Next Article in Journal
An OX-Tra’Ordinary Tale: The Role of OX40 and OX40L in Atopic Dermatitis
Next Article in Special Issue
Allogeneic Stem Cell Transplantation in Refractory Acute Myeloid Leukaemia
Previous Article in Journal
Controlling Recombination to Evolve Bacteriophages
Previous Article in Special Issue
Cellular Strategies for Separating GvHD from GvL in Haploidentical Transplantation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Stem Cells Collection and Mobilization in Adult Autologous/Allogeneic Transplantation: Critical Points and Future Challenges

by
Michele Prisciandaro
1,*,
Enrico Santinelli
2,3,
Valeria Tomarchio
2,
Maria Antonietta Tafuri
2,
Cecilia Bonchi
1,
Gloria Palazzo
1,
Carolina Nobile
1,
Alessandra Marinucci
1,
Marcella Mele
2,
Ombretta Annibali
2,
Luigi Rigacci
2 and
Michele Vacca
1,*
1
Operative Research Unit of Transfusion Medicine and Cellular Therapy, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy
2
Operative Research Unit of Hematology and Stem Cell Transplantation, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy
3
Program in Immunology, Molecular Medicine and Applied Biotechnologies, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
*
Authors to whom correspondence should be addressed.
Cells 2024, 13(7), 586; https://doi.org/10.3390/cells13070586
Submission received: 5 February 2024 / Revised: 23 March 2024 / Accepted: 26 March 2024 / Published: 28 March 2024

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.

1. Introduction

Successful hematopoietic stem cell transplantation hinges on two critical steps: effective mobilization and efficient collection by the apheresis unit to achieve the optimal transplant dose. These key processes unlock the door to improved transplant outcomes.
The main challenges faced by the clinical unit and collection facility in a transplant program include enhancing mobilization efficiency to optimize the collection of target components, monitoring and predicting mobilization, streamlining the apheresis procedure to reduce the number of days required for adequate collection, enhancing patient comfort by minimizing the overall collection duration, ensuring the quality and purity of stem cell products to optimize graft quality and transplantation outcome, and coordinating between the various facilities involved in stem cell transplantation.
In this review, we provide answers to questions about the crucial aspects of mobilizing and collecting hematopoietic stem cells (HSCs) for transplantation purposes.
The most common cell source for autologous and allogeneic transplantations is peripheral blood stem cells (PBSCs). As it is well known, the number of PBSCs in the peripheral blood is 0.1–0.5% [1]; therefore, it is necessary to implement mobilization strategies to increase the number and allow cell collection. What are the current mobilization strategies and future challenges?

1.1. Allogeneic Mobilization

Allogeneic hematopoietic stem cell transplantation (allo-HSCT), despite the introduction of several new drugs and cellular therapies that efficiently improve the outcome of high-risk hematological diseases, still represents the curative strategy for many of these life-threatening disorders [2].
In allo-HSCT, the three sources to collect the stem cell yield are the bone marrow (BM), the peripheral blood (PB), and the cord blood unit (CBU).
BM was the first stem cell source adopted for allo-HSCT. BM can be collected by multiple aspirations (each one not exceeding a 5 mL volume) from the posterior superior iliac crest while the donor is under general anesthesia. The maximum volume that can be collected is 20 mL/kg of donor weight, hopefully reaching at least 3 × 108 nucleated cells/kg of recipient weight [3].
The PB source for allo-HCT was introduced in the early 1990s as an effective stem cell source [4]. Due to the faster engraftment, the improved “graft versus leukemia” effect, the avoidance of donor exposure to general anesthesia and BM harvesting, and the favorable logistic aspects, in a few years, PB established itself as the preferred SC source for allo-HSCT [5].
Cord blood transplantation was first described in the 1990s [6,7] as an attractive alternative source, particularly in case of related or unrelated matched donor unavailability. The fast access to stem cell yields and the less stringent HLA-matching requirements favored the spread of CBU transplantation as a valid alternative option during the early 2000s, despite several disadvantages like the limited cell dose, the prolonged aplasia period, and the unavailability of the donor for further donations [8]. However, the introduction and the diffusion of the haploidentical donor as a valid option in case of an HLA-fully matched donor unavailability set off the near disappearance of CBU utilization [5].

1.2. Stem Cell Mobilization in Healthy Donors

After a successful but not reproducible attempt to collect peripheral blood stem cells (PBSCs) without stimulation [9], several studies were conducted to establish the optimal mobilization strategy to adopt. Hence, the possibility of collecting an adequate stem cell yield in a suitable time through the subcutaneous administration of granulocyte colony-stimulating factor (G-CSF) alone was confirmed [4,10]. G-CSF was chosen over GM-CSF for the lower incidence of side effects [11].
G-CSF favors mobilization, promoting the degradation of adhesion molecules such as VCAM-1 that tie stem cells to the bone marrow microenvironment and by reducing the interaction between CXCR4 (a transmembrane receptor expressed on CD34+ cells) and its chemokine CXCL12, through its transcription repression, inducing peripheral blood cell migration [12,13]. Furthermore, G-CSF interacts with osteoclasts, which may promote cellular mobilization [11,14].
Furthermore, G-CSF induces the expansion of specific lymphocytic subsets that enrich the stem cell product. In particular, these subpopulations may provide the “graft versus host” effect, like the naïve CD4+ T cells, but may also have an immunoregulatory function associated with promoting the engraftment, like the Tregs. These concepts are the basis of graft manipulation, like T cell depletion, applied generally to avoid graft-versus-host disease. However, this topic is beyond the scope of this review, and it is deepened in specific articles [15].
Several experiments were initially conducted with different doses of G-CSF [16,17,18,19,20]; however, a G-CSF dose of 10 µg/kg for 5 consecutive days was associated with a better CD34+ blood peak and a higher level of CD34+ cells in the yield, compared to a G-CSF lower dose. Moreover, the peak of CD34+ level has been shown to occur 24 h following the fourth dose of G-CSF [21]. Using this evidence, the first EBMT consensus set the G-CSF dose for PBSCs mobilization in healthy donors at 10 µg/kg for 5 consecutive days; furthermore, this paper highlighted the indications to perform leukapheresis (processing up to 15 L of blood per leukapheresis by continuous flow) and the optimal CD34+ target dose to reach (2–3 × 106/kg of recipient weight) [22].
Applying this mobilization policy, a high frequency of achieving a CD34+ level above 4 × 106/kg of recipient weight through a maximum of two leukaphereses, which was associated with faster engraftment, was noted [23].
Some experiments applying a higher dose of G-CSF did not result in a higher CD34+ cell yield [24,25].
Currently, the EBMT recommends the employment of filgrastim or lenograstim 10 µg/kg/day for at least 4 days prior to stem cell collection [3].
Biosimilar versions of filgrastim, as the recombinant human granulocyte colony-stimulating factor (rhG-CSF), did not show any difference in mobilization in terms of efficacy and adverse events compared to the originator product; therefore, they can be safely administered for this purpose [26,27].
Recent studies have highlighted the feasibility of starting the stem cell collection on the 4th, instead of the 5th, day of G-CSF administration, showing no differences in terms of the CD34+ cell yield and the number of leukaphereses required to obtain the adequate cell product, but preserving donors from adverse events exposure [28,29]. However, starting the collection on the 5th day may also be appropriate, as shown both in clinical experience and experimental models [30,31].
The dosing schedule of G-CSF was evaluated between a single or a split daily dose to achieve a better CD34+ yield through a single leukapheresis. The split dose was assumed to be more tolerable in terms of adverse events. Although there is no clear evidence of a better result with the two daily administrations, in some cases, it proved its efficacy or at least a non-inferiority compared to the single dose [32,33,34,35].
Plerixafor, a CXCL12-CXCR4 antagonist, was evaluated in combination with G-CSF and alone for mobilization of hematopoietic progenitor cells (HPCs) in healthy donors. While several studies demonstrated the efficacy of the combination in achieving target CD34+ cell yields, concerns have emerged regarding the impact of plerixafor on graft composition. Specifically, concerns exist about potential alterations in immune reconstitution and associated immune processes following allogeneic hematopoietic cell transplantation (allo-HCT) due to changes in the mobilized cell population [36,37,38]. Furthermore, plerixafor alone seems to be unable to induce a successful mobilization in a significant proportion of donors [39], but a randomized trial showed that a higher dose (480 µg/kg) may ensure an adequate collection [40]. Plerixafor showed its efficacy in preventing mobilization failure in healthy, poor mobilizer donors without inducing adverse events [41,42]. Using this evidence, the Italian scientific organization GITMO introduced a specific procedure to include Plerixafor for the mobilization of healthy donors at high risk of failure [43].
Moreover, the G-CSF and Plerixafor combination applied as a frontline mobilization strategy appeared feasible and successful in healthy donor mobilization [44]; however, mainly due to the high rate of satisfactory grafts obtained by the G-CSF alone, this strategy cannot be pursued.
Events of unsuccessful mobilization in healthy donors are anecdotal, despite that, in these cases, especially if an alternative donor is unavailable, stem cell collection through a BM harvest could be considered.

1.3. Adverse Events and Long-Term Complications

During G-CSF stimulation, the main symptoms reported by donors are bone pain, headache, and flu-like symptoms. Bone pain and apheresis side effects are generally more frequently reported by female donors and after the first day of collection [45,46,47,48]. Despite rare cases of malignancy diagnosed during post-donation follow-up, two large prospective studies found no association between these malignancies and G-CSF stimulation, supporting the safety of G-CSF administration in healthy stem cell donors [48,49].

1.4. Autologous PBSCS Mobilization Strategies

There are two general approaches for autologous PBSCs mobilization: steady-state mobilization using growth factors such as granulocyte colony-stimulating factor (G-CSF) alone ± CXCR4 antagonist, and chemotherapy mobilization using chemotherapy either as a part of or apart from these disease-specific treatment protocols followed by G-CSF. These strategies differ in stem cell yields, safety considerations, resource utilization, and levels of contamination of the apheresis product with tumor cells.
Nowadays, the optimal strategy is still a matter of debate; protocols vary according to the center policy, and no definitive conclusions have emerged in recent years [50,51].

1.5. Chemotherapy Mobilization

The choice of a chemotherapy-based mobilization regimen depends on the disease entity and institutional guidelines.
In contrast with malignant lymphoma, where chemotherapy-based stem cell mobilization plays a significant role in disease control, the impact of chemotherapy-based stem cell mobilization seems to be negligible in MM.
In the treatment of non-Hodgkin lymphoma (NHL), autologous stem cell transplantation (ASCT) plays a dual role: as a consolidative therapy for newly diagnosed high-risk patients and as a salvage therapy for relapsed or refractory disease. Additionally, it is employed in Hodgkin lymphoma (HL) for specific situations [52,53].
An ideal salvage regimen should provide sufficient disease control with acceptable hematologic and nonhematologic toxicity, and it should not negatively affect the mobilization of peripheral blood stem cells (PBSC). The mobilization and collection of adequate CD34+ PBSCs is crucial for supporting ASCT, of which G-CSF plus chemotherapy with different schemes and, in some selected cases, with the only use of G-CSF, have been the most common mobilization methods [54,55]. For patients with NHL, the infused dose of hematopoietic SCs has an important impact on engraftment kinetics; a higher mobilization target instead of MM, with an optimal dose of ≥5.0 × 106/kg CD34+ HSCs, could improve engraftment and reduce complications, respiratory and infective [56,57].
For patients with MM in Europe and Western countries, a cyclophosphamide (CTX)-based mobilization strategy is widely used. Different dose levels are employed, from low dose (CTX 1.5–2 g/m2) to intermediate high dose (CTX 3–4 g/m2).
Adequate hematopoietic progenitor cell (HPC) collection is necessary to proceed to transplantation. The target for CD34+ cell collection for a single ASCT has generally been accepted to be 3 to 6 × 106 CD34+ cells/kg. Also, a dose < 2 × 106 CD34+ cells/kg can have a deleterious effect on engraftment [58].
Following myelosuppressive chemotherapy, granulocyte colony-stimulating factor (G-CSF) is administered at 5–10 micrograms per kilogram of body weight (μg/kg bw) per day. Therapy typically begins 1 to 7 days after initiating chemotherapy and continues until the last day of apheresis. A new open question is about the use of novel agents such as lenalidomide (R) and anti-CD38 immunotherapy (daratumumab) during the induction phase that may impact stem-cells collections. In a recent retrospective study of 325 patients with MM who received either VTD (velcade, thalidomide, and dexamethasone) or VRD induction before ASCT, in comparison with VTD, VRD induction was associated with more frequent use of plerixafor (19.3% versus 5.4%, p = 0.004), which is a CXC chemokine receptor 4 (CXCR4) antagonist that improves the release of stem cells from marrow into peripheral blood [59]. In another study, the analysis of MASTER and GRIFFIN trials showed that the use of daratumumab as induction therapy in MM patients determined a 2-fold increase in the use of plerixafor [60]. In an Italian retrospective study in which patients with MM were treated with new agents (lenalidomide, carfilzomib, and daratumumab), the mobilization strategy with cyclophosphamide plus G-CSF and plerixafor “on demand” resulted in high success rate (95%) of autologous stem cell collections [61]. However, randomized clinical studies investigating standard induction triplets with or without daratumumab in MM patients showed higher use of plerixafor and lower stem cell yields in patients receiving daratumumab, regardless of the mobilization strategy adopted [62].

1.6. Steady-State Mobilization (Chemotherapy-Free)

A steady-state mobilization with G-CSF is an effective and appealing strategy compared with a chemotherapy-based approach, particularly related to the availability of plerixafor. Retrospective and prospective studies showed the feasibility and efficacy of HSC mobilization with G-CSF-only plus ‘on-demand’ plerixafor in MM patients receiving 3–4 drugs as induction regimens [63].
Some studies already cited the proportion of patients with MM who successfully collected the goal target of stem cells needed to proceed to ASCT with only the use of G-CSF plus plerixafor. It was 95% in the study group of Mina et al. and 94% and 100% in the GRIFFIN and MASTER trials [60,61].
However, the median stem cells obtained with G-CSF in these trials and studies were lower than in the groups mobilized with chemotherapies and G-CSF; therefore, the steady-state approach should not be indicated in patients at high risk that may benefit from tandem ASCT or salvage transplant or for those who are at high risk of mobilization failure due to the presence of multiple risk factors such as bone marrow infiltration > 60% of plasma cells at diagnosis, the occurrence of grade 3–4 hematologic toxicities during induction, and lenalidomide/daratumumab-based induction regimens.
In a study conducted in 118 European patients with hematological malignancies (90 with MM, 25 with NHL, 3 with HL), the combination of plerixafor + GCS-F was used to mobilize hematopoietic stem cells; the results showed the minimum cell yield (≥2 × 106 CD34+ cells/kg) was harvested in 98% of patients with MM and in 80% of those with lymphoma in a median of one apheresis [64]. In another Asiatic study focused on 43 lymphoma patients, an acquisition success rate of frontline steady-state mobilization was 100%. The number of CD34+ cells in peripheral blood on the day before collection was a predictable index for the evaluation of stem cell collection [65].
Recently, a retrospective analysis conducted at a single center involving 101 individuals newly diagnosed with multiple myeloma (MM) was published. The patients underwent mobilization after treatment with lenalidomide-bortezomib-dexamethasone (RVD) and daratumumab-RVD (DRVD), with the administration of pegylated granulocyte colony-stimulating factor (G-CSF) on day 3, along with plerixafor on day 1, as a preemptive mobilization strategy. In the DRVd and RVd groups, the median number of collected CD34+ cells was 6.54 × 106/kg and 6.78 × 106/kg, respectively. Both groups achieved the target CD34+ stem cell collection within a median of 1 day (range: 1–4 days). However, more patients in the DRVd group required additional interventions compared to the RVd group: 51% vs. 43% needed extra plerixafor doses, and 19% vs. 14% required additional G-CSF. Notably, no mobilization failures or severe (grade 3+) mobilization-related adverse events were reported in either group. Importantly, incorporating daratumumab into the RVd induction regimen did not significantly impact stem cell yield or collection time, provided patients received preemptive G-CSF and plerixafor administration [66]. Therefore, chemotherapy-free mobilization could be an option in some cases of MM and lymphoma patients.

1.7. Poor Mobilizer

Although the majority of patients are able to mobilize a sufficient quantity of CD34+ cells to collect a sufficient transplant dose with one or two collection procedures, approximately 15% fail to reach this target; these are the so-called poor mobilizers (PM) [67]. In 2012, Olivieri et al. proposed the definition of two groups of poor mobilizers: proven PM, in cases of peripheral blood CD34+ cell count never exceeding 20 cells/microliter or in cases of total collection less than 2 × 106/kg in 3 procedures and predicted PM, in the presence of a previous failed mobilization, previous radiotherapy to hematopoietic sites, or previous aplastic chemotherapy. A patient who meets two of the following criteria is also a predicted PM: the presence of advanced or refractory disease, marrow involvement, severe reduction in marrow cellularity, and age over 65 years [68].
These two categories of patients may benefit from the use of CXCR4 antagonists.

1.8. Old and New Generation of CXCR4 Antagonists

Different strategies concerning the use of plerixafor for stem cell mobilization have been adopted by different institutions, from its ‘on-demand’ or ‘just-in-time’ use (plerixafor administered according to a risk-adapted strategy based on either the number of PB CD34+ cells before the apheresis or the first CD34+ stem cell yield) to a ‘pre-emptive’ strategy in patients at high risk of stem cell mobilization failure [69]. Actually, considering the increasingly frequent use of novel agents and the diffusion of chemotherapy-free mobilization, the use of CXCR4 antagonists is in constant development; for these reasons, there are some new molecules being studied. GCP-100 is a novel CXCR4 antagonist that, in a recent in vivo study in mice, mobilized more white blood cells into peripheral blood compared to plerixafor. GPC-100-induced mobilization was further amplified by propranolol pretreatment and was comparable to mobilization by G-CSF [70]. An open-label, phase II pilot trial assessed the safety and stem cell mobilization efficacy of burixafor (GCP-100) combined with G-CSF in patients with MM, NHL, and HL. Among the nine treated patients, six (66.7%) successfully mobilized over 10 × 106 CD34+ cells/kg in a single leukapheresis session. Two patients required two leukapheresis days, while one patient who had recently received lenalidomide failed mobilization initially but succeeded after a two-week recovery period. This is significantly faster than historical control patients, who typically required a median of 2–3 days of leukapheresis sessions for adequate mobilization [71]. Based on these results, an ongoing multicenter trial aimed to assess the safety and efficacy of burixafor (GPC-100) and propranolol with and without G-CSF for the mobilization of stem cells in patients with MM undergoing ASCT is now recruiting. Another molecule being studied is YF-H-2015005, a novel CXCR4 antagonist proven to increase the quantities of circulating hematopoietic stem cells in NHL. In a recent multicenter, phase III clinical trial, 101 NHL patients were randomized to receive G-CSF plus YF-H-2015005 or placebo. The primary endpoint was the proportion of NHL patients procuring ≥ 5 × 106/kg CD34+ HSCs within ≤4 apheresis sessions. The proportions of patients achieving the primary endpoint were 57% and 12% in the YF-H-2015005 and placebo groups, respectively (p < 0.001) [72]. The GENESIS trial, a large-scale study, compared motixafortide + G-CSF to placebo + G-CSF for stem cell mobilization in multiple myeloma patients undergoing transplantation. Motixafortide significantly increased the success rate of collecting sufficient stem cells (≥6 × 106 CD34+ cells/kg) within two procedures, compared to placebo (92.5% vs. 26.2%). Notably, many patients in the motixafortide group even achieved this goal in just one procedure (88.8% vs. 9.5%). Motixafortide was safe and well-tolerated, with mostly mild and temporary injection site reactions [73]. Finally, considering the more frequent use of novel agents and antibodies in MM and lymphoma, now and in the future, it is necessary to improve the efficiency of stem cell mobilization, reducing days of leukapheresis and targeting CD34+ collected with cost-effective agents.
The GENESIS study population reported encouraging results but excluded patients with risk factors for poor mobilization. It remains to be seen whether G-CSF + motixafortide will establish itself as the new standard for mobilization in multiple myeloma or if motixafortide might replace plerixafor in routine practice for patients with poor mobilization. The data for other agents is too preliminary, and some were collected in non-oncological settings, so it is still early to understand their potential positioning in clinical practice.
Future head-to-head studies will be necessary to determine the true superiority of motixafortide over plerixafor in poorly mobilized patients.

1.9. Current Technologies in Stem Cell Collection

Apheresis involves three fundamental steps: collection of whole blood with the addition of an anticoagulant and subsequent separation of blood components; removal of the desired component; return of the remaining components to the donor/patient with or without replacement fluids [74]. The techniques employed by apheresis instrumentation must enable these steps to be performed efficiently, ensuring high product purity and safety for the donor/patient and the operator. What are the current technologies in use for the collection of PBSCs?
The selection of specific cellular components for clinical transplantation via apheresis procedures has been extensively investigated over several years, aiming to establish product standardization and universally accepted quality control measures. The intrinsic variability of patients or donors, operator skill, organizational quality management systems, and interface with the processing facility all significantly impact successful collection [50].
In healthy individuals, the ratio of red blood cells (RBCs) to white blood cells (WBCs) is approximately 1000:1, and the ratio of platelets to WBCs is 30–60:1. As is known, PBSCs constitute only a small fraction of WBCs, typically ranging from 0.1% to 0.5% [1]. Despite mobilization strategies that elevate PBSC numbers, their isolation remains a technical challenge due to contamination from more abundant blood cells. Furthermore, the subtle differences in density between various blood cell components, including leukocyte subpopulations, coupled with the inherent heterogeneity of leukocytes, add to the complexity of the process. This poses a significant challenge for mobilization and PBSC collection, as it requires isolating a high concentration of PBSCs while minimizing contamination with other blood cells. To achieve this, all cell separators employ centrifugal force to fractionate whole blood into various components, including red blood cells, granulocytes, mononuclear cells, platelets, and plasma, based on their specific gravities. Each component can then be selectively collected using a mechanical pump. Cellular components are arranged in a stratified manner, from highest to lowest specific gravity, with PBSCs residing in the mononuclear cell layer, as shown in Figure 1. Two critical factors govern this mechanism: G-force, determined by the rotor radius, and dwell time, which represents the duration of exposure to the centrifugal field [74].
Some MNC collection procedures, after initial separation using centrifugal force (specific gravity), employ elutriation, a technique that utilizes two opposing forces—centrifugation and pump flow—to separate blood components based on size [74,75].
Cell separators employ two different flow systems to separate blood components: intermittent and continuous flow, see Table 1. The intermittent flow system extracts the desired component in cycles, emptying the separation chamber and transferring blood between the chamber and reservoir before proceeding. This cyclical approach lengthens the procedure due to repeated transfers. On the other hand, the continuous flow system extracts the desired component without interruption, maintaining a continuous blood flow. This streamlined process results in shorter procedure times and a lower extracorporeal volume compared to intermittent flow systems [74]. Extracorporeal volume (ECV) considers the total blood volume that is outside of the body, including any volume within the extracorporeal circuit. It is crucial to understand the ECV associated with cell separators and procedure protocols to determine how much blood can safely be removed from the body for instrument priming. Typically, ECV is kept below 15% of the patient’s total blood volume to minimize the risk of adverse events. This is especially important in the pediatric setting, as children have smaller blood volumes than adults [74].
Potential technical issues that may negatively impact PBSC collection include platelet loss, granulocyte content, and red blood cell content.
Although platelets and MNCs have similar specific gravities, some apheresis devices allow for the adjustment of the platelet content in the product, potentially reducing platelet loss for the patient or donor [76,77].
Another issue with PBSC collection is the presence of excessive granulocyte and RBC content.
High circulating WBC counts can adversely affect PBSC collection. Therefore, in this case, inlet flow rates should be limited to increase centrifuge dwell time to allow for adequate separation and subsequent PBSC collection [50].
Excessive granulocyte and RBC content typically arise from harvesting too deeply into the RBC layer. In some cases, such as low red cell MCV, it may be necessary to go deeper into the RBC layer to obtain PBSCs because low MCV is associated with poor PBSC yields, but the collection will also contain an increased number of RBC and granulocytes [78,79,80]. As suggested by Trébéden-Negre et al., an excess of granulocyte content in the collected product has been associated with delayed engraftment. They hypothesized that this detrimental effect is caused by the impairment of stem cell homing due to the pro-inflammatory cytokines and metalloproteases generated by granulocytes [81], while RBCs are susceptible to hemolysis during the freezing process.
Therefore, it is crucial to carefully monitor the PBSC collection process and adjust the collection parameters as needed to minimize the granulocyte and RBC content in the collected product.
Each cell separator and protocol collect PBSCs in a slightly different manner, resulting in differences in product content and in the way it is influenced by patient and donor characteristics. These differences can be utilized to tailor the collection to specific patient characteristics [50].

1.10. Timing and Tools for Stem Cell Collection Prediction

Effective management of stem cell collection timing is crucial for optimizing procedure scheduling within the collection facility, promoting seamless collaboration among all entities involved in a transplant program (clinical unit, collection, and processing facilities). What constitutes optimal timing for stem cell collection, and what tools are available to determine the ideal time to initiate a stem cell apheresis procedure?
The optimal timing of hematopoietic stem cell collection is a complex decision that requires careful consideration of multiple factors, including the expertise and experience of the collection team, the patient’s unique characteristics, and the efficiency of the collection system. Considering the accreditation standards, such as JACIE-FACT, this decision is increasingly guided by a collaborative approach between clinical units and collection facilities.
In the autologous setting with a CHT-GCSF mobilization, it is well-established that there is extreme variability among patients in reaching the peak concentration of CD34+ cells post-mobilization. All the factors that cause negative changes in the integrity of the perivascular bone marrow niches where hematopoietic stem cells reside or that always interfere negatively with chemotaxis have been extensively studied [82,83,84,85]. These factors include age > 70 years, bone marrow involvement, previous irradiation, infections, and, as recently reported, the use of daratumumab [62]. Despite this, it is common that more than 90% of patients reach the peak of hematopoietic stem cells between the 10th and 20th day of mobilization, regardless of the diagnosis and previous cycles of chemotherapy or radiotherapy [86]. However, it is also a well-established opinion that there is a huge individual difference in reaching this, for reasons that are not yet fully understood, probably of genetic origin [87,88]. In order to accurately determine the optimal timing for initiating stem cell collection, a combination of benchmarks, including best practices or consensus guidelines, should be considered [54,89]. These guidelines may recommend targeting a CD34 cell count of 20 cells/µL in mobilized peripheral blood as the starting threshold. Alternatively, time-based targets, such as initiating collection on day 11 from the start of mobilization with endonxan and GCSF, may also be employed [90]. When we introduce plerixafor into the mobilization cycle as a rescue therapy for patients who mobilize poorly after G-CSF or prophylactically when the risk of poor mobilization is high [91,92], we must ensure that the CD34 peak coincides with the collection. This introduces another variable, the timing of plerixafor infusion, which is also subject to individual variability and a highly variable growth kinetics from infusion to collection. As reported by colleagues at Mount Sinai, apheresis of 2–3 TBV (performed over 3–4 h) should be initiated 15 h after plerixafor infusion for good mobilizers and 8 h for poor mobilizers, respectively. [93]. Alternatively, as suggested by the Californian Duarte Center, which has a substantial transplant volume, apheresis should be initiated 11 h after the administration of plerixafor [94].
In the allogeneic setting with growth factor-only mobilization, one aspect to consider is the timing of stem cell collection—specifically, whether to initiate it on day 4 or day 5 following the initiation of mobilization therapy.
Most international organizations and registries, including the National Marrow Donor Program (NMDP), recommend day 5 for collection initiation. However, recent scientific evidence suggests that there is no significant difference in terms of the collected dose or purity of the product between day 4 and day 5 collection [29,95,96]. Moreover, donors who collect on day 4 do not appear to have an increased risk of requiring a second collection procedure compared to those who start on day 5 [97]. Finally, in the case of a need for high doses of CD34+ cells, such as in the case of haploidentical transplantation or a significant weight difference between the recipient and the donor, or if the donor is found to be a poor mobilizer, conducting the first apheresis on the 4th day would allow for immediate rescue therapy with plerixafor (between the 4th and 5th day and not between the 5th and 6th day, as reported by current studies and guidelines [29,89], thereby increasing the likelihood of achieving the target cell dose.
All these variables suggest that it is necessary to develop additional tools to predict the optimal time for initiating stem cell collection to ensure the target dose for successful transplantation. Humpe et al. [98] investigated the decline in stem cells in the peripheral blood and the subsequent increase in the collection bag through prospective studies of kinetics between the four compartments (bone marrow, peripheral blood, cell separator, and collection bag) that make up a collection system. These findings laid the groundwork for the development of prediction algorithms. The balance between these four compartments ensures accurate prediction. The parameters that represent these four compartments and form the basis for most prediction models are identified in the hematologic and physical characteristics of the donor, the donor’s blood volume, the processed blood volume, the duration of the collection procedure, and the number of CD34+ cells present before collection.
Several prediction algorithms have been proposed; here, we present a few examples. Delamain et al. determined the optimal timing for initiating apheresis procedures by identifying the day of the CD34 peak, calculated using the following parameters: hemoglobin concentration on the day mobilization therapy commences and the day when the CD34+ value in the peripheral blood reaches 10 cells/µL. This model proved highly effective for individuals with low mobilization potential, significantly reducing the frequency of apheresis procedures by delaying their initiation [99]. Pierelli et al. published an algorithm based on a multicenter prospective study [100]. The model employs simple parameters, some of which are known prior to collection, such as the donor’s weight or the CD34+ cell count in the peripheral blood prior to collection and the transplant dose required. Other parameters are predicted, such as the blood volume to be processed. The algorithm relies on the efficiency of the apheresis procedure. This is a crucial aspect, as no model can function effectively without knowledge of the collection system’s efficiency. The high correlation between the predicted and actual data confirms the algorithm’s accuracy.
The application of predictive algorithms provides a valuable tool to aid decision-making in various aspects of stem cell collection, including the timing of collection, procedure scheduling, processing requirements, the number of required procedures, procedure duration, and minimizing the storage of surplus product.

1.11. Management Strategies for Intra- and Peri-Procedural Adverse Events in Stem Cell Collection

Collection Units prioritize patient and donor safety. What are the key principles and management strategies for intra- and peri-procedural adverse events (AEs)?
Apheresis procedures are generally well-tolerated for both therapeutic and collection purposes. However, the potential for adverse events, expected and unexpected, exists. These side effects are categorized by severity as mild (tolerated without medications), moderate (need for medication), severe (interruption due to the AE), or death (due to AE).
The 2023 update of the World Apheresis Association (WAA) apheresis registry analyzed data from 58,355 procedures performed on 9500 patients, examining both therapeutic and collection purposes. During the period 2018–2022, 3% of procedures reported adverse events (AEs), which included technical difficulties and problems with vascular access. Among these AEs, the most frequent were mild (1.5%) and moderate (1.4%). Tingling, pricking sensations, hives (urticaria), and low blood pressure (hypotension) were the main types of mild and moderate AEs. Severe AEs were very rare, occurring in only 0.15% of procedures. Excluding access and technical issues, the update reported the following breakdown by severity. Mild AEs were 1.5% (hypotension 13.2%, tingling/pricking 38.2%, nausea/vomiting 10.3%. Moderate AEs were 1.4% (tingling/pricking 74.3%, urticaria 8.4%, hypotension 5.2%, nausea 3.9%. Severe AEs were 0.1% (syncope/hypotension 25.4%, urticaria 25.4%, arrhythmia/asystole 4.8%, nausea/vomiting 3.2%, chills/fever 1.6%) [101].
Considering the above, PBSC collection by apheresis can be performed safely in an outpatient setting.
One of the most frequent PBSC adverse events is citrate-related hypocalcemia.
The preferred anticoagulant for PBSC is a citrate solution usually containing sodium citrate dihydrate 22 g/L, glucose monohydrate 24.5 g/L, and citrate acid monohydrate 8 g/L, namely, ACD-A—Anticoagulant Citrate Dextrose Solution A [102].
Citrate exerts its action through the chelation of divalent cations, primarily calcium and magnesium. This chelation, occurring during apheresis due to citrate infusion, can lead to reductions in serum concentrations of ionized calcium and magnesium. Ionized calcium levels can decrease by 25% or more during an apheresis procedure [50]. Mild reactions to citrate chelation during apheresis may manifest as metallic taste, perioral, and/or acral paresthesia. Moderate reactions are characterized by the persistence of these symptoms despite supportive measures, such as reduced blood flow rate, increased anticoagulant-to-whole blood ratio, or calcium supplementation. Additionally, nausea, vomiting, abdominal pain, shivering, lightheadedness, tremors, and hypotension reminiscent of hypovolemia or vasovagal reactions may occur. In severe cases, symptoms can escalate to carpopedal spasm, tetanic seizures, and cardiac arrhythmias, particularly QT prolongation. However, hypocalcemia is usually mild and easily managed with oral or intravenous calcium supplements.
Citrate-based anticoagulation also reduces plasma potassium and free magnesium levels. Patients undergoing PBSC collection should have an electrolyte panel drawn within 24–74 h of the procedure, and low serum potassium and magnesium levels should be replaced pre-procedure with oral or IV electrolyte supplements [74].
During apheresis procedures, regardless of the device or collection program employed, careful consideration must be given to the extracorporeal blood volume (ECV) in low-weight patients to mitigate potential hemodynamic alterations. These alterations can manifest as hypotension or vasovagal reactions.
Two distinct mechanisms can lead to hypotension, requiring different management approaches.
Hypotension due to intravascular volume depletion typically presents accompanied by tachycardia and tachypnea. To address this, the apheresis procedure is usually paused, and the patient receives a fluid bolus followed by evaluation by a physician.
Hypotension due to vasovagal reaction exhibits concurrent bradycardia, differentiating it from hypotension due to hypovolemia. This reflex response of the parasympathetic nervous system leads to vasodilation, causing significant hypotension (blood pressure as low as 50/20 mmHg) without the reflex tachycardia typically seen in hypovolemia. This is a key distinction between the two conditions.
The overactive parasympathetic response can be triggered by anxiety, pain, or hypocalcemia during the procedure. Management involves stopping the apheresis and implementing supportive measures to improve patient comfort and hemodynamics.
Beyond vital sign changes, vasovagal reactions can manifest with pallor, diaphoresis, nausea and vomiting, syncope, potential convulsions, and urinary or fecal incontinence.
In some cases, this presentation can closely resemble epileptic seizures, highlighting the importance of accurate diagnosis.
With good surveillance, it is often possible to prevent a vasovagal reaction with distraction techniques. Engaging the patient in conversation or other distracting activities often results in a return to the baseline state [74]. Upon the onset of a vasovagal reaction during apheresis, initial management focuses on alleviating patient distress and promoting hemodynamic stability. This often involves implementing non-pharmacological interventions such as deep breathing exercises, coughing, laughing, or repositioning the patient to reduce discomfort and enhance comfort. These measures can effectively resolve mild vasovagal reactions.
However, in more severe cases, escalation of therapy may be necessary. This may involve procedural intervention, including a temporary pause in donors or patients. Additionally, Trendelenburg positioning, which elevates the lower extremities to facilitate venous return and improve blood flow to the heart, can be implemented. Furthermore, fluid resuscitation through the administration of a fluid bolus can help restore intravascular volume and blood pressure.
Additional adverse reactions are associated with the type of venous access used for apheresis procedures.
Peripheral veins are the preferred choice for cell collection. However, between 0.6% and 20% of donors, depending on the apheresis center, may not have suitable peripheral veins to achieve the necessary blood flow for collecting PBSCs [49,103,104,105,106]. For autologous patients, if venous access is deemed questionable, a CVC is the predominant type of vascular access used to harvest HSCs. Current policies and practices regarding the assessment, placement, and management of venous access for HPC collections for healthy donors are based on institutional preferences and/or National Health Service-mandated policies.
A number of these regulatory agencies recommend the use of peripheral venous access for healthy donors, especially unrelated donors.
If peripheral venous access is unavailable, the use of a CVC should be considered based on appropriate clinical indication and it shall be placed by qualified personnel.
However, CVCs should be a last resort for healthy stem cell donors. Peripheral access should always be prioritized whenever possible to ensure donor safety. Bone marrow harvest can be considered as an alternative option. [107,108].
Another option is the use of midlines, which have been shown to be effective in collecting HSCs in adult donors who do not have suitable peripheral vascular access [43,109,110].
It is important to note that both peripheral and central venous access methods carry a potential risk of AEs.
Peripheral access may be associated with bruising, hematomas, nerve injuries, infections, phlebitis, and/or deep vein thrombosis. CVCs can be associated with several potential complications, including infection, thrombosis, hemorrhage, air embolism, pneumothorax, hemothorax, and arrhythmias.

2. Conclusions

Continuous optimization of stem cell mobilization and collection protocols can lead to a substantial improvement in this therapeutic approach, particularly for allogeneic stem cell transplantation. Increased efficiency, streamlined patient scheduling and processing, and prioritized patient comfort will not only enhance graft quality and ensure transplant success but also facilitate broader patient access and improve quality of life.
In addition to the described scenarios concerning stem cell collection, novel challenges are emerging with respect to the collection of cellular effectors, such as CAR T cells and other immunotherapeutic agents. These challenges are related to the production of biological drugs and gene editing, further emphasizing the critical importance of product quality, including high purity, viability, and specific functionalities. Nevertheless, these new scenarios further solidify the leading role of the collection unit in the field of precision medicine.

Author Contributions

M.P., E.S., V.T., M.A.T., C.B., G.P., C.N., A.M., M.M., O.A., L.R. and M.V. wrote and prepared the draft manuscript; M.P., O.A., L.R. and M.V. reviewed and edited the final version; L.R., supervision; M.V., supervision and validation. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Moog, R. Management strategies for poor peripheral blood stem cell mobilization. Transfus. Apher. Sci. 2008, 38, 229–236. [Google Scholar] [CrossRef] [PubMed]
  2. Snowden, J.A.; Sánchez-Ortega, I.; Corbacioglu, S.; Basak, G.W.; Chabannon, C.; de la Camara, R.; Dolstra, H.; Duarte, R.F.; Glass, B.; Greco, R.; et al. Indications for haematopoietic cell transplantation for haematological diseases, solid tumours and immune disorders: Current practice in Europe, 2022. Bone Marrow Transplant. 2022, 57, 1217–1239. [Google Scholar] [CrossRef]
  3. Carreras, E.; Dufour, C.; Mohty, M.; Kröger, N. The EBMT Handbook: Hematopoietic Stem Cell Transplantation and Cellular Therapies; Springer Nature: Cham, Switzerland, 2019. [Google Scholar]
  4. Russell, N.H.; Hunter, A.; Rogers, S.; Hanley, J.; Anderson, D. Peripheral blood stem cells as an alternative to marrow for allogeneic transplantation. Lancet 1993, 341, 1482. [Google Scholar] [CrossRef]
  5. Passweg, J.R.; Baldomero, H.; Chabannon, C.; Basak, G.W.; de la Cámara, R.; Corbacioglu, S.; Dolstra, H.; Duarte, R.; Glass, B.; Greco, R.; et al. Hematopoietic cell transplantation and cellular therapy survey of the EBMT: Monitoring of activities and trends over 30 years. Bone Marrow Transplant. 2021, 56, 1651–1664. [Google Scholar] [CrossRef]
  6. Gluckman, E.; Devergié, A.; Bourdeau-Esperou, H.; Thierry, D.; Traineau, R.; Auerbach, A.; Broxmeyer, H.E. Transplantation of umbilical cord blood in Fanconi’s anemia. Nouv. Rev. Fr. Hematol. 1990, 32, 423–425. [Google Scholar]
  7. Laporte, J.P.; Gorin, N.C.; Rubinstein, P.; Lesage, S.; Portnoi, M.F.; Barbu, V.; Lopez, M.; Douay, L.; Najman, A. Cord-blood transplantation from an unrelated donor in an adult with chronic myelogenous leukemia. N. Engl. J. Med. 1996, 335, 167–170. [Google Scholar] [CrossRef] [PubMed]
  8. Sauter, C.; Barker, J.N. Unrelated donor umbilical cord blood transplantation for the treatment of hematologic malignancies. Curr. Opin. Hematol. 2008, 15, 568–575. [Google Scholar] [CrossRef]
  9. Kessinger, A.; Smith, D.M.; Strandjord, S.E.; Landmark, J.D.; Dooley, D.C.; Law, P.; Coccia, P.F.; Warkentin, P.I.; Weisenburger, D.D.; Armitage, J.O. Allogeneic transplantation of blood-derived, T cell-depleted hemopoietic stem cells after myeloablative treatment in a patient with acute lymphoblastic leukemia. Bone Marrow Transplant. 1989, 4, 643–646. [Google Scholar]
  10. Dreger, P.; Suttorp, M.; Haferlach, T.; Löffler, H.; Schmitz, N.; Schroyens, W. Allogeneic granulocyte colony-stimulating factor-mobilized peripheral blood progenitor cells for treatment of engraftment failure after bone marrow transplantation. Blood 1993, 81, 1404–1407. [Google Scholar] [CrossRef]
  11. Cashen, A.F.; Lazarus, H.M.; Devine, S.M. Mobilizing stem cells from normal donors: Is it possible to improve upon G-CSF? Bone Marrow Transplant. 2007, 39, 577–588. [Google Scholar] [CrossRef]
  12. Cottler-Fox, M.H.; Lapidotm, T.; Petitm, I.; Kollet, O.; DiPersio, J.F.; Link, D.; Devine, S. Stem cell mobilization. Am. Soc. Hematol. Educ. Program 2003, 419–437. [Google Scholar] [CrossRef]
  13. Winkler, I.G.; Lévesque, J.P. Mechanisms of hematopoietic stem cell mobilization: When innate immunity assails the cells that make blood and bone. Exp. Hematol. 2006, 34, 996–1009. [Google Scholar] [CrossRef] [PubMed]
  14. Kollet, O.; Dar, A.; Shivtiel, S.; Kalinkovich, A.; Lapid, K.; Sztainberg, Y.; Tesio, M.; Samstein, R.M.; Goichberg, P.; Spiegel, A.; et al. Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat. Med. 2006, 12, 657–664. [Google Scholar] [CrossRef] [PubMed]
  15. Melve, G.K.; Ersvaer, E.; Eide, G.E.; Kristoffersen, E.K.; Bruserud, Ø. Peripheral Blood Stem Cell Mobilization in Healthy Donors by Granulocyte Colony-Stimulating Factor Causes Preferential Mobilization of Lymphocyte Subsets. Front. Immunol. 2018, 9, 845. [Google Scholar] [CrossRef]
  16. Matsunaga, T.; Sakamaki, S.; Kohgo, Y.; Ohi, S.; Hirayama, Y.; Niitsu, Y. Recombinant human granulocyte colony-stimulating factor can mobilize sufficient amounts of peripheral blood stem cells in healthy volunteers for allogeneic transplantation. Bone Marrow Transplant. 1993, 11, 103–108. [Google Scholar]
  17. Schmitz, N.; Dreger, P.; Suttorp, M.; Rohwedder, E.B.; Haferlach, T.; Löffler, H.; Hunter, A.; Russell, N.H. Primary transplantation of allogeneic peripheral blood progenitor cells mobilized by filgrastim (granulocyte colony-stimulating factor). Blood 1995, 85, 1666–1672. [Google Scholar] [CrossRef]
  18. Russell, J.A.; Luider, J.; Weaver, M.; Brown, C.; Selinger, S.; Railton, C.; Karlsson, L.; Klassen, J. Collection of progenitor cells for allogeneic transplantation from peripheral blood of normal donors. Bone Marrow Transplant. 1995, 15, 111–115. [Google Scholar]
  19. Körbling, M.; Przepiorka, D.; Huh, Y.O.; Engel, H.; van Besien, K.; Giralt, S.; Andersson, B.; Kleine, H.D.; Seong, D.; Deisseroth, A.B.; et al. Allogeneic blood stem cell transplantation for refractory leukemia and lymphoma: Potential advantage of blood over marrow allografts. Blood 1995, 85, 1659–1665. [Google Scholar] [CrossRef]
  20. Bensinger, W.I.; Weaver, C.H.; Appelbaum, F.R.; Rowley, S.; Demirer, T.; Sanders, J.; Storb, R.; Buckner, C.D. Transplantation of allogeneic peripheral blood stem cells mobilized by recombinant human granulocyte colony-stimulating factor. Blood 1995, 85, 1655–1658. [Google Scholar] [CrossRef] [PubMed]
  21. Dreger, P.; Haferlach, T.; Eckstein, V.; Jacobs, S.; Suttorp, M.; Löffler, H.; Müller-Ruchholtz, W.; Schmitz, N. G-CSF-mobilized peripheral blood progenitor cells for allogeneic transplantation: Safety, kinetics of mobilization, and composition of the graft. Br. J. Haematol. 1994, 87, 609–613. [Google Scholar] [CrossRef]
  22. Russel, N.; Gratwohl, A.; Schmitz, N. The place of blood stem cells in allogeneic transplantation. Br. J. Haematol. 1996, 93, 747–753. [Google Scholar] [CrossRef] [PubMed]
  23. Miflin, G.; Charley, C.; Stainer, C.; Anderson, S.; Hunter, A.; Russell, N. Stem cell mobilization in normal donors for allogeneic transplantation: Analysis of safety and factors affecting efficacy. Br. J. Haematol. 1996, 95, 345–348. [Google Scholar] [CrossRef] [PubMed]
  24. Kröger, N.; Renges, H.; Sonnenberg, S.; Krüger, W.; Gutensohn, K.; Dielschneider, T.; Cortes-Dericks, L.; Zander, A.R. Stem cell mobilisation with 16 microg/kg vs 10 microg/kg of G-CSF for allogeneic transplantation in healthy donors. Bone Marrow Transplant. 2002, 29, 727–730. [Google Scholar] [CrossRef] [PubMed]
  25. Majolino, I.; Scimé, R.; Vasta, S.; Cavallaro, A.M.; Fiandaca, T.; Indovina, A.; Catania, P.; Santoro, A. Mobilization and collection of PBSC in healthy donors: Comparison between two schemes of rhG-CSF administration. Eur. J. Haematol. 1996, 57, 214–221. [Google Scholar] [CrossRef]
  26. Reményi, P.; Gopcsa, L.; Marton, I.; Réti, M.; Mikala, G.; Pető, M.; Barta, A.; Bátai, Á.; Farkas, Z.; Borbényi, Z.; et al. Peripheral blood stem cell mobilization and engraftment after autologous stem cell transplantation with biosimilar rhG-CSF. Adv. Ther. 2014, 31, 451–460. [Google Scholar] [CrossRef] [PubMed]
  27. Sivgin, S.; Karakus, E.; Keklik, M.; Zararsiz, G.; Solmaz, M.; Kaynar, L.; Eser, B.; Cetin, M.; Unal, A. Evaluation of the efficacy and safety of original filgrastim (Neupogen®), biosimilar filgrastim (Leucostim®) and Lenograstim (Granocyte®) in CD34(+) peripheral hematopoietic stem cell mobilization procedures for allogeneic hematopoietic stem cell transplant donors. Transfus. Apher. Sci. 2016, 54, 410–415. [Google Scholar] [CrossRef] [PubMed]
  28. Goren Sahin, D.; Arat, M. Peripheral blood stem cell collection for allogeneic hematopoietic stem cell transplantation: Practical implications after 200 consequent transplants. Transfus. Apher. Sci. 2017, 56, 800–803. [Google Scholar] [CrossRef]
  29. Passeri, C.; Iuliani, O.; Di Ianni, M.; Sorrentino, C.; Giancola, R.; Abbruzzese, L.; Dallavalle, F.M.; Gattillo, S.; Mariano, M.T.; Martino, M.; et al. Comparison between peripheral blood progenitor cell collection on the 4(th) or 5(th) day of granulocyte colony-stimulating factor treatment in allogeneic stem cell donors: Implications for hematopoietic progenitor cell apheresis guidelines. Blood Transfus. 2023, 21, 37–41. [Google Scholar] [CrossRef] [PubMed]
  30. Akizuki, S.; Mizorogi, F.; Inoue, T.; Sudo, K.; Ohnishi, A. Pharmacokinetics and adverse events following 5-day repeated administration of lenograstim, a recombinant human granulocyte colony-stimulating factor, in healthy subjects. Bone Marrow Transplant. 2000, 26, 939–946. [Google Scholar] [CrossRef]
  31. Winkler, I.G.; Wiercinska, E.; Barbier, V.; Nowlan, B.; Bonig, H.; Levesque, J.P. Mobilization of hematopoietic stem cells with highest self-renewal by G-CSF precedes clonogenic cell mobilization peak. Exp. Hematol. 2016, 44, 303–314.e1. [Google Scholar] [CrossRef]
  32. Arbona, C.; Prosper, F.; Benet, I.; Mena, F.; Solano, C.; Garcia-Conde, J. Comparison between once a day vs twice a day G-CSF for mobilization of peripheral blood progenitor cells (PBPC) in normal donors for allogeneic PBPC transplantation. Bone Marrow Transplant. 1998, 22, 39–45. [Google Scholar] [CrossRef] [PubMed]
  33. Kröger, N.; Renges, H.; Krüger, W.; Gutensohn, K.; Löliger, C.; Carrero, I.; Cortes, L.; Zander, A.R. A randomized comparison of once versus twice daily recombinant human granulocyte colony-stimulating factor (filgrastim) for stem cell mobilization in healthy donors for allogeneic transplantation. Br. J. Haematol. 2000, 111, 761–765. [Google Scholar] [PubMed]
  34. Anderlini, P.; Donato, M.; Lauppe, M.J.; Huh, Y.O.; Martin, T.G.; Chan, K.W.; Champlin, R.E.; Körbling, M. A comparative study of once-daily versus twice-daily filgrastim administration for the mobilization and collection of CD34+ peripheral blood progenitor cells in normal donors. Br. J. Haematol. 2000, 109, 770–772. [Google Scholar] [CrossRef] [PubMed]
  35. Yano, T.; Katayama, Y.; Sunami, K.; Deguchi, S.; Nawa, Y.; Hiramatsu, Y.; Nakayama, H.; Arakawa, T.; Ishimaru, F.; Teshima, T.; et al. G-CSF-induced mobilization of peripheral blood stem cells for allografting: Comparative study of daily single versus divided dose of G-CSF. Int. J. Hematol. 1997, 66, 169–178. [Google Scholar] [CrossRef]
  36. Rutella, S.; Filippini, P.; Bertaina, V.; Li Pira, G.; Altomare, L.; Ceccarelli, S.; Brescia, L.P.; Lucarelli, B.; Girolami, E.; Conflitti, G.; et al. Mobilization of healthy donors with plerixafor affects the cellular composition of T-cell receptor (TCR)-αβ/CD19-depleted haploidentical stem cell grafts. J. Transl. Med. 2014, 12, 240. [Google Scholar] [CrossRef]
  37. Kean, L.S.; Sen, S.; Onabajo, O.; Singh, K.; Robertson, J.; Stempora, L.; Bonifacino, A.C.; Metzger, M.E.; Promislow, D.E.; Mattapallil, J.J.; et al. Significant mobilization of both conventional and regulatory T cells with AMD3100. Blood 2011, 118, 6580–6590. [Google Scholar] [CrossRef] [PubMed]
  38. Lundqvist, A.; Smith, A.L.; Takahashi, Y.; Wong, S.; Bahceci, E.; Cook, L.; Ramos, C.; Tawab, A.; McCoy, J.P., Jr.; Read, E.J.; et al. Differences in the phenotype, cytokine gene expression profiles, and in vivo alloreactivity of T cells mobilized with plerixafor compared with G-CSF. J. Immunol. 2013, 191, 6241–6249. [Google Scholar] [CrossRef] [PubMed]
  39. Devine, S.M.; Vij, R.; Rettig, M.; Todt, L.; McGlauchlen, K.; Fisher, N.; Devine, H.; Link, D.C.; Calandra, G.; Bridger, G.; et al. Rapid mobilization of functional donor hematopoietic cells without G-CSF using AMD3100, an antagonist of the CXCR4/SDF-1 interaction. Blood 2008, 112, 990–998. [Google Scholar] [CrossRef]
  40. Pantin, J.; Purev, E.; Tian, X.; Cook, L.; Donohue-Jerussi, T.; Cho, E.; Reger, R.; Hsieh, M.; Khuu, H.; Calandra, G.; et al. Effect of high-dose plerixafor on CD34(+) cell mobilization in healthy stem cell donors: Results of a randomized crossover trial. Haematologica 2017, 102, 600–609. [Google Scholar] [CrossRef]
  41. Hölig, K.; Schmidt, H.; Hütter, G.; Kramer, M.; Teipel, R.; Heidrich, K.; Zimmer, K.; Heidenreich, F.; Blechschmidt, M.; Torosian, T.; et al. Salvage treatment with plerixafor in poor mobilizing allogeneic stem cell donors: Results of a prospective phase II-trial. Bone Marrow Transplant. 2021, 56, 635–645. [Google Scholar] [CrossRef]
  42. Zhuang, L.; Lauro, D.; Wang, S.; Yuan, S. Addition of plerixafor in poorly mobilized allogeneic stem cell donors. J. Clin. Apher. 2022, 37, 388–394. [Google Scholar] [CrossRef]
  43. Ciceri, F.; Botti, S.; Cioce, M. Handbook GITMO; GITMO Nurses Group: Italy, 2023; Volume III, p. 622. [Google Scholar]
  44. Hauge, A.W.; Haastrup, E.K.; Sengeløv, H.; Minulescu, L.; Dickmeiss, E.; Fischer-Nielsen, A. Addition of plerixafor for CD34+ cell mobilization in six healthy stem cell donors ensured satisfactory grafts for transplantation. Transfusion 2014, 54, 1055–1058. [Google Scholar] [CrossRef] [PubMed]
  45. Anderlini, P.; Rizzo, J.D.; Nugent, M.L.; Schmitz, N.; Champlin, R.E.; Horowitz, M.M. Peripheral blood stem cell donation: An analysis from the International Bone Marrow Transplant. Registry (IBMTR) and European Group for Blood and Marrow Transplant (EBMT) databases. Bone Marrow Transplant. 2001, 27, 689–692. [Google Scholar] [CrossRef] [PubMed]
  46. Hsu, J.W.; Shaw, B.E.; Kim, S.; Logan, B.R.; Sees, J.A.; Confer, D.L.; Pulsipher, M.A.; Shah, N.; Switzer, G.E.; Abidi, M.H.; et al. Collection of Peripheral Blood Progenitor Cells in 1 Day Is Associated with Decreased Donor Toxicity Compared to 2 Days in Unrelated Donors. Biol. Blood Marrow Transplant. 2020, 26, 1210–1217. [Google Scholar] [CrossRef] [PubMed]
  47. Rinaldi, C.; Savignano, C.; Pasca, S.; Sperotto, A.; Patriarca, F.; Isola, M.; Fanin, R.; De Angelis, V. Efficacy and safety of peripheral blood stem cell mobilization and collection: A single-center experience in 190 allogeneic donors. Transfusion 2012, 52, 2387–2394. [Google Scholar] [CrossRef]
  48. Pulsipher, M.A.; Chitphakdithai, P.; Miller, J.P.; Logan, B.R.; King, R.J.; Rizzo, J.D.; Leitman, S.F.; Anderlini, P.; Haagenson, M.D.; Kurian, S.; et al. Adverse events among 2408 unrelated donors of peripheral blood stem cells: Results of a prospective trial from the National Marrow Donor Program. Blood 2009, 113, 3604–3611. [Google Scholar] [CrossRef] [PubMed]
  49. Hölig, K.; Kramer, M.; Kroschinsky, F.; Bornhäuser, M.; Mengling, T.; Schmidt, A.H.; Rutt, C.; Ehninger, G. Safety and efficacy of hematopoietic stem cell collection from mobilized peripheral blood in unrelated volunteers: 12 years of single-center experience in 3928 donors. Blood 2009, 114, 3757–3763. [Google Scholar] [CrossRef]
  50. Abutalib, S.A.; Padmanabhan, A.; Pham, H.P.; Worel, N. Best Practices of Apheresis in Hematopoietic Cell Transplantation; Springer: Cham, Switzerland, 2020. [Google Scholar]
  51. Lanza, F.; Marchetti, M.; Zannetti, B.A. Overview on novel strategies and current guidelines for hematopoietic stem cell mobilisation and collection. Transfus. Apher. Sci. 2023, 62, 103830. [Google Scholar] [CrossRef]
  52. Kanate, A.S.; Majhail, N.S.; Savani, B.N.; Bredeson, C.; Champlin, R.E.; Crawford, S.; Giralt, S.A.; LeMaistre, C.F.; Marks, D.I.; Omel, J.L.; et al. Indications for Hematopoietic Cell Transplantation and Immune Effector Cell Therapy: Guidelines from the American Society for Transplantation and Cellular Therapy. Biol. Blood Marrow Transplant. 2020, 26, 1247–1256. [Google Scholar] [CrossRef]
  53. Liu, W.; Ji, X.; Song, Y.; Wang, X.; Zheng, W.; Lin, N.; Tu, M.; Xie, Y.; Ping, L.; Ying, Z.; et al. Improving survival of 3760 patients with lymphoma: Experience of an academic center over two decades. Cancer Med. 2020, 9, 3765–3774. [Google Scholar] [CrossRef]
  54. Giralt, S.; Costa, L.; Schriber, J.; Dipersio, J.; Maziarz, R.; McCarty, J.; Shaughnessy, P.; Snyder, E.; Bensinger, W.; Copelan, E.; et al. Optimizing autologous stem cell mobilization strategies to improve patient outcomes: Consensus guidelines and recommendations. Biol. Blood Marrow Transplant. 2014, 20, 295–308. [Google Scholar] [CrossRef] [PubMed]
  55. Mohty, M.; Hübel, K.; Kröger, N.; Aljurf, M.; Apperley, J.; Basak, G.W.; Bazarbachi, A.; Douglas, K.; Gabriel, I.; Garderet, L.; et al. Autologous haematopoietic stem cell mobilisation in multiple myeloma and lymphoma patients: A position statement from the European Group for Blood and Marrow Transplantation. Bone Marrow Transplant. 2014, 49, 865–872. [Google Scholar] [CrossRef] [PubMed]
  56. Scarlata, S.; Annibali, O.; Santangelo, S.; Tomarchio, V.; Ferraro, S.; Armiento, D.; Scardocci, A.; Arcese, W.; Antonelli Incalzi, R.; Avvisati, G. Pulmonary complications and survival after autologous stem cell transplantation: Predictive role of pulmonary function and pneumotoxic medications. Eur. Respir. J. 2017, 49, 1601902. [Google Scholar] [CrossRef] [PubMed]
  57. Annibali, O.; Piccioni, L.; Tomarchio, V.; Circhetta, E.; Sarlo, C.; Franceschini, L.; Cantonetti, M.; Rizzo, E.; Angeletti, S.; Tirindelli, M.C.; et al. Impact of IFN lambda 3/4 single nucleotide polymorphisms on the cytomegalovirus reactivation in autologous stem cell transplant patients. PLoS ONE 2018, 13, e0200221. [Google Scholar] [CrossRef] [PubMed]
  58. Arora, S.; Majhail, N.S.; Liu, H. Hematopoietic Progenitor Cell Mobilization for Autologous Stem Cell Transplantation in Multiple Myeloma in Contemporary Era. Clin. Lymphoma Myeloma Leuk. 2019, 19, 200–205. [Google Scholar] [CrossRef] [PubMed]
  59. Laurent, V.; Fronteau, C.; Antier, C.; Dupuis, P.; Tessoulin, B.; Gastinne, T.; Mahé, B.; Blin, N.; Dubruille, V.; Lok, A.; et al. Autologous stem-cell collection following VTD or VRD induction therapy in multiple myeloma: A single-center experience. Bone Marrow Transplant. 2021, 56, 395–399. [Google Scholar] [CrossRef] [PubMed]
  60. Chhabra, S.; Callander, N.; Watts, N.L.; Costa, L.J.; Thapa, B.; Kaufman, J.L.; Laubach, J.; Sborov, D.W.; Reeves, B.; Rodriguez, C.; et al. Stem Cell Mobilization Yields with Daratumumab- and Lenalidomide-Containing Quadruplet Induction Therapy in Newly Diagnosed Multiple Myeloma: Findings from the MASTER and GRIFFIN Trials. Transplant. Cell. Ther. 2023, 29, 174.e1–174.e10. [Google Scholar] [CrossRef]
  61. Mina, R.; Petrucci, M.T.; Bonello, F.; Bongarzoni, V.; Saccardi, R.; Bertuglia, G.; Mengarelli, A.; Spadaro, A.; Lisi, C.; Curci, P.; et al. A prospective, multicenter study on hematopoietic stem-cell mobilization with cyclophosphamide plus granulocyte colony-stimulating factor and ‘on-demand’ plerixafor in multiple myeloma patients treated with novel agents. Haematologica 2023, 1–17. [Google Scholar] [CrossRef] [PubMed]
  62. Lemonakis, K.; Tatting, L.; Lisak, M.; Carlson, K.; Crafoord, J.; Blimark, C.H.; Santamaria, A.I.; Wichert, S.; Lenhoff, S.; Hansson, M. Impact of daratumumab-based induction on stem cell collection parameters in Swedish myeloma patients. Haematologica 2023, 108, 610–614. [Google Scholar] [CrossRef]
  63. Johnsrud, A.; Ladha, A.; Muffly, L.; Shiraz, P.; Goldstein, G.; Osgood, V.; Shizuru, J.A.; Johnston, L.; Arai, S.; Weng, W.K.; et al. Stem Cell Mobilization in Multiple Myeloma: Comparing Safety and Efficacy of Cyclophosphamide +/- Plerixafor versus Granulocyte Colony-Stimulating Factor +/- Plerixafor in the Lenalidomide Era. Transplant. Cell. Ther. 2021, 27, 590.e1–590.e8. [Google Scholar] [CrossRef]
  64. Russell, N.; Douglas, K.; Ho, A.D.; Mohty, M.; Carlson, K.; Ossenkoppele, G.J.; Milone, G.; Pareja, M.O.; Shaheen, D.; Willemsen, A.; et al. Plerixafor and granulocyte colony-stimulating factor for first-line steady-state autologous peripheral blood stem cell mobilization in lymphoma and multiple myeloma: Results of the prospective PREDICT trial. Haematologica 2013, 98, 172–178. [Google Scholar] [CrossRef] [PubMed]
  65. Guan, F.S.; He, D.H.; Li, Y.; Zhang, Y.; Zheng, G.F.; Zhu, Y.Y.; He, J.S.; Zhang, E.F.; Cai, Z.; Zhao, Y. Efficacy and Safety of Plerixafor Combined with G-CSF for Autologous Peripheral Blood Hematopoietic Stem Cell Mobilization in Lymphoma Patients. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2023, 31, 1056–1060. [Google Scholar] [CrossRef] [PubMed]
  66. Thurlapati, A.; Roubal, K.; Davis, J.A.; Shah, S.Z.; Smith, D.; McGann, M.; Gaffney, K.; Cendagorta, A.; Maldonado, A.; Weeda, E.; et al. Stem Cell Mobilization for Multiple Myeloma Patients Receiving Daratumumab-Based Induction Therapy: A Real- World Experience. Transplant. Cell. Ther. 2023, 29, 340.e1–340.e4. [Google Scholar] [CrossRef]
  67. Wuchter, P.; Ran, D.; Bruckner, T.; Schmitt, T.; Witzens-Harig, M.; Neben, K.; Goldschmidt, H.; Ho, A.D. Poor mobilization of hematopoietic stem cells-definitions, incidence, risk factors, and impact on outcome of autologous transplantation. Biol. Blood Marrow Transplant. 2010, 16, 490–499. [Google Scholar] [CrossRef] [PubMed]
  68. Olivieri, A.; Marchetti, M.; Lemoli, R.; Tarella, C.; Iacone, A.; Lanza, F.; Rambaldi, A.; Bosi, A. Proposed definition of ‘poor mobilizer’ in lymphoma and multiple myeloma: An analytic hierarchy process by ad hoc working group Gruppo ItalianoTrapianto di Midollo Osseo. Bone Marrow Transplant. 2012, 47, 342–351. [Google Scholar] [CrossRef] [PubMed]
  69. Costa, L.J.; Alexander, E.T.; Hogan, K.R.; Schaub, C.; Fouts, T.V.; Stuart, R.K. Development and validation of a decision-making algorithm to guide the use of plerixafor for autologous hematopoietic stem cell mobilization. Bone Marrow Transplant. 2011, 46, 64–69. [Google Scholar] [CrossRef] [PubMed]
  70. Sukhtankar, D.D.; Fung, J.J.; Kim, M.N.; Cayton, T.; Chiou, V.; Caculitan, N.G.; Zalicki, P.; Kim, S.; Jo, Y.; Lee, J.M.; et al. GPC-100, a novel CXCR4 antagonist, improves in vivo hematopoietic cell mobilization when combined with propranolol. PLoS ONE 2023, 18, e0287863. [Google Scholar] [CrossRef] [PubMed]
  71. Setia, G.; Hagog, N.; Jalilizeinali, B.; Funkhouser, S.; Pierzchanowski, L.; Lan, F.; Gabig, T.G.; Kiner-Strachan, B.; Kelleher, K.; Hsu, M.-C.; et al. A phase II, open-label pilot study to evaluate the hematopoietic stem cell mobilization of TG-0054 combined with G-CSF in 12 patients with multiple myeloma, non-Hodgkin lymphoma or Hodgkin lymphoma-an interim analysis. Blood 2015, 126, 515. [Google Scholar] [CrossRef]
  72. Liu, W.; Li, Y.; Wang, Q.; Su, H.; Ding, K.; Shuang, Y.; Gao, S.; Zou, D.; Jing, H.; Chai, Y.; et al. YF-H-2015005, a CXCR4 Antagonist, for the Mobilization of Hematopoietic Stem Cells in Non-Hodgkin Lymphoma Patients: A Randomized, Controlled, Phase 3 Clinical Trial. Front. Med. 2021, 8, 609116. [Google Scholar] [CrossRef]
  73. Crees, Z.D.; Rettig, M.P.; Jayasinghe, R.G.; Stockerl-Goldstein, K.; Larson, S.M.; Arpad, I.; Milone, G.A.; Martino, M.; Stiff, P.; Sborov, D. Motixafortide and G-CSF to mobilize hematopoietic stem cells for autologous transplantation in multiple myeloma: A randomized phase 3 trial. Nat. Med. 2023, 29, 869–879. [Google Scholar] [CrossRef]
  74. Balogun, R.A.; Aqui, N.; Alicia, G.; Pham Huy, P.; Torloni, A.S.; Wrhri, G.; Yamada, C. Principles of Apheresis Technology. Techinical Principles of Apheresis Medicine, 7th ed.; American Society for Apheresis: Vancouver, BC, Canada, 2020. [Google Scholar]
  75. Maitta, R.W. Current state of apheresis technology and its applications. Transfus. Apher. Sci. 2018, 57, 606–613. [Google Scholar] [CrossRef] [PubMed]
  76. Sputtek, A.; Schubert, C.; Peine, S.; Rowe, A.W. Safe collection of peripheral blood stem cells in patients and donors using the Amicus (R) separator and the Spectra Optia (R) apheresis system. In Vox Sanguinis; Wiley-Blackwell: Hoboken, NJ, USA, 2013; pp. 294–295. [Google Scholar]
  77. Burgstaler, E.A.; Porrata, L.F.; Markovic, S.N.; Winters, J.L. Use of various offset settings in the Fenwal Amicus during hematopoietic progenitor cell collection to increase lymphocyte yield and reduce cross-cellular contamination. J. Clin. Apher. 2010, 25, 301–309. [Google Scholar] [CrossRef] [PubMed]
  78. Panch, S.R.; Yau, Y.Y.; Kang, E.M.; De Ravin, S.S.; Malech, H.L.; Leitman, S.F. Mobilization characteristics and strategies to improve hematopoietic progenitor cell mobilization and collection in patients with chronic granulomatous disease and severe combined immunodeficiency. Transfusion 2015, 55, 265–274. [Google Scholar] [CrossRef] [PubMed]
  79. Wang, T.F.; Chen, S.H.; Yang, S.H.; Su, Y.C.; Chu, S.C.; Li, D.K. Poor harvest of peripheral blood stem cell in donors with microcytic red blood cells. Transfusion 2013, 53, 91–95. [Google Scholar] [CrossRef] [PubMed]
  80. Leitman, S.F.; Yau, Y.; Matthews, C.L.; Hopkins, J.A.; Min, K. Optimization of Unstimulated Mononuclear Cell Collections Using the Amicus Continuous-Flow Apheresis Device. In Transfusion; Wiley-Blackwell Publishing, Inc.: Malden, MA, USA, 2010; p. 15A. [Google Scholar]
  81. Trébéden-Negre, H.; Rosenzwajg, M.; Tanguy, M.L.; Lefrere, F.; Azar, N.; Heshmati, F.; Belhocine, R.; Vernant, J.P.; Klatzmann, D.; Norol, F. Delayed recovery after autologous peripheral hematopoietic cell transplantation: Potential effect of a high number of total nucleated cells in the graft. Transfusion 2010, 50, 2649–2659. [Google Scholar] [CrossRef] [PubMed]
  82. Perseghin, P.; Terruzzi, E.; Dassi, M.; Baldini, V.; Parma, M.; Coluccia, P.; Accorsi, P.; Confalonieri, G.; Tavecchia, L.; Verga, L.; et al. Management of poor peripheral blood stem cell mobilization: Incidence, predictive factors, alternative strategies and outcome. A retrospective analysis on 2177 patients from three major Italian institutions. Transfus. Apher. Sci. 2009, 41, 33–37. [Google Scholar] [CrossRef]
  83. Miyazaki, K.; Suzuki, K. Poor mobilizer and its countermeasures. Transfus. Apher. Sci. 2018, 57, 623–627. [Google Scholar] [CrossRef]
  84. To, L.B.; Levesque, J.P.; Herbert, K.E. How I treat patients who mobilize hematopoietic stem cells poorly. Blood 2011, 118, 4530–4540. [Google Scholar] [CrossRef] [PubMed]
  85. Ataca Atilla, P.; Bakanay Ozturk, S.M.; Demirer, T. How to manage poor mobilizers for high dose chemotherapy and autologous stem cell transplantation? Transfus. Apher. Sci. 2017, 56, 190–198. [Google Scholar] [CrossRef]
  86. Seggewiss, R.; Buss, E.C.; Herrmann, D.; Goldschmidt, H.; Ho, A.D.; Fruehauf, S. Kinetics of peripheral blood stem cell mobilization following G-CSF-supported chemotherapy. Stem Cells 2003, 21, 568–574. [Google Scholar] [CrossRef]
  87. Roberts, A.W.; Foote, S.; Alexander, W.S.; Scott, C.; Robb, L.; Metcalf, D. Genetic influences determining progenitor cell mobilization and leukocytosis induced by granulocyte colony-stimulating factor. Blood 1997, 89, 2736–2744. [Google Scholar] [CrossRef]
  88. Soukup, A.A.; Bresnick, E.H. Gata2 noncoding genetic variation as a determinant of hematopoietic stem/progenitor cell mobilization efficiency. Blood Adv. 2023, 7, 7564–7575. [Google Scholar] [CrossRef]
  89. Pierelli, L.; Perseghin, P.; Marchetti, M.; Accorsi, P.; Fanin, R.; Messina, C.; Olivieri, A.; Risso, M.; Salvaneschi, L.; Bosi, A. Best practice for peripheral blood progenitor cell mobilization and collection in adults and children: Results of a Società Italiana Di Emaferesi e Manipolazione Cellulare (SIDEM) and Gruppo Italiano Trapianto Midollo Osseo (GITMO) consensus process. Transfusion 2011, 52, 893–905. [Google Scholar] [CrossRef]
  90. Bashey, A.; Donohue, M.; Liu, L.; Medina, B.; Corringham, S.; Ihasz, A.; Carrier, E.; Castro, J.E.; Holman, P.R.; Xu, R.; et al. Peripheral blood progenitor cell mobilization with intermediate-dose cyclophosphamide, sequential granulocyte-macrophage-colony-stimulating factor and granulocyte-colony-stimulating factor, and scheduled commencement of leukapheresis in 225 patients undergoing autologous transplantation. Transfusion 2007, 47, 2153–2160. [Google Scholar] [CrossRef]
  91. Spoerl, S.; Peter, R.; Wäscher, D.; Götze, K.; Verbeek, M.; Peschel, C.; Krackhardt, A.M. Patients’ outcome after rescue plerixafor administration for autologous stem cell mobilization: A single-center retrospective analysis. Transfusion 2017, 57, 115–121. [Google Scholar] [CrossRef]
  92. Andritsos, L.A.; Huang, Y.; Abraham, I.; Huff, K.; Scrape, S.R.; Fan, T.; Alkhatib, N.; Hofmeister, C.C.; Drea, E.; McBride, A. Clinical and cost outcomes of pre-emptive plerixafor administration in patients with multiple myeloma undergoing stem cell mobilization. Leuk. Res. 2019, 85, 106215. [Google Scholar] [CrossRef]
  93. Shi, P.A.; Miller, L.K.; Isola, L.M. Prospective study of mobilization kinetics up to 18 h after late-afternoon dosing of plerixafor. Transfusion 2014, 54, 1263–1268. [Google Scholar] [CrossRef]
  94. Yuan, S.; Wang, S. How do we mobilize and collect autologous peripheral blood stem cells? Transfusion 2017, 57, 13–23. [Google Scholar] [CrossRef]
  95. Flommersfeld, S.; Sohlbach, K.; Jaques, G.; Bein, G.; Hoffmann, J.; Kostrewa, P.; Sachs, U.J. Collection of peripheral blood progenitor cells on Day 4 is feasible and effective while reducing granulocyte-colony-stimulating factor exposure to healthy donors. Transfusion 2015, 55, 1269–1274. [Google Scholar] [CrossRef]
  96. Kimura, S.; Ohkawara, H.; Minakawa, K.; Fukatsu, M.; Mori, H.; Takahashi, H.; Harada-Shirado, K.; Ohara, Y.; Takahashi, N.; Mochizuki, K.; et al. Optimal timing of apheresis for the efficient mobilization of peripheral blood progenitor cells recruited by high-dose granulocyte colony-stimulating factor in healthy donors. Transfus. Apher. Sci. 2020, 59, 102737. [Google Scholar] [CrossRef]
  97. Kaul, E.; Kothari, S.; Setia, R.; Sharma, S.; Suleiman, S.; Khandelwal, V.; Kharya, G.; Sharma, B.; Handoo, A.; Choudhary, D. Peripheral Blood Stem Cell Collection on Day 4 Is Feasible and Safe in a Majority of Allogeneic Stem Cell Transplant Donors. Biol. Blood Marrow Transplant. 2016, 22, S328. [Google Scholar] [CrossRef]
  98. Humpe, A.; Riggert, J.; Meineke, I.; Kurz, M.; Eil, A.; Storkebaum, B.; Binder, C.; Munzel, U.; Funke, I.; Höcker, P.; et al. A cell-kinetic model of CD34+ cell mobilization and harvest: Development of a predictive algorithm for CD34+ cell yield in PBPC collections. Transfusion 2000, 40, 1363–1370. [Google Scholar] [CrossRef] [PubMed]
  99. Delamain, M.T.; Marques, J.F., Jr.; de Souza, C.A.; Lorand-Metze, I.; Metze, K. An algorithm based on peripheral CD34+ cells and hemoglobin concentration provides a better optimization of apheresis than the application of a fixed CD34 threshold. Transfusion 2008, 48, 1133–1137. [Google Scholar] [CrossRef]
  100. Pierelli, L.; Maresca, M.; Piccirillo, N.; Pupella, S.; Gozzer, M.; Foddai, M.L.; Vacca, M.; Adorno, G.; Coppetelli, U.; Paladini, U. Accurate prediction of autologous stem cell apheresis yields using a double variable-dependent method assures systematic efficiency control of continuous flow collection procedures. Vox Sang. 2006, 91, 126–134. [Google Scholar] [CrossRef]
  101. Vrielink, H.; Le Poole, K.; Stegmayr, B.; Kielstein, J.; Berlin, G.; Ilhan, O.; Seval, G.C.; Prophet, H.; Aandahl, A.; Deeren, D.; et al. The world apheresis association registry, 2023 update. Transfus Apher. Sci. 2023, 62, 103831. [Google Scholar] [CrossRef]
  102. Bueno, J.L.; Alegre, A.; López-Villar, O.; Querol, S.; Arroyo, J.L.; Goterris, R.; Sureda, A.; García-Gala, J.M.; Amunarriz, C.; Albo, C.; et al. Agreements and uncertainties in autologous haematopoietic stem cell mobilization and collection. A Spanish consensus document. Bone Marrow Transplant. 2020, 55, 811–817. [Google Scholar] [CrossRef]
  103. Couzin, C.; Manceau, S.; Diana, J.S.; Joseph, L.; Magnani, A.; Magrin, E.; Amrane, H.; Dupont, E.; Raphalen, J.; Sibon, D.; et al. Vascular access for optimal hematopoietic stem cell collection. J. Clin. Apher. 2021, 36, 12–19. [Google Scholar] [CrossRef] [PubMed]
  104. Rhodes, B.; Anderlini, P. Allogeneic peripheral blood stem cell collection as of 2008. Transfus. Apher. Sci. 2008, 38, 219–227. [Google Scholar] [CrossRef] [PubMed]
  105. Hölig, K.; Blechschmidt, M.; Kramer, M.; Zimmer, K.; Kroschinsky, F.; Poppe-Thiede, K.; Bornhäuser, M.; Ehninger, G. Peripheral blood stem cell collection in allogeneic donors: Impact of venous access. Transfusion 2012, 52, 2600–2605. [Google Scholar] [CrossRef]
  106. Leitner, G.C.; Baumgartner, K.; Kalhs, P.; Biener, D.; Greinix, H.T.; Hoecker, P.; Worel, N. Regeneration, health status and quality of life after rhG-CSF-stimulated stem cell collection in healthy donors: A cross-sectional study. Bone Marrow Transplant. 2009, 43, 357–363. [Google Scholar] [CrossRef]
  107. Vacca, M.; Perseghin, P.; Accorsi, P.; Pierelli, L. Central venous catheter insertion in peripheral blood hematopoietic stem cell sibling donors: The SIdEM (Italian Society of Hemapheresis and Cell Manipulation) point of view. Transfus. Apher. Sci. 2014, 50, 200–206. [Google Scholar] [CrossRef] [PubMed]
  108. O’Leary, M.F.; Dunbar, N.M.; Kim, H.C.; Draper, N.L.; Linenberger, M.; Schwartz, J.; Miller, Y.; Murtaugh, A.; West, F.B.; Fernando, L.P.; et al. Venous access for hematopoietic progenitor cell collection: An international survey by the ASFA HPC donor subcommittee. J. Clin. Apher. 2016, 31, 529–534. [Google Scholar] [CrossRef] [PubMed]
  109. Caime, A.; Piredda, A.; Lucchetti, B.; Magarò, A.; Zencovich, C.; Clerici, M.; Laszlo, D. Midline catheter as effective device in healthy allogeneic donors and patients without an adequate peripheral venous access for HPC collection by apheresis: Preliminary experience at IEO. Transfus. Apher. Sci. 2020, 59, 102740. [Google Scholar] [CrossRef]
  110. Casacchia, C.; Lozano, M.; Schomberg, J.; Barrows, J.; Salcedo, T.; Puthenveetil, G. Novel use of a midline catheter for therapeutic and donor apheresis in children and adults. J. Clin. Apher. 2021, 36, 711–718. [Google Scholar] [CrossRef]
Figure 1. Specific gravity of blood cells and plasma. PLTs, Platelets; LYMs, Lymphocytes, MNCs, Mononuclear cells; CD34+, Hematopoietic stem cells; RBCs, Red blood cells.
Figure 1. Specific gravity of blood cells and plasma. PLTs, Platelets; LYMs, Lymphocytes, MNCs, Mononuclear cells; CD34+, Hematopoietic stem cells; RBCs, Red blood cells.
Cells 13 00586 g001
Table 1. MNC collection instruments.
Table 1. MNC collection instruments.
MNC COLLECTION
INSTRUMENTS
TYPE OF
SYSTEM
NEEDLE SYSTEM
(ECV Approx. [50,74])
KIT AND PROTOCOL
FRESENIUS-KABI
AMICUS®CFCDouble needle
(163 mL)
MNC Kit (works in cycles, centrifuge)
COM.TEC®CFCDouble needle
P1YA, P1Y, (177 mL) *
C4Y, RVY (193 mL) *
P1YA, P1Y, C4Y, RVY
(works in cycles, centrifuge)
HAEMONETICS CORP
MCS+® 9000IFCSingle needle
(Variable
125 bowl 380 (38% HCT)-259 (52% HCT)/82–87 mL) *
PROTOCOL PBSC 0971E-00
(works in cycles, centrifuge)
TERUMO BCT INC
SPECTRA OPTIA®CFCDouble needle
MNC (191 mL)
CMNC (297 mL)
MNC protocol (works in cycles, centrifuge + elutriation)
CMNC protocol (centrifuge)
ECV approx.: extracorporeal volume approximately; IFC = intermittent flow centrifugation; CFC = continuous flow centrifugation; * User manual.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Prisciandaro, M.; Santinelli, E.; Tomarchio, V.; Tafuri, M.A.; Bonchi, C.; Palazzo, G.; Nobile, C.; Marinucci, A.; Mele, M.; Annibali, O.; et al. Stem Cells Collection and Mobilization in Adult Autologous/Allogeneic Transplantation: Critical Points and Future Challenges. Cells 2024, 13, 586. https://doi.org/10.3390/cells13070586

AMA Style

Prisciandaro M, Santinelli E, Tomarchio V, Tafuri MA, Bonchi C, Palazzo G, Nobile C, Marinucci A, Mele M, Annibali O, et al. Stem Cells Collection and Mobilization in Adult Autologous/Allogeneic Transplantation: Critical Points and Future Challenges. Cells. 2024; 13(7):586. https://doi.org/10.3390/cells13070586

Chicago/Turabian Style

Prisciandaro, Michele, Enrico Santinelli, Valeria Tomarchio, Maria Antonietta Tafuri, Cecilia Bonchi, Gloria Palazzo, Carolina Nobile, Alessandra Marinucci, Marcella Mele, Ombretta Annibali, and et al. 2024. "Stem Cells Collection and Mobilization in Adult Autologous/Allogeneic Transplantation: Critical Points and Future Challenges" Cells 13, no. 7: 586. https://doi.org/10.3390/cells13070586

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop