scholarly journals Time Dependent Decrease in Efficacy of Second G-CSF Mobilization in Healthy Unrelated Donors

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1895-1895 ◽  
Author(s):  
Helmuth Schmidt ◽  
Johannes Schetelig ◽  
Karin Buhrmann ◽  
Anna Kozlova ◽  
Gero Hütter ◽  
...  

Abstract Abstract Introduction: Graft failure or a second allogeneic hematopoietic stem cell transplantation or HLA haplotypes leading to donor requests for two patients are reasons for a second collection of peripheral stem cells of the same donor. Few reports about second stimulation showed conflicting results. Higher G-CSF doses or the addition of plerixafor has been described either in autologous or allogenic settings. In order to get more information about second stimulation with G-CSF, we pooled the data from three collection centers in Germany. Donors and Methods: The collection centers of Dresden, Hameln and Cologne performed between December 1991 and May 2015 18,124 collections of peripheral stem cells. We identified 351 of donors who donated twice. All collections were performed after G-CSF stimulation (Lenograstim) of 7.5 to 10 µg / kg body weight. The same apheresis machine was used in 80 % of donors. Besides Spectra and Optia from Terumo ComTec machines from Fresenius were used. We compared leukocytes, platelets, hemoglobin and CD 34 positive cells in the blood before apheresis, yield of stem cells, and blood volume processed as endpoint of interest. There are differences in the number of donor results since the reported data in the first years were incomplete. We created 4 groups of donors depending on the interval between the two donations (Group A: interval <= 90 days, group B 91 to 180 days, group C 181 to 360 days, group D > 360 days). The minimum difference between two apheresis was 20 days, the maximum 4,436 days. The data of the donors were analyzed using NCSS as statistical program. To determine significance in the data paired t tests were performed. Results: Platelet counts, hemoglobin and erythrocyte counts at the time of apheresis was similar at first and second time point of collection. CD34-positive cells in the blood on the 1st day of apheresis were significantly higher with 77.9/µl at 1st apheresis compared to 67.9/µl at 2nd apheresis. Leukocyte counts were also higher at first donation date (42,954/µl compared to 40,330/µl). Considering the product, total collected CD34 pos. cells were lower at 2nd apheresis (617 * 106 compared to 566 * 106) but there was no significant difference in the CD34 pos. cells per kg BW of the patient which might be due to the observation that the body weight of the patients were lower at 2nd transplantation. We were interested in the time-dependence of the second mobilization capacity. The details are shown in the table. Leukocyte counts in the blood remained lower after 2nd G-CSF stimulation even after more than one year. In contrast, CD34 stimulation returned to values measured at the first stem cell collection. To achieve the requested amount of CD 34 pos. cells a higher blood volume had to be processed if the two collections are less than 6 months apart. Discussion: Our data are in accordance with earlier observations showing that at a second stimulation with G-CSF is less affective. The data of 351 donors indicate that this difference lasts for up to 1 year for stimulation of CD34 pos. cells. Only for leukocytes, there is still a significant difference also if restimulated after more than 1 year . In contrast, this decreased restimulation of stem cells has no important clinical effect on the possibility to get suffient numbers of stem cells for a transplantation. A second treatment of a donor at least with 7.5 to 10 µg G-CSF/kg body weight does not harm the donor since baseline hematologic parameters were the same at time of medical assessment. Table 1. CD 34 pos. cells/µl (blood) Leukocytes/µl (blood) Total CD 34 pos. cell in the product (*10^6) CD 34 pos. cells / BW patient (*10^6) Processed blood volume of the donor (l) Interval between 2 collections < =90 days 1st apheresis 77.0* (69) 42,772* (69) 598* (71) 8.5 (70) 13.8* (55) 2nd apheresis 59.2* 38,612* 558* 8.06 16.1* Interval between 2 collections 91 - 180 days 1st apheresis 76.6* (94) 42,872* (95) 567* (95) 7.27 (95) 14.4* (80) 2nd apheresis 64.7* 40,886* 517* 7.22 15.6* Interval between 2 collections 181 - 360 days 1st apheresis 81.3* (85) 43,643* (86) 654* (87) 9.33 (86) 14.6 (79) 2nd apheresis 77.6* 41,388* 572* 8.68 14.9 Interval between 2 collections > 360 days 1st apheresis 76.8 (98) 42,557* (98) 646 (98) 9.66 (98) 15.5 (89) 2nd apheresis 73.2 40,071* 615 10.33 14.9 *Difference significant (p<0.01) in paired t-test, in brackets number of donors Disclosures Schetelig: GSK and Sanofi: Research Funding; Janssen, Sanofi and Neovii: Membership on an entity's Board of Directors or advisory committees. Ehninger:Cellex GmbH: Equity Ownership.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4390-4390 ◽  
Author(s):  
Abhinav Deol ◽  
Judith Abrams ◽  
Ashiq Masood ◽  
Zaid Al-Kadhimi ◽  
Muneer H. Abidi ◽  
...  

Abstract Abstract 4390 Background: Plerixafor is a CXCR 4 antagonist which is now approved for use for stem cell (SC) mobilization with granulocyte colony stimulating factor (GCSF) in patients with non Hodgkin lymphoma (NHL) or multiple myeloma (MM). Prior to the approval of plerixafor, we enrolled 49 patients in a compassionate use protocol at our institution to mobilize SC for patients who previously failed at least one mobilization attempt. Methods: Patients received 0.24 mg/kg of plerixafor subcutaneously 9 –11 hrs prior to apheresis in addition to twice daily GCSF. Results: Median age of the patients was 64 years (range, 23–74 years). NHL was the most common diagnosis in 27 (55%) patients, followed by MM with 17(35%) patients and HD with 5 (10%) patients. Thirty nine patients (80%) had been treated with more than 2 chemotherapeutic regimens prior to the first attempt at stem cell collection. Thirty seven patients (76%) failed one previous mobilization attempt, while 12 (24%) had failed 2 or more previous attempts. Using the combination of Plerixafor and GCSF we collected ≥ 2.5 × 106 CD34+ cells/Kg in 33 patients (67%). The median days for pheresis were 1 day with a range of 1 to 3 days. The median SC dose collected was 4 × 106 CD34+ cells/Kg, with a range 2.5 – 14.3. The median CD-34+ peripheral blood count on the 1st day of their collection with plerixafor was 22.4/uL. In contrast the median peripheral blood CD-34+ cell count in these patients on the day of their first collection which failed was 6.2 /uL. The median increase using G-CSF and plerixafor was 14.9 CD-34+ cells/uL. We collected ≥ 2.5 × 106 CD34+ cells/Kg on 4/5 (80%) patients with HD, 13/17 (76%) patients with MM and 16/27 (59%) patients with NHL. Sixteen patients (33%) collected < 2.5 × 106 CD34+ cells/Kg. The median cell dose collected in these patients was 1.4 × 106 CD34+ cells/Kg with a range, 0.4–2.2. The median number of days of pheresis was 2 days (range, 1–4 days). In these16 patients the median CD-34+ count on the day of their previous failed collection was11.2/uL. Their CD-34+ cell count on their first day of collection after the use of G-CSF and plerixafor was 8.3/ul. Figure 1 shows the change in peripheral CD34 counts with the prior mobilization attempt and after plerixafor mobilization, for 38 patients in whom data was available. The most common side effects attributed to plerixafor were diarrhea, fatigue, thrombocytopenia and bone pain; observed in 12%, 8%, 8% and 6% patients, respectively. Forty three of the 49 patients proceeded to an autologus peripheral blood SC transplant, 34 patients received ≥ 2.5 × 106 CD34+ cells/Kg. Thirty two of these patients used the plerixafor collection as the only source of SC. Two patients had their plerixafor mobilized SC combined with a previous suboptimal SC collection. Nine patients received < 2.5 × 106 CD34 + cells/Kg; 4 patients received plerixafor mobilized SC alone, 5 patients received plerixafor mobilized SC combined with their previously mobilized SC. The preparative regimens used were R- BEAM (20 patients), Melphalan (16 patients), BEAM (6 patients) and Etoposide+TBI (1 patient). All patients received GCSF from day +6 till WBC engraftment. The median days of WBC and platelet engraftment were day +11 (range, 9–13 days) and day +16 (range, 11–77 days), respectively. There was no significant difference in days to engraftment between the patients who collected greater or less than 2.5 × 106 CD34 + cells/Kg. With a median follow up 13.7 months, long term engraftment data is available on 27 patients. The median white cell count, hemoglobin and platelet count 1 year after transplant was 4.7 × 109/L, 12.2 g/dL and 109 ×109/L, respectively. There was no significant difference in counts at the 1 year mark between patients who collected more or less than 2.5 × 106 CD34 + cells/Kg. To date 15 patients have evidence of disease progression. Two patients have developed MDS/AML post transplant. Conclusion: Overall, plerixafor leads to mobilization of sufficient stem cells in a vast majority of patients who have failed previous mobilization attempts and allows more patients to proceed to an autologous SC transplant. Plerixafor is well tolerated with minimal side effects, acceptable time to engraftment and acceptable peripheral blood counts at 1 yr after the transplant. Our analysis suggests that failure to increase peripheral CD34 count after plerixafor when compared to previous attempts may predict unsuccessful mobilization. Disclosures: Lum: Transtarget Inc: Equity Ownership, Founder of Transtarget.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5248-5248
Author(s):  
Fuyu Pei ◽  
Qi Li ◽  
Wenfeng Xu ◽  
Zhiyong Peng ◽  
Xuedong Wu ◽  
...  

Abstract Objective:To evaluate the effect of hematopoietic stem cell transplantation (HSCT) for children with leukemia in our center in recent years. Methods: We retrospectively analyzed data of 87 patients with leukemia underwent HSCT at a median age of 8 years from February 2006 to December 2013 in our center. The median follow-up time was 28 months (range, 2-96), the ratio of male to female patients was 59:28. Conditioning regimen included cyclophosphamide, fludarabine, busulfan with or without (w/o) thiotepa. Anti-thymocyte globulin and cytarabine were individually used for the patients with lymphoid leukemia and myeloid leukemia. GVHD prophylaxis included tacrolimus, mycophenolate mofetil w/o post-transplant cyclophosphamide. Median nucleated cells: 3.75 (1.16`7.56) × 107/Kg. Patients with BCR/ABL+ acute lymphoblastic leukemia (ALL) received imatinib before and after transplant over 6 months per each one. Twenty-six patients received transplant from sibling donors, 31 from haploidentical donor, 30 from unrelated donors; Status before transplant were grouped as CR1 (n= 57), CR 2 (n=13), CR 3 (n=1) and NR (n=16). Source of stem cells included PBSC in 40 cases, UCB in 3 cases, BM in 24 cases, BM+PBSC in 9 cases, and mixed stem cells (BM /PBSC+ UCB) in 11 cases. Results: The estimated 5-year overall survival (OS) was 56.8 ± 5.8% in total.Among them, OS was 54.3 ± 8.0% in 45 patients with ALL; 85.7 ± 13.2% in 8 patients with BCR/ABL+ALL; 48.6 ± 8.7% in 37 patients with BCR/ABL-ALL. 32.8 ± 15% in 29 patients with acute myeloid leukemia and 82.5 ± 11.3% in 13 patients with chronic myelogenous leukemia, respectively. Single factor analysis showed there was no significant difference for OS in comparison of BCR/ABL+ALL, BCR/ABL-ALL, AML and CML (P=0.057), but patients with BCR/ABL+ALL had higher OS compared to those with BCR/ABL-ALL (P=0.048) and to AML (P=0.040). In comparison of difference status before transplant, OS were 55.2 ± 11.6%, 54.9 ± 15.6%, 0,and 27.5 ± 11.6% in CR1, CR2, CR3 and NR, respectively (P=0.025). OS was higher in CR1 than NR (P=0.005). When comparing stem cell source, OS was 65.5 ± 8.5%, 0%, 41.7 ± 11.4%, 33.3 ± 15.7%, and 72 ± 17.8% in PBSC, unrelated CB (UCB), BM, BM+PBSC, and BM/PBSC+UCB transplants, respectively (P=0.003); PBSC transplant associated with higher OS than BM (P=0.049) and BM+PBSC (P=0.009); and BM/PBSC+UCB mixed transplant had highest OS (P=0.026). Multivariate analysis showed Risk factors for OS only remained stem cell source (P=0.046) and status before transplantation (P=0.048). the transplant types (P=0.023), and follow up time(P=0.017). Conclusion: Comparing with data reported in literature we have similar outcomesin total for childhood with leukemia. Use of imatinib pre-/post-transplant for patients with BCR/ABL+ALL conduces to the highest OS in current study. Stem cell sources and the status before transplant have a significant effect on OS. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 19 (2) ◽  
pp. 152-159
Author(s):  
E. E. Kurnikova ◽  
I. G. Khamin ◽  
V. V. Shchukin ◽  
T. V. Shamanskaya ◽  
M. S. Fadeeva ◽  
...  

Polychemotherapy, accompanied by autologous hematopoietic stem cell transplantation, can improve the results of long-term survival of patients with cancer and some non-cancer diseases. Mobilizing and collecting hematopoietic stem cells in children with very low body weight can be a difficult task. The study was approved by the Independent Ethics Committee and the Scientific Council of the Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology. 19 children with extremely low body weight was included in the current study. The median age was 8 (5–14) months, the median of body weight 7.5 (5.8–8.8) kg. Apheresis was performed in an ICU, using sedative therapy and in compliance with the conditions for the prevention of anemia, hypovolemia, hypothermia. 19 hematopoietic stem cell apheresis were performed using the Spectra Optia MNC separator program. Mobilization of CD34+ cells was performed with filgrastim; three children were additionally given plerixaphor. All 19 hematopoietic stem cell apheresis were successful: the median of collected CD34+ cells was 18.7 × 106/kg (8.6– 60.6 × 106/kg), the median apheresis duration was 204 (161–351) min. Serious side effects during apheresis were not recorded, however, in 6 children (31%) we encountered difficulties in the process of installing central venous access. The collection of hematopoietic stem cells for the future high-dose chemotherapy with autologous hematopoietic stem cells is a feasible task even for very young children with extremely low body weight. Correct preparation for manipulation, taking into account all possible risk factors and technical features, can avoid serious complications.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1438-1438
Author(s):  
Natallia Mikhalkevich ◽  
Michael W. Becker

Abstract Abstract 1438 Poster Board I-461 We previously demonstrated the loss of expression of alpha-E-Catenin, the product of the CTNNA1 gene, in primary leukemic stem cells isolated from patients with advanced Myelodysplastic Syndrome (MDS) and Acute Myeloid Leukemia (AML) associated with loss of all or part of the long arm of chromosome 5. To formally assess the impact of loss of Ctnna1 expression on hematopoiesis, we employed a murine model for the hematopoietic specific conditional loss of Ctnna1 expression. We demonstrate that Ctnna1 deficiency is associated with normal hematopoietic maturation and proliferation as assessed by peripheral blood examination and methycellulose colony assays. We assessed stem cell and early progenitor frequencies using both flow cytometry and functional assays. Ctnna1 deficiency was associated with equivalent frequencies of Sca1+C-Kit+CD135-Lineage- HSCs in both experimental animals and controls. Short term HSC and MPP frequencies were likewise unaltered. We assessed HSC function using transplantation studies. In competitive repopulation experiments, HSCs deficient for Ctnna1 maintained stable engraftment of recipient mice for up to 1 year. Limiting dilution analyses detected no significant difference in HSC frequency between wild type and Ctnna1 deficient mice. We examined the potential role of Ctnna1 deficient hematopoietic stem cells in two murine models for myeloid neoplasms 1.) exposure to mutagen ENU and 2.) a model for murine AML driven by the HoxA9-Nup98 fusion product. Following exposure of HSCs to ENU, loss of Ctnna1 was not associated with an increased risk of development of a myeloid neoplasm. Expression of the HoxA9-Nup98 fusion product by retroviral infection of Ctnna1 deficient and wild type Sca1+C-Kit+Lineage- cells resulted in no difference in time to development of the previously characterized myeloproliferative disorder or acute leukemia. Taken together, these data demonstrate that in the absence of specific genetic abnormalities, loss of Ctnna1 expression in primary murine HSCs is not associated with aberrant HSC function or the development of myeloid neoplasms. Further studies are necessary to define a role for of loss of Ctnna1 expression in human myeloid malignancies. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3693-3693 ◽  
Author(s):  
YiYing Xiong ◽  
Fan Qian ◽  
Fen Huang ◽  
Yu Zhang ◽  
Yu Wang ◽  
...  

Abstract Background Engraftment failure (EF) is a formidable complication after autologous hematopoietic stem cell transplantation (auto-HSCT). Mesenchymal stem cells (MSCs) and cord blood (CB) have been found to support hematopoiesis. Thus, we designed a multicenter randomized clinical trial to investigate the effects and safety of MSCs alone or combined with CB infusion for patients with EF. Methods Twenty-two patients were randomly assigned to receive the treatment with MSCs alone (MSCs group, n=11) or MSCs combined with CB (CB group, n=11). MSCs were administered once every 2 weeks (2 doses were a cycle) in both groups, and single-unit CB was administered at the same day with the first application of MSCs in CB group; After one cycle of treatments (within 28 days), the patients who did not response to MSCs would receive the therapeutic schedule in CB group, and those patients with partial response (PR) in MSCs group and those without complete response (CR) in CB group would continue another cycle of MSCs treatment. If patients did not obtain CR after two cycles of treatments (within 56 days), they would receive other treatments including allogeneic HSCT. Results After the first treatment cycle, the effect rates were not significant difference in MSCs and CB groups (7/11 vs. 9/11, P=0.635), and the median time of hematopoietic reconstruction was 22 (18-28) and 17 (13-22) days, respectively (P=0.036) in MSCs and CB group. There was statistically significant difference regarding neutrophil engraftment, with 17 (range 9-28) and 8 (range 6-14) days respectively (P=0.030), but no difference regarding platelet engraftment, with 21 (range 18-28) and 18 (range 11-21) days respectively (P=0.092) between MSCs and CB groups. After two cycles of treatments, 17 patients obtained CR, 2 PR and 3 NR. CB chimerisms were not detected by short tandem repeat (STR) at +15 and +30 days after CB infusion. None of the patients experienced any adverse events of grade 3/4 with the Common Terminology Criteria for Adverse Events v3.0 (CTCAE v3.0) and acute GVHD or chronic GVHD during the period of study treatment and follow-up. One patient with PR in MSCs group and 1 NR in CB group received allogeneic HSCT at +249 and +273 days after auto-HSCT because of EF and primary disease relapse, respectively. At a median follow-up time of 345 (range 129–784) days post-transplantation, 16 patients remained alive, 3 died of relapse of primary diseases and 1 died of CMV pneumonia following allo-HSCT. None of patients developed EBV-DNA viremia and EBV-associated diseases in two groups. The 2-year overall survival, disease-free survival and tumor relapse post-transplantation were 75.2% (95% CI, 63.2-87.2%), 79.5% (95% CI, 70.1-88.9%) and 20.5% (95% CI, 11.1-29.9%) respectively. Conclusions Our data suggest that ex-vivo-expanded MSCs derived from HLA-mismatched BM alone or combined with unrelated CB are effective to EF after auto-HSCT. CB can facilitate the effect of MSCs to EF. Both two strategies do not result in GVHD or increase the risk of primary diseases relapse in patients with EF. This trial was registered at www.clinicaltrials.govas#NCT01763099. Disclosures: Liu: It was supported by 863 Program (No. 2011AA020105) and National Public Health Grand Research Foundation (Grant No. 201202017).: Research Funding; It was supported by National Natural Science Foundation of China (Grant No.81000231, No.81270647) and Science and Technology Program of Guangzhou of China (11A72121174). : Research Funding.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5121-5121 ◽  
Author(s):  
Souad Talhi ◽  
Soufi Osmani ◽  
Mohamed Brahimi ◽  
Kamila Amani ◽  
Hafida Ouldjeriouat ◽  
...  

Abstract INTRODUCTION: Autologous stem cell transplant (ASCT) is the standard of care in transplant-eligible multiple myeloma (MM) patients and is associated witha significant improvement in progression-free survival (PFS), complete remission rates (CR), and overall survival (OS). However, the majority ofpatientsrelapse. This study compares the efficacy of autologous hematopoietic stem cells followed by consolidation bybortezomibbased regimens to the no consolidation therapy in adult patients. PATIENTS AND METHODS: This is a retrospective study over a period of 7 years (2009-2015). All patients less than 65 years with a newly MM diagnosis were included. The protocol used in induction was VD (n=47) treatment whichconsisted of four 3-week cycles of 1.3 mg/m2 bortezomib administered subcutaneously (SC) on days 1, 4, 8, and 11 and 40 mg dexamethasone on days 1Ð4 and 9Ð12. Therapy with VTD was composed of four 3-week cycles of SC bortezomib and dexamethasone at the same doses and schedules as for the VD regimen plus 100 mg/day thalidomide administered orally. Therapy with VCD was composed of four 3-week cycles of SC bortezomib and dexamethasone at the same doses and schedules as for the VD regimen plus 500 mg/m2 cyclophosphamide administered orally on days 1, 8, and 15. Recommended concomitant medications included bisphosphonates, antibiotics, and antiviral prophylaxis. Acetyl salicylic acid was systematically used in the VTD arm. Stem cells were mobilized with 15 or 10 microg/kg G-CSF alone. Leukapheresis to harvest stem cells was performed on day -2 and -1. The grafts were kept in a conventional blood bank refrigerator at 4¡C until reinfusion on day 0. The target yield was 2 x106 CD34+ cells/kg. Following induction therapy, all patients had to proceed to ASCT. The conditioning regimen consisted of melphalan 200 mg/m2 in all patients.The consolidation regimen consisted of two cycles of VD or VCD or VTD after autologous stem- cell transplantation. In our study patients were divided into two groups: Group1 (ASCT plus consolidation) and Group 2 (ASCT alone). The therapeutic evaluation focused on the overall response (CR + VGPR) and progression free survival (PFS) and overall survival (OS) calculated by the Kaplan-Meier method. RESULTS: Over a period of 7 years, 153 patients were collected divided in two group: G1 (n=71) and G2 (n=82). Baseline characteristics are summarized in Table 1. No significant difference was observed between the 2 groups. In terms of CR, 58% of the patients in the G1 achieved a CR after consolidation vs 33% in the G2 after ASCT alone (p=0.007). In terms of VGPR, 31% of the patients in the G1 achieved a least a VGPR vs 17% in the G2 (p=0.04). The relapse rate was significantly lower in the G1 (10%) than the G2 (39%), (p=0.0001). The median follow-up period was 23,4 months. PFS was significantly higher in the G1, median no reached vs 37 months in the G2 (p=0.02) but no significant difference was observed in terms of OS rate between the 2 groups, 91% (G1) versus 82% (G2) at 27 months (p=0.7). CONCLUSION: We conclude thatbortezomib-based regimens as consolidation therapy after ASCT in patients with MM was effective in the improvement of PFS and response rate. Table Patients characteristics. Table. Patients characteristics. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 9 (2) ◽  
pp. 73-85
Author(s):  
Alicja Szubarga ◽  
Marta Kamińska ◽  
Wiktoria Kotlarz ◽  
Stefan Malewski ◽  
Wiktoria Zawada ◽  
...  

Abstract Stem cells are an important subject of research, and are increasingly used in the treatment of various diseases. Due to the development of advanced in vitro techniques, they have become an integral part of modern medicine. The sources of human stem cells are primarily bone marrow and adipose tissue, although non – embryonic stem cells are also scattered throughout the body. Notably, recent research has focused on stem cells found in the oral cavity, both in the dental pulp and oral mucosa. Furthermore, isolation of stem cells from umbilical cord blood is also becoming increasingly popular, while wharton’s jelly and amniotic fluid also seem to be an interesting source of stem cells. The safety and efficacy of stem cells use can be established by animal studies, which are a key element of preclinical research. Mouse, rat and pig models allow for testing of stem cell therapies. Recent studies primarily use mesenchymal stem cells such as mouse – adipose derived mesenchymal stem cells and mouse and rat hematopoietic stem cells. Great hope for future therapies is the use of bioengineering to program cells into induced stem cells, which have the biggest ability for differentiation and transdifferentiation, which carries no risk of teratogenesis. Stem cells are used in many areas of medicine, especially in regenerative medicine, with a growing interest in orthopedics and in the treatment of heart failure. Mesenchymal stem cells are the most used stem cell type, which despite their limited ability to differentiate, give great therapeutic results, mainly due to their immunomodulating effect. Recent studies have even shown that the use of mesenchymal stem cells may be useful in the treatment of COVID-19. Moreover, Research on the use of mesenchymal stem cells in the treatment of Crohn’s disease, acute-graft-versus-host disease and type I diabetes are also promising. The aim of the current review is to present and systematize current knowledge about stem cells, their use and related in vitro research. Running title: Research and use of human stem cells


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4443-4443
Author(s):  
Farrukh Awan ◽  
David Deremer ◽  
Elaine Mebel ◽  
Samith Thomas Kochuparambil ◽  
Anand P. Jillella

Abstract Abstract 4443 Introduction: Various chemotherapeutic agents particularly cyclophosphamide (CY) are utilized in combination with growth factors in an attempt to increase the number of stem cells available for collection in the peripheral blood. Plerixafor (P) is a reversible antagonist of CXCR4 and interrupts its interaction with SDF-1. This results in a rapid release of hematopoietic stem cells from the marrow to the circulation. Recent pivotal phase III trial data has established the efficacy of P in combination with G-CSF (G) in patients who had failed prior attempts at stem cell collection. However, there is limited data about the utility of plerixafor in patients who are being mobilized with chemotherapy and G. Method: In this single institution study of uniformly treated patients we describe our experience with the use of P as a salvage option in patients who fail to optimally mobilize CD34+ cells (>5 × 106 CD34+ cells/kg). Patients received CY (3-4 g/m2) followed by GCSF (10 mcg/kg) from day 1 to day 10. Thirteen patients (6 NHL, 4 MM, 2 Hodgkin lymphoma, 1 Ewings sarcoma) received salvage P from 2008–2010. Their outcomes were compared with 10 matched, historic controls mobilized with (CY n=8; CY + etoposide n=1; CY + topotecan n=1) plus G-CSF (10mcg/kg/d) identified from our institutional database. Data was collected on mobilization and transplant outcomes and analyzed utilizing SPSS version 13.0. Patients receiving P were closely matched to historic controls (CY+G). Result: Both groups were similar with regards to age, gender, disease type, prior therapies and performance status (p>0.05 for all). Patients in the P arm received a median of 2.5 doses (range 1–8). The mean CD34+ count was 21.5cells/ul in the P arm and 32.5 cells/ul in the CY+G arm (p=0.2). Similarly, no significant difference was observed in the average number of apheresis sessions in the P vs. CY+G arms (4.2 vs. 4.4, p=0.8) or the total number of CD34+ stem cells collected (4.0×106/kg vs. 3.9×106/kg, p=0.9). However, 7 out of the 13 patients who received P did have an increase of >10 CD34+ cells/ul in their peripheral blood. Utilizing a cut-off of 5×106 CD34+/kg, 3 (23%) patients in the P arm and 3 (30%) patients in the CY+G arm had a successful harvest. Three NHL patients required >4 doses of P, but all eventually collected >2 × 106 CD34+ cells/kg. Neutrophil and platelet engraftment dynamics were similar in both groups of patients. Median time to neutrophil engraftment was 10 days for both groups, p=0.8, and to platelet engraftment was 22 days vs. 20.5 days, p=0.1, respectively for P vs. CY+G. Conclusion: Our limited single-center retrospective case-controlled outcomes data, suggests that when compared with CY+G, the addition of P as a salvage agent does not significantly improve mobilization outcomes. Further evaluation is needed to combine P with CY+G in terms of optimal timing and potentially dosing of chemotherapy agents utilized. We suggest that the combination P+G would provide better potential outcomes such as improved collection and less hospitalization and reduce the use of chemo-mobilization prior to an Autologous Hematopoietic Stem Cell Transplant. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1205-1205
Author(s):  
Hirofumi Toyama ◽  
Kentaro Hosokawa ◽  
Yoshiko Matsumoto Ikushima ◽  
Toshio Suda ◽  
Fumio Arai

Abstract Abstract 1205 The interaction of stem cells with their supportive microenvironmental niche is critical for sustaining stem cell pools in tissues over long periods of time. Cell-cell and cell-extracellular matrix interactions between hematopoietic stem cells (HSCs) and their niches contribute to the maintenance of stem cell properties. We previously demonstrated that N-cadherin mediated cell adhesion plays a critical role in the HSC engraftment and slow cell division of HSCs. Furthermore, in vitro culture of HSCs with bone-derived osteoblasts that expressed high levels of N-cadherin enhanced the LTR activity of HSCs. However, the expression and function of N-cadherin in HSCs is still controversial. A major problem is that there have been no specific anti-N-cadherin antibodies (Abs) that can be used for the detection of N-cadherin on the surface of living cells. To address this problem, we produced a new anti-N-cadherin Ab. For the production of anti-N-cadherin Abs, we used the phage display library and isolated recombinant Ab clones against mouse N-cadherin. After screening of the phage library and performing quality control ELISA with positive and negative control proteins, we found that three of the seven newly-developed Ab clones were suitable for FACS. FACS analysis with a new N-cadherin Ab showed that BM LSK cells expressed low levels of N-cadherin protein. Furthermore, we confirmed that the reactivity of the new N-cadherin Ab was significantly reduced in N-cadherin deficient LSK cells compared to the wild-type LSK cells. RT-PCR and Q-PCR analysis revealed significantly higher levels of N-cadherin mRNA in N-cadherin+ LSK cells compared with N-cadherin– LSK cells. Next, we performed BMT assays with adult BM-derived N-cadherin+ and N-cadherin– LSK cells isolated by using the new N-cadherin Ab, clone AbD13081, and found that N-cadherin+ LSK cells showed higher BM reconstitution compared with N-cadherin– cells. Furthermore, one of our N-cadherin Ab clones, AbD13077, has neutralizing activity and the use of this clone in cell sorting reduces the LTR activity of N-cadherin+ LSK cells. These data suggested that adult BM HSCs express N-cadherin. Next we examined the expression of N-cadherin in the fetal HSCs using a new N-cadherin Ab. We found that a large number (29.3 ± 2.6 %) of LSK cells in E12.5 fetal liver (FL) expressed N-cadherin. Interestingly, N-cadherin expression was drastically decreased in E15.5 and E18.5 FL LSK cells (13.2 ± 1.9 % in E15.5 and 16.5 ± 1.4 % in E18.5). Immunohistochemical staining revealed that N-cadherin+c-Kit+ cells/N-cadherin+EPCR+ hematopoietic cells adhered to Lyve-1+ endothelial cells in E12.5 FL. Consistent with FACS analysis, N-cadherin expression was decreased in E15.5 and E18.5 FL. In contrast, the expression of E-cadherin in hepatic cells was significantly upregulated in E15.5 and E18.5 FL. Next we analyzed the expression of the LT-HSC marker, EPCR in N-cadherin+ and N-cadherin− LSK cells. We found that EPCR+ cells were enriched in the N-cadherin+ LSK fraction in E12.5 FL, while there was no significant difference in the frequency of EPCR+ cells between N-cadherin+ and N-cadherin− LSK in E15.5 and E18.5 FL LSK cells. Finally, we performed the BMT assay with E12.5, E15.5, and E18.5 FL-derived N-cadherin+ and N-cadherin– LSK cells isolated by AbD13081. Similar to the BM N-cadherin+ LSK cells, E12.5 FL N-cadherin+ LSK cells showed higher LTR activity than N-cadherin– LSK cells. Interestingly, the advantage of LTR in N-cadherin+ LSK cells was decreased in E15.5 and E18.5 FL compared to E12.5 N-cadherin+ LSK cells, although the reconstitution of the N-cadherin+ fraction was higher than N-cadherin– fraction. Altogether, these data suggest that N-cadherin is highly expressed in E12.5 FL HSCs and plays an important role in the HSC-niche interaction for the maintenance of HSC activity. We speculated that the decrease of N-cadherin in HSC and FL cells during FL development might contribute to the migration of HSCs from FL to BM. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 31 (10) ◽  
pp. 981-991 ◽  
Author(s):  
Barclay J. Lee ◽  
Emily M. Mace

Lymphocyte development is a complex and coordinated pathway originating from pluripotent stem cells during embryogenesis and continuing even as matured lymphocytes are primed and educated in adult tissue. Hematopoietic stem cells develop in a specialized niche that includes extracellular matrix and supporting stromal and endothelial cells that both maintain stem cell pluripotency and enable the generation of differentiated cells. Cues for lymphocyte development include changes in integrin-dependent cell motility and adhesion which ultimately help to determine cell fate. The capacity of lymphocytes to adhere and migrate is important for modulating these developmental signals both by regulating the cues that the cell receives from the local microenvironment as well as facilitating the localization of precursors to tissue niches throughout the body. Here we consider how changing migratory and adhesive phenotypes contribute to human natural killer (NK)- and T-cell development as they undergo development from precursors to mature, circulating cells and how our understanding of this process is informed by in vitro models of T- and NK cell generation.


Sign in / Sign up

Export Citation Format

Share Document