Fact-checked by Grok 2 weeks ago

Plerixafor

Plerixafor, marketed under the brand name Mozobil, is a small-molecule mobilizer approved for use in combination with (G-CSF) to enhance the mobilization of peripheral blood s to facilitate their collection by in patients with (MM) or non-Hodgkin's lymphoma (NHL) who are undergoing autologous transplantation. It functions as a selective of the , reversibly inhibiting the interaction between and its , stromal cell-derived factor-1α (SDF-1α), which disrupts the homing and retention of s (HSCs) within the bone marrow niche, leading to their rapid release into the peripheral circulation. Originally developed in the early as an antiretroviral agent (AMD3100) targeting HIV entry via , its stem cell-mobilizing properties were serendipitously identified during phase I trials, prompting a pivot in research focus. The U.S. (FDA) granted accelerated approval to plerixafor in December 2008 based on pivotal phase III clinical trials demonstrating superior efficacy over G-CSF alone in achieving adequate + cell yields for transplantation. In these double-blind, -controlled studies involving patients with (n=302) and (n=298), plerixafor G-CSF enabled 71.6% of patients to ≥6 × 10^6 + cells/ within ≤2 sessions (. 34.3% with G-CSF) and % of patients to ≥5 × 10^6 + cells/ within ≤4 sessions (. 20%), with more patients in the plerixafor groups proceeding to successful engraftment post-transplant. Administered subcutaneously at a dose of 0.24 mg/ (up to 40 mg maximum) approximately 11 hours prior to following 4–5 days of G-CSF priming, plerixafor exhibits linear with a of about 5 hours and is primarily cleared renally, necessitating dose adjustments in patients with moderate to severe renal impairment. Plerixafor is particularly valuable for poor mobilizers—such as heavily pretreated patients or those over 65 years old—who fail to achieve sufficient yields with G-CSF alone, with compassionate-use data showing success rates exceeding 50% in this subgroup. Common adverse effects include gastrointestinal disturbances (e.g., , in ≥25% of patients), injection-site reactions, , and , though these are generally mild to moderate and self-limiting, with no evidence of increased or secondary malignancies in long-term follow-up. Contraindicated in individuals with to the drug, plerixafor carries warnings for potential risks such as splenic rupture, hyperleukocytosis, and embryo-fetal toxicity, underscoring the need for close monitoring during . Ongoing research explores its utility in allogeneic transplantation, other hematologic disorders like Hodgkin's , and non-malignant conditions such as , highlighting its expanding role in .

Medical uses

Stem cell mobilization in transplantation

Plerixafor was approved by the U.S. (FDA) in December 2008 for use in combination with granulocyte-colony stimulating factor (G-CSF) to mobilize + hematopoietic s to the peripheral blood for collection and subsequent autologous transplantation in patients with or non-Hodgkin's lymphoma. This approval was based on two pivotal phase III randomized, double-blind, placebo-controlled trials demonstrating superior mobilization efficacy compared to G-CSF alone. Plerixafor acts by reversibly blocking the receptor on hematopoietic stem cells, thereby disrupting the binding of stromal cell-derived factor-1 (SDF-1) and promoting the rapid release of + cells from the into the bloodstream. It is administered subcutaneously at a dose of 0.24 mg/kg of actual body weight (maximum 40 mg per day), typically 11 hours prior to the start of , following 4 days of G-CSF priming (10 mcg/kg daily). This regimen may be repeated for up to 4 consecutive days to optimize collection. In clinical trials, the addition of plerixafor to G-CSF increased the median fold change in circulating + cells by approximately 2- to 3-fold compared to G-CSF alone, with median peripheral blood + counts reaching 60-80 cells/μL versus 20-30 cells/μL. For the multiple myeloma trial (NCT00103662), 72% of patients achieved collection of at least 6 × 10^6 + cells/kg in ≤2 apheresis sessions with plerixafor plus G-CSF, compared to 34% with plus G-CSF. A similar phase III trial in non-Hodgkin's lymphoma showed 59% success in collecting ≥5 × 10^6 + cells/kg in ≤4 apheresis sessions with plerixafor plus G-CSF, compared to 20% with plus G-CSF. This enhanced mobilization has proven particularly beneficial for poor mobilizers, who constitute 15-20% of patients failing to meet collection targets with G-CSF monotherapy, enabling successful in the majority through rescue or preemptive use. As of 2025, updated protocols incorporating plerixafor with salvage have shown promise in (DLBCL), a subtype of non-Hodgkin's , by improving + cell yields and reducing collection failures prior to autologous transplantation. For instance, a study reported that adding plerixafor to regimens like R-DHAP or R-ICE increased successful collections from 60% to over 85% in relapsed/refractory DLBCL patients.

Other approved indications

Plerixafor has limited approvals for uses beyond its primary role in stem cell mobilization for autologous transplantation in multiple myeloma and non-Hodgkin lymphoma. In the European Union, the European Medicines Agency (EMA) authorized its use in 2009 in combination with granulocyte colony-stimulating factor (G-CSF) to enhance mobilization of hematopoietic stem cells in patients with lymphoma and multiple myeloma whose cells mobilize poorly with G-CSF alone. In pediatric patients (aged 1 to less than 18 years), it is approved in the for use with G-CSF to mobilize s in and solid malignant tumors, either preemptively when circulating counts are expected to be insufficient or in patients who previously failed to collect sufficient cells. As of 2025, plerixafor receives guideline support as an adjunct in combination with and G-CSF for poor mobilizers with relapsed or refractory (DLBCL) to improve yields for autologous transplantation. A multicenter phase II study demonstrated that adding plerixafor to R-DHAP and G-CSF achieved the minimum + target cell count of 6 × 10^6/kg in 81% of patients, with 66% requiring only one session. This approach is particularly beneficial in enhancing success prior to therapies like CAR-T cell or autologous transplantation in this setting. Plerixafor has no broad approvals for solid tumors or non-hematologic uses as of 2025, with applications confined to niche enhancements in autologous collection. Patient selection is typically reserved for individuals with prior exposure to regimens that compromise reserve and lead to poor with G-CSF alone.

Contraindications and precautions

Pregnancy and breastfeeding

Plerixafor is contraindicated in due to evidence of embryo-fetal from animal studies, with limited human data available. In rats and rabbits, administration during resulted in post-implantation loss, decreased fetal body weights, and malformations primarily affecting the vascular and skeletal systems, observed at doses approximately 10 times the recommended human dose on a mg/m² basis. Although no adequate and well-controlled studies exist in pregnant women, the potential for fetal harm warrants avoidance unless the clinical benefit justifies the risk. The similarly advises against use during except when clinically necessary, citing teratogenic effects in animal models including increased resorptions, retarded skeletal development, and fetal abnormalities. Prior to initiating plerixafor , pregnancy status should be verified in females of reproductive potential, and effective contraception is recommended during treatment and for at least one week following the final dose to prevent unintended exposure. This guidance aligns with recommendations from the American Society for Transplantation and Cellular Therapy (ASTCT, formerly ASBMT) and the , which emphasize avoidance in reproductive-age females unless benefits outweigh potential risks, with no significant changes from post-marketing surveillance as of 2025. Regarding , it is unknown whether plerixafor is excreted in human , but indicate presence in , posing potential risks to infants due to possible serious adverse reactions. should therefore be discontinued during plerixafor treatment and for at least one week after the final dose, as advised by the ; the recommends discontinuation during therapy due to the inability to exclude risks to the suckling child.

Other contraindications

Plerixafor is contraindicated in patients with a known history of to the drug itself or any of its excipients, as serious reactions, including anaphylactic shock, have been reported. These reactions may manifest as , urticaria, , or dyspnea, necessitating immediate discontinuation and appropriate medical intervention if they occur. In patients with moderate (CrCl 31–50 mL/min) or severe (CrCl <31 mL/min) renal impairment, the dose should be reduced to 0.16 mg/kg (maximum 27 mg daily), with close monitoring for excessive leukocytosis to prevent complications. For end-stage renal disease (CrCl <15 mL/min) or patients on dialysis, use with caution as data are limited; no specific dosing recommendation is available per EMA for CrCl <20 mL/min. Pharmacokinetic data indicate prolonged exposure in such patients, increasing the risk of adverse effects without adjusted dosing. Plerixafor is not recommended for use in patients with leukemia, as it may mobilize leukemic cells into the peripheral blood, potentially contaminating the apheresis product and worsening disease prognosis. This precaution is specific to patients with leukemia. Plerixafor is approved for use in non-leukemic hematologic malignancies, such as or , in transplantation settings. Caution is advised in patients with underlying splenic disorders, as rare cases of splenic enlargement and rupture have been associated with plerixafor administration, often presenting with left upper abdominal or shoulder pain. Prompt evaluation, including imaging if symptoms arise, is essential to rule out rupture. As of 2025, no new contraindications have emerged from recent clinical trials, maintaining the established profile focused on hypersensitivity and leukemia avoidance, with ongoing emphasis on renal and splenic precautions.

Adverse effects

Common and mild effects

Plerixafor, when administered in combination with granulocyte colony-stimulating factor () for hematopoietic stem cell mobilization, is associated with several common and mild adverse effects, primarily gastrointestinal disturbances, injection site reactions, and general symptoms. These effects are typically transient, self-limiting, and do not require discontinuation of therapy. Gastrointestinal effects are among the most frequent, affecting a substantial proportion of patients in clinical trials. Diarrhea occurs in approximately 37% of patients, nausea in 34%, vomiting in 10%, and flatulence in 7%. These symptoms are generally mild to moderate, often resolving within 24 hours without specific intervention, and are attributed to the drug's impact on chemokine receptor signaling. Injection site reactions, reported in 34% of patients, manifest as local erythema, pain, or swelling following subcutaneous administration. These reactions are localized, self-limiting, and usually resolve spontaneously within a few days, with no evidence of long-term sequelae. General mild effects include fatigue (27%), headache (22%), and arthralgia (13%), which occur in 10-30% of patients during mobilization and apheresis procedures. These symptoms are not dose-limiting and do not lead to persistent issues post-treatment. In controlled clinical trials involving patients with multiple myeloma and non-Hodgkin lymphoma, adverse effects with an incidence greater than 10% when plerixafor was combined with G-CSF included diarrhea, nausea, injection site reactions, fatigue, headache, vomiting, arthralgia, and dizziness. Management of these common effects focuses on supportive care, such as hydration and symptomatic relief for gastrointestinal symptoms, with no routine premedication required. Mild injection site reactions may occasionally be treated with topical measures or antihistamines if needed, but most resolve without intervention.

Serious and rare effects

Serious adverse effects of plerixafor are infrequent but can be life-threatening, often occurring in conjunction with granulocyte colony-stimulating factor () administration or underlying patient conditions. Cardiovascular events, including hypotension and tachycardia, have been reported in less than 5% of patients, primarily as part of hypersensitivity reactions or vasovagal responses. Rare cases of myocardial infarction have been observed in approximately 1% of oncology patients (7 out of 679) following stem cell mobilization with plerixafor and , particularly in those with pre-existing cardiovascular risk factors. Hematologic effects include leukocytosis, with white blood cell counts increasing by up to 50% or more, and hyperleukocytosis (WBC ≥100,000/mcL) occurring in about 7% of patients during clinical trials. Plerixafor may also mobilize tumor cells in patients with malignancies, potentially leading to their reinfusion during transplantation, though the clinical impact remains uncertain. Splenic rupture, a rare post-marketing event reported in less than 0.1% of cases, necessitates prompt evaluation for left upper quadrant pain or referred shoulder discomfort. Allergic reactions, such as anaphylaxis and angioedema, occur in less than 1% of patients and require immediate discontinuation of the drug and supportive care, including monitoring for at least 30 minutes post-injection due to the risk of life-threatening hypotension and shock. Most serious effects are attributed to the underlying disease, concurrent G-CSF use, or patient comorbidities rather than plerixafor alone.

Drug interactions

Pharmacokinetic considerations

Plerixafor demonstrates negligible interaction with cytochrome P450 () enzymes, as it is not metabolized via these pathways and shows no significant induction or inhibition of major isoforms, including , , , and . This profile substantially lowers the risk of metabolic drug-drug interactions compared to agents reliant on hepatic cytochrome-mediated clearance. The drug's elimination is predominantly renal, with approximately 70% excreted unchanged in the urine within 24 hours, highlighting the importance of renal function in its pharmacokinetics. Co-administration with nephrotoxic drugs, such as or , warrants caution, as these may reduce creatinine clearance and thereby extend plerixafor exposure by impairing renal excretion. Dose adjustments are recommended for patients with moderate to severe renal impairment (creatinine clearance ≤50 mL/min), further emphasizing the need to monitor renal status during combination therapy. Plerixafor is not subject to hepatic metabolism, which eliminates potential interactions with hepatically cleared pharmaceuticals that might otherwise compete for metabolic enzymes or transporters. Its low plasma protein binding, ranging from 37% to 58% across concentrations, further mitigates risks of displacement from binding sites when used alongside highly bound agents like .

Clinical interactions

Plerixafor exhibits synergistic effects with granulocyte colony-stimulating factor () in mobilizing hematopoietic stem cells () to the peripheral blood, leading to higher CD34+ cell yields compared to G-CSF alone, without requiring dose adjustments to either agent. This combination is standard in protocols for patients with multiple myeloma or non-Hodgkin lymphoma prior to autologous stem cell transplantation, though it carries an increased risk of transient leukocytosis due to elevated neutrophil, lymphocyte, and monocyte counts. In transplant settings, plerixafor may enhance HSC release when used alongside immunosuppressants, potentially improving engraftment in autologous procedures; however, in off-label allogeneic uses, close monitoring for graft-versus-host disease is advised due to altered immune cell dynamics. No significant clinical interactions have been observed with chemotherapy agents, as plerixafor is typically administered sequentially following chemotherapy-based mobilization regimens, allowing safe integration without overlap-related complications. Rare reports suggest enhanced gastrointestinal effects, such as nausea and diarrhea, when plerixafor is combined with opioids, likely due to overlapping mechanisms affecting gut motility. Theoretical concerns exist regarding immune modulation when plerixafor, a CXCR4 antagonist, is co-administered with other CXCR4 agonists, potentially altering chemokine signaling and HSC trafficking.

Pharmacology

Mechanism of action

Plerixafor acts as a selective antagonist of the C-X-C chemokine receptor type 4 (CXCR4), a G protein-coupled receptor expressed on hematopoietic stem cells (HSCs) and progenitor cells. By reversibly binding to CXCR4 with high affinity, plerixafor prevents the interaction between the receptor and its primary ligand, stromal cell-derived factor-1 (SDF-1, also known as CXCL12), which is produced in the bone marrow niche. This blockade disrupts the SDF-1/CXCR4 signaling axis that normally retains HSCs in the bone marrow through chemotaxis and adhesion mechanisms. The antagonism leads to rapid mobilization of CD34+ HSCs and progenitor cells into the peripheral circulation, typically within hours of administration, by interrupting homing and retention signals in the bone marrow microenvironment. This process also mobilizes endothelial progenitor cells, contributing to enhanced vascular repair potential in certain contexts. The mobilization is dose-dependent and synergistic with granulocyte colony-stimulating factor (G-CSF), significantly increasing circulating CD34+ cell counts compared to G-CSF alone. Plerixafor's binding to CXCR4 may be enhanced by metal ions such as zinc in vitro, which can improve receptor affinity through coordination with the bicyclam structure. It exhibits an IC₅₀ of approximately 50 nM for CXCR4 inhibition and high selectivity, showing no agonistic effects or significant binding to other chemokine receptors such as CCR5, CXCR1-3, or CXCR7, with IC₅₀ values exceeding 100 μM for these off-targets. Although originally discovered for its ability to inhibit HIV-1 entry by blocking CXCR4-mediated viral fusion (particularly for X4-tropic strains), this effect is not clinically relevant at therapeutic doses used for stem cell mobilization due to factors such as short half-life and potential adverse effects observed in antiviral trials. This mechanism also underpins its approval for treating WHIM syndrome.

Pharmacokinetics

Plerixafor is administered subcutaneously and exhibits rapid absorption, with peak plasma concentrations (C_max) occurring approximately 30 to 60 minutes after dosing. The pharmacokinetics demonstrate linear kinetics over the dose range of 0.04 to 0.24 mg/kg, and the drug follows a two-compartment model with first-order absorption and elimination. Following absorption, plerixafor distributes primarily into the extravascular space, with an apparent volume of distribution of 0.3 L/kg. Plasma protein binding is moderate, up to 58%. Metabolism of plerixafor is minimal, as evidenced by in vitro studies using human liver microsomes and hepatocytes, which showed no significant biotransformation or inhibitory effects on major cytochrome P450 enzymes (CYP1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, 3A4/5). No major metabolites have been identified, and the drug does not induce CYP1A2, CYP2B6, or CYP3A4 in human hepatocytes. Elimination occurs primarily via renal excretion, with approximately 70% of the administered dose recovered unchanged in the urine within 24 hours in individuals with normal renal function. The terminal half-life ranges from 3 to 5 hours, and total plasma clearance is approximately 4.4 L/h (about 73 mL/min). In patients with renal impairment, clearance is reduced and correlates with creatinine clearance (CrCl); for those with CrCl of 15-50 mL/min, the dose should be reduced to 0.16 mg/kg (not exceeding 27 mg/day) to maintain exposure similar to patients with normal renal function. Pharmacokinetic profiles of plerixafor remain consistent across diverse patient populations, including those with non-Hodgkin lymphoma (such as DLBCL) and multiple myeloma, as well as healthy volunteers, with no clinically significant differences based on age, gender, or race.

Chemistry

Chemical structure and properties

Plerixafor, also known as AMD3100, has the IUPAC name 1,1'-[1,4-phenylenebis(methylene)]bis[1,4,8,11-tetraazacyclotetradecane] and the molecular formula C<sub>28</sub>H<sub>54</sub>N<sub>8</sub>, with a molecular weight of 502.79 g/mol. It is a symmetric , consisting of two cyclam (1,4,8,11-tetraazacyclotetradecane) macrocyclic rings connected by a 1,4-xylylene bridge, and it exerts its activity as a (II) complex. As a physical entity, plerixafor appears as a white to off-white crystalline solid. It exhibits high in , exceeding 50 mg/mL across a range of 1 to 10, reflecting its nature. The compound's basicity is indicated by values ranging from 8.5 to 11.5 for its secondary and groups, with additional lower values below 2.4. Plerixafor has no stereoisomers of clinical relevance due to its achiral, symmetric structure. The commercial formulation of plerixafor is provided as a sterile, clear, colorless to pale-yellow injectable solution containing 24 mg of plerixafor (equivalent to 20 mg/mL of the base) in 1.2 mL single-dose vials, with each milliliter also including 5.9 mg of as an isotonicity agent and , adjusted to a of 6.0–7.5. For stability, the drug substance and product are suitable for storage at controlled (25°C or 77°F), with excursions permitted between 15°C and 30°C (59°F and 86°F); the assigned for the formulated product is 36 months under these conditions.

Synthesis

The synthesis of plerixafor, chemically known as 1,1'-[1,4-phenylenebis(methylene)]bis(1,4,8,11-tetraazacyclotetradecane), was first reported in as part of studies on dimetallic coordination complexes. The original method involved protecting three of the four nitrogen atoms in cyclam (1,4,8,11-tetraazacyclotetradecane) with tosyl groups using p-toluenesulfonyl chloride in the presence of a base such as triethylamine in at 20–45°C. This tris-tosyl-protected cyclam was then subjected to with 1,4-bis(bromomethyl)benzene (α,α'-dibromo-p-xylene) in a like DMF or , facilitated by at 50–85°C for 18–48 hours, to form the linked bis-macrocycle intermediate. Deprotection of the tosyl groups followed via treatment with a mixture of 48% hydrobromic acid and glacial acetic acid at reflux (100–115°C) for 20–48 hours, yielding plerixafor as the octahydrobromide salt after basification and extraction. The overall yield for these key steps—protection, substitution, and deprotection—was approximately 60%, though early laboratory-scale processes often achieved lower totals due to purification challenges. The high basicity of the cyclam nitrogens necessitated careful pH control during basification to prevent side products from over-alkylation or incomplete deprotection. For industrial-scale production, the process was optimized by Genzyme (following acquisition of AnorMED in 2007) to improve scalability and purity, with the substitution step conducted in DMF at elevated temperatures and deprotection sometimes adapted using high-pressure hydrogenation over palladium catalysts to cleave tosyl groups more selectively and avoid corrosive HBr. Purification typically involved extraction of the free base into toluene or dichloromethane, followed by crystallization of the octahydrochloride salt from methanol-acetone mixtures, achieving >99% HPLC purity without column chromatography. The synthesis is covered by patents from AnorMED and Genzyme (e.g., US5583131, which expired on December 10, 2013), with other related patents expiring later; following these expirations, generic versions of plerixafor have been approved and utilize similar synthetic processes, with the first generic entering the U.S. market in 2023. No major methodological changes have been reported post-regulatory approval of Mozobil in 2008.

Development and history

Discovery and early research

Plerixafor, originally designated as AMD3100, emerged from research at the Rega Institute for Medical Research at in during the early . The compound's development began with the serendipitous isolation of an impurity, JM1657, in commercial cyclam preparations, which demonstrated anti- activity in 1992. This led to the synthesis of optimized bicyclam analogs, culminating in AMD3100 by 1994, initially explored for its potential as a metal chelator in studies but quickly recognized for its antiviral potential against T-tropic HIV strains. In the mid-1990s, studies by Dominique Schols and colleagues at revealed that AMD3100 potently inhibits CXCR4-mediated -1 entry and membrane fusion , with effective concentrations in the 1-10 nM range and a selectivity index exceeding 100,000. Funded in part by NIH grants for research, this work advanced AMD3100 into phase I and II clinical trials by the late 1990s, where it showed modest antiviral effects but development for was halted in 2001 following observation of cardiac adverse effects in a phase II trial, compounded by its poor oral bioavailability. During these HIV-focused investigations, researchers observed an unanticipated rapid increase in peripheral counts, including + hematopoietic stem and progenitor cells, following AMD3100 administration in early clinical and preclinical settings. This prompted efforts in the early , with animal model studies confirming AMD3100's ability to mobilize stem cells from the . In models, subcutaneous doses of 5-20 mg/kg induced dose-dependent release of + cells within hours, mediated by blockade of the SDF-1/ axis, without initial evidence of direct anti-cancer activity. AMD3100 was licensed to AnorMED Inc. in 1997 for antiviral development, shifting focus to mobilization after the trials. In 2006, Corporation acquired AnorMED, accelerating preclinical validation of plerixafor's mobilizing effects observed in prior monkey and mouse studies. The acquisition was completed in late 2006; was later acquired by in 2011, continuing plerixafor's development.

Regulatory approvals and milestones

Plerixafor's clinical development advanced through two pivotal phase III trials conducted between 2005 and 2007, evaluating its efficacy in combination with (G-CSF) for hematopoietic stem cell mobilization. In the trial (NCT00103662), plerixafor plus G-CSF enabled 71.6% of patients to achieve the target collection of at least 6 × 10^6 + cells/kg in ≤2 sessions, compared to 34.4% with plus G-CSF, demonstrating superior mobilization. Similarly, the non-Hodgkin lymphoma trial (NCT00103610, AMD3100-3101) showed that 59% of patients receiving plerixafor plus G-CSF met the target of at least 5 × 10^6 + cells/kg in ≤4 sessions, versus 20% in the group, confirming its effectiveness in this population. These results supported the , leading to U.S. (FDA) approval on December 15, 2008, for use with G-CSF to mobilize autologous hematopoietic stem cells in patients with and prior to transplantation. Following FDA approval, plerixafor received designation in the United States for mobilization, granting market exclusivity and development incentives. Internationally, the () authorized Mozobil (plerixafor) on July 31, 2009, for the same indications in combination with G-CSF. approved it on December 8, 2011, further expanding access for eligible patients. Plerixafor also obtained status in the , recognizing its role in addressing unmet needs in rare conditions involving transplantation. Post-approval milestones included the entry of generic versions following patent expiration, with the FDA approving the first for plerixafor injection by on July 24, 2023, enhancing affordability and availability. In 2021, a phase II trial (NCT04552743) initiated evaluation of plerixafor combined with MGTA-145, a CXCR2 agonist, for improved mobilization without G-CSF, which was completed, demonstrating promising rapid and reliable collection in patients. By 2025, the (NCCN) guidelines incorporated plerixafor for poor mobilizers in (DLBCL), reflecting its established utility in challenging cases, though no new indications have received regulatory approval. continues through clinical trials exploring its potential in other mobilization scenarios. Development faced early setbacks from initial focus on HIV treatment, where phase I/II trials in the late 1990s and early 2000s demonstrated limited antiviral efficacy due to insufficient blockade , delaying pivot to stem cell applications by approximately 10 years until preclinical data emerged around 2002.

Society and culture

Plerixafor is classified as a prescription-only medication in the United States, , and the , requiring a physician's for use due to its role in hematopoietic stem cell mobilization. It is not designated as a under schedules in these jurisdictions, as it lacks significant potential for misuse beyond its approved therapeutic applications in . The drug received designation from the U.S. (FDA) in 2003 for enhancing yields in patients with non-Hodgkin's lymphoma and prior to autologous transplantation. This status granted seven years of market exclusivity from its approval in December 2008, extending through 2015, along with tax credits for costs to offset development expenses for treatments. Pediatric exclusivity extensions were not applicable, given the drug's primary indications in adult populations. Economically, plerixafor remains costly, with a wholesale acquisition cost of approximately $9,300 per dose in the U.S. as of 2024. The introduction of generic versions following FDA approvals in 2023 has helped reduce costs compared to the branded product. Globally, the plerixafor market is projected to reach $885 million by the end of 2025, driven by increasing demand in . Access to plerixafor is supported under Part B in the U.S. for FDA-approved indications, covering outpatient administration when used in combination with granulocyte-colony stimulating factor for mobilization. However, significant disparities persist in low-income regions, where limited transplantation and high out-of-pocket costs hinder availability, particularly in parts of and low- to middle-income countries. As of 2025, generic competition in the has further lowered prices, with multiple manufacturers offering equivalents that enhance without ongoing legal disputes over patents or approvals. Additional generic approvals, such as by Gland Pharma in May 2024 and Hetero Labs in June 2025, have expanded availability in the U.S. No major legal challenges related to plerixafor's regulatory status or are currently active.

Brand names and availability

Plerixafor is primarily marketed under the brand name Mozobil by , a subsidiary of . It is formulated as a 24 mg/1.2 mL solution for subcutaneous injection in a pre-filled . Generic versions of plerixafor became available in the United States in 2023, including Plerixafor Injection by Fresenius Kabi and Meitheal Pharmaceuticals, offered in similar 24 mg/1.2 mL subcutaneous formulations. Internationally, plerixafor is available under the generic name Plerixafor Accord in the , approved for subcutaneous use in combination with granulocyte-colony stimulating factor for mobilization. Generic formulations are also marketed in by manufacturers such as Fresenius Kabi Canada Ltd. and JAMP Pharma Corporation, and in under brand names including Mozifor by Hetero Drugs Ltd. and Celrixafor by Celon Laboratories. Plerixafor is widely distributed in developed countries, where it is routinely supplied to and transplant centers. In contrast, availability remains limited in low-resource settings, particularly those without established transplant programs, due to and constraints. As of 2025, more than 10 generic manufacturers produce plerixafor globally, with the drug administered exclusively via subcutaneous injection and no oral available.

Research directions

Oncology and anti-cancer applications

Plerixafor, functioning as a antagonist, disrupts the /SDF-1 axis within the , thereby impeding , invasion, and metastatic dissemination in preclinical models. In preclinical studies using mouse models, this inhibition has demonstrated reduced metastatic spread in , particularly when combined with agents like combretastatin A4, which enhanced anti-tumor and anti-metastatic effects. Similarly, in models, plerixafor modulation of the pathway has altered reactivity and limited tumor progression and dissemination. Beyond its approved role in stem cell mobilization, plerixafor has been investigated in combination therapies for hematologic malignancies. A phase I/II trial combining plerixafor with in patients with relapsed or refractory targeted microenvironment interactions to enhance chemosensitization, resulting in increased and an overall response rate of 48.5%. In solid tumors, plerixafor has shown preliminary promise in early-phase trials. Despite these findings, plerixafor's direct anti-cancer applications face significant hurdles, with no successful phase III trials establishing it as monotherapy or standard adjunct therapy due to limited in advanced settings and dose-dependent , including gastrointestinal effects and potential cardiac risks at higher doses. As of 2025, research continues to explore adjunctive roles for plerixafor in post-transplant strategies, though it remains investigational and not integrated into routine care.

Neurological and other therapeutic areas

Plerixafor, as a , has been explored in preclinical and early clinical investigations for neurological applications, primarily through its influence on the CXCR4/ axis, which regulates , immune cell trafficking, and potential remyelination processes. In (), plerixafor facilitates (HSC) mobilization for autologous (AHSCT), offering a non-myeloablative alternative to traditional or (G-CSF) regimens without significant toxicity. However, its use in requires caution, as disruption of the CXCR4/SDF-1 interaction may impair protective mechanisms against observed in experimental models, with limited published specific to MS patients as of 2025. Beyond , the / pathway's role in modulating activation and has prompted preclinical interest in plerixafor for conditions like , though direct studies remain limited to animal models demonstrating microglia modulation. No neurological indications have received regulatory approval, and most derives from foundational on the rather than plerixafor-specific trials. In stem cell research, plerixafor enhances mobilization of mesenchymal stem cells (MSCs) when combined with vascular endothelial growth factor (VEGF), promoting endogenous recruitment for tissue repair applications such as fracture healing. In a rat model of femoral osteotomy, VEGF pretreatment followed by plerixafor (AMD3100) increased circulating MSCs, leading to improved bone volume, trabecular thickness, and radiographic union scores compared to controls, with reduced variability in healing outcomes. However, a phase IIa randomized, double-blind, placebo-controlled trial in 26 patients with diabetic ischemic wounds found that a single dose of plerixafor (0.24 mg/kg subcutaneously) successfully mobilized hematopoietic stem/progenitor cells (HSPCs) but failed to accelerate wound healing, with only 38.5% complete closure at 6 months versus 69.2% in placebo (P=0.115), and larger residual wound sizes in the plerixafor arm. Plerixafor's historical exploration in immune modulation includes early investigations for , where it was identified as an anti- agent due to of CXCR4-mediated entry, but clinical development was halted owing to limited oral and short , rendering it ineffective for antiviral . Emerging applications in non-malignant immune conditions focus on autoimmune diseases, where plerixafor supports mobilization for AHSCT in refractory cases like systemic lupus erythematosus (SLE), potentially enabling immune reset through depletion of autoreactive cells, though evidence remains confined to broader rheumatic disease protocols. Recent developments, including a 2024 American Society of (ASH) abstract, highlight plerixafor's role in enhancing transplant outcomes when added to G-CSF, achieving target + cell yields (4-6 × 10^6/kg) in pediatric patients with refractory malignancies in 1-4 days, with minimal adverse effects like , suggesting broader utility in transplant optimization beyond . As of 2025, no neurologic approvals exist, and investigational progress emphasizes non-cancer and immune applications. Limitations of plerixafor in these areas include off-target (CNS) effects observed in preclinical studies, such as dose-dependent , tremors, , and convulsions in rats, dogs, and rabbits, attributed to blood-brain barrier penetration and binding to adrenergic and , with variable suppression or stimulation across species. Additionally, increased neurological symptoms linked to leukemic CNS infiltration have been noted in models, underscoring risks in neuroinflammatory contexts. Overall, data remain predominantly preclinical or early-phase, with challenges in translating mobilization benefits to functional therapeutic gains in non-oncologic settings. As of November 2025, ongoing trials continue to evaluate plerixafor in expanded indications, including combination therapies for autoimmune disorders and tissue repair, but no new approvals have been granted beyond mobilization.

References

  1. [1]
    [PDF] MOZOBIL (plerixafor) Injection, Solution for subcutaneous use
    Plerixafor is not metabolized as shown in assays using human liver microsomes or human primary hepatocytes and does not exhibit inhibitory activity in vitro ...
  2. [2]
    Plerixafor - LiverTox - NCBI Bookshelf - NIH
    Apr 12, 2019 · Plerixafor is a small molecular antagonist of the cell-surface CXCR4 receptor that plays an important role in mobilization of hematopoietic ...
  3. [3]
    Plerixafor - PMC - NIH
    Plerixafor is a novel agent for use in combination with G-CSF for the mobilization of peripheral blood stem cells in patients with MM and NHL.
  4. [4]
    Mozobil in combination with granulocyte colony-stimulating factor to ...
    The US Food and Drug Administration approved plerixafor (Mozobil; Genzyme Corp.), a new small-molecule inhibitor of the CXCR4 chemokine receptor.
  5. [5]
    Plerixafor: a review of its use in stem-cell mobilization in ... - PubMed
    Aug 20, 2011 · Plerixafor (Mozobil®) is a CXCR4 chemokine receptor antagonist that is indicated for use in combination with granulocyte colony-stimulating ...
  6. [6]
    [PDF] December 2008 Proprietary and Confidential Page 1 This label may ...
    Mozobil, a hematopoietic stem cell mobilizer, is indicated in combination with granulocyte-colony stimulating factor (G-CSF) to mobilize hematopoietic stem ...
  7. [7]
    Plerixafor and G-CSF versus placebo and G-CSF to ... - PubMed
    Jun 4, 2009 · This phase 3, multicenter, randomized (1:1), double-blind, placebo-controlled study evaluated the safety and efficacy of plerixafor with ...
  8. [8]
    Phase III prospective randomized double-blind placebo ... - PubMed
    This study evaluates the safety and efficacy of plerixafor (AMD3100), a CXCR4 antagonist, in mobilizing hematopoietic stem cells for autologous stem-cell ...Missing: NCT00103662 | Show results with:NCT00103662
  9. [9]
    A prospective, multicenter study on hematopoietic stemcell ...
    Nov 16, 2023 · The addition of plerixafor to various chemotherapy regimens and G-CSF led to a median 2-fold increase in the number of circulating CD34+ cells ...Results · Hematopoietic Stem Cell... · Discussion
  10. [10]
    Plerixafor May Improve CD34-Positive Cell Collection Prior to ...
    Jan 29, 2025 · Researchers determined adding plerixafor to salvage therapy may help to improve outcomes for patients with relapsed or refractory DLBCL ...
  11. [11]
    [PDF] Mozobil, INN-plerixafor - European Medicines Agency
    Paediatric patients (1 to less than 18 years). Mozobil is indicated in combination with G-CSF to enhance mobilisation of haematopoietic stem cells.
  12. [12]
  13. [13]
    [PDF] 1 This label may not be the latest approved by FDA. For current ...
    Mozobil is indicated in combination with filgrastim to mobilize hematopoietic stem cells (HSCs) to the peripheral blood for collection and subsequent autologous ...Missing: 2025 | Show results with:2025
  14. [14]
    Plerixafor Monograph for Professionals - Drugs.com
    Aug 10, 2025 · Plerixafor is not recommended as part of a hematopoietic stem cell mobilization regimen in patients with leukemia. Possible mobilization of ...
  15. [15]
    Efficacy of chemotherapy with G-CSF versus plerixafor with G-CSF ...
    Oct 21, 2025 · The most common adverse events related to plerixafor were gastrointestinal disorders and injection site reactions. Plerixafor and G-CSF were ...
  16. [16]
    [PDF] CLINICAL PHARMACOLOGY AND BIOPHARMACEUTICS REVIEW ...
    Jun 16, 2008 · The proposed indication is for plerixafor, administered in conjunction with a G-CSF, to enhance the mobilization of hematopoietic stem cells ( ...<|control11|><|separator|>
  17. [17]
    Pharmacokinetics and pharmacodynamics of plerixafor in patients ...
    The purpose of this study was to evaluate the safety and the PK and PD of plerixafor with G-CSF in patients with non-Hodgkin lymphoma (NHL) and multiple myeloma ...Missing: trials interactions 2023<|separator|>
  18. [18]
    Plerixafor added to chemotherapy plus G-CSF is safe and ... - PubMed
    Plerixafor added to chemotherapy plus G-CSF is safe and allows adequate PBSC collection in predicted poor mobilizer patients with multiple myeloma or lymphoma.
  19. [19]
    Plerixafor and granulocyte colony stimulating factor for poor ...
    Conclusion: Plerixafor is effective in increasing the PSC yield in poor mobilizers. Low BMI and hematologic malignancies other than Multiple Myeloma are ...<|control11|><|separator|>
  20. [20]
    Pre-emptive plerixafor injection increases blood neutrophil ...
    Pre-emptive plerixafor injection increases blood neutrophil, lymphocyte and monocyte counts in addition to CD34+ counts in patients with non-Hodgkin lymphoma ...Missing: immunosuppressants | Show results with:immunosuppressants
  21. [21]
    Plerixafor (a CXCR4 antagonist) following myeloablative allogeneic ...
    Aug 17, 2016 · Our study demonstrated that plerixafor can be given safely following myeloablative HSCT. It provides proof of principle that blocking CXCR4 after HSCT enhances ...
  22. [22]
    Salvage treatment with plerixafor in poor mobilizing allogeneic stem ...
    Oct 7, 2020 · Plerixafor offers a promising opportunity to increase PBSC mobilization associated with little physical stress for the donors. Initial clinical ...Donors And Methods · Results · Discussion
  23. [23]
    Biased action of the CXCR4-targeting drug plerixafor is essential for ...
    May 12, 2021 · Here we show that while AMD11070 acts as a full antagonist, plerixafor acts biased by stimulating β-arrestin recruitment while fully antagonizing G protein.
  24. [24]
    EBMT Releases 2025 Practice Recommendations for HCT and CAR ...
    Sep 15, 2025 · The EBMT has published its highly anticipated 2025 practice recommendations for haematopoietic cell transplantation (HCT) and CAR-T therapy.
  25. [25]
    [PDF] Hematopoietic Stem Cell Transplantation – Clinical Guideline
    Jul 21, 2025 · NCCN Guidelines for Hematopoietic Cell Transplantation (V1.2025) indicate there is insufficient evidence to recommend one of these alternative ...
  26. [26]
    [PDF] PHARMACOLOGY REVIEW(S) - accessdata.fda.gov
    Dec 3, 2008 · Plerixafor was teratogenic in rats; therefore, Pregnancy Category D is recommended for Mozobil label. Teratogenic effects were also observed in ...
  27. [27]
    Physiology and Pharmacology of Plerixafor - PMC - NIH
    Plerixafor is a potent, selective inhibitor of CXCR4 with a unique binding mode. The interaction of CXCR4 with its ligand CXCL12 is essential to the retention ...
  28. [28]
    Metal ion containing CXCR4 chemokine receptor antagonists
    Plerixafor is a cyclam based compound that contains two macrocyclic rings with an ideal ring size cavity for binding to first row transition metals. It was ...Missing: constant | Show results with:constant<|separator|>
  29. [29]
    Mozobil® (Plerixafor, AMD3100), 10 years after its approval by the ...
    Feb 18, 2019 · AMD3100 (plerixafor, Mozobil®) was first identified as an anti-HIV agent specifically active against the T4-lymphotropic HIV strains, ...
  30. [30]
    Plerixafor: Uses, Interactions, Mechanism of Action | DrugBank Online
    Sep 14, 2010 · It is used to stimulate the release of stem cells from the bone marrow into the blood in patients with non-Hodgkin's lymphoma (NHL) and multiple myeloma to ...
  31. [31]
    Plerixafor | C28H54N8 | CID 65015 - PubChem - NIH
    It is used to stimulate the release of stem cells from the bone marrow into the blood in patients with non-Hodgkin's lymphoma (NHL) and multiple myeloma to ...
  32. [32]
    [PDF] CHEMISTRY REVIEW(S) - accessdata.fda.gov
    Dec 12, 2008 · Plerixafor is soluble, to levels over in aqueous solution, when the pH is adjusted to between 1 and 10. The submitted stability data ...Missing: physical | Show results with:physical<|separator|>
  33. [33]
    Dinickel and dicopper complexes with N,N-linked bis(cyclam ...
    Methods for the synthesis of immunostimulant plerixafor. Chemistry of Heterocyclic Compounds 2020, 56 (1) , 30-35. https://doi.org/10.1007/s10593-020-02617 ...
  34. [34]
  35. [35]
    WO2014125499A1 - Improved and commercially viable process for ...
    Tosyl moieties of compound (1 1) were deprotected upon treating with mixture of acetic acid and 48% aq. HBr at 100°C for 48h to yield plerixafor 8 HBr 2H 20 ( ...
  36. [36]
    Methods for the synthesis of immunostimulant plerixafor | Request PDF
    This minireview discusses methods for the synthesis of the immunostimulant plerixafor (Mozobil®, AMD3100). The information presented covers the period from ...Missing: original | Show results with:original
  37. [37]
  38. [38]
    The bicyclam AMD3100 story - PubMed
    Here, I describe the development process of AMD3100, which began about 15 years ago with the isolation of an impurity, and the basis for the clinical ...Missing: synthesis | Show results with:synthesis
  39. [39]
    Chemokine receptor inhibition by AMD3100 is strictly confined to ...
    AMD3100 potently inhibited CXCR4-mediated calcium signaling and chemotaxis in a concentration-dependent manner in different cell types.Missing: first synthesis 1987 David Catholic
  40. [40]
    AMD3100, a small molecule inhibitor of HIV-1 entry via the CXCR4 ...
    The bicyclam AMD3100 (formula weight 830) blocks HIV-1 entry and membrane fusion via the CXCR4 co-receptor, but not via CCR5.Missing: 1990s phase
  41. [41]
    Multiple-Dose Escalation Study of the Safety, Pharmacokinetics, and ...
    Despite promising anti-HIV effects, the desire to use an orally bioavailable compound led to the halt of AMD3100 development for treatment of HIV infection.
  42. [42]
    Pharmacokinetics and Safety of AMD-3100, a Novel Antagonist of ...
    This work was partially supported by NIH General Clinical Research Center grant RR00052-38. We recognize the significant contributions to the study by ...
  43. [43]
    Rapid mobilization of murine and human hematopoietic stem and ...
    Apr 18, 2005 · Human CD34+ cells isolated after treatment with G-CSF plus AMD3100 expressed a phenotype that was characteristic of highly engrafting mouse HSCs ...
  44. [44]
    AMD3100 mobilizes hematopoietic stem cells with long-term ...
    AMD3100, a bicyclam antagonist of the chemokine receptor CXCR4, has been shown to induce rapid mobilization of CD34+ hematopoietic cells in mice, dogs, and ...Missing: CD34+ | Show results with:CD34+
  45. [45]
    Plerixafor: AMD 3100, AMD3100, JM 3100, SDZ SID 791 - PubMed
    AnorMED (now Genzyme) decided to pursue a full Marketing Authorisation Application (MAA) in Europe for plerixafor in stem cell transplant. Previously, the ...
  46. [46]
    (PDF) Mozobil® (Plerixafor, AMD3100), 10 years after its approval ...
    AMD3100 (plerixafor, Mozobil®) was first identified as an anti-HIV agent specifically active against the T4-lymphotropic HIV strains, as it selectively ...
  47. [47]
  48. [48]
  49. [49]
    Drug Approval Package: Mozobil (plerixa) Injection NDA 22311
    Mar 24, 2009 · Mozobil (plerixa) Injection Company: Genzyme Corporation Application No.: 022311. Approval Date: 12/15/2008 ; Company · Genzyme CorporationMissing: details | Show results with:details
  50. [50]
    Search Orphan Drug Designations and Approvals - FDA
    Marketing approved: 1, Generic Name: plerixafor. Trade Name: Mozobil (r). Marketing Approval Date: 12/15/2008. Approved Labeled Indication: To enhance ...Missing: details | Show results with:details
  51. [51]
    Mozobil | European Medicines Agency (EMA)
    Mozobil received a marketing authorisation valid throughout the EU on 31 July 2009. Mozobil : EPAR - Medicine overview. Reference Number: EMA/254533/2019.
  52. [52]
    Summary Basis of Decision - Mozobil TM - Health Canada
    2 Notice of decision. On December 8, 2011, Health Canada issued a Notice of Compliance to Genzyme Canada Inc. for the drug product, Mozobil.
  53. [53]
  54. [54]
    NCCN Guidelines for B-Cell Lymphomas
    No information is available for this page. · Learn whyMissing: plerixafor poor mobilizers
  55. [55]
  56. [56]
    Efficacy and Cost Analysis of a Plerixafor Protocol for Peripheral ...
    The average wholesale package price for one 1.2-mL vial of plerixafor is $8652.68.7 Costa and colleagues found that using a risk-adaptive protocol for poor ...
  57. [57]
    2023 First Generic Drug Approvals - FDA
    Mar 8, 2024 · 2023 First Generic Drug Approvals. Each year, FDA's Center for Drug Evaluation and Research (CDER) approves a wide range of new drug products.
  58. [58]
    Pharmaceutical Grade Plerixafor 2025-2033 Trends
    Jun 18, 2025 · The global market for Pharmaceutical Grade Plerixafor is projected to reach $884.9 million in 2025, exhibiting a Compound Annual Growth Rate ...
  59. [59]
  60. [60]
    Treatment Accessibility, Availability, and Healthcare Costs for ...
    Sep 3, 2025 · These disparities highlight the critical need to better understand the availability and accessibility of MM treatments across Asian countries.
  61. [61]
    Simplification of Hematopoietic Stem Cell Transplantation and ...
    Sep 12, 2025 · Simplification of Hematopoietic Stem Cell Transplantation and Continued FACT Accreditation: Strategies for Low- and Middle-Income Countries.
  62. [62]
  63. [63]
    North America Comprehensive Analysis of North America Plerixafor ...
    Oct 27, 2025 · Regulatory approval processes: While the FDA has approved Plerixafor, evolving regulatory requirements demand rigorous clinical data, post- ...
  64. [64]
    Fresenius Kabi Launches Plerixafor Injection
    Plerixafor Injection is a hematopoietic stem cell mobilizer used in combination with granulocyte-colony stimulating factor in patients with non-Hodgkin's ...
  65. [65]
    Comparison of the efficacy of a generic plerixafor versus Mozobil as ...
    Oct 4, 2024 · The recent availability of much lower-costing generic forms of plerixafor offers the potential to dramatically lower the economic barrier to ...Missing: disparities income regions
  66. [66]
  67. [67]
    Plerixafor Injection - Mozifor Injection Latest Price, Manufacturers ...
    17,000/Vial. Packaging Type: Vial. Strength: 24 mg / 1.2 ml. About Mostim 24Mg/1.2Ml InjIt is used in the treatment of Non-Hodgkin's Disease (a blood cancer ...
  68. [68]
    Generic Mozobil Availability - Drugs.com
    Oct 8, 2025 · Yes, the FDA has approved a generic version of Mozobil. However, FDA approval doesn't guarantee the generic will be available in pharmacies.Missing: price reduction
  69. [69]
    Plerixafor for Stem Cell Mobilization in Autologous Haematopoietic ...
    Apr 30, 2025 · Plerixafor is not only highly effective but also safe for use in lymphoma patients. These case series findings underscore its value as a key ...Missing: effects | Show results with:effects
  70. [70]
    PLERIXAFOR - Uses, DMF, Dossier, Manufacturer, Supplier ...
    PharmaCompass offers a list of Plerixafor API manufacturers, exporters & distributors, which can be sorted by GMP, USDMF, JDMF, KDMF, CEP (COS), WC, ...
  71. [71]
    Targeting the CXCR4/CXCL12 Axis in Cancer Therapy
    This review aims to summarize the recent advancements in the design and development of CXCR4 antagonists as potential anticancer agents.
  72. [72]
    Combretastatin A4 nanodrug combined plerixafor for inhibiting ...
    Dec 1, 2019 · This cooperative strategy provides a useful method for inhibiting tumor growth and metastasis simultaneously, and gives the evidence to support ...
  73. [73]
    Targeting CXCR4 by a selective peptide antagonist modulates ...
    Mar 25, 2016 · Targeting CXCR4 by a selective peptide antagonist modulates tumor microenvironment and microglia reactivity in a human glioblastoma model.<|control11|><|separator|>
  74. [74]
    Phase I/II trial of the CXCR4 inhibitor plerixafor in combination with ...
    In the phase I study, the MTD was plerixafor 0.32 mg/kg, and bortezomib 1.3 mg/m2 . The overall response rate for the phase II study was 48.5%, and the clinical ...Missing: 2022 | Show results with:2022
  75. [75]
    A phase 2 trial of CXCR4 antagonism and PD1 inhibition in ... - NIH
    Aug 15, 2025 · This was an investigator-initiated, single-center, phase 2 clinical trial designed to evaluate the clinical activity and safety of the ...Missing: NCT02757892 | Show results with:NCT02757892
  76. [76]
    CXCR4 antagonist AMD3100 (plerixafor): From an impurity to a ...
    AMD3100 (plerixafor), a CXCR4 antagonist, has opened a variety of avenues for potential therapeutic approaches in different refractory diseases.
  77. [77]
    October 2025 – Myeloma News from the Journals
    Oct 29, 2025 · For October 2025, the IMF team of medical editors has created overviews of myeloma studies from top medical journals.
  78. [78]
    Hematopoietic Stem Cell Therapy for Multiple Sclerosis: Top 10 ...
    Although plerixafor is not toxic in the manner of chemotherapy, caution is warranted in MS patients, as CXCR4-SDF-1 plays a role in protecting against ...
  79. [79]
    Up-regulation of microglial chemokine CXCL12 in anterior cingulate ...
    Up-regulation of microglial chemokine CXCL12 in anterior cingulate cortex mediates neuropathic pain in diabetic miceMissing: rodents | Show results with:rodents
  80. [80]
    VEGF with AMD3100 endogenously mobilizes mesenchymal stem ...
    VEGF with AMD3100 endogenously mobilizes mesenchymal stem cells and improves fracture healing.Missing: repair | Show results with:repair
  81. [81]
    Stem cell mobilization with plerixafor and healing of diabetic ... - NIH
    Jun 2, 2020 · This study tested whether HSPCs mobilization with plerixafor improved the healing of ischemic diabetic wounds. Successful HSPC mobilization ...
  82. [82]
    Plerixafor - Medical Clinical Policy Bulletins - Aetna
    The requested medication will not be used beyond 4 consecutive days or after completion of stem cell harvest/apheresis. Aetna considers all other indications as ...
  83. [83]
    Autologous haematopoietic stem cell transplantation for rheumatic ...
    Aug 20, 2025 · Autologous stem cell transplantation for systemic lupus erythematosus. ... Depletion of autoreactive immunologic memory followed by ...
  84. [84]
    Plerixafor, Breaking Barriers and Moving the World of Bone Marrow Transplant
    ### Summary of 2024 ASH Abstract on Plerixafor for Bone Marrow Transplant Enhancements