GM-CSF Secreting Leukemia Cell Vaccination after Allogeneic Reduced Intensity Hematopoietic Stem Cell Transplantation for Advanced Myeloid Malignancies

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 825-825 ◽  
Author(s):  
Vincent Ho ◽  
Glenn Dranoff ◽  
Haesook Kim ◽  
Matthew Vanneman ◽  
Mildred Pasek ◽  
...  

Abstract Studies have shown that leukemia specific donor immune responses can be elicited by cancer vaccination after allogeneic SCT. Given the likelihood of disease progression in patients with active leukemia at the time of transplant, potential anti-tumor vaccines must be administered early post-transplant if they are to exert a meaningful effect. Early vaccination after SCT could capitalize upon the rapid homeostatic lymphoid expansion associated with post conditioning lymphopenia. However, there is concern about the efficacy of vaccinations early after allogeneic transplant when patients remain on immune suppression to prevent GVHD. GVAX, a cancer vaccine composed of leukemia cells genetically modified to secrete GM-CSF, has demonstrated activity in MDS and AML. We completed a trial investigating the feasibility and safety of administering GVAX early after allogeneic HLA matched reduced intensity conditioning (RIC) SCT for patients with MDS-RAEB or active AML. Prior to SCT, autologous myeloblasts were harvested and transfected with an adenovirus vector bearing the GM-CSF gene to generate the GVAX vaccine. Conditioning consisted of fludarabine 30mg/m2/d IV × 4, and busulfan 0.8mg/kg IV q12H × 8 doses prior to allogeneic PBSC infusion. GVHD prophylaxis included tacrolimus and mini-methotrexate. Vaccination started between day +30 to +45 post SCT if there was adequate count recovery and no grade II–IV acute GVHD. GVAX was administered SC/ID weekly × 3 doses, then q2 wks × 3 doses. Taper of tacrolimus began after vaccine completion (» d+120). Twenty four patients (13 URD, 11 MRD) were transplanted: 16 AML, 6 MDS/RAEB, 2 CML myeloid blast crisis. Median age was 62 (range, 41–71 years). Median marrow blast content at transplant was 22% (range, 6–91%). GVAX was successfully generated for all patients. Of the 24 patients transplanted, 9 could not start vaccine due to rapid leukemia progression (4); acute GVHD requiring systemic steroids (3); sepsis (1) and IPS (1); Among the 15 patients who started vaccination per protocol, 10 completed all 6 vaccines. Mild injection site reaction with induration, erythema, and pruritus was the only common side effect. After vaccination, 3/15 patients developed grade II acute GVHD and 7/15 had cGVHD. Relapse free (RFS) and overall survival (OS) at 2 years for patients who started GVAX were 46% and 56%, respectively. This is superior to the 2-yr DFS and OS of 12% and 16% (p=0.02), respectively, in a matched cohort of 34 patients with the same disease receiving RIC SCT during the same time period. Among the patients who completed all 6 vaccines, 9/10 remain in complete remission (6 AML, 3 MDS-RAEB) with median follow-up of 22.5 months post SCT (range, 7–38 mos). Three patients with disease relapse/progression early post SCT entered CR after vaccination and taper of tacrolimus. Pathologic examination of vaccination and leukemia cell DTH sites revealed significant infiltration with inflammatory cells and eosinophils in all patients who responded. Concordant with prior studies showing that anti-cancer activity after GM-CSF secreting tumor cell vaccines is associated with NKG2D-target-cell interactions mediated by NK and NK-T cells, our immunologic studies revealed progressively decreasing levels of soluble NKG2D ligands in patients with sustained remission after GVAX. Our results demonstrate that GVAX vaccination early after RIC SCT elicits important biologic activity despite administration during full immune suppression with tacrolimus. Given that all of the patients had active disease at transplant and received a reduced intensity conditioning regimen, we would have expected few to enter complete and sustained remission. Our encouraging results suggest GVAX vaccination is safe and may have anticancer activity in patients with MDS/AML after allogeneic SCT.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 5311-5311
Author(s):  
Xiaohua Chen ◽  
Gregory A. Hale ◽  
Raymond C. Barfield ◽  
Ely Benaim ◽  
Wing H. Leung ◽  
...  

Abstract Haploidentical hematopoietic stem cell transplantation (HaploHSCT) from a mismatched family member (MMFM) donor offers an alternative option for patients who lack an HLA-matched donor. The main obstacles to successful haploidentical hematopoietic stem cell transplantation from a mismatched family member donor are delayed immune reconstitution, vulnerability to infections, and severe graft-versus-host disease (GvHD). Method: We designed a reduced-intensity conditioning regimen that excluded total body irradiation and anti-thymocyte globulin. The graft was immunomagnetically depleted of CD3+ T-cells (CD3 negative selection) and contained a large number of both CD34+ and CD34− stem cells and most other immune cells especailly NK cells. This protocol was used to treat 22 pediatric patients with refractory hematologic malignancies. Results and Discussion: After transplantation, 91% of the patients achieved full donor chimerism. They also showed rapid recovery of CD3+ T-cells, T-cell receptor excision circle counts, TCRβ repertoire diversity and NK-cells during first four months post-transplantation. The incidence and extent of viremia were limited and no lethal infection was seen. Only 9% of patients had grade 3 acute GvHD, while 27% patients had grade 1 and another 27% had grade 2 acute GvHD. This well-tolerated regimen appears to accelerate immune recovery and shorten the duration of early post-transplant immunodeficiency, thereby reducing susceptibility to viral infections. Rapid T-cell reconstitution, retention of NK-cells in the graft, and induction of low grade GvHD may also enhance the potential anti-cancer immune effect.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 873-873
Author(s):  
Andrea Toma ◽  
Marie-Lorraine Balère-Appert ◽  
Jean-Michel Boiron ◽  
Pierre Bordigoni ◽  
Gerard Socie ◽  
...  

Abstract Abstract 873 The use of peripheral blood stem cells (PBSC) for hematopoietic stem cell transplantation (HSCT) is associated with a higher risk of chronic graft versus host disease (GvHD) but its impact on survival is not clear since it may favor a greater graft versus leukemia (GvL) effect. However, in the context of HSCT from unrelated donors (UD), the balance between GvH and GvL may differ from the context of sibling donors and thus the use of PBSC may be deleterious. In this retrospective study, we analyzed 103 patients from the french registry who received a graft from an UD after a reduced intensity conditioning regimen (RIC) to evaluate the role of various parameters including the source of stem cells on the outcome. Seventy-one D/R pairs (69%) were 10/10 HLA match at the allelic level. Mismatches concerned 5, 6, 15, 2 and 7 D/R pairs for HLA-A, -B, -C, -DRB1 and -DQB1, respectively. The median age was 46 years (18-67). All patients had hematologic malignancies: AL (n=35), MM (n=18), CLL (n=5), NHL (n=11), HD (n=9), CML (n=12), MDS (n=9), and MPS (n=4). 39% of the patients were in an advanced phase of the disease at time of HSCT. The conditioning regimen was Fluda/TBI 2Gys for 26 patients, Bu/Fluda/ATG for 24 patients, Fluda/Melph for 16 patients and others for 37 patients. Overall, anti-thymocytes globulins (ATG) were part of the conditioning regimen for 77% of patients. The source of stem cells was PBSC for 65 patients and bone marrow (BM) for 38 patients. The median follow up of the cohort is 61,3 months (1,2-113,7). The results showed that 95% of patients engrafted. Five patients did not engraft (4 in the BM group and 1 in the PBSC group). Acute GvHD grade II to IV and grade III/IV occurred in 47% and 19% of patients, respectively. The risk of developing chronic GvHD was 49% at 2 years. Overall survival (OS) was 36% at five years. The median disease free survival (DFS) was 55 months among the 36 patients alive. We performed univariate and multivariate analysis of factors susceptible to impact on GvHD and survival. The multivariate analysis included the impact of HLA mismatch, disease status, diagnosis, source of stem cells, patient's and donor's ages. This multivariate analysis performed on the global population shows a trend towards an improved OS with the use of BM instead of PBSC. However, when focusing the multivariate analysis on the 71 patients transplanted with a 10/10 match donor, the most potent factor influencing the outcome is the use of BM which is associated with an improved OS (p=0.03) and DFS (p=0.02), less acute GvHD grade II-IV (p=0.05), or grade III/IV (p=0.05) and less chronic GvHD (p=0.05). These results suggest that the use of BM as the source of stem cells should be reconsidered in the context of matched UD after RIC transplantation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3869-3869
Author(s):  
Kinjal Sanghavi ◽  
Anthony C. Wiseman ◽  
Qing Cao ◽  
Erica D. Warlick ◽  
Claudio G Brunstein ◽  
...  

Abstract Fludarabine (FLU) is a chemotherapeutic and immunosuppressive agent used in reduced intensity conditioning (RIC) regimens prior to hematopoietic stem cell transplantation. High exposure to F-ara-A, the active metabolite of FLU, has been associated with more treatment related mortality (TRM). No standard dosing models allow estimation of exposure and individualized dosing. We developed a population pharmacokinetic model for adults that estimates F-ara-A clearance (Cl) from which area under the curve (AUC0-∞) is calculated.(Clin Pharmacol Ther, Vol95, S87, March 2014) We retrospectively tested the performance of the model by associating individual predicted AUC 0-∞to clinical outcomes. TV F-ara-A CL (L/hr) = (7.04 + 3.9 * (CrCl/85) * (70/IBW)) * (IBW/70)0.75 AUC 0-∞ (μg-hr/ml) = Administered dose in F-ara-A equivalents (mg)/Estimated TV F-ara-A Cl (L/hr) Adult HCT patients (n=301, 2008 to 2014) who received IV FLU in their conditioning regimen were included. FLU doses, actual body weight, height, serum creatinine, along with TRM, engraftment and acute GVHD were reviewed. TRM was defined as death without relapse or disease progression. GVHD was staged and graded according to the standard GVHD criteria. The median age (range) was 58 yrs. (18-75). 131(43.5%) received PBSCT, 31(10.3%) bone marrow and cord blood in 139(46.2%). FLU doses were 25-40 mg/m2/day; nearly all x 5 days. The median (range) daily dose was 67 mg (38 mg-100 mg). The pharmacokinetic model was used to estimate the F-ara-A Cl and then predict their AUC0-∞ from the first dose and the total cumulative AUC0-∞. Recursive partitioning regression analysis was used to determine the optimal cut points for the first dose AUC0-∞ and cumulative AUC0-∞towards clinical outcomes. The cumulative incidence of engraftment, TRM and acute GVHD (grades II-IV and III-IV) was calculated using death prior to event as a competing risk. The proportional hazards model of Fine and Gray was used to assess the association of F-ara-A exposures towards TRM, acute GVHD and engraftment. The median (range) F-ara-A Cl, AUC0-∞ and cumulative AUC0-∞ predicted from the model were 10.92 L/hr (7.51-15.37), 4.79 μg-hr/ml (2.40 -7.52) and 23.93 μg-hr/ml (11.20-37.62), respectively. Patients with a higher first dose AUC0-∞ ≥6 μg-hr/ml had a higher incidence [95%CI] of day 100 TRM (20% [7-33%] vs 6% [4-9%], p<0.01) compared to those with <6 μg-hr/ml. Similarly, higher cumulative AUC0-∞ ≥30 μg-hr/ml was also associated with higher risk of day 100 TRM (21% [7-34%] vs 6% [3-9%], p<0.01) compared to those <30 μg-hr/ml. Both PK measures [first dose AUC0-∞ ≥6 μg-hr/ml and cumulative AUC0-∞≥30 μg-hr/ml] had higher risks of 12 month TRM (32% [16-48%] vs 15% [11-20%], p=0.02) and (34% [17-51%] vs 15% [11-20%], p=0.02), respectively. An AUC0-∞ ≥4 μg-hr/mL led only to a marginally higher risk of 6 month GVHD grades II-IV and III-IV (42% [35-48%] vs 28% [14-41%] p=0.07 and 43% [37-50] vs. 30%[16-44%], p=0.06, respectively) compared to <4 μg-hr/ml. However a cumulative AUC0-∞≥20 μg-hr/ml was associated with a significantly greater risk of acute GVHD grades II-IV (43% [36-49%] vs. 26% [14-38%], p= 0.02) and grades III-IV (44%, [38-51%] vs, 28% [15-41%], p=0.03). Lower F-ara-A clearance and higher AUC 0-∞ were associated with greater risks of TRM and acute GVHD. These data support personalizing FLU dose rather than using empirical body surface area based dosing. Targeting FLU therapy may improve outcomes after HCT. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2305-2305
Author(s):  
Mauricette Michallet ◽  
Quoc-Hung Le ◽  
Anne-Sophie Michallet ◽  
Anne Thiebaut ◽  
Emannuelle Tavernier ◽  
...  

Abstract Management of donor lymphocyte infusions (DLI) after reduced intensity conditioning regimen allogeneic hematopoietic stem cell transplantation (RICT) remains not clear and needs prospective studies. We performed a retrospective analysis on 47 patients (29 males and 18 females) who received 94 DLI among 96 patients who underwent RICT in our institution. Twenty-four patients received 1 DLI (D) [mean dose: 0.3x108 CD3 /Kg (0.01–1)] and 70 escalating doses (ED) [2 DLI (n=10), 3 DLI (n=9), 4 DLI (n=1), 5 DLI (n=1), 6 DLI (n=1) and 8 DLI (n=1)] from 0.1 to 5.6x108 CD3/Kg. The diagnosis pretransplant was acute leukemias (n=10) and myelodysplasia (n=3), chronic myeloid leukemia (n=2), Hodgkin (n=7) and non Hodgkin lymphomas (n=6), multiple myeloma (n=14) and solid tumours (n=5). Twenty patients have already been transplanted before RICT and 9 patients were in complete remission (CR), 19 in partial response (PR), 18 in evolutive disease (EDis). As hematopoietic stem cells, 22 patients received peripheral blood and 25 bone marrow and as conditioning 26 patients received busulfan, fludarabine and anti-thymocytes globulines (ATG), 13 patients TBI 2 grays associated to fludarabine (n=8) and 6 grays associated with cyclophosphamide (n=5), 5 patients received cyclophosphamide and ATG and 3 patients aracytine, idarubicine and fludarabine. After transplant and before any DLI, 21 developped acute GVHD (11 grade I, 8 grade II and 2 grade III). The indications and results after DLI are given in the Table 1. Only 4 patients received DLI for two different indications within time: 2 for relapse firstly and partial chimerism secondly and 2 according to protocol requirement firstly and for relapse secondly. After DLI, we noted 18 acute GVHD (7 grade I, 4 grade II, 6 grade III and 1 grade IV), 9 were resolutive after specific therapy and 18 patients developed chronic GVHD (13 limited and 5 extensive). Probability of overall survival at 2 years of patients who underwent RICT including DLI (n=47) was significantly better than for patients undergoing RICT without DLI (n=57) [43% (95%CI 30.5–60.4) vs 31.5 (95%CI 18.8–52.8) (p=0.01)] but there was no difference when we consider EFS [20.7% (95%CI 11.6–36.8) vs 21.6% (11–42.5) (p=0.49)]. We performed for patients receiving DLI a multivariate analysis stratified on diagnosis studying sex, age, status at transplant and mean dose of infused lymphocytes and we demonstrated that lymphocyte dose had a significant negative impact on EFS (HR= 1.06 95% CI 1.01–1.10) (p=0.01). To better understand the real place of DLI within allogeneic immunotherapy against malignancies, we need more details about the results of DLI in retrospective analysis but principally prospective studies in the future. TABLE 1 Indication of DLI Nb of patients Nb of DLI DLI (D) DLI (ED) Dose x 10 exp8 CD3/Kg Response to DLI Evolutive Disease N = 34 23 2.26 10 13 1.46 EDis 5 1.6 2 3 0.66 CR 6 1.5 2 4 0.45 PR Mixed Chimerism N = 7 6 1.5 5 1 0.7 Total Donor 1 1 1 0 0.7 Mixed Chimerism Protocol N = 6 1 1 1 0 0.1 CR 3 1.6 2 1 0.34 PR 2 2 1 2 0.52 EDis


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3680-3680 ◽  
Author(s):  
Vincent Ho ◽  
Glenn Dranoff ◽  
Haesook Kim ◽  
Mildred Pasek ◽  
Corey Cutler ◽  
...  

Abstract Disease relapse is a frequent cause of treatment failure in patients undergoing reduced intensity allogeneic stem cell transplantation for advanced MDS and AML. Prior studies with GVAX, a cancer vaccine composed of irradiated autologous myeloblasts modified to secrete GM-CSF, suggested anticancer activity in MDS and AML after autologous SCT or in primary therapy. We investigated the feasibility and safety of administering GVAX after allogeneic SCT. Patients with MDS-RAEB or AML with >5% marrow blasts and with a donor matched at HLA-A,B, DRB1 were eligible. Prior to transplant, autologous myeloblasts were collected from the marrow or blood and transfected with an adenovirus vector bearing the GM-CSF gene to generate the GVAX vaccine. Patients were conditioned with fludarabine 30mg/m2/d IV days -6 to -3, and busulfan 0.8mg/kg IV q12H days -6 to -3 prior to allogeneic PBSC infusion. GVHD prophylaxis included tacrolimus and mini-methotrexate. GM-CSF (Leukine) 250 mg/m2 SC QD was administered from day +1 until neutrophil engraftment. GVAX was administered SC/ID weekly for the first three doses, then q2 weeks for the last three doses starting between day +30 to +45 if there was neutrophil recovery and no grade II-IV acute GVHD. Taper of tacrolimus began after vaccine completion. Eighteen patients (10 URD, 8 MRD) have been transplanted to date: 12 AML, 4 MDS/RAEB, 2 CML myeloid blast crisis. Median age was 63 (range, 41–71 years). Median marrow blast content at transplant was 31% (range, 6–91%). GVAX was successfully generated for all 18 patients. Median vaccine cell dose was 1.0 × 107 cells (range, 0.1–1.0 ×107), and median 24-hour GM-CSF secretion by the vaccine was 8.2 ng/ml/106 cells (range 0.4 – 195). Seven patients did not initiate vaccination due to: inadequate neutrophil recovery/leukemia progression (4); acute GVHD (2); sepsis/death (1). Two patients were recently transplanted and have not reached day 30 to start vaccination. Among 9 patients who received GVAX, vaccination was well tolerated. Mild injection site reaction with induration, erythema, or pruritus was the only common side effect. After vaccination, 1 patient developed grade II skin acute GVHD and 3 had cGVHD that resolved with steroids. Donor chimerism was not adversely affected by vaccination. Six of 9 patients who started GVAX are alive at a median follow up of 7.5 months post transplant (range 1–16 mos). These include 4 patients in complete remission (3 AML, 1 MDS-RAEB) between 5 and 16 months after transplant, and 2 patients with minimal persistent AML early after SCT. Four of 5 patients who completed 6 vaccines remain progression free. Histologic examination of vaccination and leukemia cell DTH sites revealed significant infiltration with inflammatory cells and eosinophils. These preliminary results suggest GVAX vaccination is safe and may have anticancer activity in patients with MDS/AML after reduced intensity allogeneic SCT.


Author(s):  
Michael H. Albert ◽  
Mehtap Sirin ◽  
Manfred Hoenig ◽  
Fabian Hauck ◽  
Catharina Schuetz ◽  
...  

AbstractGraft failure requires urgent salvage HSCT, but there is no universally accepted approach for this situation. We investigated T-cell replete haploidentical HSCT with post-transplantation cyclophosphamide following serotherapy-based, radiation-free, reduced intensity conditioning in children with non-malignant disorders who had rejected their primary graft. Twelve patients with primary or secondary graft failure received T-cell replete bone marrow grafts from haploidentical donors and post-transplantation cyclophosphamide. The recommended conditioning regimen comprised rituximab 375 mg/m2, alemtuzumab 0.4 mg/kg, fludarabine 150 mg/m2, treosulfan 20–24 g/m2 and cyclophosphamide 29 mg/kg. After a median follow-up of 26 months (7–95), eleven of twelve patients (92%) are alive and well with complete donor chimerism in ten. Neutrophil and platelet engraftment were observed in all patients after a median of 18 days (15–61) and 39 days (15–191), respectively. Acute GVHD grade I was observed in 1/12 patients (8%) and mild chronic GVHD in 1/12 patients (8%). Viral reactivations and disease were frequent complications at 75% and 42%, respectively, but no death from infectious causes occurred. In summary, this retrospective analysis demonstrates that a post-transplantation cyclophosphamide-based HLA-haploidentical salvage HSCT after irradiation-free conditioning results in excellent engraftment and overall survival in children with non-malignant diseases.


Sign in / Sign up

Export Citation Format

Share Document