1,294
Views
0
CrossRef citations to date
0
Altmetric
Hematology

G-CSF + plerixafor versus G-CSF alone mobilized hematopoietic stem cells in patients with multiple myeloma and lymphoma: a systematic review and meta-analysis

, , &
Article: 2329140 | Received 06 Aug 2023, Accepted 27 Feb 2024, Published online: 12 Mar 2024

Abstract

Aim

The combination of granulocyte-colony stimulating factor (G-CSF) and plerixafor is one of the approaches for hematopoietic stem cell mobilization in patients with multiple myeloma (MM), non-Hodgkin’s lymphoma (NHL), and Hodgkin’s lymphoma (HL). This systematic review and meta-analysis aimed to determine the ability of G-CSF + plerixafor to mobilize peripheral blood (PB) CD34+ cells and examine its safety profile.

Methods

We performed a database search using the terms ‘granulocyte colony stimulating factor’, ‘G-CSF’, ‘AMD3100’, and ‘plerixafor’, published up to May 1, 2023. The methodology is described in further detail in the PROSPERO database (CRD42023425760).

Results

Twenty-three studies were included in this systematic review and meta-analysis. G-CSF + plerixafor resulted in more patients achieving the predetermined apheresis yield of CD34+ cells than G-CSF alone (OR, 5.33; 95%, 4.34–6.55). It was further discovered that G-CSF + plerixafor could mobilize more CD34+ cells into PB, which was beneficial for the next transplantation in both randomized controlled (MD, 18.30; 95%, 8.74–27.85) and single-arm (MD, 20.67; 95%, 14.34–27.00) trials. Furthermore, G-CSF + plerixafor did not cause more treatment emergent adverse events than G-CSF alone (OR, 1.25; 95%, 0.87–1.80).

Conclusions

This study suggests that the combination of G-CSF and plerixafor, resulted in more patients with MM, NHL, and HL, achieving the predetermined apheresis yield of CD34+ cells, which is related to the more effective mobilization of CD34+ cells into PB.

1. Introduction

Multiple myeloma (MM), non-Hodgkin’s lymphoma (NHL), and Hodgkin’s lymphoma (HL) are common hematological malignancies, which often lead to complications and a decreased quality of life for patients [Citation1–3]. To treat the myelosuppression caused by these disorders, autologous hematopoietic stem cell transplantation (AHSCT) is recommended to restore bone marrow and hematopoietic functions after high-dose chemotherapy, as well as to generate immunity [Citation4–6]. Compared to conventional chemotherapy, AHSCT improves patient remission and overall survival [Citation7, Citation8]. Clinically, isolating sufficient CD34+ cells from peripheral blood (PB) for transplantation is necessary, among which hematopoietic stem cells (HSCs) account for more than 90% of the CD34+ cells [Citation9].

In clinical practice, PB CD34+ cells mobilized by Granulocyte colony-stimulating factor (G-CSF) are often used for AHSCT, revolutionizing stem cell transplantation [Citation10, Citation11]. Different forms of recombinant human G-CSF, such as filgrastim, lenograstim, and pegfilgrastim, are widely used in clinical practice, reducing febrile neutropenia and other adverse reactions [Citation12]. After the patent period expired, some biosimilars have been developed, but their efficacy and safety still need to be further verified [Citation10]. However, the administration of G-CSF alone may lead to insufficient collection of CD34+ cells, causing mobilization failure. The risk factors for mobilization failure include age of the patient, the presence of T-cell and NK/T-cell lymphoma, and high-dose cyclophosphamide treatment [Citation13]. In recent years, plerixafor, a CXCR4 receptor-specific inhibitor, has been proposed for administration in combination with G-CSF for HSC mobilization, and that this combination resulted in a higher collection of CD34+ cells in patients with MM, NHL, and HL compared to G-CSF alone [Citation14–16]. Studies have shown that G-CSF + plerixafor cannot achieve the desired therapeutic effect in patients who have already undergone mobilization without success using G-CSF alone [Citation17–19]. Therefore, whether G-CSF + plerixafor has clinical value for mobilizing CD34+ cells in patients with MM, NHL, and HL and for patients with mobilization failure warrants further investigation.

Accordingly, we conducted a systematic review and meta-analysis to evaluate the clinical efficacy and safety of G-CSF + plerixafor for CD34+ cell mobilization in patients with MM, NHL, and HL.

2. Methods

2.1. Search strategy and selection criteria

The study protocol followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses-Diagnostic Test Accuracy (PRISMA-DTA) statement and was registered in the PROSPERO database (ID: CRD42023425760) in May 2023. We searched the database using the following term: ‘plerixafor’, ‘AMD3100’, ‘G-CSF’, and ‘granulocyte colony-stimulating factor’. PubMed, Embase, Cochrane, and Web of Science databases were searched for potentially relevant studies in English-language articles (up to May 1, 2023). References to the identified articles were also searched to supplement the data sources.

2.2. Study selection

Titles and abstracts were independently screened by two reviewers after removing duplicate studies retrieved from the four databases and other sources [Citation20]. Disagreements were resolved through consultation, and third-party opinions were consulted when necessary. Studies that met the following criteria were included in the meta-analysis: (1) Full-text studies published in peer-reviewed journals. (2) Patients who were diagnosed with MM, NHL, or HL; and (3) received G-CSF + plerixafor to mobilize CD34+ cells. (4) Patients who also had at least one of the following indicators: those who achieved the predetermined apheresis CD34+ cell yield (cells × 106/kg) and those with available CD34+ cell counts (cells/μL). The exclusion criteria were as follows: (1) reviews, abstracts, comments, and case reports; (2) non-English language publications; and (3) studies consisting of less than five patients.

2.3. Risk of bias assessment

Two reviewers independently assessed the risk of bias in eligible studies using the Cochrane ‘risk-of-bias’ assessment tool.

2.4. Data extraction and statistical analysis

Two reviewers independently extracted data from eligible studies, including the following: author, year, trial ID, patients (age and disease), administration (G-CSF and plerixafor), predetermined apheresis yield of CD34+ cells, confounding variables, and previous treatment of the patients. Data analysis was performed using the RevMan software. A meta-analysis was carried out on the data about the administration of ‘G-CSF + plerixafor’ versus ‘G-CSF alone’ or before and after the addition of plerixafor: (1) number of patients who achieved the predetermined apheresis yield of CD34+ cell count (cells × 106/kg), (2) CD34+ cell count (cells/μL), and (3) number of adverse events. Subgroup analysis was performed according to the disease type. An I2 > 50% was considered inappropriate heterogeneity, in which case the random-effects model was used. Statistical significance was set at p < 0.05.

3. Results

3.1. Search results

A total of 4,363 records were retrieved from the four databases according to the search terms, and two records were retrieved from other sources. Duplicate records were excluded, and a total of 23 articles were finally included in this systematic review and meta-analysis after applying the inclusion and exclusion criteria () [Citation17–19, Citation21–40].

Figure 1. Search strategy and study selection.

Figure 1. Search strategy and study selection.

3.2. Studies characterisations

lists the main characteristics of the 23 included articles. The articles were published between 2008 and 2023, were single- or multicenter, prospective, or retrospective studies, and were listed with clinical trial IDs. All patients were adults with MM, NHL, or HL. The dose of G-CSF was mainly 10 μg/kg, and the dose of plerixafor was mainly 0.24 mg/kg. The predetermined apheresis yield of CD34+ cells was between 2 × 106 and 6 × 106 cells/kg. In addition, Supplementary Table S1 lists the confounding variables and previous treatment of the patients.

Table 1. Main characteristics of included studies.

3.3. Risk of bias in studies

We assessed the risk of bias in the included articles (), and the results are summarized in Supplementary Figure S1. ‘Blinding of patients and personnel’ and ‘selective reporting’ of eight articles [Citation17–19, Citation21–39] were assessed as low risk. In ‘random sequence generation’, bias in ten articles [Citation17–19, Citation23, Citation24, Citation28, Citation35, Citation38–40] was assessed as unclear risk. In ‘allocation concealment’, bias in five articles [Citation17–19, Citation28, Citation35, Citation38] was assessed as unclear risk. In ‘blinding of outcome assessment’, bias in one article [Citation33] was assessed as unclear risk. In ‘incomplete outcome data’, bias in four articles [Citation17, Citation19, Citation30, Citation39] was assessed as unclear risk. Among ‘other biases’, bias in two articles [Citation31, Citation40] was assessed as high risk, and bias in six articles [Citation17, Citation18, Citation26, Citation29, Citation32, Citation39] was assessed as unclear risk. In general, the risk of bias was low.

Figure 2. Risk of bias graph in the included studies.

Figure 2. Risk of bias graph in the included studies.

3.4. G-CSF + plerixafor enabled more patients to achieve the predetermined apheresis yield of CD34+ cells

For patients requiring AHSCT, the clinical efficacy of G-CSF + plerixafor was evaluated based on the proportion of patients who achieved the predetermined apheresis yield of CD34+ cells. In twelve articles, 923 patients in the G-CSF + plerixafor group and 1214 patients in the G-CSF group were included in the meta-analysis [Citation22, Citation26–33, Citation36, Citation37, Citation40]. Overall, more patients in the G-CSF + plerixafor group (64.9%, 599/923) achieved the predetermined apheresis yield of CD34+ cells compared to G-CSF alone (33.8%, 410/1214) (OR, 5.33; 95% CI, 4.34–6.55; p < 0.00001) (). Subgroup analysis demonstrated significantly higher odds ratios for achieving the predetermined apheresis yield of CD34+ cells in the MM (OR, 4.18; 95% CI, 3.04–5.74; p < 0.00001), NHL (OR, 6.38; 95% CI, 4.73–8.60; p < 0.00001), HL (OR, 11.86; 95% CI, 4.14–33.96; p < 0.00001), and (MM, NHL, and HL) groups (OR, 4.58; 95% CI, 2.15–9.74; p < 0.0001) when administering G-CSF + plerixafor compared to G-CSF alone. The above results suggest that the addition of plerixafor could enhance the activity of G-CSF to isolate more CD34+ cells, which is conducive to the successful AHSCT of patients.

Figure 3. Meta-analysis of the proportion of patients achieving the predetermined apheresis yield of CD34+ cells (cells × 106/kg) in the G-CSF + plerixafor group versus the G-CSF group. HL, Hodgkin’s lymphoma; MM, multiple myeloma; NHL, non-Hodgkin’s lymphoma; PB, peripheral blood.

Figure 3. Meta-analysis of the proportion of patients achieving the predetermined apheresis yield of CD34+ cells (cells × 106/kg) in the G-CSF + plerixafor group versus the G-CSF group. HL, Hodgkin’s lymphoma; MM, multiple myeloma; NHL, non-Hodgkin’s lymphoma; PB, peripheral blood.

3.5. G-CSF + plerixafor was not effective in increasing CD34+ cells in patients who had failed to mobilize with G-CSF alone

Owing to its high price and other factors, plerixafor is not frequently used as a primary therapy, but is often used as a salvage therapy (also known as ‘just in time’) after mobilization failure using G-CSF alone [Citation41]. In five articles, 396 patients in the G-CSF + plerixafor group and 328 patients in the G-CSF group were included in the meta-analysis [Citation17–19, Citation22, Citation35, Citation41]. Overall, even in patients whose mobilization failed using G-CSF alone (89.9%, 295/328), salvage addition of plerixafor (57.1%, 226/396) resulted in a lower population ratio of achieved predetermined apheresis CD34+ cells than in patients who successfully mobilized with G-CSF alone (OR, 0.14; 95% CI, 0.06–0.29; p < 0.00001) (Supplementary Figure S2). Subgroup analysis showed that in both the MM (OR, 0.16; 95% CI, 0.04–0.68; p = 0.001) and (NHL and HL) groups (OR, 0.09; 95% CI, 0.03–0.29; p < 0.0001), the number of people who achieved the predetermined apheresis yield of CD34+ cells was still lower after adding plerixafor to the mobilization of those who failed compared to the mobilization of those who succeeded with G-CSF alone. In the NHL group, adding plerixafor could improve mobilization (OR, 0.29; 95% CI, 0.08–1.13; p = 0.07); however, only one study investigated this. These results suggest that the salvage addition of plerixafor does not contribute to achieving the predetermined apheresis CD34+ cell count in patients who had already failed to mobilize with G-CSF alone.

3.6. G-CSF + plerixafor increased the PB CD34+ cell count

We further investigated the enhancing effect of plerixafor on the proliferation of PB CD34+ cells regardless of patient mobilization failure. In four articles, 193 patients in the G-CSF + plerixafor group and 214 patients in the G-CSF group were included in the meta-analysis [Citation26, Citation27, Citation32, Citation33]. Overall, we found that the PB CD34+ cell count was significantly higher in the G-CSF + plerixafor group than in the G-CSF group (MD, 28.11; 95% CI, 14.07–42.14; p = 0.004) (). Subgroup analysis showed that PB CD34+ cells in the MM (MD, 46.17; 95% CI, 19.25–73.09; p = 0.0008), NHL (MD, 31.27; 95% CI, 14.01–48.54; p = 0.0004), and (MM, NHL, and HL) groups (MD, 10.80; 95% CI, 6.51–15.09; p < 0.00001) were higher with G-CSF + plerixafor than with G-CSF alone; however, no difference was observed in the MM group (MD, 22.30; 95% CI, −20.09–64.69; p = 0.30). These results suggest that, compared to G-CSF alone, G-CSF + plerixafor could improve the PB CD34+ cell count.

Figure 4. Meta-analysis of PB CD34+ cell count (cells/μL) between the G-CSF + plerixafor and G-CSF groups. HL, Hodgkin’s lymphoma; MM, multiple myeloma; NHL, non-Hodgkin’s lymphoma; PB, peripheral blood.

Figure 4. Meta-analysis of PB CD34+ cell count (cells/μL) between the G-CSF + plerixafor and G-CSF groups. HL, Hodgkin’s lymphoma; MM, multiple myeloma; NHL, non-Hodgkin’s lymphoma; PB, peripheral blood.

3.7. PB CD34+ cell count was higher after G-CSF combined with plerixafor

To effectively compare the changes in PB CD34+ cell counts before and after the addition of plerixafor, 228 patients (one withdrawn) from eight single-arm studies were included [Citation22–25, Citation28, Citation34, Citation38, Citation39]. In general, the PB CD34+ cell count was significantly higher after G-CSF treatment combined with plerixafor compared to the initial count (MD, 20.67; 95% CI, 14.34–27.00; p < 0.00001) (). Similar results were obtained for the MM (MD, 18.54; 95% CI, 11.24–25.85; p < 0.00001), NHL (MD, 30.79; 95% CI, 14.38–47.21; p = 0.0002), and (MM, NHL, and HL) groups (MD, 21.58; 95% CI, 1.58–41.59; p = 0.03) in subgroup analyses. These results suggest that G-CSF combined with plerixafor increased the PB CD34+ cell count.

Figure 5. Meta-analysis of PB CD34+ cell count (cells/μL) before and after plerixafor administration. PB, peripheral blood. HL, Hodgkin’s lymphoma; MM, multiple myeloma; NHL, non-Hodgkin’s lymphoma.

Figure 5. Meta-analysis of PB CD34+ cell count (cells/μL) before and after plerixafor administration. PB, peripheral blood. HL, Hodgkin’s lymphoma; MM, multiple myeloma; NHL, non-Hodgkin’s lymphoma.

3.8. G-CSF combined with plerixafor did not increase treatment emergent adverse events (TEAEs)

The evaluation of the safety of plerixafor mainly depends on treatment emergent adverse events (TEAEs). In eight articles, 528 patients in the G-CSF + plerixafor group and 458 patients in the G-CSF group were included in the meta-analysis [Citation17, Citation19, Citation21, Citation27, Citation30, Citation32, Citation36, Citation37]. In general, G-CSF + plerixafor (78.4%, 414/528) did not cause more TEAEs than G-CSF alone (80.1%, 367/458) (OR, 1.25; 95% CI, 0.87–1.80; p = 0.23) (). Subgroup analysis found that the addition of plerixafor in the MM (OR, 1.27; 95% CI, 0.74–2.20; p = 0.38), NHL (OR, 1.39; 95% CI, 0.75–2.55; p = 0.29), and (NHL and HL) groups (OR, 1.00; 95% CI, 0.45–2.24; p = 0.99) did not affect the incidence of TEAEs. These results suggest that plerixafor did not cause more TEAEs during the mobilization of CD34+ cells using G-CSF.

Figure 6. Meta-analysis of the incidence of TEAEs between the G-CSF + plerixafor group versus the G-CSF group. HL, Hodgkin’s lymphoma; MM, multiple myeloma; NHL, non-Hodgkin’s lymphoma; TEAEs, treatment emergent adverse events.

Figure 6. Meta-analysis of the incidence of TEAEs between the G-CSF + plerixafor group versus the G-CSF group. HL, Hodgkin’s lymphoma; MM, multiple myeloma; NHL, non-Hodgkin’s lymphoma; TEAEs, treatment emergent adverse events.

4. Discussion

This systematic review and meta-analysis found that G-CSF administered in combination with plerixafor mobilized CD34+ cells more effectively than G-CSF alone in patients with MM, NHL, and HL. G-CSF + plerixafor increased the number of patients reaching the predetermined apheresis yield of CD34+ cells, increased the PB CD34+ cell count, and did not cause additional TEAEs, thus increasing the success rate of AHSCT. In addition, timely plerixafor supplementation also helped to increase PB CD34+ cell count in patients who failed mobilization with G-CSF alone.

For patients with malignant hematological diseases, such as MM, NHL, and HL, AHSCT can potentially prolong survival [Citation42–44]. Whether sufficient HSCs can be mobilized is the key to the success of transplantation. G-CSF is a commonly used pre-transplant to mobilize HSCs because it is safe and produces rapid and sustained engraftment [Citation45–47]. However, some patients cannot be effectively mobilized using G-CSF alone. Recently, plerixafor has been recommended for administration in combination with G-CSF, which has been shown to mobilize HSCs to the PB more efficiently [Citation48, Citation49]. However, due to its high price, plerixafor is rarely used in combination with G-CSF during the first mobilization [Citation50]. Therefore, evaluating whether the combination of plerixafor and G-CSF is more capable of mobilizing HSCs is necessary, along with determining whether it is safe.

The combination of G-CSF and plerixafor demonstrated a higher proportion of patients achieving the predetermined apheresis yield of CD34+ cells compared to G-CSF alone, regardless of whether the patients were refractory, relapsed, or failed to mobilize (). This directly benefits the subsequent AHSCT. In addition, that the predetermined apheresis yield of CD34+ cells was lower in patients with NHL and HL compared to patients with MM was noteworthy, which may be due to the fact that patients with NHL and HL tend to have longer treatment lines, which may affect the effectiveness of mobilization. However, for poor mobilizers using G-CSF alone, isolating the predetermined apheresis yield of CD34+ cells was difficult, even after salvage addition of plerixafor (Supplementary Figure S2). These results suggest that the timely administration of plerixafor can better mobilize CD34+ cells than salvage administration. The failure of salvage administration may be attributed to medical policy. Due to the need to limit medical expenses, plerixafor is often administered to the poorest mobilizers at the highest risk [Citation51]. Many patients with significant mobilization deficits do not achieve the predetermined apheresis yield of CD34+ cells after receiving salvage plerixafor [Citation17–19, Citation23, Citation35]. Notably, the bias variable was that different original studies used different predetermined apheresis CD34+ cell yields. In general, 2 × 106 CD34+ cells/kg is considered the minimum acceptable cell yield, and 4 × 106 CD34+ cells/kg or higher is considered optimal [Citation52]. The predetermined apheresis CD34+ cell yields for MM is generally higher than those for NHL or HL [Citation18, Citation22, Citation31, Citation39].

G-CSF promotes the release of CD34+ cells from the bone marrow niche to the PB by interrupting signaling pathways involved in motility such as CXCL12/CXCR4 [Citation53]. Plerixafor further mobilizes CD34+ cells into the PB, which is conducive for collection [Citation52]. In our study, compared to the G-CSF only group, PB CD34+ cells were increased in the G-CSF + plerixafor group (). After G-CSF treatment alone failed to collect enough PB CD34+ cells, timely plerixafor supplementation contributed to a subsequent increase in PB CD34+ cell numbers (). However, determining the optimal timing of plerixafor supplementation and the mode of administration require further clinical research. Moreover, we found that PB CD34+ cells mobilized in patients with MM were consistently lower than those in patients with NHL regardless of the mobilization protocol, which may be related to the high cytogenetic or molecular genetic risk present in patients with MM [Citation54]. We confirmed from multiple sources that the combination of G-CSF and plerixafor could mobilize PB CD34+ cells better.

The combination of G-CSF and plerixafor was also promising in terms of safety, as it caused no additional TEAEs than G-CSF alone (). Overall, the grades of plerixafor-associated TEAEs were low; no grade 4 or 5 TEAEs were reported. In addition, no deaths were reported. Grades 1 and 2 TEAEs included gastrointestinal disorders and injection site reactions, whereas grade 3 TEAEs included peripheral neuropathy and pleural effusion. In addition, that plerixafor did not cause poor engraftment of HSCs, but rather improved bone marrow regeneration was gratifying. Results indicated that the combined mobilization due to G-CSF and plerixafor resulted in rapid and durable engraftment of HSCs.

However, our systematic review and meta-analysis study still has some limitations. One limitation is that NHL includes a variety of disease entities. However, due to the difficulty in extracting data from the original studies, making a more detailed classification was not possible, which limited the precision of our study.

5. Conclusions

This study found that the combination of G-CSF and plerixafor was more likely to achieve a predetermined apheresis yield of CD34+ cells than G-CSF alone in patients with MM, NHL, and HL. This is mainly related to the fact that plerixafor can mobilize HSCs to the PB more effectively. Early combined administration of plerixafor and G-CSF may be helpful for mobilization, but it is of limited help to poor mobilizers who already received G-CSF alone. Moreover, the combination of G-CSF and plerixafor did not cause severe TEAEs. Therefore, the combination of G-CSF and plerixafor is beneficial to AHSCT.

Authors contributions

YYL, XQ, YPL, and RXZ conceived the study. XQ and RXZ conducted the literature search. YYL, YPL, and RXZ screened the full-text papers and extracted the data. YYL, XQ, and YPL run the analysis. XQ and RXZ drafted the manuscript, and all authors provided input into revisions and approved the final draft for submission.

Ethical approval

This is a review study, and no ethical approval is required.

Abbreviations
G-CSF=

Granulocyte colony-stimulating factor

HSC=

Hematopoietic stem cells

MM=

Multiple myeloma

NHL=

Non-Hodgkin’s lymphoma

HL=

Hodgkin’s lymphoma

PB=

Peripheral blood

AHSCT=

Autologous hematopoietic stem cell transplantation

TEAE=

Treatment emergent adverse events

Supplemental material

Supplemental Material

Download Zip (324.1 KB)

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

Data are available to investigators after request.

Additional information

Funding

The author(s) reported there is no funding associated with the work featured in this article.

References

  • McDuffie HH, Pahwa P, Karunanayake CP, et al. Clustering of cancer among families of cases with hodgkin lymphoma (HL), multiple myeloma (MM), non-Hodgkin’s lymphoma (NHL), soft tissue sarcoma (STS) and control subjects. BMC Cancer. 2009;9(1):1. doi:10.1186/1471-2407-9-70.
  • Zhang L, Ghielmini M, Cheson BD, et al. Pros and cons of rituximab maintenance in follicular lymphoma. Cancer Treat Rev. 2017;58:34–11. doi:10.1016/j.ctrv.2017.05.007.
  • Goodman KA, Riedel E, Serrano V, et al. Long-term effects of high-dose chemotherapy and radiation for relapsed and refractory hodgkin’s lymphoma. J Clin Oncol. 2008;26(32):5240–5247. doi:10.1200/JCO.2007.15.5507.
  • Dimopoulos MA, Richardson PG, Bahlis NJ, et al. Addition of elotuzumab to lenalidomide and dexamethasone for patients with newly diagnosed, transplantation ineligible multiple myeloma (ELOQUENT-1): an open-label, multicentre, randomised, phase 3 trial. Lancet Haematol. 2022;9(6):e403–e414. doi:10.1016/S2352-3026(22)00103-X.
  • Lemieux C, Ahmad I, Bambace NM, et al. Evaluation of the impact of autologous hematopoietic stem cell transplantation on the quality of life of older patients with lymphoma. Biol Blood Marrow Transplant. 2020;26(1):157–161. doi:10.1016/j.bbmt.2019.09.007.
  • Moskowitz CH, Walewski J, Nademanee A, et al. Five-year PFS from the AETHERA trial of brentuximab vedotin for hodgkin lymphoma at high risk of progression or relapse. Blood. 2018;132(25):2639–2642. doi:10.1182/blood-2018-07-861641.
  • Phillips GL, Meisenberg BR, Reece DE, et al. Activity of single-agent melphalan 220-300 mg/m2 with amifostine cytoprotection and autologous hematopoietic stem cell support in non-Hodgkin and hodgkin lymphoma. Bone Marrow Transplant. 2004;33(8):781–787. doi:10.1038/sj.bmt.1704424.
  • Schuler E, Boughoufala S, Rautenberg C, et al. Relapse patterns and treatment strategies in patients receiving allogeneic hematopoietic stem cell transplantation for myeloid malignancies. Ann Hematol. 2019;98(5):1225–1235. doi:10.1007/s00277-019-03670-6.
  • Boitano AE, Wang J, Romeo R, et al. Aryl hydrocarbon receptor antagonists promote the expansion of human hematopoietic stem cells. Science. 2010;329(5997):1345–1348. doi:10.1126/science.1191536.
  • Welte K. G-CSF: filgrastim, lenograstim and biosimilars. Expert Opin Biol Ther. 2014;14(7):983–993. doi:10.1517/14712598.2014.905537.
  • García-García I, Cid J, Carbassé G, et al. Comparison between standard and high dose of G-CSF for mobilization of hematopoietic progenitors cells in patients and healthy donors. Transfus Med Rev. 2022;36(3):159–163. doi:10.1016/j.tmrv.2022.06.004.
  • Kuderer NM, Dale DC, Crawford J, et al. Impact of primary prophylaxis with granulocyte colony-stimulating factor on febrile neutropenia and mortality in adult cancer patients receiving chemotherapy: a systematic review. J Clin Oncol. 2007;25(21):3158–3167. doi:10.1200/JCO.2006.08.8823.
  • Rossi G, Skert C, Morello E, et al. PBSC mobilization in lymphoma patients: analysis of risk factors for collection failure and development of a predictive score based on the kinetics of circulating CD34+ cells and WBC after chemotherapy and G-CSF mobilization. Hematol Oncol. 2015;33(3):125–132. doi:10.1002/hon.2148.
  • Shah H, Kim S, Singh P, et al. Clinical outcomes of multiple myeloma patients who undergo autologous hematopoietic stem cell transplant with G-CSF or G-CSF and plerixafor mobilized grafts. Am J Hematol. 2020;95(2):198–204. doi:10.1002/ajh.25672.
  • Keating GM. Plerixafor: a review of its use in stem-cell mobilization in patients with lymphoma or multiple myeloma. Drugs. 2011;71(12):1623–1647. doi:10.2165/11206040-000000000-00000.
  • Calandra G, McCarty J, McGuirk J, et al. AMD3100 plus G-CSF can successfully mobilize CD34+ cells from non-Hodgkin’s lymphoma, hodgkin’s disease and multiple myeloma patients previously failing mobilization with chemotherapy and/or cytokine treatment: compassionate use data. Bone Marrow Transplant. 2008;41(4):331–338. doi:10.1038/sj.bmt.1705908.
  • Sureda A, Chabannon C, Masszi T, et al. Analysis of data collected in the european society for blood and marrow transplantation (EBMT) registry on a cohort of lymphoma patients receiving plerixafor. Bone Marrow Transplant. 2020;55(3):613–622. doi:10.1038/s41409-019-0693-z.
  • Steiner N, Göbel G, Mauser L, et al. Poor mobilizers in lymphoma but not myeloma patients had significantly poorer Progression-Free survival after autologous stem cell transplantation: results of a large retrospective, single-Center observational study. Cancers (Basel). 2023;15(3):608. doi:10.3390/cancers15030608.
  • Morris C, Chabannon C, Masszi T, et al. Results from a multicenter, noninterventional registry study for multiple myeloma patients who received stem cell mobilization regimens with and without plerixafor. Bone Marrow Transplant. 2020;55(2):356–366. doi:10.1038/s41409-019-0676-0.
  • Ruan T, Yue Y, Lu W, et al. Association between low ambient temperature during pregnancy and adverse birth outcomes: a systematic review and meta-analysis. Chin Med J (Engl). 2023;136(19):2307–2315. doi:10.1097/CM9.0000000000002361.
  • Zhu J, Huang H, Chen H, et al. Plerixafor and granulocyte-colony-stimulating factor for mobilization of hematopoietic stem cells for autologous transplantation in chinese patients with non-Hodgkin’s lymphoma: a randomized phase 3 study. Transfusion. 2018;58(1):81–87. doi:10.1111/trf.14426.
  • Worel N, Rosskopf K, Neumeister P, et al. Plerixafor and granulocyte-colony-stimulating factor (G-CSF) in patients with lymphoma and multiple myeloma previously failing mobilization with G-CSF with or without chemotherapy for autologous hematopoietic stem cell mobilization: the Austrian experience on a named patient program. Transfusion. 2011;51(5):968–975. doi:10.1111/j.1537-2995.2010.02896.x.
  • Varmavuo V, Mäntymaa P, Kuittinen T, et al. Pre-emptive plerixafor injection increases blood neutrophil, lymphocyte and monocyte counts in addition to CD34(+) counts in patients with non-Hodgkin lymphoma mobilizing poorly with chemotherapy plus G-CSF: potential implications for apheresis and graft composition. Transfus Apher Sci. 2012;46(3):257–262. doi:10.1016/j.transci.2012.03.011.
  • Stiff P, Micallef I, McCarthy P, et al. Treatment with plerixafor in non-Hodgkin’s lymphoma and multiple myeloma patients to increase the number of peripheral blood stem cells when given a mobilizing regimen of G-CSF: implications for the heavily pretreated patient. Biol Blood Marrow Transplant. 2009;15(2):249–256. doi:10.1016/j.bbmt.2008.11.028.
  • Stewart DA, Smith C, MacFarland R, et al. Pharmacokinetics and pharmacodynamics of plerixafor in patients with non-Hodgkin lymphoma and multiple myeloma. Biol Blood Marrow Transplant. 2009;15(1):39–46. doi:10.1016/j.bbmt.2008.10.018.
  • Sanchez-Ortega I, Querol S, Encuentra M, et al. Plerixafor in patients with lymphoma and multiple myeloma: effectiveness in cases with very low circulating CD34 + cell levels and preemptive intervention vs remobilization. Bone Marrow Transplant. 2015;50(1):34–39. doi:10.1038/bmt.2014.196.
  • Ri M, Matsue K, Sunami K, et al. Efficacy and safety of plerixafor for the mobilization/collection of peripheral hematopoietic stem cells for autologous transplantation in japanese patients with multiple myeloma. Int J Hematol. 2017;106(4):562–572. doi:10.1007/s12185-017-2255-8.
  • Ray GK, Jena RK, Panda T, et al. Prospective identification of potential factors influencing stem cell mobilization and the necessity for plerixafor use in newly diagnosed multiple myeloma patients undergoing autologous stem cell transplantation. Hematol Transfus Cell Ther. 2021;43(4):402–409. doi:10.1016/j.htct.2020.04.011.
  • Pusic I, Jiang SY, Landua S, et al. Impact of mobilization and remobilization strategies on achieving sufficient stem cell yields for autologous transplantation. Biol Blood Marrow Transplant. 2008;14(9):1045–1056. doi:10.1016/j.bbmt.2008.07.004.
  • Nahi H, Celanovic M, Liu Q, et al. A pilot, exploratory, randomized, phase II safety study evaluating tumor cell mobilization and apheresis product contamination in patients treated with granulocyte colony-stimulating factor alone or plus plerixafor. Biol Blood Marrow Transplant. 2019;25(1):34–40. doi:10.1016/j.bbmt.2018.08.020.
  • Micallef IN, Stiff PJ, Stadtmauer EA, et al. Safety and efficacy of upfront plerixafor + G-CSF versus placebo + G-CSF for mobilization of CD34(+) hematopoietic progenitor cells in patients ≥60 and <60 years of age with non-Hodgkin’s lymphoma or multiple myeloma. Am J Hematol. 2013;88(12):1017–1023. doi:10.1002/ajh.23561.
  • Maziarz RT, Nademanee AP, Micallef IN, et al. Plerixafor plus granulocyte colony-stimulating factor improves the mobilization of hematopoietic stem cells in patients with non-hodgkin lymphoma and low circulating peripheral blood CD34(+). Biol Blood Marrow Transplant. 2013;19(4):670–675. doi:10.1016/j.bbmt.2013.01.005.
  • Matsue K, Kumagai K, Sugiura I, et al. Plerixafor for mobilization and collection of haematopoietic stem cells for autologous transplantation in japanese patients with non-Hodgkin lymphoma: a randomized phase 2 study. Int J Hematol. 2018;108(5):524–534. doi:10.1007/s12185-018-2505-4.
  • Horwitz ME, Chute JP, Gasparetto C, et al. Preemptive dosing of plerixafor given to poor stem cell mobilizers on day 5 of G-CSF administration. Bone Marrow Transplant. 2012;47(8):1051–1055. doi:10.1038/bmt.2011.217.
  • Haverkos BM, Huang Y, Elder P, et al. A single center’s experience using four different front line mobilization strategies in lymphoma patients planned to undergo autologous hematopoietic cell transplantation. Bone Marrow Transplant. 2017;52(4):561–566. doi:10.1038/bmt.2016.304.
  • DiPersio JF, Stadtmauer EA, Nademanee A, et al. Plerixafor and G-CSF versus placebo and G-CSF to mobilize hematopoietic stem cells for autologous stem cell transplantation in patients with multiple myeloma. Blood. 2009;113(23):5720–5726. doi:10.1182/blood-2008-08-174946.
  • DiPersio JF, Micallef IN, Stiff PJ, et al. Phase III prospective randomized Double-Blind Placebo-Controlled trial of plerixafor plus granulocyte colony-stimulating factor compared with placebo plus granulocyte colony-stimulating factor for autologous stem-cell mobilization and transplantation for patients with non-Hodgkin’s lymphoma. J Clin Oncol. 2009;27(28):4767–4773. doi:10.1200/JCO.2008.20.7209.
  • Cooper DL, Pratt K, Baker J, et al. Late afternoon dosing of plerixafor for stem cell mobilization: a practical solution. Clin Lymphoma Myeloma Leuk. 2011;11(3):267–272. doi:10.1016/j.clml.2011.03.014.
  • Andreola G, Babic A, Rabascio C, et al. Plerixafor and filgrastim XM02 (tevagastrim) as a first line peripheral blood stem cell mobilisation strategy in patients with multiple myeloma and lymphoma candidated to autologous bone marrow transplantation. Eur J Haematol. 2012;88(2):154–158. doi:10.1111/j.1600-0609.2011.01719.x.
  • Cashen A, Lopez S, Gao F, et al. A phase II study of plerixafor (AMD3100) plus G-CSF for autologous hematopoietic progenitor cell mobilization in patients with hodgkin lymphoma. Biol Blood Marrow Transplant. 2008;14(11):1253–1261. doi:10.1016/j.bbmt.2008.08.011.
  • Hübel K, Ostermann H, Glaß B, et al. Plerixafor in non-Hodgkin’s lymphoma patients: a german analysis of time, effort and costs. Bone Marrow Transplant. 2019;54(1):123–129. doi:10.1038/s41409-018-0228-z.
  • Kvistad SAS, Burman J, Lehmann AK, et al. Impact of previous disease-modifying treatment on safety and efficacy in patients with MS treated with AHSCT. J Neurol Neurosurg Psychiatry. 2022;93(8):844–848. doi:10.1136/jnnp-2022-328797.
  • Nie M, Bi XW, Zhang WW, et al. Consolidative treatment after salvage chemotherapy improves prognosis in patients with relapsed extranodal natural killer/T-cell lymphoma. Sci Rep. 2016;6(1):23996. doi:10.1038/srep23996.
  • Steinhardt MJ, Krummenast FC, Rosenwald A, et al. R-CHOP intensification with mid-cycle methotrexate and consolidating AraC/TT with BCNU/aHSCT in primary aggressive lymphoma with CNS involvement. J Cancer Res Clin Oncol. 2022;148(1):205–214. doi:10.1007/s00432-021-03663-x.
  • Uy GL, Goyal SD, Fisher NM, et al. Bortezomib administered pre-auto-SCT and as maintenance therapy post transplant for multiple myeloma: a single institution phase II study. Bone Marrow Transplant. 2009;43(10):793–800. doi:10.1038/bmt.2008.384.
  • Musto P, Simeon V, Grossi A, et al. Predicting poor peripheral blood stem cell collection in patients with multiple myeloma receiving pre-transplant induction therapy with novel agents and mobilized with cyclophosphamide plus granulocyte-colony stimulating factor: results from a gruppo italiano malattie EMatologiche dell’Adulto multiple myeloma working party study. Stem Cell Res Ther. 2015;6(1):64. doi:10.1186/s13287-015-0033-1.
  • Schmitt M, Publicover A, Orchard KH, et al. Biosimilar G-CSF based mobilization of peripheral blood hematopoietic stem cells for autologous and allogeneic stem cell transplantation. Theranostics. 2014;4(3):280–289. doi:10.7150/thno.7752.
  • Karponi G, Psatha N, Lederer CW, et al. Plerixafor + G-CSF-mobilized CD34+ cells represent an optimal graft source for thalassemia gene therapy. Blood. 2015;126(5):616–619. doi:10.1182/blood-2015-03-629618.
  • Karres D, Ali S, van Hennik PB, et al. EMA recommendation for the pediatric indications of plerixafor (mozobil) to enhance mobilization of hematopoietic stem cells for collection and subsequent autologous transplantation in children with lymphoma or malignant solid tumors. Oncologist. 2020;25(6):e976–e981. doi:10.1634/theoncologist.2019-0898.
  • Afifi S, Adel NG, Devlin S, et al. Upfront plerixafor plus G-CSF versus cyclophosphamide plus G-CSF for stem cell mobilization in multiple myeloma: efficacy and cost analysis study. Bone Marrow Transplant. 2016;51(4):546–552. doi:10.1038/bmt.2015.322.
  • Veltri L, Cumpston A, Shillingburg A, et al. Hematopoietic progenitor cell mobilization with "just-in-time" plerixafor approach is a cost-effective alternative to routine plerixafor use. Cytotherapy. 2015;17(12):1785–1792. doi:10.1016/j.jcyt.2015.09.002.
  • Mohty M, Duarte RF, Croockewit S, et al. The role of plerixafor in optimizing peripheral blood stem cell mobilization for autologous stem cell transplantation. Leukemia. 2011;25(1):1–6. doi:10.1038/leu.2010.224.
  • Li Z, Hardij J, Evers SS, et al. G-CSF partially mediates effects of sleeve gastrectomy on the bone marrow niche. J Clin Invest. 2019;129(6):2404–2416. doi:10.1172/JCI126173.
  • Estey E. High cytogenetic or molecular genetic risk acute myeloid leukemia. Hematology Am Soc Hematol Educ Program. 2010;2010(1):474–480. doi:10.1182/asheducation-2010.1.474.