A Feasibility Study of Rapamycin with Hyper-CVAD Chemotherapy in Adults with Acute Lymphoblastic Leukemia (ALL) and Other Aggressive Lymphoid Malignancies and Evaluation of mTOR Signaling Using Phosphoflow

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4245-4245
Author(s):  
Emma C Scott ◽  
Alexander Perl ◽  
Selina M. Luger ◽  
Martin Carroll ◽  
Margaret Kasner

Abstract Abstract 4245 Background. Therapies for adults with acute lymphocytic leukemia (ALL) fail to cure the majority of patients. The mTOR inhibitor, rapamycin, is a potent chemosensitizer in ALL cells and combination rapamycin- methotrexate is curative in an ALL xenotransplantation model. We therefore explored the feasibility of adding rapamycin to a multi-drug ALL regimen in subjects with de novo or relapsed Philadelphia chromosome negative ALL or other aggressive lymphoid malignancies. Additionally we performed pharmacodynamic analysis of peripheral blood blasts to estimate mTOR activation at baseline and with rapamycin. Methods. Subjects were treated with rapamycin 12 mg on day 1, followed by 4 mg daily on days 2–7. To allow steady state inhibition of mTOR, systemic therapy with hyper-CVAD (A cycles) alternating with high dose methotrexate and high dose cytarabine (B cycles) was given following the day 3 rapamycin dose of each cycle, with rituximab if CD20 positive and intrathecal prophylaxis if indicated. Peripheral blood specimens were collected serially during the first cycle of chemotherapy and were aliquoted, incubated with signal modulators as controls for 30 minutes then fixed with 4% formaldehyde and permeabilized. No ficoll separation was performed. Thereafter, cell samples from all time points for a subject were thawed, denatured with ice-cold methanol, and stained for cytometer analysis using a uniform antibody cocktail including alexa fluor-488 conjugated to phosphorylated S6 kinase. Results. We report results on the first 7 subjects, median age 45, (range 26–65) of which 3 had relapsed B-precursor ALL and the remainder were newly diagnosed T lymphoblastic lymphoma, adult T- cell lymphoma (ATLL), mantle cell (MCL) and Burkitt’s lymphoma (BL). Feasibility. Thus far 16 ‘A’ and 15 ‘B’ cycles have been administered. The median time to next rapamycin was 23 days (range 19–57) and to chemotherapy was 25 days (range 21–59). Median time to recovery of absolute neutrophil count of > 500 and platelet count of >50 was 16 (range 0–27) and 17 (range 0–46) days, respectively. Two subjects never had count recovery in the setting of persistent disease. The following > grade 3 non- hematologic toxicities were observed: 5 neutropenic fevers, 5 infections, 4 non-neutropenic fevers, 1 ataxia (cytarabine related), psychosis, and hypophosphatemia. No fungal infections were noted. No treatment-related mortality has been observed. Responses. Of 7 subjects, 4 achieved complete responses (CR) after cycle 2B (MCL, BL, ATLL and T- lymphoblastic lymphoma). The ATLL and MCL patients completed 5 and 4 cycles respectively and remain in CR after allogeneic BMT. The T-lymphoblastic lymphoma and BL patients completed 8 and 7 cycles respectively and remain in CR. All three subjects with relapsed ALL were taken off study with persistent disease after 1 to 4 cycles, and have subsequently died. Pharmacokinetics. Rapamycin levels fell within the range typically targeted for transplant immunosuppression -mean (SD) 8.57 (3.33), range 4.7–14.7. Pharmacodynamics. Two of 3 subjects with pre B- ALL had sufficient peripheral blast count of >200/μ l to perform intracellular phosphoflow. Both subjects’ blasts displayed constitutive phosphorylation of the ribosomal S6 protein (25% & 31%) at baseline with maximal inhibition at 72 hours of in vivo rapamycin in the 1st sample and submaximal inhibition in the 2nd sample (2.9% & 12.4%). Rapamycin trough levels were 9.5 and 8.7 respectively. The addition of 1000nM ex vivo rapamycin produced further reduction (0.68% & 2.07%). Despite exhibiting varying degrees of rapamycin sensitivity, both subjects had persistent disease after only 1 and 2 cycles of therapy respectively. Conclusion. We show that the addition of rapamycin to Hyper-CVAD in adults with ALL and other aggressive lymphoid malignancies is feasible and results in similar toxicities to Hyper-CVAD alone without increased myelosuppression or treatment related mortality. At basal state, pS6, measured by flow cytometry, is heterogeneous in primary ALL samples and likely only demonstrable in a subset of blasts. Whole blood intracellular flow cytometry is a novel, feasible and potentially powerful technique to monitor pharmacodynamic response to novel therapeutics that inhibit mTOR signaling in ALL. Expansion of this trial to better characterize toxicity, response rates and the feasibility of performing PD/PK correlation is planned. Disclosures: Off Label Use: Rapamycin. This is FDA approved for rejection prevention in solid organ transplant. It has been used investigationaly in this study for treating ALL. Carroll:Glaxo Smith Kline, Inc.: Research Funding; Sanofi Aventis Corporation: Research Funding; TetraLogic Pharmaceuticals: Research Funding; Agios Pharmaceuticals: Research Funding.

2016 ◽  
Vol 22 (3) ◽  
pp. S397-S398
Author(s):  
Nasheed Mohammad Hossain ◽  
Patricia Lamont Kropf ◽  
Stefan Klaus Barta ◽  
Mary Ellen Martin ◽  
John Ulicny ◽  
...  

1993 ◽  
Vol 16 (5_suppl) ◽  
pp. 80-82 ◽  
Author(s):  
M. Di Nicola ◽  
S. Siena ◽  
M. Bregni ◽  
F. Peccatori ◽  
M. Magni ◽  
...  

After myeloablative regimens, combined reinfusion of peripheral blood hematopoietic circulating progenitor cells (CPC) and bone marrow, yields a very rapid hematopoietic recovery. Therefore, based on the knowledge that CPC express the CD34 and CD33 differentiation antigen, we have developed a direct immunofluorescence flow cytometry assay to detect the peak of CPC in the peripheral blood of patients treated with high dose chemotherapy and growth factors. This assay, compared to CFU-GM assay, has the following advantages: 1) easy to do 2) standardized method 3) real time information on CPC number. This work illustrates the practical aspects of this assay and substantiate the widespread use of the CD34/33 flow cytometry assay to guide harvesting of circulating hematopoietic progenitors for autologous transplantation.


Cancers ◽  
2020 ◽  
Vol 12 (6) ◽  
pp. 1678
Author(s):  
Panagiotis Karagiannis ◽  
Lena Sänger ◽  
Winfried Alsdorf ◽  
Katja Weisel ◽  
Walter Fiedler ◽  
...  

High dose chemotherapy (HDT) followed by autologous peripheral blood stem cell transplantation (ASCT) is standard of care including a curative treatment option for several cancers. While much is known about the management of patients with allogenic SCT at the intensive care unit (ICU), data regarding incidence, clinical impact, and outcome of critical illness following ASCT are less reported. This study included 256 patients with different cancer entities. Median age was 56 years (interquartile ranges (IQR): 45–64), and 67% were male. One-year survival was 89%; 15 patients (6%) required treatment at the ICU following HDT. The main reason for ICU admission was septic shock (80%) with the predominant focus being the respiratory tract (53%). Three patients died, twelve recovered, and six (40%) were alive at one-year, resulting in an immediate treatment-related mortality of 1.2%. Independent risk factors for ICU admission were age (odds ratio (OR) 1.05; 95% confidence interval (CI) 1.00–1.09; p = 0.043), duration of aplasia (OR: 1.37; CI: 1.07–1.75; p = 0.013), and Charlson comorbidity score (OR: 1.64; CI: 1.20–2.23; p = 0.002). HDT followed by ASCT performed at an experienced centre is generally associated with a low risk for treatment related mortality. ICU treatment is warranted mainly due to infectious complications and has a strong positive impact on intermediate-term survival.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1759-1759 ◽  
Author(s):  
Teresa Field ◽  
Janelle Perkins ◽  
Melissa Alsina ◽  
Ernesto Ayala ◽  
Karen Fancher ◽  
...  

Abstract Myeloablative doses of intravenous busulfan in combination with fludarabine have been employed as conditioning before hematopoietic cell allografts with reduced treatment-related toxicity and mortality. In this report, we describe the early results of a targeted busulfan pharmacokinetic dosing strategy (tBuFlu) used in combination with fludarabine before either related or unrelated grafts We treated 43 pts with tBuFlu prior to allogeneic peripheral blood stem cell transplantation. The median patient age was 48 (range 22–67) years. Patient diagnoses included AML (8 de novo, 6 with prior MDS, and 2 treatment related), MDS (6 pts), MF (5), NHL (5 pts), ALL (4 pts), CML (3 pts), CLL (2), MM (1) and PNH (1). Three patients had received a prior autologous HCT. Donors were HLA-A, B, C, DRB1, DQB1 matched siblings (24), matched unrelated donors (13), or unrelated donors mismatched for one HLA antigen (3), one HLA allele (2), or two HLA alleles (1). Fludarabine 40 mg/m2 was given intravenously daily for four days, with each infusion followed immediately by intravenous busulfan. The dose of busulfan for days 1 and 2 was 130 mg/m2. Pharmacokinetic analysis was performed after the first infusion of busulfan; in 41 pts, the goal was to adjust busulfan doses for days 3 and 4 to achieve an average targeted Css level of 800–1000 ng/ml. Levels were drawn incorrectly in 4 of these pts and doses were not changed. Twenty-five (61%) pts had their doses adjusted, increased by 42% (± 28%) in 18 and decreased by 27% (± 14%) in 7, while 12 pts had Css within the desired range without adjustment. Patients received tacrolimus and standard doses of methotrexate for GVHD prophylaxis, with the exception of three patients treated with tacrolimus and mycophenolate mofetil or cyclosporine and prednisone. Engraftment occurred in 41 (95%) pts. Median day to ANC > 500/uL was 16 days, range (range 12–28); median day to platelet count > 20,000/uL untransfused was 18 days (range 8–45). Thirteen (52%) of 25 pts followed for at least 100 days experienced acute GVHD requiring treatment. Two pts have died of transplant-related complications (aplasia and suspected fungal pneumonitis), and 4 pts have failed to achieve remission or have relapsed. Median follow-up is 115 days (range 17–361 days). The 100-day K-M estimate of survival for the whole cohort is 94%, and event-free survival 86%. The 100-day mortality in this study compares well with the 100-day mortality reported to the IBMTR for patients with AML, ALL, MDS, and CML transplanted from either HLA-matched siblings or unrelated donors. These preliminary results indicate that tBuFlu is a promising myeloablative regimen that can be utilized in older patients with low early treatment-related mortality. We plan to escalate the busulfan dose based on targeted Css to explore a potential dose-response relationship and improve control of malignancy.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 947-947
Author(s):  
Jean El-Cheikh ◽  
Elias Kfoury ◽  
Christian Chabannon ◽  
Anne-Marie Stoppa ◽  
Reda Bouabdallah ◽  
...  

Abstract Auto-SCT for MM can provide superior outcome to standard treatments. Since its introduction, auto-SCT has usually been limited to MM patients aged up to 60–65 years. However, traditional upper age limits for auto-SCT are being currently challenged along with the definition of “elderly” itself, especially that no obvious differences in MM biology has been elucidated to justify an arbitrary cut-off of 65 years. This retrospective single centre analysis assessed the outcome of 186 consecutive MM patients aged over 60 years treated with auto-SCT, with the specific aim to compare the outcome of the 82 “elderly” (age>65 y.) patients subgroup, with their 104 “younger” mates aged between 60 and 65 years treated in the same period and in the same auto-SCT program. Median age among the total 186 patients population was 64 (range, 60–77). Except for age, both groups were comparable (P=NS) as for demographic features, disease characteristics (S&D stage, monoclonal component), and prognostic factors (b2-microglobulin). The majority of patients (91%) received homogeneous “induction” VAD chemotherapy, with this being comparable between the “elderly” (87%) and “younger” (94%) group. In this population, and prior to auto-SCT, the calculated hematopoietic cell transplantation-specific comorbidity index (adapted from the Charlson Comorbidity Index) was also comparable between both groups (77% of the “younger” patients with a 0–1 index, vs. 74% in the “elderly” group; P=NS). The peripheral blood stem cells mobilization procedures (G-CSF with or without chemotherapy) were also comparable between both groups. 97% of the patients received high-dose melphalan conditioning for auto-SCT. 33% of the “younger” and 28% of the “older” group (P=NS) completed a second auto-SCT. ANC and platelets recovery were comparable between both groups (P=NS), and the median length of hospitalization for the first auto-SCT was not different between the two groups: 19 (range, 2–32) days in the “younger” group vs. 17 (range, 2–39) in the “older” group; P=NS). Infectious and other “serious” auto-SCT-related complications were also comparable between groups (P=NS). With a median follow-up of 41 (range, 5–227) months after auto-SCT, 120 patients are still alive. Disease progression (n=40; 61%) was the main cause of death, with this being comparable between both groups. Auto-SCT-related mortality was 3.8% (n=4/104) in “younger” and 3.7% (n=3/82) among “older” subjects. Comparing “younger”/”older” subjects, progression-free survival was significantly higher in the younger group (P<10e-4). However, disease response rate after the first auto-SCT was comparable (CR, VGPR and PR rates: 88% vs. 90%, P=NS), and overall survival (OS) was also comparable (57% vs. 54% at 5 years, P=NS; 32% vs. 24% at 10 years, P=NS). In a Cox multivariate analysis model, none of the relevant characteristics was shown to be a critical prognostic features for OS. Of note, age was insignificant for both OS and transplant-related mortality. We conclude that there is no clear justification for an age-discriminant policy for MM therapy. “Physiologic” aging is likely more important than “chronologic” aging. Thus, all treatment options, including auto-SCT in the “elderly” population, must be rigorously evaluated, as age does not appear to be an adverse parameter for selected MM patients receiving high-dose melphalan therapy with peripheral blood stem cell support.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3103-3103
Author(s):  
Marie Sebert ◽  
Raphaël Porcher ◽  
Marie Robin ◽  
Lionel Ades ◽  
Emmanuel Raffoux ◽  
...  

Abstract Abstract 3103 Introduction: Allogeneic Hematopoietic Stem Cell Transplantation (HSCT) provides the best chance of long-term survival for patients with intermediate or high-risk acute myeloid leukemia (AML). The major limitation of this procedure is the risk of treatment related mortality (TRM). Use of reduced intensity conditioning (RIC) regimen has become standard practice among older candidates with comorbidities. Although RIC regimen have been used for over a decade in older patients, the benefit of this approach in younger patients with AML compared with the risk of toxicity of standard regimen (MAC) is still discussed. We compared the outcomes for patients with AML over 35 years using RIC or MAC HSCT. Patients, methods, and transplantation characteristics: From January 2000 to December 2010, 132 consecutive patients older than 35 years with AML (18 secondary AML) received HSCT in our center, either from siblings (n=87) or HLA 10/10 allele-matched donors (n=45). MAC (n=72) and RIC (n=60) regimens were defined as previously described (Bacigalupo, 2009). Seventy-three patients were in first complete remission (CR1); 30% of patients had poor risk cytogenetics (MRC classification). Karnofsky performance status was scored at time of HSCT. Engraftment, acute and chronic graft-versus-host disease (GvHD), transplantation-related mortality (TRM), relapse rate as well as overall survival (OS) at 4 years were compared according to the intensity of the conditioning regimen. First a classical multivariable Cox analysis was conducted. In a second step, baseline confounding factors were adjusted for using inverse probability-of-treatment weighting (IPTW). Results of the comparison: Patient characteristics according to the intensity of the conditioning regimen were similar for AML type (de novo versus secondary), gender, karnofsky performance status, CR#, donor type and number of CD34+ infused. Particularly, cytogenetic risks were comparable in both groups. Patients were younger in the MAC group (median age 44 years [range 35 to 56 years] vs 54[37 to 66] for RIC, p<0,0001), received mainly bone marrow as source of stem cells (54% versus 2% for RIC, p<0,0001) and GvHD prophylaxis using cyclosporine plus methotrexate (89% versus 5% for RIC, p<0,0001). Moreover, ATG in the conditioning regimen (more ATG in RIC: 51 vs. 14%, p<0.0001), donor age (older for RIC: 49 vs. 39 years, p=0.002) and number of nucleated cells infused (higher in RIC: 11 vs. 4 × 108/kg, p<0.0001) were also different. The median follow-up was 47 months (10 to 134), and 25% of patients had a follow-up of at least 74 months. During evolution, all patients engrafted. The cumulative incidence (CIf) of acute GVHD grade II-IV was 49% (35% after RIC vs 61% after MAC, p=0.001). The 5-year CIf of chronic GVHD was 37% (40% after RIC vs 30% after MAC, p=0.32). During FU, 71 patients died. The 5-year CIf of TRM was 21% (13% after RIC vs 28% after MAC, p=0.009). Adjusting for cytogenetic risk, gender donor/recipient mismatch and infused nucleated cells, no difference was observed between RIC and MAC (HR 0.9, p=0.16). The 5-year CIf of relapse was 42% (51% after RIC vs 35% after MAC (p=0.22)). Adjusting for gender donor/recipient mismatch, donor/recipient CMV serostatus and infused CD34+ cells, no marked difference was observed between RIC and MAC (HR 0.8, 95%CI 0.4–1.5, p=0.50). The 5-year OS was 39% (50% after RIC vs 34% after MAC, p=0.38). Using both Cox regression and IPTW to account for imbalance in patients characteristics, similar OS was found after RIC and MAC (Figure 1), with adjusted HRs for MAC vs RIC of 0.9 (95%CI 0.4–1.8, p=0.68) with Cox regression and 0.9 (95%CI 0.4–1.8, p=0.76) with IPTW. Conclusion: In patients with AML over 35 years, MAC regimen lead to a non significant higher rate of treatment related mortality with no benefit in terms of relapse when compared with RIC regimen. Until prospective trials are completed, this study supports the use of a RIC regimen for patients with AML older than 35 years who are transplanted either from siblings or matched unrelated donors. Disclosures: Fenaux: Amgen: Honoraria, Research Funding; Celgene: Honoraria, Research Funding; Janssen: Honoraria, Research Funding; Roche: Honoraria, Research Funding; GSK: Honoraria, Research Funding; Novartis: Honoraria, Research Funding.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2273-2273
Author(s):  
Victor Jimenez-Zepeda ◽  
Peter Duggan ◽  
Paola Neri ◽  
Ahsan Chaudhry ◽  
Jason Tay ◽  
...  

Abstract Introduction Recent data suggests that bortezomib, a proteasome inhibitor, in combination with high-dose melphalan (Bor-HDM) provides with a synergistic effect able to improve the level of response for MM patients undergoing auto-SCT. In the present study, patients receiving induction followed by ASCT with Bor-HDM and HDM alone were evaluated. Methods All consecutive patients undergoing ASCT from 01/2004 to 03/2016 were evaluated. All patients received induction chemotherapy before undergoing auto-SCT. Patients received conditioning with either HDM at 200 mg/m2 (or adjusted as per renal failure) or HDM with Bortezomib (Bor-HDM). Most of patients received Bortezomib conditioning at 1.3 mg/m2. As per physician discretion, the dose of 1 mg/m2 was also employed in 30% of cases. Definitions of response and progression were used according to the EBMT modified criteria. MRD negativity was assessed by flow cytometry at day-100 post-ASCT. Results Clinical characteristics are shown in Table 1. Among 301 cases, 129 were treated with Bor-HDM while 172 patients went onto receive HDM alone as part of the conditioning regimen. Induction regimens are shown in Table 1. At the time of analysis, 83% and 58% of patients in the Bor-HDM and HDM group are still alive and 34% and 69.1% of patients have already progressed, respectively. At day-100 post ASCT, ORR of 97%, with CR/VGPR rate of 84.2% was seen in the Bor-HDM group compared to 94.2% and 68.6% in the HDM group (p=0.001). MRD negativity was higher in the Bor-HDM group (33.3%) compared to HDM (12.2%) (p=0.001). Median OS was similar for Bor-HDM and HDM (p=0.864) (Fig 1a). In addition, median PFS did not differ among patients receiving HDM or Bor-HDM (37.7months vs 29.3 months, p=0.2) (Fig1b) In conclusion,Bor-HDMis a conditioning regimen able to provide higher rates ofnCR/CR, as well as MRD negativity compared to HDM alone. Further studies are warranted to explore this regimen, especially when other upfront therapies are employed. Overall Survival according to the conditioning regimen employed for patients with MM undergoing ASCT Overall Survival according to the conditioning regimen employed for patients with MM undergoing ASCT Figure 1 Progression-Free Survival according to the conditioning regimen employed for patients with MM undergoing ASCT Figure 1. Progression-Free Survival according to the conditioning regimen employed for patients with MM undergoing ASCT Disclosures Jimenez-Zepeda: Takeda: Honoraria; Amgen: Honoraria; Janssen: Honoraria; Celgene, Janssen, Amgen, Onyx: Honoraria. Neri:Celgene and Jannsen: Consultancy, Honoraria. Bahlis:Onyx: Consultancy, Honoraria; Janssen: Consultancy, Honoraria, Other: Travel Expenses, Research Funding, Speakers Bureau; Amgen: Consultancy, Honoraria; BMS: Honoraria; Celgene: Consultancy, Honoraria, Other: Travel Expenses, Research Funding, Speakers Bureau.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3419-3419
Author(s):  
Ramzi Abboud ◽  
Michael Slade ◽  
Wenners Ballard ◽  
Jesse Keller ◽  
John F DiPersio ◽  
...  

Abstract Background:Allogeneic hematopoietic cell transplantation is a cornerstone of therapy for hematologic malignancies and often a patient's only curative intent treatment. Following development of post-transplant cyclophosphamide (PTCy) regimens, the use of haploidentical hematopoietic cell transplantation (haplo-HCT) has expanded. While overall outcomes for haploidentical transplantation appear to be excellent, its novel approach brings toxicities that are particular to its biological and clinical milieu. We recently described occurrence of severe cytokine release syndrome (CRS) after haplo-HCT. We further reported that severe CRS was associated with poor clinical outcomes, including transplant related mortality (TRM), overall survival (OS), and neutrophil engraftment (Abboud et al, BBMT, 2016). However, the factors predicting the occurrence of and long-term outcomes of patients who develop severe CRS after haplo-HCT is currently not known. Objective: To describe our clinical experience with CRS in an expanded cohort of haplo-HCT patients, its implication on clinical outcomes and elucidation of possible risk factors for the development of severe CRS. Patients and Methods: We performed a retrospective review of patients who had undergone peripheral blood T-Cell replete haplo-HCT with PTCy from July 2009 through March 2016 at our institution. A total of 137 patients were identified, 51% (74) were male, with a median age at transplant of 52 (19-73), a total of 40% (57) had active disease at the time of transplant. The most common diagnosis was AML (93 pts), followed by ALL (16 pts) and MDS (15 pts). Thirty-one percent (44 pts) had undergone prior transplant. In grading CRS, we used our approach modified from by Lee et al (Blood, 2014). Twenty-two patient, donor and disease characteristics were examined to identify predictors for the development of severe CRS. Results:One hundred and twenty-four (90%) of patients met criteria for CRS, and 26 (19%) suffered from severe (grade 3-4) CRS. Virtually all patients (99%) with CRS suffered from fevers. Patients with severe CRS had a significant delay in neutrophil (p < 0.0001) and platelet (p < 0.0001) engraftment compared to the patients who developed mild or no CRS (Figure 1A and 1B). Severe CRS was also associated with a high early transplant related mortality; the rate of death before post-transplant day 28 was 6.9 times higher for patients with grade 3-4 CRS compared with those with mild CRS (p < 0.0001, Figure 1C). Consistent with these findings, the development of severe CRS was associated with extremely poor survival. Median survival was 3 months for grade 3-4 CRS, 15 months for grade 1-2 CRS, and 13 months for no CRS. One-year OS was 4% for grade 3-4 CRS, 55% for grade 1-2 CRS, and 50% for no CRS (Figure 1D). There was no difference in the cumulative incidence of relapse, acute graft versus host disease, and chronic graft versus host disease (data not shown). A total of nine patients received Anti-IL-6 Therapy with tociluzimab (4 mg/kg of actual body weight), 4 of which suffered from severe CRS. In terms of predictive factors, the development of severe CRS was associated with disease risk index (p=0.037), HCT-CI score (p=0.005) and presence of a previous transplant (p=0.026) by univariate analysis. Risk and severity of CRS did not differ by age, ABO mismatch, age, CMV status of donor, donor sex, T-cell or CD34 cell dose. There was no difference in rates of CRS among patients in remission or with active disease at the time of transplant. Conclusions: Severe CRS after peripheral blood haplo-HCT is associated with high early TRM, poor OS and delayed neutrophil and platelet engraftment. Furthermore, patients with high DRI, high HCT-CI and prior HCT are at a higher risk for the development of severe CRS after haplo-HCT. We have previously shown the safety and potential efficacy of using anti-IL-6 therapy in these patients. Our current results suggest potential benefit to targeting this pathway prophylactically in patients at high risk for the development of severe CRS. Table Patient Characteristics Table. Patient Characteristics Figure CRS impacts neutrophil (A) and platelet (B) engraftment and is associated with high TRM (C) and poor OS (D). Figure. CRS impacts neutrophil (A) and platelet (B) engraftment and is associated with high TRM (C) and poor OS (D). Disclosures DiPersio: Incyte Corporation: Research Funding. Abboud:Gerson and Lehman Group: Consultancy; Merck: Research Funding; Teva: Research Funding, Speakers Bureau; Novartis: Research Funding; Pfizer: Research Funding; Seattle Genetics: Research Funding; Alexion: Honoraria; Baxalta: Honoraria; Pharmacyclics: Honoraria; Takeda: Honoraria; Cardinal: Honoraria. Fehniger:Affimed: Consultancy; Celgene: Research Funding; Fortress Biotech: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document