235
Views
28
CrossRef citations to date
0
Altmetric
Review

Hematopoietic stem and progenitor cell harvesting: technical advances and clinical utility

Pages 55-67 | Published online: 18 Feb 2015

Abstract

Hematopoietic stem and progenitor cell (HSPC) transplantations require prior harvesting of allogeneic or autologous HSPCs. HSPCs are usually present in bone marrow (BM) during the entire life, in cord blood (CB) at birth, or in peripheral blood (PB) under particular circumstances. HSPCs were first harvested in BM and later in CB and PB, as studies showed interesting features of such grafts. All harvesting methods were in use throughout the years, except BM harvesting for HSPC autologous transplantation, which was replaced by PB harvesting. BM, CB, and PB harvesting methods have been developed, and materials and devices technically improved to increase the number of HSPCs harvested. In parallel, knowing the features of the donors or patients associated with successful numbers of HSPCs allows the adaptation of appropriate harvesting methods. Moreover, it is important to ensure the safety of donors or patients while harvesting. This review describes the methods used for harvesting based on recent studies or developments around these methods, and more particularly, the means developed to increase the numbers of HSPCs harvested in each method. It also explains briefly the influence of technical improvements in HSPC harvesting on potential changes in HSPC graft composition.

Introduction

Hematopoietic stem and progenitor cell (HSPC) transplantation, which was initially considered as an experimental therapy, has been performed and studied over the last 40 years. It has become a referent treatment of severe hematological diseases.

As HSPCs are localized in the bone marrow (BM), the first HSPC transplantations in the 1950s used that as source of cells.Citation1 Over the last three decades, allogeneic BM transplantations have become a referent therapy for severe malignant or nonmalignant hematologic diseases.Citation2

HSPC transplantations evolved after HSPCs were detected in other sites such as peripheral blood (PB) or cord blood (CB).Citation3Citation5 The first allogeneic CB transplantation was successfully performed at the end of the 1980s.Citation5 In parallel, the development of apheresis devices enabled teams to harvest sufficient PB HSPCs for transplantation. Over the last 20 years, numerous HSPC transplantations have been performed. In all types of HSPC transplantations (BM, CB, PB), it was demonstrated that the outcome for the transplanted patients depended on the number of HSPCs contained in the graft. HSPC harvesting methods have, therefore, been improved to transplant higher numbers of HSPCs. In this review, we focus on the recent technical advances in HSPC harvesting, recent studies or developments that have brought new knowledge, and their consequences on the graft composition and their clinical utility.

HSPC harvesting methods

BM harvesting

Although HSPC harvesting was performed for the first time more than 50 years ago, BM harvesting was developed mainly to perform allogeneic HSPC transplantations and later autologous transplantations. Nowadays, BM is harvested to perform only allogeneic HSPC transplantation.

Protocol for BM harvesting

The current protocol recommended for BM harvesting consists in aspirating BM from the posterior iliac crest in a donor under general anesthesia using a needle with multiple side holes, which should be performed by one or two hematologists. The level of aspiration is restricted to 15–20 mL per puncture into sterile syringes previously rinsed with a heparin/saline solution. While harvesting, regular gentle agitation of the harvesting bag containing an anticoagulant solution prevents clotting. A total nucleated cell (TNC) count performed at midway predicts the optimal BM volume to be harvested within the limit of the maximum volume. The BM harvested is sent to the cell therapy unit where it is filtered and processed in case of ABO incompatibility.

The acceptable cell dose harvested in BM and required for allogeneic transplantation is 3–5×108 TNCs per kilogram of recipient body weight (BW). However, when harvesting and transplanting higher numbers of TNCs, better outcomes, such as improved overall survival, were shown in patients.Citation6 This occurred particularly in patients allogeneically transplanted for acute myeloid leukemia (AML).Citation6 Therefore, hematological teams have developed strategies to harvest higher numbers of TNCs.

How to increase numbers of HSPCs harvested in BM

It was suggested that priming donors with granulocyte colony-stimulating factor (G-CSF) enhanced the number of TNCs harvested, but that approach was not developed.Citation7 Two other ways to harvest higher numbers of BM TNCs and HSPCs, ie, by harvesting larger volumes of BM or by increasing the cell density of the BM harvested, have been developed. The total volume of BM harvested, within the limit of 20 mL/kg to prevent excessive blood loss, depends on the donor’s BW. In standard procedures, hematologists usually harvest the highest possible volume, which could be deleterious inducing a hemodilution of the BM harvested. Indeed, it was clearly shown that the volume of BM harvested was inversely correlated to the cell density.Citation8

To obtain a higher cell density and higher number of cells, it is necessary to change the needle position at short intervals. It is also recommended to optimize the level of aspiration at each site, repetitive aspirations of small volumes of BM enhancing the numbers of TNCs and HSPCs harvested.Citation9 Moreover, using needles with multiple side holes combined with harvesting small, repetitive BM volumes induced a high BM cell yield.Citation10,Citation11

Parameters other than harvesting techniques, such as the characteristics of donors, could influence the cell density and numbers of TNCs in the BM harvested. Among them, the donor BW and baseline white blood cell (WBC) levels were correlated to the cell density in BM harvests.Citation8 Other characteristics, including lower age and cytomegalovirus-negative donors, smoking, higher hemoglobin and mononuclear cell blood levels, higher number of whole blood donations in the year preceding the BM harvest, and higher body surface area, were associated with higher numbers of TNCs harvested in BM.Citation12 A higher hemoglobin level, higher number of whole blood donations, and smoking were probably associated with a more active hematopoiesis.

CB harvesting

CB at delivery was found to contain HSPCs.Citation4,Citation13 These HSPCs displayed interesting features such as high-potential clonogenicity, but their absolute numbers were, however, not sufficient for allogeneic transplantation in adults and were only appropriate to transplant low-BW recipients, ie, children.Citation5 Such allogeneic transplantations displayed advantages over allogeneic BM and PB HSPC transplantations, such as low incidence of graft-versus-host-disease. Considering the advantages of CB transplantations, hematologic teams created a European organization for CB banking.Citation14 The aim was to allow transplantation from unrelated children and possibly from low-BW adults if the numbers of cells contained in CB units (CBU) were sufficient for transplantation. For that purpose, the numbers of HSPCs harvested in CBUs had to be increased by optimizing harvesting methods or obstetrical conditions. In 2005, an American team developed the concept of transplanting two CBUs partially matched together, which induced lower duration of aplasia and fewer infectious complications than after single CBU transplantations in adults.Citation15 This reinforced the need for standardizations in the stages of the CBU banking process and for studies to increase the number of HSPCs in CBUs.

Protocol for CB harvesting

The initial method for harvesting CB was described at the end of the 1980s.Citation13 Although a few attempts were made to improve it, the current protocol recommended is very similar to the initial protocol and is described as follows: After the birth of the infant, the umbilical cord is double-clamped from the umbilicus and transacted between the clamps. The umbilical cord vein is punctured under sterile conditions, and the blood flows freely by gravity into an anticoagulated sterile closed harvesting system.Citation16,Citation17 Birth unit staff should be trained in CB harvesting to reduce the rejection rate due to labeling problems, bacterial contamination, and clotting.Citation16 The mean volume and numbers of HSPCs harvested in CBU can vary from one study to another, but can be described as follows: 108±28 mL, 12.5±5.3×108 TNC, and 3.6±3.3×106 CD34+ cells.Citation18 The harvest must be stored and transported under controlled temperature. The time interval between CBU harvesting and processing is limited to 24–36 hours corresponding to sufficient cell viability.

How to increase numbers of HSPCs harvested in CB

Initially, the numbers of HSPCs contained in CBUs or transplanted were defined by measuring the numbers of TNCs and colony forming unit-granulocyte macrophage (CFU-GM).Citation5,Citation13 After 2000, the CD34+ immunomarker was used to characterize the population of HSPCs contained in BM, PB, and in CBU, in addition to TNCs.Citation19 A strong correlation was found between the numbers of HSPCs and the CB volume harvested.Citation20,Citation21 Obstetrical and technical methods were then developed to harvest the highest possible volume of CB. CB volume was initially considered as – and was later proven to be – one of the best predictive data for acceptable CBU banking.Citation20,Citation21

A variety of potential CB harvesting methods have been described to harvest large volumes of CB. Closed, semiclosed, and open systems were developed using blood bags, syringes/flushed or not, or drains/flushed or not, with no significant difference in the harvested volume.Citation22,Citation23 Since 1998, the US Food and Drug administration (FDA) invited professional groups to submit proposed standards, data, and information to have available CBUs appropriate for allogeneic transplantation. Providers developed systems to obtain more CBU volume and TNCs harvested (). Some systems were likely to increase the volume or the numbers of cells harvested, but these were not extensively used, and the protocol for CB harvesting remained as described above.

Table 1 Recent studies performed to improve amounts of hematopoietic stem and progenitor cell (HSPC) harvested in cord blood (CB)

In order to increase the volumes and numbers of cells harvested, obstetrical teams assessed and compared CB harvesting methods. CB can be harvested before or after the placenta delivery, ie, in utero or ex utero, and during cesarean. In utero, harvesting is done by midwives in the delivery room, the placenta being compressed by the uterus at the third stage of labor. Ex utero, harvesting is done by cord bank employees in an adjacent room right after the placental delivery. Therefore, the obstetrical CB harvesting methods vary from one team to another (). Some studies have shown higher volumes and HSPC numbers when harvested in utero, while other studies have shown comparable volumes and HSPCs numbers in both conditions.Citation18,Citation29Citation31 Both methods have advantages and disadvantages, which generated controversies. Therefore, early cord clamping is associated with higher volumes harvested, but this may hamper the normal process of delivery. Harvesting CB after delivery is easier, but only lower volumes can be harvested. Moreover, comparisons between harvests performed during vaginal and cesarean delivery showed either identical numbers or higher numbers of HSPCs harvested during the latter.Citation32,Citation33

The factors that influence the yield of CB volume and cells have been studied. Therefore, primigravidae, higher birth weight, Caucasian race, young (34–37 weeks) or old (40 weeks), gestational age, and female sex were associated with higher volumes and numbers of CD34+ cells harvested.Citation20,Citation33,Citation34 Prenatal sonografic parameters can estimate fetal weight and are correlated with CB hematological parameters.Citation35 Knowledge of these factors will help in banking and harvesting CB efficiently, but the banking levels depend on the cell threshold and strategy of each CB bank. The main reason for excluding the CBUs harvested is the low volume or low numbers of cells contained in a CBU. The other reasons for excluding the CBU are preparation or logistical complications, abnormal biological result in donor, and CBU microbial infection ().Citation18,Citation36,Citation37 The rate of CBU microbial contamination varies from 0% to 48%.Citation23,Citation24,Citation38

Table 2 Reasons for excluding the CBUs harvested

In order to limit the exclusion of the CBUs, it is necessary to select suitable donors. Successful selection requires the collection of accurate information when gathering and obtaining informed consent from the mother. Parents with previous history of cancers or hematologic, genetic, or autoimmune diseases are excluded from donation. Serological tests (for hepatitis, human immunodeficiency virus [HIV], cytomegalovirus, syphilis, and Epstein–Barr virus) must be performed in the mother’s blood at delivery. However a genetic, hematologic, or oncological disease can occur in newborns several years after CB harvesting and storing, which requires information about the CB donor’s health before sending the CBU for transplantation.

In 1998, the foundation NetCord was developed to establish an international registry for CB banks and procedures with standards for the safe exchange and transplantation use of CBU. In 2000, the NetCord Foundation for the Accreditation of Cellular Therapy released international standards for accreditation not only for harvesting but also for testing, processing, and storing CBU.Citation39

PB HSPC harvesting: mobilization and apheresis techniques

The presence of HSPCs in PB was detected in 1971.Citation3 In parallel, over the last decades, the development of apheresis devices has allowed the harvesting of PB HSPCs. As no HSPCs are present or detected in PB under normal conditions, it is necessary to mobilize HSPCs from BM to PB. These mobilization treatments are not the same in donors for allogeneic transplantations and in patients for autologous transplantations.

HSPC mobilization in healthy donors

Related or unrelated donors usually receive G-CSF (filgrastim or lenograstim) 10 μg/kg/day from 4–5 days before apheresis.Citation40 The factors associated with a better efficiency in mobilizing CD34+ cells are male sex, higher body mass index, higher G-CSF dosage, higher premobilization WBC, and the use of lenograstim rather than filgrastim.Citation41,Citation42 Higher age, female sex, white ethnicity, and donors lighter than their recipient are factors associated with a poorer mobilization.Citation43,Citation44 Knowing the risk factors for poor mobilization allows processing larger blood volume or anticipating a possible rescue by BM harvesting. It is difficult to determine the percentage of donors who fail to mobilize an adequate number of HSPCs for harvest because this minimal number is different among centers. In case of insufficient mobilization and, therefore, insufficient HSPCs harvest, the BM harvest rescue can be replaced by using a new mobilization agent named plerixafor, but this treatment is not approved by the FDA and in most countries.Citation45

HSPC mobilization in patients

The following three broad strategies are usually followed to mobilize HSPCs from BM to PB in patients:

  1. Combined chemotherapies associated with the hematopoietic growth factor, ie, G-CSF currently used to treat the underlying disease, ie, Hodgkin’s disease (HD), non-Hodgkin lymphoma (NHL), or solid tumors, and inducing aplasia allow mobilization of CD34+ cells into PB.Citation46Citation48 Randomized studies have shown that doubling the dose of filgrastim improved the CD34+ cell harvest and decreased the median number of apheresis procedures.Citation49

  2. Cyclophosphamide associated with hematopoietic growth factors can be used in the treatment of multiple myeloma (MM) and HSPC mobilization. Mobilization by cyclophosphamide after new chemotherapy regimen is possible, although some of these agents (thalidomide, lenalidomide) were suspected to cause harvest failure.Citation50,Citation51 Randomized studies have shown that the addition of growth factors (GM-CSF or G-CSF) to cyclophosphamide resulted in a significant increase in the numbers of CD34+ cells harvested.Citation52 The use of biosimilar G-CSF instead of G-CSF seemed to induce the same levels of CD34+ HSPCs harvested and the same harvesting duration.Citation53

  3. Mobilization by hematopoietic growth factors alone can be efficiently used to mobilize HSPCs. Randomized studies compared growth factors, ie, filgrastim to molgramostim and to pegfilgrastim, without demonstrating any superiority for a growth factor or a scheme.Citation54,Citation55

Over the last 15 years, numerous studied have shown the factors that affect HSPC mobilization, ie, age, sex, underlying disease, interval between diagnosis and harvest, exposure to alkylating agents, prior irradiation, marrow involvement, blood-platelets baseline, and cancer relapse ().Citation56Citation62 Megakaryocyte-platelet lineage is particularly sensitive to damage in the BM microenvironment. Therefore, premobilization blood platelet baseline appears to be an indicator for autologous HSPC mobilization.Citation59,Citation61 Moreover, agents (thalidomide, bortezomib or lenalidomide, and fludarabine) used in new therapeutic schemes could affect HSPC mobilization. Indeed, the number of HSPCs harvests after short courses of chemotherapy using these agents is lower than when control groups receive older combination of chemotherapies. Moreover, fludarabine exposure hampers adequate HSPC mobilization probably by causing stem cell damage.Citation58,Citation62 However, the regimen used to treat MM does not significantly hamper HSPC mobilization during G-CSF or during cyclophosphamide mobilization.Citation63,Citation64 Knowing all these factors will enable teams to feature poor mobilizers (PMs).

Table 3 Main factors that may negatively affect successful HSPC harvest

There is no consensus on the definition of PM patients. The patients in whom poor mobilization induces HSPC harvest failure for low blood peak of circulating CD34+ cells associated or not with factors likely to affect HSPC mobilization were named PMs. The patients suspected to become PMs were named predicted PMs. The criteria used for defining the status of PM were different among different studies (). Whatever the definition or the criteria chosen, the overall incidence of mobilization failure varies from 5% to 40%.Citation67Citation69

Table 4 Definition and criteria of poor mobilizers (PMs)

How to manage poor mobilizers

After revealing a PM patient, different solutions can be used to perform appropriate mobilization.

  1. BM harvesting is no more used to perform autologous HSPC harvests because it was proven to be ineffective in patients with insufficient PB HSPC mobilization and/or harvest.Citation70

  2. Remobilization can be performed with high-dose chemotherapy associated with growth factors or with growth factors only.Citation71

  3. High-dose administration of growth factors can be used following the principle of dose-dependent response to mobilization.Citation72,Citation73

  4. Stem cell factor has been successfully used in combination with G-CSF to mobilize HSPCs.Citation74 Stem cell factor is the ligand for c-kit, which is a membrane receptor with tyrosine kinase activity expressed on several tissues including the hematopoietic system. This treatment induced adequate harvest in 40% of PMs.Citation75

  5. More recently, the use of plerixafor has demonstrated encouraging results. Plerixafor is a reversible inhibitor of HSPC adhesion to stromal cells by CXCR4 binding to stromal-derived factor-1. The best scheme for using plerixafor consists in combining it with mobilization by G-CSF. This treatment induces rates of successful CD34+ cell harvests in 70% of NHL, HD, and MM PMs.Citation76 Combining plerixafor with pegfilgrastim or with mobilizing chemotherapy seems safe and effective in PMs, but these results require confirmative data.Citation77 The main disadvantage of plerixafor is its cost, requiring the development of algorithms for the use of plerixafor in autologous HSPC mobilization.Citation78

After mobilizing HSPCs from BM to PB, the apheresis teams have to harvest them from PB.

PB HSPC harvesting by apheresis

The purpose of apheresis sessions during treatment for NHL, HD, or MM is to harvest enough HSPCs to perform autologous transplantation in the patient. The most relevant number of HSPCs to be harvested is 4×106 CD34+ cells/kg BW for treating NHL patients and 4×106 CD34+ cells/kg BW per graft for treating MM patients by harvesting one or two grafts. In all diseases, the minimum number of HSPCs to be harvested is 2×106 CD34+/kg BW for each graft.

To perform allogeneic HSPC transplantations, apheresis sessions are performed in an human leukocyte antigen (HLA)-compatible donor after checking the absence of infectious, oncological, autoimmune, and vascular diseases. The number of HSPCs to be harvested varies depending on the teams and nature of the transplantation (non-myelo, myelo-ablative, or haplo-identical), ie, from 4 to 10×106 CD34+ cells/kg recipients’ BW.

A strong correlation was found between pre-apheresis PB CD34+ cell counts combined or not with the numbers of CD34+ cells harvested at mid-point and numbers of CD34+ cells harvested after the corresponding apheresis sessions.Citation79,Citation80 The analysis of these pre-apheresis blood levels allows apheresis teams to adapt the blood volume processed and potentially perform large-volume leukapheresis.Citation81

Apheresis techniques

Different apheresis devices have been developed to harvest PB HSPCs. All these techniques share a common process, ie, separating blood components in layers by centrifugation and harvesting blood-mobilized HSPCs in a particular layer associated with other blood cells. The principles of the main and current apheresis techniques are summarized in . Over the last 30 years, the COBE Spectra has been considered as the main apheresis device used to harvest PB HSPCs by most apheresis teams.Citation82,Citation83 This technique was extensively used with the Manual Collection Protocol. Later, an automated version of COBE Spectra, named Auto-PBSC, was developed and operated with cyclical harvest of a mononuclear cell fraction.Citation84

Table 5 Main current apheresis techniques used for hematopoietic stem and progenitor cell (HSPC) harvest

Studies have compared the performances of the devices ().Citation82,Citation85Citation90 Indexes were developed to characterize the performances of the devices during harvesting. The main index is the collection efficiency (CE). Other indexes (platelets or hemoglobin losses, apheresis duration, contamination with nontargeted cells) were analyzed.Citation88Citation90 CEs were similar in both protocols (manual and Auto-PBSC) developed for COBE Spectra.Citation84 The platelet loss was lower with the Auto-PBSC than with the Manual COBE Spectra technique.Citation84 The comparison of the performances of Fenwal Amicus and COM.TEC showed identical CE.Citation85 Compared to the Haemonetics MCS+ or to the Baxter Amicus, a shorter apheresis duration and a better correlation between PB pre-apheresis CD34+ cell counts and numbers of CD34+ cells harvested were shown with COBE Spectra.Citation82,Citation83

Table 6 Recent studies comparing the performances of apheresis devices for HSPC harvesting

The COBE Spectra technique is being replaced by a new technique, the Spectra Optia.Citation86 A comparison of the performances between COBE Spectra, Spectra Optia, and COM. TEC techniques showed higher CE in both Spectra Optia and COM.TEC techniques, but Spectra Optia sessions required longer durations.Citation87,Citation88

How to manage poor HSPC harvest by apheresis

The HSPC harvest depends on the pre-apheresis circulating CD34+ cell counts, performances of the apheresis techniques, and the blood volume processed. Each of these elements can be improved. After unsuccessful mobilization, the unfavorable situation can be quickly corrected by using plerixafor in a salvage administration.Citation65,Citation91 The old devices gave the opportunity to improve the harvest manually, while new devices perform the task automatically. Another solution consists in processing more blood volumes during the apheresis sessions. In the past, apheresis sessions consisted of processing large volumes.Citation81 Such approach was safe, but a significant decrease in blood electrolyte concentration and platelets had to be prevented.Citation92 Studies considering the possibility to perform large-volume leukapheresis with the new devices should be carried out.

How to ensure safety of donors

BM harvest

The safety of BM donors must be ensured during and after harvesting. Knowing donors’ medical history and clinical evaluation enables medical teams to prevent occurrence of complications during general anesthesia. It is possible to harvest BM after local anesthesia associated or not with analgesia.Citation93 Decreases in blood red cell levels are linked to the volume of BM harvested and can be treated by iron supplementation or by autologous red cell transfusion. After the donation, donors usually complain of bone pain. The complications and quality of life in adult and pediatric BM donors must be evaluated.Citation94

CB harvest

When harvesting CB, it is essential to ensure safety to both the mother and the infant. The CB harvest must be performed after the delivery. The safe management of obstetric delivery should never be compromised to facilitate CB harvest.Citation16 The umbilical CB clamping should not be performed too early after delivery to prevent deprivation in blood volume and hemodynamic disturbances in the infant.Citation95

PB harvest

Ensuring safety is important for allogeneic donors of PB HSPCs. In this setting, three elements must be considered, ie, the immediate side effects after mobilization by G-CSF or new agents, side effects during apheresis, and long-term side-effects of the mobilizing agents. G-CSF stimulation induces pain (bone, muscle, headache) until the end of stimulation. Donors with higher WBC levels experience more fatigue; females experience headache, nausea, and fever; and higher G-CSF dosage is associated with bone pain.Citation96 G-CSF induces sometimes high WBC levels, which could generate vascular complications. Indeed, splenic rupture, which is a rare complication, has been observed in allogeneic and autologous donors.Citation97 When WBC is higher than 60,000/μL, a GSF dose adaptation can be proposed to prevent vascular complications.

The second element concerns the apheresis itself. In a minor proportion of donors (0.6%–20% depending on the apheresis center), a central venous catheter may be necessary.Citation98 Pain at the site of puncture occurs more frequently in donors with a central (58%) than peripheral vein access (38%).Citation99 The occurrence of acute side effects in large series of BM and PB HSPC harvests in donors has been compared, which showed that peak levels of pain and toxicities were comparable in both harvesting methods.Citation100

The incidence of complications (cancer, autoimmune disease, and thrombosis) was similar after BM and PB HSPC donation (0.99% vs 0.31%, respectively).Citation101 The long-term safety of G-CSF in donors is still being debated. G-CSF induces epigenetic and cytogenetic abnormalities, which persist for several months.Citation102 The donor immune response is disturbed during 6 months, with decrease in T-cell PB counts due to the apheresis, and also in interleukin (IL)-2 and IL-10 production due to G-CSF.Citation103 Long-term medical and biological follow-up are necessary to confirm the safety of G-CSF in donors.

When harvesting PB in patients, the intensity of the main symptoms or complications from G-CSF stimulation and apheresis can be confused with the symptoms from the chemotherapy-induced aplasia. Plerixafor can induce transient side effects (diarrhea, nausea, injection site erythema, headache, paresthesia). The long-term side effects of plerixafor should be studied in particular, if this treatment is to be used in routine or in donors.

Cell composition of graft and influence on HSPC transplantations

Clinical consequence of BM graft composition

BM contains HSPCs that are the main cell population to be transplanted, but other cells must be considered for hematopoietic transplantations.

In harvested BM, stromal cells and the marrow microenvironment are associated with HSPCs, and both are important to reconstitute BM in nononcological hematological diseases and diseases with marrow abnormalities. When the main aim is to obtain a graft-versus-leukemia (GVL) effect, PB and not BM HSPCs are preferentially harvested. The recent development of haplo-identical transplantations followed by posttransplantation cyclosphamide injection has reintroduced the interest for using BM.Citation104 BM was also used after solid-organ or composite-tissue transplantation to induce immune tolerance.Citation105

Clinical consequence of CBU composition

The use of CBUs for allogeneic HSPC transplantation is associated with lower incidence of graft-versus-host-disease with slow immune and hematological recovery, inducing high incidence of opportunistic or severe viral infections. The transplantations of two CBUs decrease the incidence of these complications.Citation15 The numbers of HSPCs transplanted are associated with the outcome. The relation between lymphocyte composition in transplanted CB and outcome after transplantation is currently being studied.Citation106

Clinical consequence of PB graft composition

During allogeneic transplantation, a PB graft is harvested when a GVL effect is aimed. Indeed, during apheresis, both HSPCs and lymphocytes (which participate in the GVL effect) are harvested in the same layer. This feature is used when harvesting allogeneic donor lymphocytes for infusion to reinforce the GVL effect. This principle has been developed in reduced intensity conditioning of allogeneic transplantations.

The PB autologous HSPC harvesting process is performed either in first-line or first-relapse poor-prognosis NHL, in HD, and in first-line treatment for MM or some solid tumors (neuroblastoma or brain tumors). The number of HSPCs transplanted is associated with a better outcome in patients treated for NHL and MM.Citation107,Citation108 In addition to HSPCs, the PB harvests contain significant numbers of lymphocytes that influence the immune reconstitution after transplantation. The numbers of T and NK cells are higher in grafts after mobilization with plerixafor than after standard mobilization.Citation109 Studies should be carried out to determine whether autologous grafts mobilized with plerixafor are associated with a better outcome or to changes in immune reconstitution.

Conclusion

HSPC transplantations have become a referent treatment of severe hematological diseases and gives opportunities to obtain a long-term remission. The number of HSPCs infused during the transplantation is predictive of a better outcome in most types of diseases and hematopoietic transplantations. Therefore, as a first and main step in transplantations, the harvest must contain enough autologous or allogeneic HSPCs. Besides standard HSPC harvesting protocols, harvesting methods and techniques have been improved over the years. Studies also brought better knowledge of the factors associated with better harvests.

When considering BM transplantations with improved harvesting techniques, the latest studies have focused on the evaluation of the complications and quality of life of donors. On the other side, properties of stromal cells and their interaction with HSPCs have been characterized. Such knowledge has generated new indications for using BM transplantation to induce immune tolerance. The characterization of the interactions between HSPCs and stromal cells has led to target the pathways involved in HSPC mobilization from BM to blood. Treatments have been developed to increase mobilization targeting these interactions. Plerixafor is the prototype of these new mobilizing agents. In the next few years, other new mobilizing agents targeting such interactions will be developed, but their cost will require the development of algorithms to use them.

The development of new mobilization agents has enabled teams to harvest autologous HSPCs in almost all patients. Autologous transplantation is therefore now possible for a majority of patients, inducing very low failure of HSPC harvests. Further studies should show whether such harvesting possibilities can change the prognosis of PM patients who need an autologous HSPC transplantation. The development of CBU transplantation and banks should improve the chances of performing successful allogeneic HSPC transplantations in patients without (HLA-) related or unrelated donors.

In conclusion, to increase the success of HSPC transplantations, it is necessary to continue improving the harvesting techniques, numbers of HSPCs, and clinical consequences in patients or donors, which emphasizes the major role of the teams performing HSPC harvesting. Indeed, this purpose can be achieved with further studies based on their work and carried out on a regular basis.

Disclosure

The authors report no conflicts of interest in this work.

References

  • ThomasEDLochteHLJrCannonJHSahlerODFerrebeeJWSupralethal whole body irradiation and isologous marrow in manJ Clin Invest1959381709171613837954
  • ArmitageJOBone marrow transplantationN Eng J Med199433012827838
  • McCredieKBHershEMFreirichEJCells capable of colony formation in the peripheral blood of manScience197117139682932945538844
  • LuLXiaoMShenRNGribgsbySBroxmeyerHEEnrichment, characterization, and responsiveness of single primitive cord blood hematopoietic progenitors with high proliferative and replating potentialBlood199381141487678069
  • GluckmanEBroxmeyerHAuerbachAHematopoietic reconstitution in a patient with Fanconi’s anemia by means of umbilical-cord blood from an HLA-identical siblingN Engl J Med198932117117411782571931
  • RochaVLabopinMGluckmanEAcute Leukemia Working Party of the European Blood and Marrow Transplant RegistryRelevance of bone marrow cell dose on allogeneic transplantation outcomes for patients with acute myeloid leukemia in first complete remission: results of a European surveyJ Clin Oncol200220214324433012409331
  • IsolaLMSciglianoESkerrettDA pilot study of allogeneic bone marrow transplantation using related donors stimulated with G-CSFBone Marrow Transplant19972012103310379466275
  • KaoRHLiCCShawCKCorrelation between characteristics of unrelated bone marrow donor and cell density of total nucleated cell in bone marrow harvestInt J Hematol200989222723019130172
  • BacigalupoATongJPodestaMBone marrow harvest for marrow transplantation: effect of multiple small (2 mL) or large (20 mL) aspiratesBone Marrow Transplant1992964674701628131
  • WangTFChuSCChenSHThe effect of different harvest strategies on the nucleated cell yields of bone marrow collectionBiol Blood Marrow Transplant201117335135520553925
  • TanikawaSSakamakiHMoriSRelationship between the presence of side-holes in bone marrow aspiration needle and the number of harvested bone marrow mononuclear cellsRinsho Ketsueki19973812124912539455143
  • BouwmeesterWFechterMMHeymansMWTwiskJWEbelingLJBrandAPrediction of nucleated cells in bone marrow stem cell products by donor characteristics: a retrospective single centre analysisVox Sang2010983276283
  • BroxmeyerHEDouglasGWHangocGHuman umbilical cord blood as a potential source of transplantable hematopoietic stem/progenitor cellsProc Natl Acad Sci U S A19898610382838322566997
  • GluckmanEEuropean organization for cord blood bankingBlood Cells1994242–36016087749125
  • BarkerJNWeisdorfDJDeForTETransplantation of 2 partially HLA-matched umbilical cord blood units to enhance engraftment in adults with hematologic malignancyBlood200510531343134715466923
  • ArmsonBAMaternal/fetal medicine committee, society of obstetricians and gynaecologists of Canada. Umbilical cord blood banking: implications for perinatal care providersJ Obstet Gynaecol Can200527326329015943003
  • LauberSLattaMKlüterHMüller-SteinhardtMThe Mannheim Cord Blood Bank: experiences and perspectives for the futureTransfus Med Hemother2010372909720737051
  • SolvesPMoragaRSaucedoEComparison between two strategies for umbilical cord blood collectionBone Marrow Transplant200331426927312621461
  • AroviitaPTeramoKWestmanPHiilesmaaVKekomäkiRAssociations among nucleated cell, CD34+ cell and colony-forming cell contents in cord blood units through a standardized banking processVox Sang200384321922712670371
  • PageKMMendizabalABetz-StableinBOptimizing donor selection for public cord blood banking: influence of maternal, infant, and collection characteristics on cord blood unit qualityTransfusion201454234035223711284
  • KeersmaekersCLMasonBAKeersmaekersJPonziniMMlyanarekRAFactors affecting umbilical cord blood stem cell suitability for transplantation in an in utero collection programTransfusion201454354554923869580
  • FasouliotisSJSchenkerMDHuman umbilical cord blood banking and transplantation: a state of the artEur J Obstet Gynecol Reprod Biol2000901132510767505
  • BertoliniFLazzariLLauriEA comparative study of different procedures for the collection and banking of umbilical cord bloodJ Hematother19954129367757396
  • ElchalalUFasouliotisSJShtockheimDPostpartum umbilical cord blood collection for transplantation: a comparison of three methodsAm J Obstet Gynecol2000182122723210649183
  • BelvedereOFeruglioCMalangoneWIncreased blood volume and CD34+CD38− cell recovery using a novel umbilical cord blood collection systemStem Cells200018424525110924090
  • BornsteinRFloresAIMontalbánMADel ReyDde la SernaJGilsanzFA modified cord blood collection method achieves sufficient cell levels for transplantation in most adult patientsStem Cells200523332433415749927
  • TakebeNGageFChengXLauwMISPreliminary findings on the use of a pulsatile machine reperfusion of a placenta to improve the cord blood collection yield including primitive hematopoietic stem cell fractionsTransfusion20094991911191619497058
  • TanKKTangKZHuangSEx utero harvest of hematopoietic stem cells from placenta/umbilical cord with an automated collection systemIEEE Trans Biomed Eng20095692331233419497809
  • SurbekDVSchönfeldBTichelliAGratwohlAHolzgreveWOptimizing cord blood mononuclear cell yield: a randomized comparison of collection before vs after placenta deliveryBone Marrow Transplant19982233113129720751
  • LaskyLCLaneTAMillerJPIn utero or ex utero cord blood collection: which is better?Transfusion200242101261126712423508
  • WongAYuenPMLiKTsoiWCCord blood collection before and after placental delivery: levels of nucleated cells, haematopoietic progenitor cells, leukocyte subpopulations and macroscopic clotsBone Marrow Transplant200127213313811281381
  • OmoriAManabeMKudoKTanakaKTakahashiKKashiwakuraIInfluence of obstetric factors on the yield of mononuclear cells, CD34+ cell count and volume of placental/umbilical cord bloodJ Obstet Gynaecol Res2010361525720178527
  • CairoMSWagnerELFraserJCharacterization of banked umbilical cord blood hematopoietic progenitor cells and lymphocyte subsets and correlation with ethnicity, birth weight, sex, and type of delivery: a Cord Blood Transplantation (COBLT) Study reportTransfusion200545685686615934982
  • JanRHWenSHShyrMHChiangBLImpact of maternal and neonatal factors on CD34+ cell count, total nucleated cells, and volume of cord bloodPediatr Transplant200812886887318643913
  • CobellisLCastaldiMATrabuccoECord blood unit bankability can be predicted by prenatal sonographic parametersEur J Obstet Gynecol Reprod Biol2013170239139523958574
  • LiuJHeJChenSCord blood banking and transplantation in China: a ten years experience of a single public bankTransfus Med Hemother2012391232722896763
  • JawdatDArabSThaheryHAlmashaqbehWAlaskarAHajeerAHImproving cord blood unit quantity and quality at King Abdullah International Medical Research Center Cord Blood BankTransfusion Epub692014
  • ClarkPTrickettAStarkDVowelsMFactors affecting microbial contamination rate of cord blood collected for transplantationTransfusion20125281770177722211719
  • WarkentinPIFoundation for the accreditation of cellular therapy. Voluntary accreditation of cellular therapies: foundation for the accreditation of cellular therapy (FACT)Cytotherapy20035429930512944235
  • Anguita-CompagnonATDibarrartMTPalmaJMobilization and collection of peripheral blood stem cells: guidelines for blood volume to process, based on CD34-positive blood cell count in adults and childrenTransplant Proc201042133934420172346
  • ChenJBurnsKMBabicADonor body mass index is an important factor that affects peripheral blood progenitor cell yield in healthy donors after mobilization with granulocyte-colony-stimulating factorTransfusion201454120321023763340
  • BertaniGSantoleriLMartinoMIdentification of hematopoietic progenitor cell donor characteristics predicting successful mobilization results in an Italian multicenter studyTransfusion20145482028203324588265
  • Al-AliHKBourgeoisMKrahlRThe impact of the age of HLA-identical siblings on mobilization and collection of PBSCs for allogeneic hematopoietic cell transplantationBone Marrow Transplant201146101296130221132022
  • BillenAMadrigalJASzydloRMShawBEFemale donors and donors who are lighter than their recipient are less likely to meet the CD34+ cell dose requested for peripheral blood stem cell transplantationTransfusion Epub5272014
  • HaugeAWHaastrupEKSengeløvHMinulescuLDickmeissEFischer-NielsenAAddition of plerixafor for CD34+ cell mobilization in six healthy stem cell donors ensured satisfactory grafts for transplantationTransfusion20145441055105823944772
  • de LatourRPChaouiDBourhisJHMobilization of peripheral blood progenitor cells after DHAP regimen with or without rituximab: a large multicenter comparative study in patients with malignant lymphomaLeuk Lymphoma200748589790417487733
  • EndoTSatoNMogiYPeripheral blood stem cell mobilization following CHOP plus rituximab therapy combined with G-CSF in patients with B-cell non-Hodgkin’s lymphomaBone Marrow Transplant200433770370714743197
  • CesaroSTintoriVNesiFA prospective study on the efficacy of mobilization of autologous peripheral stem cells in pediatric oncohematology patientsTransfusion20135371501150923034006
  • SheppardDBredesonCAllanDTayJSystematic review of randomized controlled trials of hematopoietic stem cell mobilization strategies for autologous transplantation for hematological malignanciesBiol Blood Marrow Transplant20121881191120322261379
  • JungSHParkHAhnJSEfficacy of stem cell mobilization in patients with newly diagnosed multiple myeloma after a CTD (cyclophosphamide, thalidomide, and dexamethasone) regimenInt J Hematol2013971929723233155
  • BhutaniDZonderJValentJEvaluating the effects of lenalidomide induction therapy on peripheral stem cells collection in patients undergoing autologous stem cell transplant for multiple myelomaSupport Care Cancer20132192437244223591714
  • MartínezESuredaADalmasesCDMobilization of peripheral blood progenitor cells by cyclophosphamide and rhGM-CSF in multiple myelomaBone Marrow Transplant1996181178831988
  • PublicoverARichardsonDSDaviesAUse of biosimilar granulocyte colony-stimulating factor for peripheral blood stem cell mobilization: an analysis of mobilization and engraftmentBr J Haematol2013162110711123614650
  • WeisdorfDMillerJVerfaillieCCytokine-primed bone marrow stem cells vs peripheral blood stem cells for autologous transplantation: a randomized comparison of GM-CSF vs G-CSFBiol Blood Marrow Transplant1997342172239360784
  • HerbertKEGambellPLinkEKPegfilgrastim compared with filgrastim for cytokine-alone mobilization of autologous haematopoietic stem and progenitor cellsBone Marrow Transplant201348335135622858510
  • BensingerWAppelbaumFRowleySFactors that influence collection and engraftment of autologous peripheral-blood stem cellsJ Clin Oncol19951310254725557595706
  • WeaverCHSchwartzbergLSBirchRCollection of peripheral blood cells after the administration of cyclophosphamide, etoposide, and granulocyte-colony-stimulating factor: an analysis of 497 patientsTransfusion19973798969039308634
  • KettererNSallesGMoulletIFactors associated with successful mobilization of peripheral blood progenitor in 200 patients with lymphoid malignanciesBr J Haematol199810312352429792315
  • MendroneAArraisCASaboyaRChamoneDAFDulleyFLFactors affecting hematopoietic progenitor cell mobilization: an analysis of 307 patientsTransfus Apher Sci200839318719219036640
  • KoenigsmannMJentsch-UllrichKMohrenMBeckerEHeimMFrankeAThe role of diagnosis in patients failing peripheral blood progenitor cell mobilizationTransfusion200444577778415104662
  • LacativaCPLacativaPGGarnicaMRisk factors for unsuccessful peripheral blood stem cell harvesting using granulocyte-colony stimulating factor mobilization in patients with multiple myelomaTransfus Apher Sci201247333133522874435
  • WatermanJRybickiLBolwellBFludarabine as a risk factor for poor stem cell harvest, treatment-related MDS and AML in follicular lymphoma patients after autologous hematopoietic cell transplantationBone Marrow Transplant201247448849321572461
  • MoreauPHulinCMaritGStem cell collection in patients with de novo multiple myeloma treated with the combination of bortezomib and dexamethasone before autologous stem cell transplantation according to IFM 2005-01 trialLeukemia20102461233123520428201
  • CavalloFBringhenSMiloneGStem cell mobilization in patients with newly diagnosed multiple myeloma lenalidomide induction therapyLeukemia201125101627163121637283
  • LiJHamiltonEVaughnLEffectiveness and cost analysis of “just in time” salvage plerixaflor administration in autologous transplant patients with poor stem cell mobilization kineticsTransfusion201151102175218221492180
  • AttolicoIPavoneVOstuniAPlerixafor added to chemotherapy plus G-CSF is safe and allows adequate PBSC collection in predicted poor mobilizer patients with multiple myeloma or lymphomaBiol Blood Marrow Transplant201218224124921791194
  • OlivieriAMarchettiMLemoliRProposed definition of ‘poor mobilizer’ in lymphoma and multiple myeloma: an analytic hierarchy process by ad hoc working group Gruppo ItalianoTrapianto di Midollo OsseoBone Marrow Transplant201247334235121625224
  • PerseghinPTerruzziEDassiMManagement of poor peripheral blood stem cell mobilization: incidence, predictive factors, alternative strategies and outcome. A retrospective analysis on 2177 patients from three major Italian institutionsTransfus Apher Sci2009411333719540167
  • WuchterPRanDBrucknerTPoor mobilization of hematopoieic stem cells–definition, incidence, risk factors, and impact on outcome of autologous transplantationBiol Blood Marrow Transplant201016449049919925876
  • WattsMJSullivanAMLeverettDBack-up bone marrow is frequently ineffective in patients with poor peripheral-blood stem-cell mobilizationJ Clin Oncol1998164155415609552065
  • ReiserMJostingADraubeASuccessful peripheral blood stem cell mobilization with etoposide (VP-16) in patients with relapsed or resistant lymphoma who failed cyclophosphamide mobilizationBone Marrow Transplant199923121223122810414907
  • LieAKHuiCHRawlingTGranulocyte colony-stimulating factor (G-CSF) dose-dependent efficacy in peripheral blood stem cell mobilization in patients who had failed initial mobilization with chemotherapy and G-CSFBone Marrow Transplant19982298538579827812
  • Pérez-DueñasBAlcortaIEstellaJRivesSTollTTusetESafety and efficacy of high-dose G-CSF (24 microg/kg) alone for PBSC mobilization in childrenBone Marrow Transplant2002301298798812476297
  • FaconTHarousseauJLMaloiselFStem cell factor in combination with filgrastim after chemotherapy improves peripheral blood progenitor cell yield and reduces apheresis requirements in multiple myeloma patients: a randomized controlled trialBlood19999441218122510438709
  • LapierreVRossiJFHeshmatiFAncestim (r-metHuSCF) plus filgrastim and/or chemotherapy for mobilization of blood progenitors in 513 poorly mobilizing cancer patients: the French compassionate experienceBone Marrow Transplant201146793694220956952
  • ShaughnessyPUbertiJDevineSPlerixafor and G-CSF for autologous stem cell mobilization in patients with NHL, Hodgkin’s lymphoma and multiple myeloma: results from the expanded access programBone Marrow Transplant201348677778123178544
  • HerbertKEDemosthenousLWiesnerGPlerixafor plus pegfilgrastim is a safe, effective mobilization regimen for poor or adequate mobilizers of hematopoietic stem and progenitor cells: a phase I clinical trialBone Marrow Transplant20144981056106224887382
  • CostaLJAlexanderETHoganKRSchaubCFoutsTVStuartRKDevelopment and validation of a decision-making algorithm to guide the use of plerixafor for autologous hematopoietic stem cell mobilizationBone Marrow Transplant2011461646920383210
  • SchotsRVan RietIDamiaensSThe absolute number of circulating CD34+ cells predicts the number of haematopoietic stem cells that can be collected by apheresisBone Marrow Transplant19961745095158722347
  • LaneTABasheyACarrierEImproving the efficiency of PBPC collection by pre-apheresis peripheral blood and mid-apheresis product measurements of CD34 cellsCytotherapy20046431832716146884
  • MenichellaGLaiMSerafiniRLarge volume leukapheresis for collecting hemopoietic progenitors: role of CD 34+ precount in predicting successful collectionInt J Artif Organs199922533434110467933
  • WuFYHengKKSallehRBComparing peripheral blood stem cell collection using the COBE Spectra, Haemonetics MCS+, and Baxter AmicusTransfus Apher Sci201247334535023032067
  • IkedaKOhtoHNemotoKCollection of MNCs and progenitor cells by two separators for PBPC transplantation: a randomized crossover trialTransfusion200343681481912757534
  • SchreinerTWiesnethMKrugECollection of allogeneic peripheral blood progenitor cells by two protocols on an apheresis systemTransfusion19983811–12105110559838937
  • AltuntasFKocyigitIOzturkAComparison of the Fenwal Amicus and Fresenius Com.Tec cell separators for autogous peripheral blood progenitor cell collectionTransfus Apher Sci200736215916717369096
  • ReinhardtPBrauningerSBialleckHAutomatic inter-face controlled apheresis collection of stem/progenitor cells: results from an autologous donor validation trial of a novel stem cell apheresis deviceTransfusion20115161321133021155834
  • BrauningerSBialleckHThorauschKFeltTSeifriedEBonigHAllogeneic donor peripheral blood “stem cell” apheresis: prospective comparison of two apheresis systemsTransfusion20125251137114522044384
  • FlommersfeldSBakchoulTBeinGWachtelALoecheltCSachsUJA single center comparison between three different apheresis systems for autologous and allogeneic stem cell collectionsTransfus Apher Sci201349342843323827328
  • IkedaKOhtoHKannoTPeripheral blood cell progenitor cell collection by two programs for autologous and allogeneic transplantationTransfusion20145451235124224117442
  • CherqaouiBRouelNAuvrignonAPeripheral blood stem collection in low-weight children: retrospective comparison of two apheresis devicesTransfusion20145451371137824117598
  • AndreolaGVanazziARadiceDWho should be really considered as a poor mobilizer in the plerixafor era?Transfus Apher Sci2012471273222480954
  • BojanicIDubravcicKBatinicDLarge volume leukapheresis: efficacy and safety of processing patient’s total blood volume six timesTransfus Apher Sci201144213914721320801
  • NgHNSimKMBoeySKBone marrow harvesting using EMLA (eutectic mixture of local anaesthesics) cream, local anaesthesia and patient-controlled analgesia with alfentanilBone Marrow Transplant199923994194510338051
  • van WalravenSMStraathofLMSwitzerGEImmediate and long-term somatic effects, and health-related quality of life of BM donation during early childhood. A single-center report in 210 pediatric donorsBone Marrow Transplant2013481404522705802
  • LazzariLCorsiniCCurioniCThe Milan Cord Blood Bank and the Italian Cord Blood NetworkJ Hematother1996521171228723786
  • ChenSHYangSHChuSCThe role of donor characteristics and post-granulocyte colony-stimulating factor white blood cell counts in predicting the adverse events and yields of stem cell mobilizationInt J Hematol201193565265921509437
  • AkyolGPalaCYildirimAA rare but severe complication of filgrastim in a healthy donor: splenic ruptureTransfus Apher Sci2014501535524360843
  • PulsipherMAChitphakdithaiPMillerJPAdverse events among 2408 unrelated donors of peripheral blood stem cells: results of a prospective trial from the National Marrow Donor ProgramBlood2009113153604361119190248
  • HöligKBlechschmidtMKramerMPeripheral blood stem cell collection in allogeneic donors: impact of venous accessTransfusion201252122600260522500612
  • PulsipherMAChitphakdithaiPLoganBRAcute toxicities of unrelated bone marrow versus peripheral blood stem cell donation: results of a prospective trial from the National Marrow Donor ProgramBlood2013121119720623109243
  • PulsipherMAChitphakdithaiPLoganBRLower risk for serious adverse events and no increased risk for cancer after PBSC vs BM donationBlood2014123233655366324735965
  • Marmier-SavetCLarosaFLegrandFG-CSF-induced aneuploidy does not affect CD34+ cells and does not require cell divisionBlood2010115491091120110433
  • Marmier-SavetCLarosaFLegrandFPersistence of lymphocyte function perturbations after granulocyte-colony-stimulating factor mobilization and cytapheresis in normal peripheral blood stem cell donorsTransfusion201050122676268520663114
  • RaiolaAMDominiettoAdi GraziaCUnmanipulated haploidentical transplants compared with other alternative donors and matched sibling graftsBiol Blood Marrow Transplant201420101573157924910379
  • KawaiTSachsDHSprangersBLong-term results in recipients of combined HLA-mismatched kidney and bone marrow transplantation without maintenance immunosuppressionAm J Transplant20141471599161124903438
  • WikellHPonandai-SrinivasanSMattssonJGertowJUhlinMCord blood graft composition impacts the clinical outcome of allogeneic stem cell transplantationTranspl Infect Dis201416220321224456214
  • GordanLNSugrueMWLynchJWPoor mobilization of peripheral blood stem cells is a risk factor for worse outcome in lymphoma patients undergoing autologous stem cell transplantationLeuk Lymphoma200344581582012802919
  • WahlinAErikssonMHultdinMRelation between harvest success and outcome after autologous peripheral blood stem cell transplantation in multiple myelomaEur J Haematol200473426326815347313
  • VarmavuoVMäntymaaPNousiainenTValonenPKuittinenTJantunenEBlood graft composition after plerixafor injection in patients with NHLEur J Haematol201289212813522536949