scholarly journals Co-transplantation of mesenchymal stem cells makes haploidentical HSCT a potential comparable therapy with matched sibling donor HSCT for patients with severe aplastic anemia

2020 ◽  
Vol 11 ◽  
pp. 204062072096541
Author(s):  
Zenghui Liu ◽  
Xiaoxiong Wu ◽  
Shunqing Wang ◽  
Linghui Xia ◽  
Haowen Xiao ◽  
...  

The application of haploidentical hematopoietic stem cell transplantation (HSCT) with mesenchymal stem cell (MSC) infusion as a treatment regimen for severe aplastic anemia (SAA) has been reported to be efficacious in single-arm trials. However, it is difficult to assess without comparing the results with those from a first-line, matched-sibling HSCT. Herein, we retrospectively reviewed 91 patients with acquired SAA. They received HSCT from haploidentical donors combined with MSC transfer (HID group). We compared these patients with 103 others who received first-line matched-sibling HSCT (MSD group) to evaluate relative treatment efficacy. Compared with the patients in the MSD group, those in the HID group presented with higher incidences of grades II–IV and III–IV acute graft versus host disease (aGvHD) and chronic graft versus host disease (cGvHD) ( p < 0.05). However, the incidence of myeloid and platelet engraftment, graft failure, poor graft function, and extensive cGvHD were comparable for both groups. The median follow-up was 36.6 months and the 3-year overall survival rate was similar for both groups (83.5% versus 79.1%). Univariate and multivariate analyses revealed that time intervals greater than 4 months from diagnosis to transplantation, experienced graft failure, poor graft function, or grade III–IV aGvHD were significantly associated with adverse outcomes. All HID patients received MSC co-transplantation with hematopoietic stem cells. However, the infused MSCs were derived from umbilical cord (UC-MSC group; 43 patients) or bone marrow (BM-MSC group; 48 patients) and were administered at different medical centers. We first compared the outcomes between the two groups and detected that the BM-MSC group exhibited lower incidences of grade III–IV aGvHD and cGvHD ( p < 0.05). This study suggests that co-transplantation of hematopoietic and MSCs significantly reduces the risk and incidence of graft rejection and may effectively improve overall survival in patients with SAA even in the absence of closely related histocompatible donor material.

Hemato ◽  
2021 ◽  
Vol 2 (1) ◽  
pp. 43-63
Author(s):  
Masahiro Imamura

Impaired hematopoiesis is a serious complication after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Bone marrow aplasia and peripheral cytopenias arise from primary and secondary graft failure or primary and secondary poor graft function. Chimerism analysis is useful to discriminate these conditions. By determining the pathogenesis of impaired hematopoiesis, a timely and appropriate treatment can be performed. Hematopoietic system principally consists of hematopoietic stem cells and bone marrow microenvironment termed niches. Abnormality in hematopoietic stem and progenitor cells and/or abnormality in the relevant niches give rise to hematological diseases. Allo-HSCT is intended to cure each hematological disease, replacing abnormal hematopoietic stem cells and bone marrow niches with hematopoietic stem cells and bone marrow niches derived from normal donors. Therefore, treatment for graft failure and poor graft function after allo-HSCT is required to proceed based on determining the pathogenesis of impaired hematopoiesis. Recent progress in this area suggests promising treatment manipulations for graft failure and poor graft function.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3147-3147 ◽  
Author(s):  
Roni Tamari ◽  
Sheetal Ramnath ◽  
Deborah Kuk ◽  
Craig S. Sauter ◽  
Doris M Ponce ◽  
...  

Abstract Abstract 3147 Introduction: Poor graft function (PGF) without immune rejection, defined as persistent cytopenias with hypocellular marrow and full donor myeloid chimerism, can be a life-threatening complication after allogeneic HSCT. It is commonly caused by viral infectious, myelosuppressive drugs like antivirals, and graft-vs-host disease (GvHD). Treatment options include supportive therapy with transfusions and growth factors and in severe cases administration of additional hematopoietic stem cells (HSCs) from the same donor without conditioning (stem cell boost). The incidence, natural history, and the indications for stem cell boost therapy are not well defined. Aims: To assess the incidence, etiologies, and indications for stem cell boost for PGF in a homogeneous group of patients with advanced MDS and AML who underwent TCD HSCT from matched or mismatched related or unrelated donors after conditioning with the same myeloablative regimen. Patients and methods: Poor graft function was defined as persistent neutropenia (ANC <1,000 μL and G-CSF administration x3 in 30 days), thrombocytopenia (platelets <50,000 μL or platelets transfusion × 4 in 30 days), and/or hemoglobin <8 g//dL after engraftment with hypocellular BM and full donor myeloid chimerism. Severe PGF was defined as ANC <500 μL, red cell transfusion-dependent anemia with reticulocytopenia of < 20,000 μL, and platelets <20,000 μL. The patient population in which this study was done included 42 patients enrolled between 09/2009 and 05/2012 in a phase 2 trial of palifermin peri-transplant to reduce transplant-related mortality. The median age was 57.5 years (1–65). All patients received the same myeloablative conditioning regimen with busulfan, melphalan, fludarabine, rabbit ATG and palifermin peri-transplant. G-CSF mobilized donor peripheral blood stem cells underwent CD34+ selection and depletion of T cells using CliniMACS immunomagnetic selection columns (Milteny Biotec). Donors were HLA matched (31; 13 related and 18 unrelated) or mismatched unrelated (11). Chimerism was determined in bone marrow as well as neutrophils, B cells, and T cells by short tandem repeat analysis on DNA extracted from bone marrow and peripheral blood cell subsets. Results: Forty-one patients were evaluable for this analysis; 1 patient was not included as he rejected the allograft shortly after engraftment. There were 8 cases of PGF with a cumulative incidence (CI) at 1 year of 18% (13% HLA matched, 33% HLA mismatch). The etiology was infection in 7 cases, and unknown in the 8th case. This patient presented with presumed autoimmune anemia and thrombocytopenia associated with a hypercellular marrow and did not respond to multiple lines of therapies. Her marrow became later hypocellular and met the criteria for PGF. None of the PGF cases in this series was associated with GvHD at the time of diagnosis of PGF. The infectious etiologies included: 6 viral infections and 1bacterial sepsis + myelosuppressive drugs. The most common viral etiology associated with PGF was CMV (50%). The 1-year CI of PGF in CMV seropositive patients was 25% and in CMV seronegative patients was 14%. Of note, HHV6 viremia was detected in patients with PGF. HHV6 is not routinely monitored, however, making it difficult to establish a causative role. All patients had moderate PGF at diagnosis and 3 cases had worsening of cytopenias and met the criteria for severe PGF. To date, 3 PGF patients have died from EBV-PTLD, adenovirus infection or GVHD (developed after CMV treatment with liposomal cidofovir), 3 continue to suffer from PGF and 2 patients are alive with recovered good blood counts after eradication of CMV. Of the 3 patients with persistent PGF, one received a TCD boost with no response, and 2 continued to be treated for CMV viremia. A stem cell boost was indicated if pancytopenia persisted despite eradication of cause of the PGF. In this small series, there were not enough events to evaluate association between PGF and CD34 cell dose, CD3 cell dose or day 100 T-cell chimerism. Conclusions: In this homogenous population of patients with MDS who underwent TCD allogeneic HSCT, the incidence of PGF is about 20%. The most common cause was viral infection with predominance of CMV. Therefore, strategies to prevent CMV reactivation in patients undergoing allogeneic HSCT has the potential to reduce the risk of PGF and avoid the need for infusion of additional stem cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2850-2850
Author(s):  
Veena Devi Salem Fauble ◽  
James L Slack ◽  
William Hogan ◽  
Vivek Roy ◽  
Jose Leis ◽  
...  

Abstract Abstract 2850 Myelofibrosis (MF), both primary myelofibrosis (PMF) and post essential thrombocytosis/polycythemia vera myelofibrosis (post ET/PV-MF), are chronic myeloproliferative neoplasms characterized by a progressive clinical course that leads to shortened survival. The heterogeneous nature of MF lends itself to a variable clinical course that commonly includes hepatosplenomegaly, constitutional symptoms, and progressive cytopenias. Medical therapy for myelofibrosis has been effective in palliation of common symptoms related to anemia, splenomegaly but allogeneic stem cell transplantation (ASCT) remains the only potentially curative therapeutic modality. The timing of ASCT, choice of conditioning regimen, patient selection and the impact of co-morbidities are critical to optimal use of this modality. We have retrospectively analyzed our experience for patients undergoing ASCT for primary and post ET/PV myelofibrosis at Mayo Clinic across all 3 sites: Scottsdale Arizona; Rochester, Minnesota; and Jacksonville, Florida. Methods: We conducted a retrospective review of all patients from 1992 to 2012 with PMF or post ET/PV-MF who underwent an ASCT at Mayo Clinic. This retrospective review included a waiver of informed consent and was approved by the Mayo Clinic Institutional Review Board. Patients that transformed to the MPN blast phase and individuals who underwent a second transplant for either relapse or graft failure were also included. Overall survival was estimated using Kaplan-Meier. Associations between prognostic factors and overall survival were assessed using Cox regression. Results: Baseline Patient Characteristics: Forty-eight patients (29 male) with a median age of 57 yrs (range 31–73) underwent ASCT. DIPSS-plus risk score was low in 2, intermediate 1 in 2, intermediate 2 in 9 and high in 35 patients. Disease type was PMF in 24, PV-MF in 10, ET-MF in 10, and MPN/MDS overlap in 4 pts. Eight patients evolved to the MPN-blast phase prior to transplant. JAK-2 mutation was positive in 27 patients and negative in 12 patients with JAK-2 status unknown in 9 patients. The median time from PV/ET to MF was 136 months (83–189 months) and from MF diagnosis to transplant was 59 months (3–144 months). All but 2 patients were red cell transfusion dependent prior to transplant (96%). Eight patients underwent splenectomy prior to transplant. Transplantation Characteristics: Patients who underwent ASCT received either myeloablative (MA, 13 pts) or reduced-intensity conditioning (RIC, 35 pts) regimen. The graft was from an unrelated donor in 22 pts and related donor in 26 pts with 44 being matched and 4 being mismatched. The myeloablative regimens included Bu/Cy, TBI/Cy, and Bu/Flu. The RIC regimens included flu/mel, Bu/Flu, FBM, and TBI/Flu. GVHD prophylaxis was with tacrolimus/MTX, CSA/MTX, and tacrolimus/MMF. Nineteen patients received ATG. Infectious disease prophylaxis, CMV monitoring, and additional supportive care measures were according to institutional guidelines. A majority of patients received peripheral blood stem cells with only 1 patient receiving bone marrow as the source of stem cells. Outcomes: The median time to neutrophil engraftment was 17.2 days. Two patients received a second transplant for relapsed disease. The incidence of graft failure was 10%. Graft versus host disease both acute and chronic occurred in 33 patients and 15 patients respectively. Day 100 overall survival was 92% and 2 year survival was 62% (95% CI 45–75%). There were no statistically significant associations between individual prognostic factors (DIPSS-plus classification, age, and HLA match) and survival. Conclusions: The Mayo Clinic national experience of ASCT for MF across our geographically diverse Cancer Center is encouraging. Our centers have transplanted 48 patients over the past 10 years with very favorable outcomes. Our results from a multi center practice are consistent with regards to survival, graft versus host disease, and types of transplants being performed compared to currently available published data from large single location transplant centers. This data helps confirm the prevailing knowledge that ASCT is a useful treatment. Disclosures: Mesa: Incyte: Research Funding; Lilly: Research Funding; Sanofi: Research Funding; NS Pharma: Research Funding; YM Bioscience: Research Funding.


Haematologica ◽  
2019 ◽  
Vol 105 (11) ◽  
pp. 2639-2646
Author(s):  
Maria M. Cuadrado ◽  
Richard M. Szydlo ◽  
Mike Watts ◽  
Nishil Patel ◽  
Hanna Renshaw ◽  
...  

Poor graft function is a serious complication following allogeneic hematopoietic stem cell transplantation. Infusion of CD34+-selected stem cells without pre-conditioning has been used to correct poor graft function, but predictors of recovery are unclear. We report the outcome of 62 consecutive patients who had primary or secondary poor graft function who underwent a CD34+-selected stem cell infusion from the same donor without further conditioning. Forty-seven of 62 patients showed hematological improvement and became permanently transfusion and growth factor-independent. In multivariate analysis, parameters significantly associated with recovery were shared CMV seronegative status for recipient/donor, the absence of active infection and matched recipient/donor sex. Recovery was similar in patients with mixed and full donor chimerism. Five -year overall survival was 74.4% (95% CI 59-89) in patients demonstrating complete recovery, 16.7% (95% CI 3-46) in patients with partial recovery and 22.2% (CI 95% 5-47) in patients with no response. In patients with count recovery, those with poor graft function in 1-2 lineages had superior 5-year overall survival (93.8%, 95% CI 82-99) than those with tri-lineage failure (53%, 95% CI 34-88). New strategies including cytokine or agonist support, or second transplant need to be investigated in patients who do not recover.


2018 ◽  
Vol 11 (1) ◽  
pp. 191-195 ◽  
Author(s):  
Samip Master ◽  
Ashish Dwary ◽  
Richard Mansour ◽  
Glenn M. Mills ◽  
Nebu Koshy

Eltrombopag is a thrombopoietin agonist and has been used in aplastic anemia and post-transplantation thrombocytopenia. The c-MPL receptor is present on hematopoietic stem cells. There are no reports of eltrombopag utilization for improving poor graft function in the post-transplant setting. Here were report a case of a young female with post-transplant poor graft function as evident from the low absolute neutrophil count, anemia, and thrombocytopenia on day 60. Eltrombopag was started on day 72 and resulted in improvement in all 3 cell lines. The counts continued to be stable even after eltrombopag was discontinued. The patient tolerated the drug without significant side effects for 1 year.


2020 ◽  
Vol 11 ◽  
pp. 204062072094874
Author(s):  
Juan Chen ◽  
Hongtao Wang ◽  
Jiaxi Zhou ◽  
Sizhou Feng

Poor graft function (PGF) following allogeneic hematopoietic stem-cell transplantation (allo-HSCT) is a life-threatening complication and is characterized by bilineage or trilineage blood cell deficiency and hypoplastic marrow with full chimerism. With the rapid development of allo-HSCT, especially haploidentical-HSCT, PGF has become a growing concern. The most common risk factors illustrated by recent studies include low dose of infused CD34+ cells, donor-specific antibody, cytomegalovirus infection, graft versus host disease (GVHD), iron overload and splenomegaly, among others. Because of the poor prognosis of PGF, it is crucial to uncover the underlying mechanism, which remains elusive. Recent studies have suggested that the bone marrow microenvironment may play an important role in the pathogenesis of PGF. Deficiency and dysfunction of endothelial cells and mesenchymal stem cells, elevated reactive oxygen species (ROS) levels, and immune abnormalities are believed to contribute to PGF. In this review, we also discuss recent clinical trials that evaluate the safety and efficacy of new strategies in patients with PGF. CD34+-selected stem-cell boost (SCB) is effective with an acceptable incidence of GVHD, despite the need for a second donation. Alternative strategies including the applications of mesenchymal stem cells, N-acetyl-l-cysteine (NAC), and eltrombopag have shown favorable outcomes, but further large-scale studies are needed due to the small sample sizes of the recent clinical trials.


Sign in / Sign up

Export Citation Format

Share Document