Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.721
Filter
1.
J Clin Apher ; 39(3): e22127, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38803152

ABSTRACT

BACKGROUND: Increasing indications for cellular therapy collections have stressed our healthcare system, with autologous collections having a longer than desired wait time until apheresis collection. This quality improvement initiative was undertaken to accommodate more patients within existing resources. STUDY DESIGN AND METHODS: Patients with multiple myeloma who underwent autologous peripheral blood stem cell collection from October 2022 to April 2023 were included. Demographic, mobilization, laboratory, and apheresis data were retrospectively collected from the medical record. RESULTS: This cohort included 120 patients (49.2% male), with a median age of 60 years. All received G-CSF and 95% received pre-emptive Plerixafor approximately 18 hours pre-collection. Most (79%) had collection goals of at least 8 × 106/kg CD34 cells, with 63% over 70 years old having this high collection goal (despite 20 years of institutional data showing <1% over 70 years old have a second transplant). With collection efficiencies of 55.9%, 44% of patients achieved their collection goal in a single day apheresis collection. A platelet count <150 × 103/µL on the day of collection was a predictor for poor mobilization; among 27 patients with a low baseline platelet count, 17 did not achieve the collection goal and 2 failed to collect a transplantable dose. CONCLUSIONS: With minor collection goal adjustments, 15% of all collection appointments could have been avoided over this 6-month period. Other strategies to accommodate more patients include mobilization modifications (Plerixafor timing or substituting a longer acting drug), utilizing platelet counts to predict mobilization, and modifying apheresis collection volumes or schedule templates.


Subject(s)
Benzylamines , Cyclams , Granulocyte Colony-Stimulating Factor , Hematopoietic Stem Cell Mobilization , Multiple Myeloma , Transplantation, Autologous , Humans , Multiple Myeloma/therapy , Cyclams/pharmacology , Cyclams/therapeutic use , Middle Aged , Male , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Mobilization/methods , Aged , Retrospective Studies , Blood Component Removal/methods , Heterocyclic Compounds/administration & dosage , Heterocyclic Compounds/therapeutic use , Adult , Peripheral Blood Stem Cell Transplantation/methods , Platelet Count
3.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(2): 556-560, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38660866

ABSTRACT

OBJECTIVE: To investigate the efficiency and optimal time of stem cell apheresis mobilized by pegylated recombinant human granulocyte colony stimulating factor (PEG-rhG-CSF) in autologous stem cell transplantation (ASCT) for hematological malignancies without monitoring pre-collection CD34+ cells. METHODS: Forty-six patients underwent stem cell mobilization were retrospectively analyzed between August 2017 and January 2022 at the First Affiliated Hospital of Fujian Medical University. 27 patients using high dose chemotherapy combined with PEG-rhG-CSF mobilization were enrolled in the PEG-rhG-CSF group, and other 19 patients mobilized with recombinant human granulocyte colony stimulating factor (G-CSF) were enrolled in G-CSF group. The mobilization and collection effects of the patients in two groups were compared. RESULTS: A total of 46 patients underwent 86 apheresis procedures, the median amount of mononuclear cell (MNC) in the PEG-rhG-CSF group and G-CSF group was 6.54(3.85-12.61)×108/kg and 6.15(1.13-11.58)×108/kg, respectively (P >0.05), the total CD34+ cells of the grafts were 11.44(1.33-65.02)×106/kg and 4.95(0.30-24.02)×106/kg (P < 0.05), with harvest timing of 14(10-20) days and 14(4-22) days, respectively (P >0.05). In the PEG-rhG-CSF group, there was a significant difference between the number of CD34+ cells collected when white blood cells (WBC) ≥10×109/L and WBC<10×109 /L, 19.04(2.85-65.02)×106/kg and 6.22(0.81-34.86)×106/kg, respectively (P < 0.05). CONCLUSION: Stem cells mobilization with PEG-rhG-CSF was highly efficient with a median mobilization time of 14 days. In the absence of peripheral blood CD34 monitoring, peripheral blood WBC≥10×109/L can be considered as a threshold for a single stem cell apheresis to collect sufficient stem cells.


Subject(s)
Granulocyte Colony-Stimulating Factor , Hematologic Neoplasms , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Polyethylene Glycols , Recombinant Proteins , Transplantation, Autologous , Humans , Granulocyte Colony-Stimulating Factor/administration & dosage , Retrospective Studies , Hematologic Neoplasms/therapy , Antigens, CD34 , Hematopoietic Stem Cells/cytology , Female , Male
4.
Cells ; 13(7)2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38607025

ABSTRACT

Achieving successful hematopoietic stem cell transplantation (HSCT) relies on two fundamental pillars: effective mobilization and efficient collection through apheresis to attain the optimal graft dose. These cornerstones pave the way for enhanced patient outcomes. The primary challenges encountered by the clinical unit and collection facility within a transplant program encompass augmenting mobilization efficiency to optimize the harvest of target cell populations, implementing robust monitoring and predictive strategies for mobilization, streamlining the apheresis procedure to minimize collection duration while ensuring adequate yield, prioritizing patient comfort by reducing the overall collection time, guaranteeing the quality and purity of stem cell products to optimize graft function and transplant success, and facilitating seamless coordination between diverse entities involved in the HSCT process. In this review, we aim to address key questions and provide insights into the critical aspects of mobilizing and collecting hematopoietic stem cells for transplantation purposes.


Subject(s)
Blood Component Removal , Hematopoietic Stem Cell Transplantation , Adult , Humans , Hematopoietic Stem Cell Mobilization/methods , Transplantation, Homologous , Blood Component Removal/methods , Hematopoietic Stem Cells
5.
Pediatr Transplant ; 28(3): e14751, 2024 May.
Article in English | MEDLINE | ID: mdl-38659194

ABSTRACT

BACKGROUND: The published experience concerning autologous peripheral blood stem cell collection in children is very limited. METHODS: The data of pediatric patients who underwent autologous stem cell mobilization and apheresis between January 2011 and April 2020 were analyzed retrospectively. RESULTS: We studied retrospectively 64 mobilization and apheresis procedures in 48 pediatric patients (34 males, 14 females), mean age of 7.31 ± 5.38 (range, 1.5-19.7) years, the underlying disease was mostly neuroblastoma (NBL). The body weight of 21 patients (43.75%) was 15 kg or less. The targeted autologous peripheral stem cell apheresis (APSCA) was successfully achieved in 98% of patients. Neuroblastoma patients were younger than the rest of the patients and underwent apheresis after receiving fewer chemotherapy cycles than others and all of them mobilized within the first session successfully. Plerixafor was added to mobilization in nine heavily pretreated patients (18.7%), median two doses (range, 1-4 doses). 11 patients (22.9%) underwent radiotherapy (RT) before mobilization with doses of median 24 Gy (range, 10.8-54.0 Gy). Patients with RT were older at the time of apheresis and had received more chemotherapy courses than patients without RT. As a result, patients with a history of RT had significantly lower peripheral CD34+ cells and CD34+ yields than those without RT. In 17 patients (35.4%), 22 different complications were noted. The most common complications were catheter-related infections (n:10, 20.8%), followed by catheter-related thrombosis in eight patients (16.7%). CONCLUSIONS: Patients who had far less therapy before apheresis were more likely to mobilize successfully. Our study provides a detailed practice approach including complications during APSCA aiming to increase the success rates of apheresis in transplantation centers.


Subject(s)
Blood Component Removal , Hematopoietic Stem Cell Mobilization , Neoplasms , Peripheral Blood Stem Cell Transplantation , Transplantation, Autologous , Humans , Female , Male , Hematopoietic Stem Cell Mobilization/methods , Child , Retrospective Studies , Child, Preschool , Adolescent , Infant , Blood Component Removal/methods , Peripheral Blood Stem Cell Transplantation/methods , Neoplasms/therapy , Young Adult , Peripheral Blood Stem Cells
6.
Stem Cell Reports ; 19(5): 639-653, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38608679

ABSTRACT

Sepsis survivors exhibit immune dysfunction, hematological changes, and increased risk of infection. The long-term impacts of sepsis on hematopoiesis were analyzed using a surgical model of murine sepsis, resulting in 50% survival. During acute disease, phenotypic hematopoietic stem and progenitor cells (HSPCs) were reduced in the bone marrow (BM), concomitant with increased myeloid colony-forming units and extramedullary hematopoiesis. Upon recovery, BM HSPCs were increased and exhibited normal function in the context of transplantation. To evaluate hematopoietic responses in sepsis survivors, we treated recovered sham and cecal ligation and puncture mice with a mobilizing regimen of granulocyte colony-stimulating factor (G-CSF) at day 20 post-surgery. Sepsis survivors failed to undergo emergency myelopoiesis and HSPC mobilization in response to G-CSF administration. G-CSF is produced in response to acute infection and injury to expedite the production of innate immune cells; therefore, our findings contribute to a new understanding of how sepsis predisposes to subsequent infection.


Subject(s)
Granulocyte Colony-Stimulating Factor , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells , Myelopoiesis , Sepsis , Animals , Sepsis/complications , Granulocyte Colony-Stimulating Factor/pharmacology , Mice , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Disease Models, Animal , Mice, Inbred C57BL , Male , Survivors
7.
Transfus Med Rev ; 38(2): 150824, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38569349

ABSTRACT

Limited data regarding elevation of N-terminal pro-B-type natriuretic peptide (NT-proBNP) in mobilized donors with G-CSF is available. We extended these findings by examining serum NT-proBNP in a cohort study including 35 healthy donors and 69 patients who received G-CSF for CD34+ mobilization as well as 54 patients who did not receive G-CSF but who underwent collection of CD3+ cells for chimeric antigen receptor (CAR) T-cell manufacturing. No donor in the three cohorts experienced significant cardiac adverse events. NT-proBNP levels were measured before and after G-CSF administration and after finishing apheresis procedure. NT-proBNP increase was observed in mobilized healthy donors after G-CSF administration, but was not observed in mobilized or non-mobilized patients. Only in the cohort of healthy donors, pairwise comparisons using Wilcoxon signed ranks test showed a significant increase between the mean serum NT-proBNP level after G-CSF administration and the mean serum NT-proBNP level measured before G-CSF administration (231.09 ± 156.15 pg/mL vs. 58.88 ± 26.84 pg/mL; P < .01). No correlation was observed between NT-proBNP increase and G-CSF dose (rs = 0.09; n = 32; P = .6) and no other variables contributing to predict serum NT-proBNP increase were detected. In conclusion, we observed a statistically, although not clinically, significant increase of NT-proBNP in healthy donors who received G-CSF as CD34+ cell mobilization.


Subject(s)
Granulocyte Colony-Stimulating Factor , Hematopoietic Stem Cell Mobilization , Natriuretic Peptide, Brain , Peptide Fragments , Humans , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Male , Granulocyte Colony-Stimulating Factor/blood , Female , Hematopoietic Stem Cell Mobilization/methods , Middle Aged , Adult , Cohort Studies , Aged , Blood Donors , Antigens, CD34
10.
Transfus Apher Sci ; 63(3): 103934, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38678982

ABSTRACT

Autologous hematopoietic progenitor cell transplantation (ASCT) has been used for more than five decades to treat malignant and non-malignant diseases. Successful engraftment after high-dose chemotherapy relies on the ability to collect sufficient CD34 + hematopoietic progenitor cells (HPCs), typically from peripheral blood after mobilization. Commonly, either granulocyte colony-stimulating factor (G-CSF) alone as a single agent (i.e. steady-state mobilization) or G-CSF after chemotherapy is administered to collect adequate numbers of HPCs (minimum ≥2 × 106 CD34 + cells/kg for one ASCT; optimal up to 5 × 106 CD34 + cells/kg). However, a significant proportion of patients fail successful HPC mobilization, which is commonly defined as a CD34+ cell count below 10-15/µL after at least 4 days of 10 µg/kg b.w. G-CSF alone, or after chemo-mobilization in combination with 5-10 µg/kg b.w. G-CSF. In these situations plerixafor, a chemokine receptor inhibitor (CXCR4) can be used to enhance HPC collection in patients with multiple myeloma and malignant lymphoma whose cells mobilize poorly. Risk factors for poor mobilization have been evaluated and several strategies (e.g. plerixafor to rescue the mobilization approach or pre-emptive use) have been suggested to optimize mobilization, especially in patients at risk. This manuscript discusses the risk factors of poor CD34+ mobilization and summarizes the current strategies to optimize mobilization and HPC collection.


Subject(s)
Hematopoietic Stem Cell Mobilization , Humans , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Granulocyte Colony-Stimulating Factor/therapeutic use , Cyclams/pharmacology , Cyclams/therapeutic use , Benzylamines
11.
Transfusion ; 64(5): 871-880, 2024 May.
Article in English | MEDLINE | ID: mdl-38600674

ABSTRACT

BACKGROUND: Despite recent advances in the treatment of multiple myeloma, high-dose chemotherapy followed by autologous hematopoietic stem cell transplantation (ASCT) remains an essential therapeutic keystone. As for the stem cell mobilization procedure, different regimens have been established, usually consisting of a cycle of chemotherapy followed by application of granulocyte-colony stimulating factor (G-CSF), although febrile neutropenia is a common complication. Following national guidelines, our institution decided to primarily use G-CSF only mobilization during the COVID-19 pandemic to minimize the patients' risk of infection and to reduce the burden on the health system. STUDY DESIGN AND METHODS: In this retrospective single-center analysis, the efficacy and safety of G-CSF only mobilization was evaluated and compared to a historic control cohort undergoing chemotherapy-based mobilization by cyclophosphamide and etoposide (CE) plus G-CSF. RESULTS: Although G-CSF only was associated with a higher need for plerixafor administration (p < .0001) and a higher number of apheresis sessions per patient (p = .0002), we were able to collect the target dose of hematopoietic stem cells in the majority of our patients. CE mobilization achieved higher hematopoietic stem cell yields (p = .0015) and shorter apheresis sessions (p < .0001) yet was accompanied by an increased risk of febrile neutropenia (p < .0001). There was no difference in engraftment after ASCT. DISCUSSION: G-CSF only mobilization is a useful option in selected patients with comorbidities and an increased risk of serious infections, especially in the wintertime or in future pandemics.


Subject(s)
Cyclophosphamide , Etoposide , Granulocyte Colony-Stimulating Factor , Hematopoietic Stem Cell Mobilization , Multiple Myeloma , Transplantation, Autologous , Humans , Hematopoietic Stem Cell Mobilization/methods , Multiple Myeloma/therapy , Retrospective Studies , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocyte Colony-Stimulating Factor/therapeutic use , Middle Aged , Male , Female , Cyclophosphamide/therapeutic use , Cyclophosphamide/administration & dosage , Aged , Etoposide/therapeutic use , Etoposide/administration & dosage , Hematopoietic Stem Cell Transplantation/methods , Benzylamines , COVID-19 , Adult , Cyclams/therapeutic use , Cyclams/pharmacology , SARS-CoV-2 , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects
12.
Ann Med ; 56(1): 2329140, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38470973

ABSTRACT

AIM: The combination of granulocyte-colony stimulating factor (G-CSF) and plerixafor is one of the approaches for hematopoietic stem cell mobilization in patients with multiple myeloma (MM), non-Hodgkin's lymphoma (NHL), and Hodgkin's lymphoma (HL). This systematic review and meta-analysis aimed to determine the ability of G-CSF + plerixafor to mobilize peripheral blood (PB) CD34+ cells and examine its safety profile. METHODS: We performed a database search using the terms 'granulocyte colony stimulating factor', 'G-CSF', 'AMD3100', and 'plerixafor', published up to May 1, 2023. The methodology is described in further detail in the PROSPERO database (CRD42023425760). RESULTS: Twenty-three studies were included in this systematic review and meta-analysis. G-CSF + plerixafor resulted in more patients achieving the predetermined apheresis yield of CD34+ cells than G-CSF alone (OR, 5.33; 95%, 4.34-6.55). It was further discovered that G-CSF + plerixafor could mobilize more CD34+ cells into PB, which was beneficial for the next transplantation in both randomized controlled (MD, 18.30; 95%, 8.74-27.85) and single-arm (MD, 20.67; 95%, 14.34-27.00) trials. Furthermore, G-CSF + plerixafor did not cause more treatment emergent adverse events than G-CSF alone (OR, 1.25; 95%, 0.87-1.80). CONCLUSIONS: This study suggests that the combination of G-CSF and plerixafor, resulted in more patients with MM, NHL, and HL, achieving the predetermined apheresis yield of CD34+ cells, which is related to the more effective mobilization of CD34+ cells into PB.


Subject(s)
Hematopoietic Stem Cell Transplantation , Heterocyclic Compounds , Lymphoma, Non-Hodgkin , Lymphoma , Multiple Myeloma , Humans , Hematopoietic Stem Cell Mobilization/methods , Multiple Myeloma/therapy , Granulocyte Colony-Stimulating Factor , Heterocyclic Compounds/adverse effects , Lymphoma/chemically induced , Lymphoma/therapy , Lymphoma, Non-Hodgkin/chemically induced , Lymphoma, Non-Hodgkin/therapy , Hematopoietic Stem Cells , Transplantation, Autologous , Benzylamines , Hematopoietic Stem Cell Transplantation/methods
13.
Cell Commun Signal ; 22(1): 177, 2024 03 12.
Article in English | MEDLINE | ID: mdl-38475811

ABSTRACT

BACKGROUND: The incidence of multiple myeloma (MM), a type of blood cancer affecting monoclonal plasma cells, is rising. Although new drugs and therapies have improved patient outcomes, MM remains incurable. Recent studies have highlighted the crucial role of the chemokine network in MM's pathological mechanism. Gaining a better understanding of this network and creating an overview of chemokines in MM could aid in identifying potential biomarkers and developing new therapeutic strategies and targets. PURPOSE: To summarize the complicated role of chemokines in MM, discuss their potential as biomarkers, and introduce several treatments based on chemokines. METHODS: Pubmed, Web of Science, ICTRP, and Clinical Trials were searched for articles and research related to chemokines. Publications published within the last 5 years are selected. RESULTS: Malignant cells can utilize chemokines, including CCL2, CCL3, CCL5, CXCL7, CXCL8, CXCL12, and CXCL13 to evade apoptosis triggered by immune cells or medication, escape from bone marrow and escalate bone lesions. Other chemokines, including CXCL4, CCL19, and CXCL10, may aid in recruiting immune cells, increasing their cytotoxicity against cancer cells, and inducing apoptosis of malignant cells. CONCLUSION: Utilizing anti-tumor chemokines or blocking pro-tumor chemokines may provide new therapeutic strategies for managing MM. Inspired by developed CXCR4 antagonists, including plerixafor, ulocuplumab, and motixafortide, more small molecular antagonists or antibodies for pro-tumor chemokine ligands and their receptors can be developed and used in clinical practice. Along with inhibiting pro-tumor chemokines, studies suggest combining chemokines with chimeric antigen receptor (CAR)-T therapy is promising and efficient.


Subject(s)
Heterocyclic Compounds , Multiple Myeloma , Humans , Hematopoietic Stem Cell Mobilization , Chemokines , Signal Transduction , Biomarkers
14.
Curr Opin Hematol ; 31(3): 104-114, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38359264

ABSTRACT

PURPOSE OF REVIEW: Gene therapy for sickle cell disease (SCD) is advancing rapidly, with two transformative products recently approved by the US Food and Drug Administration and numerous others under study. All current gene therapy protocols require ex vivo modification of autologous hematopoietic stem cells (HSCs). However, several SCD-related problems impair HSC collection, including a stressed and damaged bone marrow, potential cytotoxicity by the major therapeutic drug hydroxyurea, and inability to use granulocyte colony stimulating factor, which can precipitate severe vaso-occlusive events. RECENT FINDINGS: Peripheral blood mobilization of HSCs using the CXCR4 antagonist plerixafor followed by apheresis collection was recently shown to be safe and effective for most SCD patients and is the current strategy for mobilizing HSCs. However, exceptionally large numbers of HSCs are required to manufacture an adequate cellular product, responses to plerixafor are variable, and most patients require multiple mobilization cycles, increasing the risk for adverse events. For some, gene therapy is prohibited by the failure to obtain adequate numbers of HSCs. SUMMARY: Here we review the current knowledge on HSC collection from individuals with SCD and potential improvements that may enhance the safety, efficacy, and availability of gene therapy for this disorder.


Subject(s)
Anemia, Sickle Cell , Hematopoietic Stem Cell Transplantation , Heterocyclic Compounds , Humans , Hematopoietic Stem Cell Mobilization/methods , Heterocyclic Compounds/therapeutic use , Heterocyclic Compounds/adverse effects , Hematopoietic Stem Cells/metabolism , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Granulocyte Colony-Stimulating Factor , Genetic Therapy/adverse effects
15.
Transfusion ; 64(4): 742-750, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38407504

ABSTRACT

Plerixafor (PLER), a reversible antagonist of the CXC chemokine receptor type 4, has been in clinical use for mobilization of blood grafts for autologous hematopoietic cell transplantation (AHCT) for about 15 years. Initially PLER was investigated in placebo-controlled trials with the granulocyte colony-stimulating factor (G-CSF) filgrastim. It has also been used in combination with chemotherapy plus G-CSF in patients who had failed a previous mobilization attempt or appeared to mobilize poorly with current mobilization (preemptive use). This review summarizes what is known regarding addition of PLER to standard mobilization regimens. PLER increases mobilization of CD34+ cells, decreases the number of apheresis sessions needed to achieve collection targets and increases the proportion of patients who can proceed to AHCT. It appears also to increase the amount of various lymphocyte subsets in the grafts collected. In general, hematologic recovery after AHCT has been comparable to patients mobilized without PLER, although slower platelet recovery has been observed in some studies of patients who mobilize poorly. In phase III studies, long-term outcome has been comparable to patients mobilized without PLER. This also appears to be the case in patients receiving plerixafor for poor or suboptimal mobilization of CD34+ cells. In practice, PLER is safe and has not been shown to increase tumor cell mobilization.


Subject(s)
Hematopoietic Stem Cell Transplantation , Heterocyclic Compounds , Multiple Myeloma , Humans , Hematopoietic Stem Cell Mobilization , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/therapeutic use , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/therapeutic use , Transplantation, Autologous , Multiple Myeloma/therapy , Antigens, CD34/metabolism
16.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(1): 322-326, 2024 Feb.
Article in Chinese | MEDLINE | ID: mdl-38387943

ABSTRACT

Plerixafor, an analog of C-X-C motif chemokine receptor 4 (CXCR4), which allows the release of stem cells from the bone marrow into peripheral blood (PB) by disrupting the interaction of CXCR4 with stromal cell-derived factor-1 (SDF-1), is effective in mobilization for peripheral blood stem cells (PBSC). Due to its market approval has not been long and its high price in China, the clinical application of plerixafor is still very limited. The clinicians are actively seeking the optimal use of plerixafor to improve the success rate of PBSC collection and reduce the cost. This article reviews the latest research progress related to plerixafor application, in order to summarize the optimal use of plerixafor in autologous hematopoietic stem cell transplantation (auto-HSCT).


Subject(s)
Cyclams , Heterocyclic Compounds , Peripheral Blood Stem Cells , Humans , Hematopoietic Stem Cell Mobilization , Transplantation, Autologous , Benzylamines
18.
Blood Cells Mol Dis ; 105: 102824, 2024 03.
Article in English | MEDLINE | ID: mdl-38262104

ABSTRACT

In preparation for hematopoietic stem cell mobilization and collection, current ex vivo gene therapy protocols for sickle cell disease require patients to undergo several months of chronic red cell transfusion. For health care equity, alternatives to red cell transfusion should be available. We examined whether treatment with GBT1118, the murine analog of voxelotor, could be a safe and feasible alternative to red cell transfusion. We found that 3 weeks of treatment with GBT1118 increased the percentage of bone marrow hematopoietic stem cells and upon plerixafor mobilization, the percentage of peripheral blood hematopoietic stem cells. Our data suggest that voxelotor should be further explored for its potential safety and utility as preparation for hematopoietic stem cell mobilization and collection.


Subject(s)
Anemia, Sickle Cell , Benzaldehydes , Hematopoietic Stem Cell Transplantation , Heterocyclic Compounds , Niacinamide/analogs & derivatives , Pyrazines , Humans , Mice , Animals , Hematopoietic Stem Cell Mobilization/methods , Bone Marrow/metabolism , Hematopoietic Stem Cells/metabolism , Heterocyclic Compounds/therapeutic use , Heterocyclic Compounds/pharmacology , Pyrazoles , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Anemia, Sickle Cell/metabolism , Genetic Therapy/adverse effects , Granulocyte Colony-Stimulating Factor/pharmacology
19.
J Clin Apher ; 39(1): e22102, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38186371

ABSTRACT

BACKGROUND: Peripheral blood stem cell (PBSC) collection in children poses challenges due to their small size, low body weight (BW), and unique pediatric physiology, especially among children weighing 20 kg (kg) or less. METHODS: PBSC collection data of both healthy children and patients with thalassemia major (TM) weighing 20 kg or less between January 2013 and December 2020 were reviewed. Moreover, PBSCs characteristics along with various aspects of efficiency and safety between healthy donors and patients with TM were compared. RESULTS: A total of 262 PBSC procedures were performed on 255 children. Of these, 91 procedures were carried out on 85 allogeneic healthy donors, and 171 auto-backup collections were performed on 170 patients with TM to ensure PBSC availability and prevent transplantation failure. A minimum pre-apheresis hemoglobin (HGB) level of 60 g/L was discovered to be safe and feasible in patients with TM. The median CD34+ cell dose in the PBSC product during the initial apheresis procedure was higher in healthy donors compared to patients with TM (7.29 ± 5.28 × 106 cells/kg vs5.88 ± 4.23 × 106 cells/kg, P = .043). The total CD34+ cells/kg recipient weight exhibited a positive correlation with pre-apheresis monocyte counts, but a negative correlation with donor weight. Apheresis significantly reduced hematocrit and platelet counts in the allogeneic group compared to the autologous group. Patients with TM experienced a higher occurrence of bone pain related to granulocyte colony-stimulating factor treatment. Notably, no serious complications related to PBSCs mobilization, central venous catheter placement, or the apheresis procedure were observed in either group. CONCLUSIONS: PBSCs collection was both safe and effective in healthy children and pediatric patients with TM weighing 20 kg or less.


Subject(s)
Blood Component Removal , Peripheral Blood Stem Cells , beta-Thalassemia , Humans , Child , beta-Thalassemia/complications , beta-Thalassemia/therapy , Hematopoietic Stem Cell Mobilization/methods , Granulocyte Colony-Stimulating Factor
20.
Blood ; 143(16): 1656-1669, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38295333

ABSTRACT

ABSTRACT: Autologous stem cell transplantation (ASCT) is the standard of care consolidation therapy for eligible patients with myeloma but most patients eventually progress, an event associated with features of immune escape. Novel approaches to enhance antimyeloma immunity after ASCT represent a major unmet need. Here, we demonstrate that patient-mobilized stem cell grafts contain high numbers of effector CD8 T cells and immunosuppressive regulatory T cells (Tregs). We showed that bone marrow (BM)-residing T cells are efficiently mobilized during stem cell mobilization (SCM) and hypothesized that mobilized and highly suppressive BM-derived Tregs might limit antimyeloma immunity during SCM. Thus, we performed ASCT in a preclinical myeloma model with or without stringent Treg depletion during SCM. Treg depletion generated SCM grafts containing polyfunctional CD8 T effector memory cells, which dramatically enhanced myeloma control after ASCT. Thus, we explored clinically tractable translational approaches to mimic this scenario. Antibody-based approaches resulted in only partial Treg depletion and were inadequate to recapitulate this effect. In contrast, a synthetic interleukin-2 (IL-2)/IL-15 mimetic that stimulates the IL-2 receptor on CD8 T cells without binding to the high-affinity IL-2Ra used by Tregs efficiently expanded polyfunctional CD8 T cells in mobilized grafts and protected recipients from myeloma progression after ASCT. We confirmed that Treg depletion during stem cell mobilization can mitigate constraints on tumor immunity and result in profound myeloma control after ASCT. Direct and selective cytokine signaling of CD8 T cells can recapitulate this effect and represent a clinically testable strategy to improve responses after ASCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Multiple Myeloma , Humans , Multiple Myeloma/pathology , T-Lymphocytes, Regulatory , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cell Mobilization/methods , Transplantation, Autologous , Stem Cell Transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...