Cost and Utilization Of Stored Autologous PBSC To Support Tandem ASCT In MM Patients In The Era Of Novel Agent Therapy

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4507-4507
Author(s):  
Colin Phipps ◽  
Michael L. Linenberger ◽  
Damian J. Green ◽  
M. Corinna Palanca-Wessels ◽  
Pamela S. Becker ◽  
...  

Introduction Historically, autologous stem cell transplantation (ASCT) for multiple myeloma (MM) improves overall survival (OS) compared to conventional chemotherapy alone. Before the introduction of novel agent therapy, tandem ASCT, defined as a second ASCT within 180 days of the first, was an important option for suboptimal responses after an initial ASCT and thus collecting adequate peripheral blood stem cells (PBSCs) for 2 transplants has been considered standard of care. However, the role of tandem transplants is being challenged by novel agent maintenance strategies. Considering this changing landscape of MM therapy, we sought to evaluate the current PBSC collection and storage practices that set the CD34+ cell dose goal to be sufficient for 2 transplants. Methods We obtained clinical, PBSC collection and storage data on MM patients who underwent ASCT from 1993 to 2011 from the Autologous Transplant and Cellular Therapy Laboratory databases at the Fred Hutchinson Cancer Research Center. We determined frequencies and trends of all second ASCTs, including tandem, costs involved in PBSC collection, storage, and utilization of the product that remained cryopreserved for a second ASCT. Cell dose target at our center is 5 x 106 CD34+ cells/kg/transplant. To analyze trends over time, we divided the sample into groups of 3 or 4 year periods. Collection and cryopreservation costs for second ASCT were calculated by first determining the number of days required to collect sufficient PBSC for one ASCT, then the number of additional days to complete the actual collection, and the cumulative costs of long-term storage in our facility. Cost was estimated per July 2012 charges: 1 day PBSC collection $3,016, 1 day processing for cryopreservation $5,955, 1 year storage fees $408 ($34/month). The cost of mobilization chemotherapy and growth factors were not included. Results From May 1993 to June 2011, 889 MM patients underwent PBSC collection and ASCT (111 of 1000 excluded due to incomplete records). Median total PBSC collection days was 2 (range 1 – 10) with median yield of 13.18 x 106/kg CD34+ cells. Median days to collect sufficient cells for one ASCT was 1 day (1 – 9) and 383 patients collected adequate cells for 2 ASCTs after 1 apheresis procedure. Of 889 patients, 135 underwent a second ASCT within a median 14 months (2.5 – 113) of the first. Number of second ASCTs per time period: 1993 to 1995 – 9 of 39 MM patients undergoing ASCT (23%), 1996 to 1999 – 18 of 100 (18%), 2000 to 2003 – 15 of 162 (9%), 2004 to 2007 – 62 of 251 (25%), 2008 to 2011 – 31 of 337 (9%). Fifty patients underwent tandem ASCTs and these accounted for 89%, 72%, 7%, 24%, and 42% of all second ASCTs during the respective periods; the other 85 occurred > 6 months after the first. Number of additional days and associated costs to collect and store PBSC for a second ASCT: 5 days ($44,855), n=1 patient; 4 days ($35,844), n=2; 3 days ($26,913), n=10; 2 days ($17,942), n=41; and 1 day ($8,971), n=211. 637 patients had unused PBSC that remained in storage for ≥ 1 year, with a rising trend over time: 1993 to 1995 – 7 (1%), 1996 to 1999 – 65 (10%), 2000 to 2003 – 77 (12%), 2004 to 2007 – 185 (29%), 2008 to 2011 – 303 (48%). Duration of storage was < 2 years for 34, 2 to 5 years for 346, and > 5 years for 257 patients. Median PBSC storage time was 40 months. PBSC products from 260 patients were discarded (or sent to repository), after a median storage of 54 months, for the following reasons: 5 had allogeneic transplant, 74 were alive and possibly concerned about costs and/or the unlikely need for a second ASCT, 3 had cell viability issues, and 178 patients had died. From January 2009, some patients received plerixafor to achieve the collection goal for 1 ASCT. Its cost was not considered additional. Estimated average cost for PBSC collection, cryopreservation and storage was at least $20,065.96/person and at least $6718.11/person was spent to collect and store PBSC for a tandem ASCT that was never performed. Conclusions Approximately 70% of patients had PBSC products that remained unused and in prolonged cryopreservation after the first ASCT. Estimated cost for collection and storage of PBSC beyond that needed for a single ASCT accounted for roughly 1/3 of total costs. This conventional practice should be reconsidered in view of rising treatment costs, the evolving role of novel agents in maintenance therapy, and the deeper responses achievable with novel agents prior to first ASCT, thus reducing the need for tandem ASCT. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 733-733 ◽  
Author(s):  
Sang Kyun Sohn ◽  
Dong Hwan Kim ◽  
Nan Young Lee ◽  
Jin Ho Baek ◽  
Jong Gwang Kim ◽  
...  

Abstract Background: Little is known about the role of the CD56+ natural killer (NK) cell dose on the outcome of allogeneic peripheral blood stem cell transplantation (PBSCT). Recently, a higher dose of NK cells has been associated with a lower incidence of severe GVHD in a PBSCT setting. Therefore, the current study attempted to evaluate the effect of the NK cell dose on transplant outcomes, including non-relapse mortality (NRM) and infectious events, in an allogeneic PBSCT setting. Methods and Materials: Sixty-one cytokine mobilized PBSC recipients from HLA-matched sibling donors were analyzed according to the infused dose of CD34+ cells and NK cells in relation to overall survival (OS), NRM, GVHD, and infectious events. Results: The group of patients that received a higher dose of NK cells (≥ 5x107/Kg) showed a lower incidence of NRM (p=0.0186) and infectious events (p=0.0107). When confining the analysis to the group that received a CD34+ cell dose of ≥ 6x106/Kg, those patients that received a higher dose of NK cells exhibited a lower incidence of extensive chronic GVHD (p=0.0704). In a multivariate analysis using Cox’s regression model, a higher dose of NK cells was significantly associated with better transplant outcomes (for NRM, NK cell dose p=0.042, for CD34+ cell dose p=0.018; for infectious events, NK cell dose p=0.013, CD34+ cell dose 0.016; for bacterial infection, NK cell dose p=0.049). The group that received a higher NK cell dose also showed a faster immune recovery (p=0.046 for NK cell recovery, p=0.034 for helper T-cell recovery) in serial measurements of peripheral lymphocyte subsets at D+90, +180, and +365. Conclusions: The present data suggests that a high dose of NK cells may play an important role in improving transplant outcomes, in terms of reducing NRM and infectious events together with CD34+ cells. The protective role of NK cells against infections may also be associated with a faster immune recovery after allogeneic PBSCT. Figure Figure Figure Figure


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2315-2315
Author(s):  
Jan Jansen ◽  
Pamela L Nolan ◽  
Margaret I Reeves ◽  
Luke Paul Akard ◽  
James M. Thompson ◽  
...  

Abstract The viability of transported PBPC products has not been studied extensively. Commonly, PBPC products are transported at a concentration of &gt;200 x109/l in containers with −20oC ice packs. Continuous temperature monitoring has shown that the temperatures of these products stays at &lt;10oC for less than 24 hours and reaches room temperature by 48 hours. Samples of freshly collected PBPC from 12 allogeneic donors were studied for various viability parameters during storage for up to 96 hours. The effects of storage time, concentration of cells, temperature, and storage in gas-permeable bags were studied. Trypan-blue exclusion and double fluorescence for 7-AAD and CD34 were used for viability assessment. Over a wide range of temperatures and storage times, the viable CD34+ assay was more sensitive to damage than trypan-blue exclusion (mean Δ 10.7%, p&lt;0.0001 in paired t-test). The viable CD34+ assay was routinely used in parallel with CFU-GM cultures. No difference in survival of viable CD34+ cells or CFU-GM was found whether cells were incubated for 48hr in test-tubes or in gas-permeable bags. When cells at 200 x 109/l were incubated for 48hr at room temperature, the mean viability decreased to 19% and 6% of starting values of viable CD34+ cells and CFU-GM, respectively. Serial dilution to 25 x 109/l improved the survival to 81% and 51% respectively. Similarly, incubation at lower temperatures led to better survival of CD34+ cells and CFU-GM: 67% and 18% at 17oC, 80% and 50% at 13oC, and 95% and 86% at 4oC. At 200 x109/l and 22oC the survivals of CD34+ cells and CFU-GM were 74% and 21% at 24hr, 19% and 7% at 48hr, 7% and 6% at 72hr, and 3% and 13% at 96hr. The effects of concentration, temperature and duration of storage were all significant (p&lt;0.05). Transportation at 4oC leads to the best survival of CD34+ cells and CFU-GM, in particular at a low concentration. If transportation at a slightly higher temperature is necessary, dilution of the PBPC product will enhance the survival of CD34+ cells and CFU-GM. Proliferative assays such as CFU-GM appear the most sensitive parameters of PBPC survival, and should be included in the validation process of PBPC transportation.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1669-1669
Author(s):  
Su Chu ◽  
Ravi Bhatia

Abstract Abstract 1669 Tyrosine kinase inhibitor (TKI) treatment inhibits proliferation in CML stem/progenitor cells, but only modestly increases apoptosis. Residual leukemia stem cells remain a potential source of disease relapse in IM-treated patients. The Bcl-2 family of anti-apoptotic proteins plays a central role in the regulation of apoptosis. Several Bcl-2 inhibitors are being evaluated in preclinical and clinical studies and there is considerable interest in evaluating their ability to induce apoptosis in CML stem and progenitor cells. However these agents have considerable toxicity possibly related to lack of selectivity for individual family members. We performed a functional siRNA screen to determine the role of individual Bcl-2 family members in maintaining survival of in CML and normal CD34+ cells. CML and normal CD34+ cells were transfected with siRNAs targeting Bcl-2, Bcl-2L1, Bcl-2L2, Bcl-2L10, Mcl-1 and Bcl2A1. In this screen Mcl-1 knockdown resulted in significant reduction in viability of CML CD34+ cells, with or without co-treatment with IM (1uM). Significant reduction in normal CD34+ viability was not seen. These results were validated using different siRNA sequences to knockdown Mcl1 expression. Increased apoptosis of CML but not normal CD34+ cells was seen (23±8% for CML vs. 4.2±1.5% for normal CD34+ cells, n=3, p<0.5). CML CD34+ cell apoptosis was further enhanced by combination of Mcl-1 inhibition with IM treatment (48±15% for CML vs. 7.2±3% for CB progenitors, p<0.1). To further evaluate the role of Mcl-1 in regulating CML CD34+ cell growth, an anti-Mcl-1 shRNA construct was cloned into the pHIV7-SF-RFP lentivirus vector. Cord blood and CML CD34+ cells were transduced with Mcl-1 specific or control, non-specific shRNA expressing vectors. Western blotting demonstrated effective knockdown of Mcl-1 protein levels in Mcl-1 shRNA transduced CD34+cells (82% reduction in CML and 78% in normal CD34+ cells). CD34+ RFP+ cells were selected by flow cytometry and cultured in presence and absence of IM. A significant increase in apoptosis was seen in Mcl-1 knockdown CML CD34+ cells compared with control shRNA-transduced cells, and further increase in apoptosis was seen following IM treatment (4.7±0.5 for control shRNA-transduced cells VS 25.7±2.1 for Mcl-1 knockdown cells). Mcl-1 knockdown CML CD34+ cells generated fewer colonies in methylcellulose progenitor culture (93 colonies for control siRNA transduced cells vs. 31 colonies for Mcl-1 knockdown cells) and demonstrated reduced cell expansion following culture with growth factor (SCF; IL3; GM-CSF and G-CSF) compared with control shRNA transduced cells (383,750± 172,476 for control shRNA-transduced cells 224,250± 87,044 for Mcl-1 knockdown cells). Cell expansion was further reduced with IM treatment. Mcl-1 knockdown resulted in complete loss of erythroid colony formation. Analysis of cell differentiation by flow cytometry after culture for 4 or 7 days revealed that Mcl-1 knockdown resulted in reduced generation of both erythroid (GPA+) and myeloid (CD33+ and CD14+) cells. In contrast to the results of the initial siRNA studies, shRNA-mediated Mcl-1 knockdown also resulted in significantly increased apoptosis of normal CD34+ cells (12.6± 1.6% for control shRNA-transduced cells and 24.5± 0.9% for Mcl-1 knockdown cells) associated with reduced colony formation and reduced growth in culture (1.265e+006± 273,892 for control shRNA-transduced cells 589,000 ± 188,082 for Mcl-1 knockdown cells). We conclude that RNAi-mediated Mcl-1 knockdown inhibits CML CD34+ cell survival and proliferation and enhances apoptosis after IM treatment, but also reduces viability of normal CD34+ cells. Since Mcl-1 protein expression is subject to multiple levels of regulation, our results suggest that strategies to selectively target Mcl-1 regulatory mechanisms active in CML but not normal progenitors may be less toxic and have greater clinical utility than direct targeting of the protein. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4847-4847
Author(s):  
Scott J Ragg ◽  
Annabella Chang ◽  
David Ma

Abstract Background The current External Quality Assurance (EQA) programs for haemopoietic progenitor cell (HPC) involve enumeration of total CD34 + cells in fixed samples, which does not align with clinical practice where analysis of viable CD34 + cells (vCD34 +) is required. The COVID-19 pandemic forced a fundamental change in the global procurement of allogeneic HPC for transplantation. To better meet the emergent challenges of transporting cryopreserved allogeneic HPC during pandemics, there is an urgent need for EQA programs to evaluate reproducibility and harmonization of vCD34 + HPC enumeration between collection and transplant centres. A successful vCD34 + EQA program will require cost-effective distribution of cryopreserved reference samples (CRS) with acceptable reproducibility and specificity. This study aims to evaluate the feasibility of distribution of CRS to participating facilities for vCD34 + enumeration using dry ice, instead of liquid nitrogen which is not suitable for CRS transport due to logistical and cost implications. Method: A 15 ml sample was cryopreserved from each of 10 HPC harvests from consented transplant donors (SVH HREC approval #10/07). Cryopreserved HPC samples were either stored on dry ice for 1-4 days, or on dry ice for one day followed by liquid nitrogen (LN 2) storage for 1-3 days to assess optimal conditions for vCD34 + EQA. For viable CD34 + measurement by flow cytometry, the single platform assay was performed using Trucount tubes containing CD45-FITC/CD34-PE and 7-AAD viability exclusion dye. The optimum transportation condition was validated in pilot and multi-center national studies which involved transport of CRS on dry ice to 12 recipient centers in 5 of the 6 Australian states. Results: Dry ice and LN 2 transport and storage conditions were simulated at the central laboratory and the vCD34 + enumerated. It was found that a combination of one day on dry ice followed by LN 2 storage stabilized the viability compared to continuous storage on dry ice. A successful pilot study confirmed the effect on vCD34 + count of shipping two CRS from central Lab to two interstate laboratories. For the national multicenter study, the transportation distances ranged from 0.5 - 4,000 km (median 513 km) with transit times ranging from 1- 26 hours (median 22.5 hours). Eight of 12 centers (67%) returned comparable results that were within ±10% of the median. There was no significant difference between samples tested immediately upon arrival or after subsequent LN 2 storage (p=0.41). There was no significant relationship between comparability of vCD34 + counts and the sample transit time (R=0.67, p=0.07) nor distance travelled (R = 0.19, p=0.55), showing that laboratory outcome was unrelated to sample transport. Conclusion: Dry ice distribution of cryopreserved HPC for up to 26 hours results in a stable CRS. The estimated cost of safer and more convenient dry ice delivery is &gt;20-fold lower than LN 2. This feasibility study illustrates that an EQA utilizing this mode of transport and storage of CRS is suitable for inter-facility harmonization and standardization forming the basis of an EQA programs for vCD34 + HPC enumeration. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1860-1860
Author(s):  
Moshe E. Gatt ◽  
Izhar Hardan ◽  
Evgene Chubar ◽  
Celia Surio ◽  
Tamar Tadmor ◽  
...  

Abstract Abstract 1860 Background: AL amyloidosis is a plasma cell dyscrasia, associated with low survival rates, especially in patients with cardiac involvement. Treatment includes chemotherapy, mostly melphalan, and steroids, aiming to suppress amyloidogenic light chain production. Hematological response (HR) is correlated with improved survival and restoration of organ function. Introduction of novel agents such as Thalidomide, Lenalidomide and Bortezomib, over the last decade was suggested to improve patients' outcome. However, these encouraging data are mainly coming from small prospective studies affected by potential selection bias due to the exclusion of patients with poor prognosis. “Real-life” reports are required to assess the efficacy of these new treatments in a non-selected patient population. Aims: The aims of this retrospective observational study was to evaluate the “real-life” response of patients with systemic AL amyloidosisto novel agents, regardless of age or disease severity. Methods: Data of patients treated with novel agents at 10 different Israeli hospitals were analyzed. Newly diagnosed and relapsing systemic AL amyloidosispatients were eligible, if they received at least one cycle of novel agent based therapy. Specific regimens and doses were at the discretion of treating physicians. Results: 89 patients treated with 114 cycles of novel agent regimens, were reported. Median age at presentation was 66 years. At baseline evaluation, the median NTproBNP and troponin were 1450 and 0.068, respectively. 85% of patients had renal or cardiac or both organ involvements (34%) at presentations. 41% had > 2 organs involved. Treatment protocols included: Thalidomide-steroid-based therapy (n=19, comprising 10 who have also received alkylator chemotherapy); Lenalidomide-steroid based therapy (n=26, 13 with alkylator chemotherapy) and Bortezomib-steroid based therapy (n=68, 43 with alkylator chemotherapy). Treatment with Thalidomide-based regimens resulted in an overall response rate (ORR) of 73%, including 26% ≥VGPRs. Treatment with Lenalidomide-based regimens resulted in an ORR of 63%, including 36% ≥VGPRs. Yet, only 5 of 26 patients received this agent as first-line therapy. Treatment with Bortezomib-based regimens resulted in an ORR of 80%, including 55% ≥VGPRs. In general, treatment of newly diagnosed patients yielded better responses in all the arms and was most prevalent in the Bortezomib group (44 of 68 patients). However, it is noteworthy that about 30% of patients experienced grade 3 adverse events (heart and renal failure, and severe infections), mostly following Bortezomib and Lenalidomidebased regimens. At a median follow up of 12 months (range1–87 months), the overall survival was 52% (35%, 65% and 68% respectively for the three treatment groups, p= n.s.). Interestingly, censoring patients without progression for more than 12 months (with a median follow up of 20 months) revealed that 2 of 11 and 5 of 13 in the Thalidomide and Lenalidomide treated patients maintained their responses, compared with 31 of 44 patients treated with Bortezomib based regimens (Figure 1). Conclusion: Treatment with novel agents, especially Bortezomib-based regimens, may result in long-term responses in a substantial number of newly diagnosed and relapsing patients with AL amyloidosis. This may be translated into a better overall survival of responding patients. Physicians caring for these patients should be aware of potential treatment-related toxicities Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 948-948
Author(s):  
Meletios A Dimopoulos ◽  
Sosana Delimpasi ◽  
Eirini Katodritou ◽  
Eleftheria Hatzimichael ◽  
Marie-Christine Kyrtsonis ◽  
...  

Abstract Abstract 948 Renal impairment (RI) is a common presenting complication of multiple myeloma (MM) and is associated with increased risk of treatment related toxicity and early death. The management of RI in patients with MM requires vigorous supportive measures and the immediate institution of antimyeloma therapy. After the introduction of novel agents a significant improvement of the survival of patients with MM has been observed; however, the impact of these therapies on the survival of MM patients who present with RI has not been extensively studied. In order to analyze the impact of RI in newly diagnosed patients with MM over the past 20 years, we analyzed 1773 patients with symptomatic myeloma who were treated within the Greek Myeloma Study Group (GMSG). Patients were divided in groups according to the date of initial treatment (1/1/1990-31/12/1994, 1/1/1995-31/12/1999, 1/1/2000-31/12/2004, after 1/1/2005). Thalidomide became available in Greece after 1/1/2000 and bortezomib after 1/1/2005. eGFR was calculated by the modified MDRD formula and the degree of RI was rated as severe when eGFR was <30 ml/min, moderate when eGFR was 30–59 ml/min and mild (or no RI) when eGFR >60 ml/min. The frequency of RI over time was similar as well as the proportion of patients who presented with severe RI (17% vs. 21% vs. 17% vs. 19%) for the respective time periods (p=0.496). More patients >65 years started therapy after 2000 (44% vs. 50% vs. 59% vs. 59%, respectively, p<0.001), especially patients >75 years (13% vs. 18% vs. 24% vs. 32%, respectively, p<0.001). Anemia (Hb <10 g/dl; p=0.007) and ISS-3 disease (p=0.001) were more common after 1/1/1995; there were no other significant differences in the characteristics of the patients during the respective time periods. No patients received upfront novel agents before 31/12/1999, while 20% received upfront novel agent in the period 2000–2004 (almost exclusively thalidomide) and 73% after 1/1/2005 (mostly thalidomide and bortezomib). Myeloma response (≥PR) to frontline therapy was achieved in 56.5% & 54% of patients in the period 1990–1994 & 1995–1999 vs. 67% and 72% of patients in the periods 2000–2004 and after 2005 (p<0.001). The median survival of patients has improved significantly during the past 20 years: 39 months (1990-1994), 31 months (1995-1999), 40.5 months (2000-2004), 54 months after 2005 (p<0.001). The median OS for patients with severe RI has improved significantly from 18 months & 19.5 months in the 1990–1994 & 1995–1999 to 29 months and 32 months for the periods 2000–2004 and after 2005 (p=0.005). For patients with moderate RI the OS improved from 33 & 26 months between 1990–1994 & 1995–1999 to 40 & 44 months in the periods 2000–2004 and after 2005 (p=0.003). For patients with an eGFR ≥60 ml/min the OS improvement was less pronounced (48.5 months vs. 45 months vs. 51 months for the periods 1990–1994 & 1995–1999 & 2000–2004 respectively (p=0.076) and only after 2005 a significant improvement in OS is observed (median OS has not been reached; 3-year OS rate is 73%, p<0.001). For patients with severe RI early death rates (<2 months from initiation of therapy) were 12% vs. 7% for patients with moderate RI vs. 3% for patients with mild or no RI (p<0.001) and remained unchanged over time. We then adjusted for differences between groups in a multivariate model: treatment after 1/1/2000 was independently associated with improved survival compared to patients treated before 31/12/1999 (p<0.001). After adjusting for the degree of RI in the model, the hazards ratios (HR) for death for patients with severe RI for the 2000–2004 and after 2005 periods were 0.485 & 0.387 respectively compared to patients treated before 2000 (p<0.001 for both comparisons). For patients with moderate RI the respective HRs were 0.65 (p=0.003) & 0.57 (p=0.001), while for patients with mild or no RI the HRs were 0.85 (p=0.1) & 0.66 (p=0.003) for the 2000–2004 and after 2005 periods, respectively. In conclusion, the incidence of RI at diagnosis of MM has remained unchanged during the past 20 years, despite the increasing numbers of older patients who are diagnosed and treated for MM. The risk of early death is almost 2 to 4-fold higher in patients with severe RI vs. patients with moderate or no RI and has not improved over time. However, after the introduction of novel therapies there has been a significant improvement of the survival of patients with RI, which is more pronounced in patients with severe RI. Disclosures: No relevant conflicts of interest to declare.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 8022-8022
Author(s):  
Binod Dhakal ◽  
Saurabh Chhabra ◽  
Mehdi Hamadani ◽  
Anita D'souza ◽  
Saad Zafar Usmani ◽  
...  

8022 Background: Given the unprecedented deep response rates with the novel agent induction, the role of high dose therapy (HDT) followed by ASCT in MM pts. has been questioned, and was re-evaluated in a number of randomized clinical trials (RCTs). Although, the results of most studies suggest the continued benefit of HDT/ASCT, some RCTs suggest no overall survival (OS) benefit. We undertook a systematic review and meta-analysis of phase III randomized RCTs evaluating the role of HDT compared to standard therapy (SDT) in the context of novel agent induction Methods: We searched the PubMed, Scopus and Cochrane Collection of Controlled Trial databases using the term myeloma combined with autologous or transplant or myeloablative or stem cell from 2000-2016. A total of 2480 articles identified, of which 4 large phase III RCTs compared upfront HDT with SDT with novel agents use. Two individuals independently extracted the data. Reported hazard ratio (HR) and survival data were pooled using random effects models (STATA v14, College Station, Tx). Heterogeneity was assessed using I2. Results: Four studies comprising 2421 patients were included (Table 1). One study did not report the HR for death and hence OS analysis was limited to 3 studies. The combined hazard for progression with HDT was 0.55 (95% CI 0.40-0.71) (p < 0.005). The combined hazard for death with HDT was 0.65 (95% CI 0.29-1.0) (p = 0.007). Sensitivity and sub-group analysis showed no difference in PFS (p = 0.06) and OS (p = 0.22) with HDT. Significant heterogeneity was demonstrated by I2 of 71.4% for PFS (p = 0.01) and 68.4% for OS (p = 0.04). Conclusions: Based on our analysis, even in the novel agent era, HDT appears to be beneficial and should be considered standard of care for all transplant eligible MM pts. [Table: see text]


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2135-2135
Author(s):  
Eliza B Geer ◽  
Lilian Varricchio ◽  
Fabrizio Martelli ◽  
Wu He ◽  
Lizette Couto ◽  
...  

Abstract Cushing's disease (CD) is a rare endocrine disorder (1.2-2.4/million/year) characterized by chronic excess endogenous glucocorticoids (GC) due to an adrenocorticotropic hormone-secreting pituitary adenoma. Untreated CD results in increased mortality and multiple morbidities (obesity, diabetes, hypertension, cardiovascular disease) and, in one case report, erythrocytosis (Gursoy et al, J End Invest. 2006;29:742). The effects of chronic GC exposure on erythropoiesis in a CD cohort have not yet been studied. We prospectively quantified hematocrit (Hct), hemoglobin (Hb) and platelets (ptl) values in CD patients before (v1) and after surgical remission (v2, mean time since surgery=14.5 months) and in matched healthy controls (HC). Frequency, antigenic profiling and erythroid (Ery) expansion potential of circulating hematopoietic progenitor cells (HPC) in the three cohorts were also evaluated. The subjects analyzed included 28 v1 [6 males, 22 females, mean age=41 years; body mass index (BMI)=33.3 kg/m2 ] and 13 HC [2 males, 11 females, mean age=41 years; BMI=30.7 kg/m2 (range 26.7-34.7, p=0.073). Eleven patients were analyzed over time in both v1 and v2. Mean cortisol in v1 (22.2±6.3 ug/dL) was higher than in HC (8.5±3.8 ug/dL, p=3.4E-07) and decreased in v2 (17.6±5.1% vs. 8.7±3.9%, N= 11, p=0.0006). Hct was higher in v1 than in HC (39.8±4.7%, vs. 38.8±2.7%, p = 0.045). In the 11 patients analyzed over time, hct decreased in v2 vs. v1 (39.2±4.7% vs. 42.3±4.4%, p=0.011). Hb in v1 was not different than in HC (13.10±1.6 vs. 13.12±3.8 g/dL, p =0.225) but decreased in patients studied both in v1 and V2 (14.1±1.5 g/dL vs. 13.2±1.8 g/dL, p=0.009). Similarly, plts were not different in v1 and HC (272.3±81.4 vs. 240.8±76.0 K/uL, p=0.274) but decreased in v2 (307.7±112.1 vs. 270.6±74.5 K/uL, p=0.021). In v1, Hct did not correlate with serum (R = 0.34, p = 0.33) or 24h urine (R = 0.072, p = 0.73) cortisol concentrations. There was no difference in frequency of HPC among v1, v2 and HC [2.6±3.0, 0.34±0.28 and 1.26±0.67% of CD34+ cells and 30.2±27.2, 23.7±13.2 and 16.5±11.5 CFC/105 mononuclear cells (MNC) in v1, v2 and HC]. CD34pos cells from all groups expressed similar levels of cKIT, IL-3Rβ and prominin1, but a greater proportion of those from v1 and v2 expressed thrombospondin and thrombopoietin (Mpl) receptors than those from HC (2 vs 0.4%), suggesting that CD HPC are biased toward erythro-megakaryocytopoiesis. Consistently, in cultures without the synthetic GC dexamethasone (Dex), MNC from v1 (12±6 %) and v2 (19%) generated in 10 days a greater proportion of Erys than MNC from HC (2±1% ). However, in cultures with Dex, MNC from v1 (48±25), but not those from v2 (70±23), generated less Erys than MNC from HC (83±26 , p=0.03), suggesting that Erys from v1 HPC respond poorly to Dex. This was tested by comparing the ability of Dex to induce biochemical (GRα phosphorylation at S211 and cell-surface expression of CXCR4/calreticulin) and biological (proliferation in synergy with growth factors, GFs) responses in Erys from CD and HC. Erys from v1, v2 and HC expressed equivalent levels of GRα but those from v1 Erys contained lower levels of pGRαS211/S203 than those from v2 or HC (Fig 1A). In contrast with HC Erys, Dex decreased GRα and did not induce pGRαS211 in v1 Erys (Fig 1B). Moreover, Dex increased cell-surface expression of CXCR4 (MFI from 580 to 700) and calreticulin (MFI from 300 to 700) and proliferation (by 30%) in HC Erys but not in those from v1 s (CXCR4 MFI from 278 to 154, calreticulin MFI from 136 to 152; proliferation increases by 6%). These results indicate that chronic GC excess increases Hct values but may also activate a post-transcriptional mechanism that reduces GRα expression inducing desensitization of erythroid cells to GC. Figure 1. A) Levels of total, pS211 and pS203 GRα in Erys from HC, v1 and v2. B) levels of GRα and GRα phosphorylated at pS203 in Erys from HC and v1 exposed from 15' to Dex alone or in combination with the GR inhibitor RU486. Figure 1. A) Levels of total, pS211 and pS203 GRα in Erys from HC, v1 and v2. B) levels of GRα and GRα phosphorylated at pS203 in Erys from HC and v1 exposed from 15' to Dex alone or in combination with the GR inhibitor RU486. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1667-1667
Author(s):  
Li-Sheng Wang ◽  
Ling Li ◽  
Liang Li ◽  
Keh-Dong Shiang ◽  
Min Li ◽  
...  

Abstract Abstract 1667 MicroRNAs are key regulators of gene expression that regulate normal differentiation and contribute to malignant transformation of hematopoietic cells. Using microRNA microarrays we identified increased expression of miR-486 in chronic myeloid leukemia (CML) compared to normal CD34+ cells. In both normal and CML cells, miR-486 expression level was significantly higher in MEP compared to HSC, GMP and CMP populations. Treatment with Imatinib resulted in reduced expression of miR-486-5p in CML CD34+ cells, suggesting that upregulation of miR-486-5p expression was at least in part BCR-ABL kinase dependent. Consistent with this ectopic expression of BCR-ABL in cord blood CD34+ cells using retroviral vectors resulted in 4.2 fold increase in miR-486-5p expression. miR-486-5p is located within the last intron of the Ankyrin-1 gene on chromosome 8 and is enriched in muscle cells. However, the role of miR-486-5p in normal and leukemic hematopoiesis has not been evaluated. To explore the role of miR-486-5p in growth and differentiation of hematopoietic progenitor cells (HSPC), we first overexpressed hsa-miR-486-5p pre-microRNA in normal CD34+ cells using lentiviral vectors. CB CD34+ cells overexpressing miRNA-486-5p generated modestly increased numbers of cells (1.22 fold) in culture with SCF, IL-3, GM-CSF, G-CSF and EPO for 6 days compared to cells expressing control vectors, with increased numbers of erythroid cells and reduced numbers of myeloid cells. We further investigated the role of miR-486-5p on growth and differentiation of normal and leukemic HSPC by inhibiting miR-486-5p expression using a modified pmiRZip lentivirus vector expressing an anti-miR-486-5p sequence and comparing to cells expressing a control scrambled anti-miRNA sequence. Expression of anti-miR-486-5p resulted in reduced proliferation of normal CD34+ cells (32±10% inhibition) and BCR-ABL transformed CD34+ cells (38±7 % inhibition) with significantly greater inhibition of erythroid compared to myeloid cells. Anti-miR486-5p expression resulted in significantly increased apoptosis of BCR-ABL-transformed CD34+ cells but not normal CD34+ cells (CML CD34+ cells: scramble 11.1±2.4%, anti-miR-486-5p 14.7±1.7 % p=0.02; Normal CD34+ cells: scramble 9.7±5.4%, anti-486-5p 13.4±7.9% p=0.15). Importantly, anti-miR-486-5p significantly enhanced the sensitivity of BCR-ABL transformed CD34+ cells to imatinib-mediated apoptosis [combination of scramble with IM: 17.7±8.1%; anti-miR-486-5p with IM: 26.4±13%]. A search for conserved miR-486-5p target genes in the TargetScan database identified the important hematopoietic negative regulatory factors Foxo1 and Pten amongst the highest ranking targets. Using pMIR-REPORT constructs containing miR-486-5p seed sites within the Foxo1 and Pten 3'-UTR we showed that Foxo1 and Pten are direct targets of miR-486-5p. Expression of anti-miR-486-5p increased Foxo1 and Pten protein expression and decreased active Akt in normal and CML CD34+ cells. Knockdown of Foxo1 using shRNA partly blocked the suppressive effects of anti-miR486-5p on the growth of CD34+ cells. In summary, we have shown that miR-486-5p expression is modulated during hematopoietic differentiation and plays an important role in regulating hematopoietic progenitor growth and differentiation towards the erythroid lineage. We further show that miR-486-5p expression is enhanced in CML CD34+ cells, related at least in part to BCR-ABL kinase activity, and contributes to enhanced progenitor growth and survival. Inhibition of miR-486-5p results in enhanced sensitivity of CML CD34+ cells to IM-induced apoptosis. miR-486-5p effects are mediated at least in part through inhibition of Foxo1 and Pten expression. We conclude that miR-486-5p represents a novel regulatory mechanism that promotes erythroid differentiation in normal hematopoiesis and modulates Bcr-Abl-mediated transformation and tyrosine kinase inhibitor sensitivity in CML progenitors. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 141-141 ◽  
Author(s):  
Konstantina Kallinikou ◽  
Fernando Anjos-Afonso ◽  
Michael P Blundell ◽  
Stuart J Ings ◽  
Deepika Kassen ◽  
...  

Abstract Abstract 141 Short term cytokine exposure reduces the engraftment potential of haematopoietic stem and progenitor cells (HSPC). We have previously shown, in MPB CD34+ cells, that this occurs in conjunction with reduced short term homing to the BM of irradiated NOD/SCID animals, and is evident by 4 hours of culture (Ahmed at al, Blood 2004; 103:2079). Homing and engraftment of HSPC is dependent on the SDF-1/CXCR4 axis, that is negatively regulated by CD26, a cell-surface peptidase that cleaves SDF-1. Thus, blockade of CD26 with diprotin A improves engraftment of cord blood HSPC. CD26 levels are low on MPB CD34+ cells, but increase on cytokine exposure, an effect that may account for reduced homing function. We tested the effect of diprotin A treatment on the homing and engraftment of MPB CD34+ progenitors in irradiated NOD/SCID mice. Cytokine exposure (SCF, FL, IL3, IL6 for 48–72 hrs) reduced BM homing of progenitors (to 32±7% of uncultured cells, p=0.031). Treatment with Diprotin A significantly improved the homing of cultured progenitors (p=0.0002 cf non-treated cells), to the levels seen in uncultured cells (NS cf homing of uncultured cells with or without Diprotin A). Despite this increase in levels of homing to the BM, long term engraftment of cultured progenitors is not rescued by CD26 blockade, suggesting a defect beyond the initial step of homing to BM. To assess the BM attachment of transplanted cells, we used the intrabone (IB) assay, injecting cells directly into the tibiae of irradiated recipients thus bypassing the need for homing from the systemic circulation. In comparison to intravenously (IV) injected cells, IB delivery improved engraftment of both cultured and uncultured MPB CD34+ cells. At a cell dose of 106, median engraftment of uncultured cells was 13.27% (range 2.43–49.1, n=5) by IB injection compared with 1.5% (0.1–4.6, n=6) by IV delivery (p<0.01). The engraftment defect of cultured cells, however, persisted in IB transplantation. At a cell dose of 106/animal, median engraftment of cultured cells was 0.73% (0.001–10.93, n=12), compared with 13.27% (2.43–49.1, n=5) for uncultured cells (p<0.05). Injecting twice the cell dose for cultured cells (expanded equivalent number) did not rescue this defect. Engraftment of cultured cells in the injected bone (0.055%, range 0.001–9.28, n=6) was significantly lower than that of uncultured cells (14.4%, 0.08–71.2, n=9, p<0.05). These findings suggest that cytokine exposure reduces the ability of MPB CD34+ cells to be retained in the BM. To study this attachment defect directly, we developed an ex-vivo model where CD34+ cells are incubated in long bones of irradiated, 3-week old Sprague Dawley rats after resident HSPC are removed by vigorous flushing. Irradiation, flushing protocols and cell doses were optimized to ensure sensitivity and reproducibility of the assay. Progenitor adherence was assessed by colony assays on infused and recovered attached cells, correcting for animal weight. In this model, cultured progenitors displayed reduced attachment: median number of attached progenitors, 182.1 (23–384, n=26), compared with 385.4 (49–1124, n=30) for uncultured cells (p<0.0001). Attachment of both uncultured and cultured progenitors was reduced by CXCR4 blockade (42%, and 53% reduction respectively, p<0.05 for both), confirming the in vivo relevance of this model. This model provides a novel system to directly study and manipulate the lodgment of HSPC in the BM. Next, we tested the effect of cytokine exposure on ligand-specific adhesion of MPB progenitors. CD34+ cells were incubated on immobilized ligands and progenitor adhesion assessed from the clonogenic output of non-adherent cells. Progenitor adhesion to several putative niche ligands was significantly reduced following cytokine exposure. Specific adhesion of uncultured progenitors to N-cadherin was 31.8±2%, compared to 19.7±3.2% for cultured progenitors (p=0.0058). Cytokine culture also reduced specific adhesion to osteopontin and VCAM-1 (p=0.0025 and p=0.0164 respectively), but not to fibronectin, suggesting that reduced adhesive function of cultured cells is not a global defect. We conclude that whilst short term homing of cultured MPB CD34+ cells to the BM can be improved by CD26 blockade, the resulting long term engraftment defect remains. This defect is at least partly related to altered adhesive interactions of cultured cells to ligands within the BM. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document