scholarly journals Differential effects of donor lymphocyte infusion upon treatment response and GVHD according to relapse level and donor sources in patients with myelodysplastic syndrome

2021 ◽  
Vol 12 ◽  
pp. 204062072110437
Author(s):  
Silvia Park ◽  
Tong Yoon Kim ◽  
Jong Hyuk Lee ◽  
Joon yeop Lee ◽  
Gi June Min ◽  
...  

Introduction: Donor lymphocyte infusion (DLI) is one of the effective options for post-transplant disease control of myelodysplastic syndrome (MDS). Its success or failure depends on the induction of antitumor immune reactions, durability of clinical responses, and severity of unwanted toxicities mainly from graft- versus-host disease (GVHD). Methods: By analyzing 61 patients receiving DLI for post-transplant MDS relapse, we assessed treatment outcomes and affecting factors, especially focusing on the level of relapse (hematological, molecular, and imminent relapse). Results: The response rate (42.1%, 36.4%, 72.7%), and overall survival (OS) at 2 years (27.8%, 45.5%, 70.1%) were different for each relapse level with imminent relapse group showing the most promising results. For OS, response to DLI or pre-DLI chemotherapy, and time to relapse were independent prognostic factors. Meanwhile, post-DLI GVHD and time to relapse were independently predictive for DLI response; post-DLI GVHD was predictive for DLI response, but not for OS, suggesting a potential detrimental impact of GVHD on survival. The incidence of GVHD and GVHD-related deaths were 37.7% and 10.0%, respectively, and CD3+ cell doses triggering GVHD tended to be lower in cases with haploidentical donor or imminent relapse. Conclusion: Despite being limited by small number of cases and its retrospective nature, this study again demonstrated the therapeutic effects of DLI in relapsed MDS, and that earlier detection and intervention at lower level relapse might possibly be associated with better results. Furthermore, we propose that tailored cell dosing schedule based on relapse level and donor source may be helpful in minimizing fatal GVHD.

2006 ◽  
Vol 6 (5p1) ◽  
pp. 933-946 ◽  
Author(s):  
Baolin Liu ◽  
Jianqiang Hao ◽  
Yisheng Pan ◽  
Bin Luo ◽  
Britt Westgard ◽  
...  

2017 ◽  
Vol 49 (6) ◽  
pp. 1388-1393
Author(s):  
D. Dondossola ◽  
M. Cavenago ◽  
B. Antonelli ◽  
P. Reggiani ◽  
U. Maggi ◽  
...  

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1650-1650
Author(s):  
Avichai Shimoni ◽  
Izhar Hardan ◽  
Noga Shem-Tov ◽  
Avital Rand ◽  
Elena Ribakovsky ◽  
...  

Abstract Relapse of AML/MDS after allogeneic SCT is associated with poor outcome. A subset of patients (pts) can be salvaged with donor lymphocyte infusion (DLI) with or without preceding chemotherapy or with a second SCT. It was previously speculated that pts given RIC may have a better chance to be salvaged than pts failing high-dose conditioning. To answer this question we retrospectively analyzed results of 171 SCTs for AML/ MDS with intravenous busulfan (ivBu) -based regimens. 58 pts were eligible for myeloablative conditioning and were given ivBu (12.8 mg/kg) and cyclophosphamide (BuCy). Among all others, 57 were given RIC consisting of fludarabine (F) and ivBu (FB2, 6.4 mg/kg) and 56 were given a modified myeloablative conditioning consisting of F and full-dose ivBu (FB4, 12.8 mg/kg). Median age was 40 (17–64), 60 (43–75) and 52 (18–66) years, respectively (p<0.001). 56% of pts had active leukemia at SCT and 47% had SCT from unrelated or mismatched donors with no difference between the 3 regimens. With a median follow-up of 31 months (4–90), 70 pts relapsed, 20 after BuCy, 25 after FB2, and 25 after FB4, cumulative incidence 38%, 49% and 48%, respectively (p=NS). The kinetics of relapse was similar among the regimens. The median time to relapse was 5.3 (1–59), 3.4 (1–34) and 2.7 (1–30) months after BuCy, FB2 and FB4, respectively (p=NS); 65%,60% and 64% of all relapses occurred within 6 months of SCT, while 20%, 12% and 8% occurred after more than 2 years, respectively. 25 pts were treated with immune-suppression withdrawal and supportive care alone, 12 pts were given DLI with or without preceding low-dose chemotherapy, 33 pts were given intensive ‘salvage’ type chemotherapy followed by donor stem-cells and lymphocytes (n=24) or a second SCT from the same (n=4) or another donor (n=5). Allocation was based on pt status and pt and physician discretion, but was not significantly different among the regimens; 60%, 44% and 40% of pts initially conditioned with BuCy, FB2 and FB4 were treated intensively, respectively (p=NS). With a median follow-up of 15 months from relapse (0.5–52), 12 pts are alive, 7 in remission, 3 with active disease and 2 still early after treatment. The most important predicting factor for survival after relapse was the duration of remission. Pts relapsing > 6 months and < 6 months after SCT had a median survival of 7.2 and 1.4 months, respectively and estimated 2-year overall survival (OS) 22% (95C.I. 2–41) and 2% (95C.I. 0–7), respectively (p< 0.001). The 2-year OS after relapse was 20%, 0% and 4% after BuCy, FB2 and FB4, respectively (p=0.04). SCT in refractory disease was also predictive of poor outcome after relapse in univariant analysis (p<0.001). Multivariable analysis determined short remission after SCT and conditioning with FB regimens as independent factors predicting short survival after relapse, with hazard ratios of 3.3 (1.9–6.3, p< 0.001) and 2.0 (1.0–4.0, p=0.04), respectively. In conclusion the notion that pts given RIC can be salvage more easily if they relapse is not substantiated and should not be a rationale to select RIC over myeloablative conditioning. Although this observation may be explained by inability of pts ineligible for myeloablative conditioning to tolerate further intensive therapies, other mechanisms related to differences in the biology of the different transplants may be possible.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3055-3055
Author(s):  
Nirali N Shah ◽  
David Loeb ◽  
Hahn Khuu ◽  
David Stroncek ◽  
Mark Raffeld ◽  
...  

Abstract Abstract 3055 Background: Treatment of relapse after allogeneic hematopoietic stem cell transplantation (alloHSCT) remains challenging. The Wilms' tumor 1 (WT1) gene product is a tumor-associated antigen that is expressed in acute leukemia and other hematologic malignancies with limited expression in normal tissues. This pilot trial incorporates antigen-specific immunotherapy and allogeneic adoptive cell transfer for pediatric and adult patients with relapsed hematologic malignancies after alloHSCT. The primary objectives are to assess safety and feasibility of a peptide-loaded donor-derived dendritic cell (DC) vaccine and donor lymphocyte infusion (DLI) designed to enhance the graft-vs -leukemia effect. Secondary objectives are to determine if immunologic and clinical responses to WT1 specific peptides can be generated by this novel vaccine strategy after alloHSCT. Design: HLA-A2+ patients with WT1-expressing hematologic malignancies that have relapsed after alloHSCT are eligible. Donor-derived DC vaccines are given every 2 weeks for 6 doses and DLI every 4 weeks for 3 doses. Peripheral blood monocyte-derived DCs are loaded with a combination of three HLA-A2 binding WT1 peptides (WT1 37–45; WT1 126–134; WT1 187–195) linked to the 11-mer HIV TAT protein transduction domain peptide (47–57) designed to enhance antigen presentation. The DCs are generated from donor peripheral blood monocytes that are separated from DLI collected by apheresis. Monocytes are incubated with GM-CSF and IL-4 followed by maturation with LPS and IFN-g. WT1 expression is assessed using immunohistochemistry and/or quantative RT-PCR. Study endpoints included toxicity, feasibility, and antigen-specific immune and clinical responses. Results: 4 patients, aged 9–19 years have been treated to date, 3 with acute lymphoblastic leukemia (ALL) and one with Hodgkin lymphoma (HL). Donors were matched siblings in 3 cases and a 10/10 HLA matched father in one case. Vaccines were successfully produced from all donors. All patients tolerated vaccine and DLI administrations well. The most common adverse events were mild, reversible pain and pruritus at vaccine administration and delayed type hypersensitivity (DTH) skin test sites. One patient developed Grade I skin GVHD that did not require treatment. Immune responses were observed in all 3 patients with ALL. ELISPOT was considered positive when it was at least two times greater than and had at least 10 spots more than background. 3 of 4 (75%) patients had positive ELISPOT responses to WT1 peptides. 3 of 4 (75%) patients had positive DTH responses to the keyhole limpet hemocyanin (KLH) control and 2 of 4 (50%) to the WT1 peptides. Median overall survival was 12 months. 1 patient remains in remission 12 months after initiation of therapy and 3 have died of disease. All donors tolerated apheresis well and there were no complications from donor procedures. Conclusions: This novel allogeneic immunotherapy regimen is feasible, well-tolerated and can induce immune responses in the allogeneic setting. Accrual is ongoing. Disclosures: Off Label Use: This is a pilot clinical trial of an investigational cancer vaccine.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3106-3106
Author(s):  
Ramzi Abboud ◽  
Jesse Keller ◽  
Michael Slade ◽  
Michael P. Rettig ◽  
Stephanie Meier ◽  
...  

Abstract Introduction: Allogeneic hematopoietic cell transplantation is a cornerstone of therapy for hematologic malignancies and often a patient's only curative intent treatment. Traditionally reserved for patients with an HLA-matched donor, advances in graft versus host disease (GVHD) prophylaxis utilizing post-transplant cyclophosphamide (PTCy) have expanded the use of haploidentical hematopoietic cell transplantation (haplo-HCT). While overall outcomes for haplo-HCT appear to be excellent, its novel approach brings toxicities that are particular to its biological and clinical milieu. The immediate post-transplant course in T cell replete haplo-HCT is often complicated by symptoms including fever, hypoxia, hypotension and organ dysfunction resembling the cytokine release syndrome (CRS) previously described in recipients of targeted cellular therapeutics such as CAR T-Cells. IL-6 is thought to be a key mediator of CRS in patients receiving novel T-cell engaging therapies, and tocilizumab has been used with success in this setting. Here we aim to describe the nature and incidence of CRS in haplo-HCT recipients, as well as its potential implications on clinical outcomes. Additionally, prospective analysis of cytokine profiles of haplo-HCT recipients and clinical responses to tocilizumab therapy are presented. Patients and Methods: We performed a retrospective review of patients who underwent haplo-HCT transplantation at our institution from July 2009 through April 2015. Patients were scored for symptoms of CRS based on established grading criteria (Lee et al, Blood 2014). Patients were stratified into three categories by grade of CRS experienced: none (grade 0), mild (grade 1-2) and severe (grade 3-4). Outcomes were assessed. A total of 84 patients were identified, 55% (46) were male, with a median age at transplant of 49 (19-73), and 49% (41) had active disease at the time of transplant. The most common diagnosis was AML (55 pts), followed by ALL (9 pts), MDS (5 pts) and NHL (2 pts). Among the patients, 26% (22 pts) had undergone prior transplant. In addition to the retrospective review, baseline and post- transplant cytokine levels were prospectively drawn in 10 patients who underwent haplo-HCT. A total of 7 additional patients who met criteria for CRS were treated prospectively with tocilizumab (dose: 4mg/kg-bw) and clinical responses were recorded. We recorded CRP levels in selected patients as part of clinical monitoring. Results: We found a high incidence, 85%, of CRS in our haplo-HCT patients. Among a total of 84 patients, 12 (14%) experienced severe CRS, 60 (72%) had mild CRS, and 12 (14%) patients had no evidence of CRS. The most common manifestations of severe CRS included: fever (100%), respiratory failure (75%), hypotension (83.3%), hepatic failure (25%) and renal failure (33.3%). The median maximum CRP (during post-transplant days 0 to 8) in all patients suffering from CRS was 155 mg/L. Of the twelve patients who suffered from severe CRS, nine (75%) died. Predicted median survival was 0.72 months for severe CRS, 12.7 months for patients with no CRS and was not reached for patients with mild CRS (fig 1). Rates of acute and chronic graft-versus-host disease did not differ by CRS status. The incidence of mild and severe CRS did not differ by ABO mismatch, age, CMV status, donor sex, T-cell or CD34 cell dose. There was no difference in rates of CRS for patients in remission versus active disease at time of transplant. There were no differences in engraftment. Cytokine profiles in haplo-HCT recipients showed significant elevation in serum IL-6 levels, most significant in patients who suffered from severe CRS (fig 2). We administered tocilizumab to 7 patients with severe CRS symptoms early after haplo-HCT (median day of treatment was day +3) resulting in the resolution of their CRS symptoms within 48-72 hours. Over the same time period CRP levels dropped below 50% of the peak value. Summary: CRS is common after T-cell replete haplo-HCT and severe CRS is potentially associated with high risk of early mortality after transplant. Cytokine profiles suggest IL-6 is a key mediator of CRS in haplo-HCT patients. Tocilizumab appears to be an effective treatment for patients who develop severe CRS early after T-cell replete haplo-HCT. Future prospective studies are warranted in studying the role of tocilizumab in the treatment and potentially prevention of severe CRS in haplo-HCT patients. Disclosures Fehniger: Celgene: Research Funding. Uy:Novartis: Research Funding. Abboud:Teva Pharmaceuticals: Research Funding; Pharmacyclics: Membership on an entity's Board of Directors or advisory committees; Novartis: Research Funding; Pfizer: Research Funding; Merck: Research Funding; Gerson Lehman Group: Consultancy.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2844-2844
Author(s):  
Yang Jo Chung ◽  
Suntae Kim ◽  
Zhenhua Zhang ◽  
Peter D. Aplan

Abstract Bone marrow (BM) contains both hematopoietic cells and non-hematopoietic, stromal cells, which support hematopoiesis. In mice and humans, CD45 is a pan-hematopoietic marker universally expressed on nucleated hematopoietic cells. Recent studies have shown that CD45-, non-hematopoietic stromal cells present in the BM of patients with MDS can display the same cytogenetic abnormalities found in the CD45+ MDS clone, suggesting that stromal cells may be part of the malignant clone in patients with MDS. Although CD45- , non-hematopoietic BM cells (such as multipotent adult progenitor cells) can produce CD45+, hematopoietic stem cells, there is little evidence that CD45+ hematopoietic cells can de-differentiate into CD45-, non-hematopoietic stromal cells. To study the possible conversion of hematopoietic (CD45+) to non-hematopoietic (CD45-) cells, we used the NUP98-HOXD13 (NHD13) mouse model of myelodysplastic syndrome (MDS). Two immortal cell lines (251 and 63B) were established from NHD13 mice with AML. These cell lines grow as a combination of adherent and suspension cells; although the suspension cells are almost entirely (>98%) CD45+, only 50% of the adherent cells are CD45+. Surprisingly, sub-culture of CD45+ suspension cells, generates CD45- adherent cells, leading to the suggestion that CD45+ hematopoietic cells can de-differentiate and produce CD45-, non-hematopoietic cells. Further immunophenotype analysis demonstrates that the CD45+ cells are also positive for myelo-monocytic markers such as CD11b, CD16/32, and F4/80, whereas the CD45- fraction is negative for these markers. To verify the conversion of CD45+ to CD45- cells at a clonal level, we sorted highly purified (>99.99%) CD45+ cells and sub-cultured CD45+ cells at a density of 1 or 10 cells per well in 96 well plates. Although there was no growth in the wells seeded at 1 cell/well, 23 out of 96 wells seeded at 10 cells/well expanded; at least 5 of these clones produced CD45- cells. Expression of the NHD13 transgene (driven by the pan-hematopoietic Vav1 promoter) was 100-fold higher in the CD45+ fraction compared to the CD45- fraction, and gene expression arrays identified differential expression of a number of hematopoietic markers, such as Pu.1 and Csf2rb. Finally, preliminary bisulfite sequencing experiments suggest that the CD45 promoter is heavily methylated in the CD45- fraction, but un-methylated in the CD45+ fraction. Taken together, these results indicate that the cell lines can cycle between non-hematopoietic and hematopoietic cells. To determine if the cell lines have retained malignant potential, we transplanted lethally irradiated recipients via intravenous injection. At 6 and 16 weeks post-transplant, there was no engraftment detected in the peripheral blood (PB). However, at 24 weeks, one of three recipients showed low level engraftment (1.7% of PB). Remarkably, this mouse became frankly leukemic 2 weeks later, with splenomegaly, anemia, monocytosis, and donor engraftment of 17-53% in the PB, BM, and spleen. To determine which cell fraction was responsible for the disease phenotype, we transplanted purified CD45+ and CD45- cells via intrafemoral (IF) injection. Interestingly, all recipients of the CD45+ cells died due to profound pancytopenia (hgb, 3.9±1.0 g/dL; WBC, 0.24±0.30 K/uL) by day 13 post-transplant; in contrast recipients of CD45- cells were healthy, with normal CBCs. Although we could not detect donor cells in recipients of CD45- cells by FACS at any time point up to 34 weeks post-transplant, engraftment of donor cells in the BM of these recipients could be detected by transgene specific genomic DNA PCR. These results indicate that the disease entity resides in the CD45+ fraction, and that long term engraftment of quiescent CD45- cells occurs. In addition, these results suggest the intriguing possibility that quiescent CD45- cells, which do not express the NHD13 transgene, can be transformed by demethylation of the CD45 promoter (and subsequent activation of the Vav promoter, which drives the NHD13 transgene). Experiments to test this hypothesis are currently in progress. Taken together, these findings suggest that malignant cells can cycle between stromal and hematopoietic cells, based at least in part on epigenetic events such as cytosine methylation, and support the hypothesis that non-hematopoietic BM cells may be an important part of the malignant clone in patients with MDS and AML. Disclosures Aplan: NIH Office of Technology Transfer: Patents & Royalties.


Blood ◽  
2012 ◽  
Vol 119 (2) ◽  
pp. 355-363 ◽  
Author(s):  
Youn H. Kim ◽  
Dita Gratzinger ◽  
Cameron Harrison ◽  
Joshua D. Brody ◽  
Debra K. Czerwinski ◽  
...  

Abstract We have developed and previously reported on a therapeutic vaccination strategy for indolent B-cell lymphoma that combines local radiation to enhance tumor immunogenicity with the injection into the tumor of a TLR9 agonist. As a result, antitumor CD8+ T cells are induced, and systemic tumor regression was documented. Because the vaccination occurs in situ, there is no need to manufacture a vaccine product. We have now explored this strategy in a second disease: mycosis fungoides (MF). We treated 15 patients. Clinical responses were assessed at the distant, untreated sites as a measure of systemic antitumor activity. Five clinically meaningful responses were observed. The procedure was well tolerated and adverse effects consisted mostly of mild and transient injection site or flu-like symptoms. The immunized sites showed a significant reduction of CD25+, Foxp3+ T cells that could be either MF cells or tissue regulatory T cells and a similar reduction in S100+, CD1a+ dendritic cells. There was a trend toward greater reduction of CD25+ T cells and skin dendritic cells in clinical responders versus nonresponders. Our in situ vaccination strategy is feasible also in MF and the clinical responses that occurred in a subset of patients warrant further study with modifications to augment these therapeutic effects. This study is registered at www.clinicaltrials.gov as NCT00226993.


Sign in / Sign up

Export Citation Format

Share Document