median follow-up of 2.3 years (range, .2 to 6.9 years). Both patients treated with eculizumab after HCT had minimal to no acute GVHD (aGVHD), with grade I skin aGVHD in 1 patient and no aGVHD in the other patient, and no chronic GVHD at 2.1 and 4.1 years post-HCT, respectively. With the approval of eculizumab, the indications for HCT include persistent hemolysis, persistent thrombosis, and associated marrow failure. Administration of eculizumab before and after HCT warrants further study, particularly considering our observation of minimal to no GVHD in 2 patients who received eculizumab after HCT.
Collapse
|
17
|
Graves SS, Parker MH, Storb R. Animal Models for Preclinical Development of Allogeneic Hematopoietic Cell Transplantation. ILAR J 2018; 59:263-275. [PMID: 30010833 PMCID: PMC6808062 DOI: 10.1093/ilar/ily006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 01/22/2018] [Accepted: 06/15/2018] [Indexed: 02/01/2023] Open
Abstract
Since its inception in the 1950s, hematopoietic cell transplantation (HCT) has become a highly effective clinical treatment for malignant and nonmalignant hematological disorders. This milestone in cancer therapy was only possible through decades of intensive research using murine and canine animal models that overcame what appeared in the early days to be insurmountable obstacles. Conditioning protocols for tumor ablation and immunosuppression of the recipient using irradiation and chemotherapeutic drugs were developed in mouse and dog models as well as postgrafting immunosuppression methods essential for dependable donor cell engraftment. The random-bred canine was particularly important in defining the role of histocompatibility barriers and the development of the nonmyeloablative transplantation procedure, making HCT available to elderly patients with comorbidities. Two complications limit the success of HCT: disease relapse and graft versus host disease. Studies in both mice and dogs have made significant progress toward reducing and to some degree eliminating patient morbidity and mortality associated with both disease relapse and graft versus host disease. However, more investigation is needed to make HCT more effective, safer, and available as a treatment modality for other non-life-threatening diseases such as autoimmune disorders. Here, we focus our review on the contributions made by both the murine and canine models for the successful past and future development of HCT.
Collapse
Affiliation(s)
- Scott S Graves
- Clinical Research Division of the Fred Hutchinson Cancer Research Center in Seattle, Washington
| | - Maura H Parker
- Clinical Research Division of the Fred Hutchinson Cancer Research Center in Seattle, Washington
| | - Rainer Storb
- Clinical Research Division of the Fred Hutchinson Cancer Research Center in Seattle, Washington
- Department of Medicine, University of Washington in Seattle, Washington
| |
Collapse
|
18
|
Zitzer NC, Garzon R, Ranganathan P. Toll-Like Receptor Stimulation by MicroRNAs in Acute Graft-vs.-Host Disease. Front Immunol 2018; 9:2561. [PMID: 30455702 PMCID: PMC6230675 DOI: 10.3389/fimmu.2018.02561] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 10/17/2018] [Indexed: 12/20/2022] Open
Abstract
Acute graft-vs.-host disease (aGVHD) is a frequent complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT), accounting for substantial morbidity and mortality associated with this treatment modality. The pathogenesis of aGVHD involves a complex cascade of humoral and cellular interactions in which donor T cells target HLA mismatched host tissues, causing tissue injury through secretion of pro-inflammatory cytokines and induction of direct cytotoxicity. Toll-like receptors (TLRs) are key components of the innate immune system that recognize endogenous danger-associated molecular patterns (DAMPs) and exogenous pathogen-associated molecular patterns (PAMPs). Patients receiving conditioning chemotherapy and/or whole-body irradiation prior to all-HSCT are prone to gastrointestinal damage and translocation of microbiota across compromised intestinal epithelium, resulting in release of PAMPs and DAMPs. These “danger signals” play critical roles in disease pathogenesis by both initiating and propagating aGVHD through dendritic cell maturation and alloreactive T cell responses. There are 10–15 TLRs identified in mammalian species, a subset of which recognize single-stranded RNA (ssRNA) and serve as a key component of viral immunity. Recently, ssRNAs other than those of viral origin have been investigated as potential ligands of TLRs. MicroRNAs (miRs) are short (19–24 nt) non-coding RNAs that play critical roles in a variety of diseases. While traditionally miRs post-translationally modulate gene expression, non-canonical functions such as regulating TLR stimulation by acting as TLR ligands have been described. Here, we review the role of TLRs in aGVHD pathogenesis, the function of miRs in TLR stimulation, and the recent literature describing miRs as TLR ligands in aGVHD.
Collapse
Affiliation(s)
- Nina C Zitzer
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Ramiro Garzon
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Parvathi Ranganathan
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
19
|
Villa NY, McFadden G. Virotherapy as Potential Adjunct Therapy for Graft-Vs-Host Disease. CURRENT PATHOBIOLOGY REPORTS 2018; 6:247-263. [PMID: 30595970 PMCID: PMC6290699 DOI: 10.1007/s40139-018-0186-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW This review discusses the pathophysiology, risk factors, and the advances in the prevention or treatment of graft-vs-host disease (GvHD) by exploiting adjunct virotherapy. In addition, nonviral adjunct therapeutic options for the prevention of GvHD in the context of allogeneic hematopoietic stem cell transplantation (allo-HSCT) are discussed. The role of oncolytic viruses to treat different HSCT-eligible hematological cancers is also considered and correlated with the issue of GvHD in the context of allo-HSCT. RECENT FINDINGS Emerging therapies focused on the prevention or treatment of GvHD include the use of regulatory T cells (Tregs), mesenchymal stem cells (MSCs), microbiome manipulation, B cell inhibitors, among others. Our lab and others have reported that an oncolytic DNA virus from the Poxviridae family, called myxoma virus (MYXV), not only exhibits oncolytic activity against various hematologic malignancies like multiple myeloma (MM) or acute myeloid leukemia (AML) but also, in addition, ex vivo MYXV treatment of human allogeneic-bone marrow transplants (allo-BMT), or allo-peripheral blood mononuclear cell (allo-PBMC) transplants can abrogate GvHD in xenografted mice without impairing graft-vs-tumor (GvT) effects against residual cancer. To date, this is the first and the only oncolytic virus with a dual potential of mediating oncolysis against a residual cancer target and also inhibiting or preventing GvHD following allo-HSCT. SUMMARY This review discusses how oncolytic virotherapy can be applied as a potential adjunct therapy for the potential treatment of GvHD. In addition, we highlight major emerging nonviral therapies currently studied for the treatment or prevention of GvHD. We also review the emerging oncolytic virotherapies against different hematological cancers currently eligible for allo-HSCT and highlight the potential role of the oncolytic virus MYXV to decrease GvHD while maintaining or enhancing the positive benefits of GvT.
Collapse
Affiliation(s)
- Nancy Y. Villa
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ 85287 USA
| | - Grant McFadden
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ 85287 USA
| |
Collapse
|
20
|
Watkins BK, Tkachev V, Furlan SN, Hunt DJ, Betz K, Yu A, Brown M, Poirier N, Zheng HB, Taraseviciute A, Colonna L, Mary C, Blancho G, Soulillou JP, Panoskaltsis-Mortari A, Sharma P, Garcia A, Strobert E, Hamby K, Garrett A, Deane T, Blazar BR, Vanhove B, Kean LS. CD28 blockade controls T cell activation to prevent graft-versus-host disease in primates. J Clin Invest 2018; 128:3991-4007. [PMID: 30102255 DOI: 10.1172/jci98793] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 06/26/2018] [Indexed: 12/30/2022] Open
Abstract
Controlling graft-versus-host disease (GVHD) remains a major unmet need in stem cell transplantation, and new, targeted therapies are being actively developed. CD28-CD80/86 costimulation blockade represents a promising strategy, but targeting CD80/CD86 with CTLA4-Ig may be associated with undesired blockade of coinhibitory pathways. In contrast, targeted blockade of CD28 exclusively inhibits T cell costimulation and may more potently prevent GVHD. Here, we investigated FR104, an antagonistic CD28-specific pegylated-Fab', in the nonhuman primate (NHP) GVHD model and completed a multiparameter interrogation comparing it with CTLA4-Ig, with and without sirolimus, including clinical, histopathologic, flow cytometric, and transcriptomic analyses. We document that FR104 monoprophylaxis and combined prophylaxis with FR104/sirolimus led to enhanced control of effector T cell proliferation and activation compared with the use of CTLA4-Ig or CTLA4-Ig/sirolimus. Importantly, FR104/sirolimus did not lead to a beneficial impact on Treg reconstitution or homeostasis, consistent with control of conventional T cell activation and IL-2 production needed to support Tregs. While FR104/sirolimus had a salutary effect on GVHD-free survival, overall survival was not improved, due to death in the absence of GVHD in several FR104/sirolimus recipients in the setting of sepsis and a paralyzed INF-γ response. These results therefore suggest that effectively deploying CD28 in the clinic will require close scrutiny of both the benefits and risks of extensively abrogating conventional T cell activation after transplant.
Collapse
Affiliation(s)
- Benjamin K Watkins
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Victor Tkachev
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute; The University of Washington; Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Scott N Furlan
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute; The University of Washington; Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Daniel J Hunt
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute; The University of Washington; Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Kayla Betz
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute; The University of Washington; Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Alison Yu
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute; The University of Washington; Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Melanie Brown
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute; The University of Washington; Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Nicolas Poirier
- Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire (CHU) Nantes, Nantes, France.,OSE Immunotherapeutics, Nantes, France
| | - Hengqi Betty Zheng
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute; The University of Washington; Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Agne Taraseviciute
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute; The University of Washington; Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Lucrezia Colonna
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute; The University of Washington; Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Caroline Mary
- Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire (CHU) Nantes, Nantes, France.,OSE Immunotherapeutics, Nantes, France
| | - Gilles Blancho
- Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire (CHU) Nantes, Nantes, France
| | - Jean-Paul Soulillou
- Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire (CHU) Nantes, Nantes, France
| | - Angela Panoskaltsis-Mortari
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Prachi Sharma
- Yerkes National Primate Research Center, Atlanta, Georgia, USA
| | | | | | - Kelly Hamby
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Aneesah Garrett
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Taylor Deane
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Bernard Vanhove
- Centre de Recherche en Transplantation et Immunologie, UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire (CHU) Nantes, Nantes, France.,OSE Immunotherapeutics, Nantes, France
| | - Leslie S Kean
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute; The University of Washington; Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
21
|
Rognoni C, Bertolani A, Jommi C. Budget impact analysis of rituximab biosimilar in Italy from the hospital and payer perspectives. GLOBAL & REGIONAL HEALTH TECHNOLOGY ASSESSMENT 2018. [DOI: 10.1177/2284240318784289] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Carla Rognoni
- Centre for Research on Health and Social Care Management (CERGAS), SDA Bocconi University, Milan, Italy
| | - Arianna Bertolani
- Centre for Research on Health and Social Care Management (CERGAS), SDA Bocconi University, Milan, Italy
| | - Claudio Jommi
- Centre for Research on Health and Social Care Management (CERGAS), SDA Bocconi University, Milan, Italy
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
22
|
Eddou H, Ennouhi A, Sina M, Zinebi A, El Benaye J, Moudden M, Doghmi K, Malfuson JV, Mikdame M, El Baaj M. Pyoderma gangrenosum après allogreffe de cellules souches hématopoïétiques. Ann Dermatol Venereol 2018; 145:445-450. [DOI: 10.1016/j.annder.2018.02.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 09/17/2017] [Accepted: 02/15/2018] [Indexed: 12/18/2022]
|
23
|
Ali R, Ramdial J, Algaze S, Beitinjaneh A. The Role of Anti-Thymocyte Globulin or Alemtuzumab-Based Serotherapy in the Prophylaxis and Management of Graft-Versus-Host Disease. Biomedicines 2017; 5:biomedicines5040067. [PMID: 29186076 PMCID: PMC5744091 DOI: 10.3390/biomedicines5040067] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/01/2017] [Accepted: 11/20/2017] [Indexed: 12/02/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplant is an established treatment modality for hematologic and non-hematologic diseases. However, it is associated with acute and long-term sequelae which can translate into mortality. Graft-versus-host disease (GVHD) remains a glaring obstacle, especially with the advent of reduced-intensity conditioning. Serotherapy capitalizes on antibodies which target T cells and other immune cells to mitigate this effect. This article focuses on the utility of two such agents: anti-thymocyte globulin (ATG) and alemtuzumab. ATG has demonstrated benefit in prophylaxis against GVHD, especially in the chronic presentation. However, there is limited impact of ATG on overall survival and it has little utility in the treatment context. There may be an initial improvement, particularly in skin manifestations, but no substantial benefit has been elicited. Alemtuzumab has shown benefit in both prophylaxis and treatment of GVHD, but at the consequence of a more profound immunosuppressive phase, mandating aggressive viral prophylaxis. There remains heterogeneity in the doses and regimens of the agents, with no standardized protocol in place. Furthermore, it seems that once steroid-refractory GVHD has been established, there is little that can be offered to offset the ultimately dismal outcome. Here we present a systematic overview of ATG- or alemtuzumab-based serotherapy in the prophylaxis and management of GVHD.
Collapse
Affiliation(s)
- Robert Ali
- Hematology/Medical Oncology Fellow, University of Miami/Miller School of Medicine, Miami, FL 33136, USA.
| | - Jeremy Ramdial
- Hematology/Medical Oncology Fellow, University of Miami/Miller School of Medicine, Miami, FL 33136, USA.
| | - Sandra Algaze
- Internal Medicine Residency Program, University of Miami/Miller School of Medicine, Miami, FL 33136, USA.
| | - Amer Beitinjaneh
- Associate Professor of Medicine, Stem Cell Transplant and Cellular Therapy Program, Sylvester Comprehensive Cancer Center, University of Miami/Miller School of Medicine; Miami, FL 33136, USA.
| |
Collapse
|
24
|
Yerushalmi R, Shem‐Tov N, Danylesko I, Shouval R, Nagler A, Shimoni A. The combination of cyclosporine and mycophenolate mofetil is less effective than cyclosporine and methotrexate in the prevention of acute graft-versus host disease after stem-cell transplantation from unrelated donors. Am J Hematol 2017; 92:259-268. [PMID: 28052467 DOI: 10.1002/ajh.24631] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/15/2016] [Accepted: 12/19/2016] [Indexed: 12/24/2022]
Abstract
Acute graft-versus-host disease (GVHD) is the major treatment-related complication after stem-cell transplantation (SCT) from unrelated-donors. Several GVHD prophylaxis regimens have been explored, but no regimen has shown superiority. We analyzed transplantation outcomes in 472 consecutive unrelated-donor SCT recipients, using cyclosporine/methotrexate (MTX, n = 314) or cyclosporine/mycophenolate-mofetil (MMF, n = 158) for GVHD prophylaxis. Neutrophil engraftment was faster after MMF, days 11 and 14, respectively (P = .001). Acute GVHD grade II-IV and III-IV occurred in 47% and 28% after MMF compared to 27% and 12% after MTX, respectively (P < .001). Nonrelapse mortality (NRM) was 44% and 24%, respectively (P < .001). Death associated with GVHD occurred in 25% and 8% (P < .0001), while other NRM causes occurred in 19% and 16%, respectively (P = .39). Relapse mortality was similar. Overall survival was better after MTX, 40% and 29%, respectively (P = .006). However, this difference had only borderline significance when adjusting for differences in patient characteristics (HR, 1.3, P = .08). To minimize potential selection bias we analyzed outcomes on the basis of an intention-to-treat like analysis. During the years 2008-2009, the leading GVHD prophylaxis regimen for unrelated-donor SCT included MMF (89% of transplants). During the other periods, MTX was used predominantly (82% of transplants). The two periods were otherwise well-matched. Acute GVHD occurred more often in 2008-2009. Death associated with GVHD occurred more often, while other NRM causes occurred less often resulting in similar NRM and overall survival. In conclusion, cyclosporine/MMF is associated with faster engraftment and possibly with less organ toxicity than cyclosporine/MTX. However, it is associated with increased rates of acute GVHD and GVHD-associated deaths.
Collapse
Affiliation(s)
- Ronit Yerushalmi
- The Division of Hematology and Bone Marrow TransplantationTel‐Aviv UniversityChaim Sheba Medical Center, Tel‐Hashomer Israel
| | - Noga Shem‐Tov
- The Division of Hematology and Bone Marrow TransplantationTel‐Aviv UniversityChaim Sheba Medical Center, Tel‐Hashomer Israel
| | - Ivetta Danylesko
- The Division of Hematology and Bone Marrow TransplantationTel‐Aviv UniversityChaim Sheba Medical Center, Tel‐Hashomer Israel
| | - Roni Shouval
- The Division of Hematology and Bone Marrow TransplantationTel‐Aviv UniversityChaim Sheba Medical Center, Tel‐Hashomer Israel
| | - Arnon Nagler
- The Division of Hematology and Bone Marrow TransplantationTel‐Aviv UniversityChaim Sheba Medical Center, Tel‐Hashomer Israel
| | - Avichai Shimoni
- The Division of Hematology and Bone Marrow TransplantationTel‐Aviv UniversityChaim Sheba Medical Center, Tel‐Hashomer Israel
| |
Collapse
|
25
|
Pharmacokinetics, Pharmacodynamics, and Pharmacogenomics of Immunosuppressants in Allogeneic Hematopoietic Cell Transplantation: Part II. Clin Pharmacokinet 2016; 55:551-93. [PMID: 26620047 DOI: 10.1007/s40262-015-0340-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Part I of this article included a pertinent review of allogeneic hematopoietic cell transplantation (alloHCT), the role of postgraft immunosuppression in alloHCT, and the pharmacokinetics, pharmacodynamics, and pharmacogenomics of the calcineurin inhibitors and methotrexate. In this article (Part II), we review the pharmacokinetics, pharmacodynamics, and pharmacogenomics of mycophenolic acid (MPA), sirolimus, and the antithymocyte globulins (ATG). We then discuss target concentration intervention (TCI) of these postgraft immunosuppressants in alloHCT patients, with a focus on current evidence for TCI and on how TCI may improve clinical management in these patients. Currently, TCI using trough concentrations is conducted for sirolimus in alloHCT patients. Several studies demonstrate that MPA plasma exposure is associated with clinical outcomes, with an increasing number of alloHCT patients needing TCI of MPA. Compared with MPA, there are fewer pharmacokinetic/dynamic studies of rabbit ATG and horse ATG in alloHCT patients. Future pharmacokinetic/dynamic research of postgraft immunosuppressants should include '-omics'-based tools: pharmacogenomics may be used to gain an improved understanding of the covariates influencing pharmacokinetics as well as proteomics and metabolomics as novel methods to elucidate pharmacodynamic responses.
Collapse
|
26
|
Pharmacokinetics, Pharmacodynamics and Pharmacogenomics of Immunosuppressants in Allogeneic Haematopoietic Cell Transplantation: Part I. Clin Pharmacokinet 2016; 55:525-50. [PMID: 26563168 DOI: 10.1007/s40262-015-0339-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although immunosuppressive treatments and target concentration intervention (TCI) have significantly contributed to the success of allogeneic haematopoietic cell transplantation (alloHCT), there is currently no consensus on the best immunosuppressive strategies. Compared with solid organ transplantation, alloHCT is unique because of the potential for bidirectional reactions (i.e. host-versus-graft and graft-versus-host). Postgraft immunosuppression typically includes a calcineurin inhibitor (cyclosporine or tacrolimus) and a short course of methotrexate after high-dose myeloablative conditioning, or a calcineurin inhibitor and mycophenolate mofetil after reduced-intensity conditioning. There are evolving roles for the antithymyocyte globulins (ATGs) and sirolimus as postgraft immunosuppression. A review of the pharmacokinetics and TCI of the main postgraft immunosuppressants is presented in this two-part review. All immunosuppressants are characterized by large intra- and interindividual pharmacokinetic variability and by narrow therapeutic indices. It is essential to understand immunosuppressants' pharmacokinetic properties and how to use them for individualized treatment incorporating TCI to improve outcomes. TCI, which is mandatory for the calcineurin inhibitors and sirolimus, has become an integral part of postgraft immunosuppression. TCI is usually based on trough concentration monitoring, but other approaches include measurement of the area under the concentration-time curve (AUC) over the dosing interval or limited sampling schedules with maximum a posteriori Bayesian personalization approaches. Interpretation of pharmacodynamic results is hindered by the prevalence of studies enrolling only a small number of patients, variability in the allogeneic graft source and variability in postgraft immunosuppression. Given the curative potential of alloHCT, the pharmacodynamics of these immunosuppressants deserves to be explored in depth. Development of sophisticated systems pharmacology models and improved TCI tools are needed to accurately evaluate patients' exposure to drugs in general and to immunosuppressants in particular. Sequential studies, first without and then with TCI, should be conducted to validate the clinical benefit of TCI in homogenous populations; randomized trials are not feasible, because there are higher-priority research questions in alloHCT. In Part I of this article, we review the alloHCT process to facilitate optimal design of pharmacokinetic and pharmacodynamics studies. We also review the pharmacokinetics and TCI of calcineurin inhibitors and methotrexate.
Collapse
|
27
|
The addition of ofatumumab to the conditioning regimen does not improve the outcome of patients with high-risk CLL undergoing reduced intensity allogeneic haematopoietic cell transplantation: a pilot trial from the GETH and GELLC (CLL4 trial). Bone Marrow Transplant 2016; 51:1404-1407. [PMID: 27214073 DOI: 10.1038/bmt.2016.145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
28
|
Mossoba ME, Halverson DC, Kurlander R, Schuver BB, Carpenter A, Hansen B, Steinberg SM, Ali SA, Tageja N, Hakim FT, Gea-Banacloche J, Sportes C, Hardy NM, Hickstein DD, Pavletic SZ, Khuu H, Sabatini M, Stroncek D, Levine BL, June CH, Mariotti J, Rixe O, Fojo AT, Bishop MR, Gress RE, Fowler DH. High-Dose Sirolimus and Immune-Selective Pentostatin plus Cyclophosphamide Conditioning Yields Stable Mixed Chimerism and Insufficient Graft-versus-Tumor Responses. Clin Cancer Res 2015; 21:4312-20. [PMID: 26071480 DOI: 10.1158/1078-0432.ccr-15-0340] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/26/2015] [Indexed: 02/07/2023]
Abstract
PURPOSE We hypothesized that lymphoid-selective host conditioning and subsequent adoptive transfer of sirolimus-resistant allogeneic T cells (T-Rapa), when combined with high-dose sirolimus drug therapy in vivo, would safely achieve antitumor effects while avoiding GVHD. EXPERIMENTAL DESIGN Patients (n = 10) with metastatic renal cell carcinoma (RCC) were accrued because this disease is relatively refractory to high-dose conditioning yet may respond to high-dose sirolimus. A 21-day outpatient regimen of weekly pentostatin (P; 4 mg/m(2)/dose) combined with daily, dose-adjusted cyclophosphamide (C; ≤200 mg/d) was designed to deplete and suppress host T cells. After PC conditioning, patients received matched sibling, T-cell-replete peripheral blood stem cell allografts, and high-dose sirolimus (serum trough target, 20-30 ng/mL). To augment graft-versus-tumor (GVT) effects, multiple T-Rapa donor lymphocyte infusions (DLI) were administered (days 0, 14, and 45 posttransplant), and sirolimus was discontinued early (day 60 posttransplant). RESULTS PC conditioning depleted host T cells without neutropenia or infection and facilitated donor engraftment (10 of 10 cases). High-dose sirolimus therapy inhibited multiple T-Rapa DLI, as evidenced by stable mixed donor/host chimerism. No antitumor responses were detected by RECIST criteria and no significant classical acute GVHD was observed. CONCLUSIONS Immune-selective PC conditioning represents a new approach to safely achieve alloengraftment without neutropenia. However, allogeneic T cells generated ex vivo in sirolimus are not resistant to the tolerance-inducing effects of in vivo sirolimus drug therapy, thereby cautioning against use of this intervention in patients with refractory cancer.
Collapse
Affiliation(s)
- Miriam E Mossoba
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - David C Halverson
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Roger Kurlander
- Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, Maryland
| | - Bazetta Blacklock Schuver
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Ashley Carpenter
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Brenna Hansen
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | | | - Syed Abbas Ali
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Nishant Tageja
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Frances T Hakim
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Juan Gea-Banacloche
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Claude Sportes
- Georgia Regents University Cancer Center, Augusta, Georgia
| | - Nancy M Hardy
- University of Maryland Greenbaum Cancer Center, Baltimore, Maryland
| | - Dennis D Hickstein
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Steven Z Pavletic
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Hanh Khuu
- Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, Maryland
| | - Marianna Sabatini
- Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, Maryland
| | - David Stroncek
- Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, Maryland
| | - Bruce L Levine
- University of Pennsylvania, Abramson Family Cancer Research Center, Philadelphia, Pennsylvania
| | - Carl H June
- University of Pennsylvania, Abramson Family Cancer Research Center, Philadelphia, Pennsylvania
| | - Jacopo Mariotti
- Fondazione IRCCS Instituto Nazionale dei Tumori, Milan, Italy
| | - Olivier Rixe
- University of New Mexico Cancer Center, Albuquerque, New Mexico
| | - Antonio Tito Fojo
- Genitourinary Malignancies Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | | | - Ronald E Gress
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Daniel H Fowler
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, NIH, Bethesda, Maryland.
| |
Collapse
|
29
|
Decker B, Parker HG, Dhawan D, Kwon EM, Karlins E, Davis BW, Ramos-Vara JA, Bonney PL, McNiel EA, Knapp DW, Ostrander EA. Homologous Mutation to Human BRAF V600E Is Common in Naturally Occurring Canine Bladder Cancer--Evidence for a Relevant Model System and Urine-Based Diagnostic Test. Mol Cancer Res 2015; 13:993-1002. [PMID: 25767210 DOI: 10.1158/1541-7786.mcr-14-0689] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 03/09/2015] [Indexed: 12/19/2022]
Abstract
UNLABELLED Targeted cancer therapies offer great clinical promise, but treatment resistance is common, and basic research aimed at overcoming this challenge is limited by reduced genomic and biologic complexity in artificially induced rodent tumors compared with their human counterparts. Animal models that more faithfully recapitulate genotype-specific human pathology could improve the predictive value of these investigations. Here, a newly identified animal model for oncogenic BRAF-driven cancers is described. With 20,000 new cases in the United States each year, canine invasive transitional cell carcinoma of the bladder (InvTCC) is a common, naturally occurring malignancy that shares significant histologic, biologic, and clinical phenotypes with human muscle invasive bladder cancer. In order to identify somatic drivers of canine InvTCC, the complete transcriptome for multiple tumors was determined by RNAseq. All tumors harbored a somatic mutation that is homologous to the human BRAF(V600E) mutation, and an identical mutation was present in 87% of 62 additional canine InvTCC tumors. The mutation was also detectable in the urine sediments of all dogs tested with mutation-positive tumors. Functional experiments suggest that, like human tumors, canine activating BRAF mutations potently stimulate the MAPK pathway. Cell lines with the mutation have elevated levels of phosphorylated MEK, compared with a line with wild-type BRAF. This effect can be diminished through application of the BRAF(V600E) inhibitor vemurafenib. These findings set the stage for canine InvTCC as a powerful system to evaluate BRAF-targeted therapies, as well as therapies designed to overcome resistance, which could enhance treatment of both human and canine cancers IMPLICATIONS This study demonstrates the activating BRAF mutation (V600E), which is found in multiple human cancers, is a driver of canine InvTCC, and highlights a urine-based test for quick diagnosis.
Collapse
Affiliation(s)
- Brennan Decker
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland. University of Cambridge, Cambridge, United Kingdom
| | - Heidi G Parker
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Deepika Dhawan
- Purdue Comparative Oncology Program, Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, Indiana
| | - Erika M Kwon
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Eric Karlins
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Brian W Davis
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - José A Ramos-Vara
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana
| | - Patty L Bonney
- Purdue Comparative Oncology Program, Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, Indiana
| | - Elizabeth A McNiel
- Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts
| | - Deborah W Knapp
- Purdue Comparative Oncology Program, Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, Indiana. Purdue University Center for Cancer Research, West Lafayette, Indiana
| | - Elaine A Ostrander
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
30
|
Tailoring the GVHD prophylaxis regimen according to transplantation associated toxicities—Substituting the 3rd dose of methotrexate to mycophenolate mofetil. Leuk Res 2014; 38:913-7. [DOI: 10.1016/j.leukres.2014.05.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 05/24/2014] [Accepted: 05/27/2014] [Indexed: 12/19/2022]
|
31
|
Next generation treatment of acute graft-versus-host disease. Leukemia 2014; 28:2283-91. [PMID: 24938648 DOI: 10.1038/leu.2014.195] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 05/23/2014] [Accepted: 06/04/2014] [Indexed: 02/07/2023]
Abstract
Despite rapid increase in the utilization of allogeneic hematopoietic stem cell transplantation, non-relapse mortality and sequela from acute graft-versus-host disease (GVHD) remain principle barriers. GVHD involves complex interactions between innate and adaptive immunity, culminating in tissue damage by inflammatory mediators and cellular effectors. Recently, our understanding of the molecular intricacies of GVHD has grown tremendously. New insights into the roles played by novel cytokines, chemokines, intracellular signaling pathways, epigenetics and post-translational modifications of proteins in GVHD biology provide numerous targets that might be therapeutically exploited. This review highlights recent advances and identifies opportunities for reshaping contemporary GVHD therapeutics.
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Although allogeneic hematopoietic stem cell transplantation (allo-SCT) is potentially curative for a number of hematologic malignancies, its use is limited by the development of acute and chronic graft-versus-host disease (GVHD). This potentially fatal complication occurs in approximately 50% of allo-SCT recipients. RECENT FINDINGS The pathogenesis of acute and chronic GVHD remains poorly understood, methods to prevent it are largely unchanged over the last two decades, and response to front-line treatment with corticosteroids is suboptimal. For patients with steroid-refractory disease, response to second-line treatment is dismal. The prospective clinical studies evaluating new agents for GVHD have been hampered by the inconsistencies in design, making generalization difficult, and few multicenter studies have been conducted. SUMMARY Advances have been made over the last decade in grading both acute and chronic GVHD, with the development of biomarkers that provide improved prognostic information in acute GVHD and National Institutes of Health Consensus Criteria for improved grading of chronic GVHD. This, along with the broad understanding of the need to conduct prospective studies with uniform inclusion criteria and endpoints leading to multicenter studies, will hopefully lead to advancements in the prevention of GVHD in the near future.
Collapse
Affiliation(s)
- Samantha M Jaglowski
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | | |
Collapse
|