MedPath

Plerixafor Advanced Drug Monograph

Published:Jul 29, 2025

Generic Name

Plerixafor

Brand Names

Mozobil, Plerixafor Accord

Drug Type

Small Molecule

Chemical Formula

C28H54N8

CAS Number

110078-46-1

Plerixafor (DB06809): A Comprehensive Monograph on a Pivotal Hematopoietic Stem Cell Mobilizer

Executive Summary

Plerixafor is a first-in-class, small-molecule, selective antagonist of the C-X-C chemokine receptor type 4 (CXCR4), which has fundamentally altered the clinical practice of hematopoietic stem cell (HSC) mobilization.[1] Marketed under the brand name Mozobil, and now available in generic formulations, it is indicated for use in combination with granulocyte-colony stimulating factor (G-CSF) to mobilize HSCs into the peripheral blood for collection and subsequent autologous transplantation in patients with non-Hodgkin's lymphoma (NHL) and multiple myeloma (MM).[1]

The development of plerixafor is a notable example of serendipity in drug discovery. Originally synthesized and investigated as an anti-HIV agent designed to block the CXCR4 co-receptor used by T-tropic viral strains, its clinical development for this indication was halted.[4] However, astute observation during early-phase trials revealed a consistent and marked elevation in circulating white blood cells, a phenomenon correctly identified as active HSC mobilization from the bone marrow.[4] This led to a strategic pivot, repositioning plerixafor as a hematologic agent.

Its primary clinical value lies in its ability to overcome the limitations of G-CSF alone, which fails to produce adequate stem cell yields in a significant percentage of patients, often termed "poor mobilizers".[2] By disrupting the CXCR4/SDF-1α axis that anchors HSCs in the marrow, plerixafor induces a rapid and predictable release of CD34+ cells into the periphery.[4] Pivotal clinical trials have demonstrated that the addition of plerixafor to a G-CSF regimen significantly increases the proportion of patients who achieve their target cell collection goals, often in fewer apheresis sessions, thereby reducing patient burden and healthcare resource utilization.[7]

Plerixafor possesses a well-characterized and generally manageable safety profile, with the most common adverse effects being transient gastrointestinal and injection-site reactions.[10] The recent introduction of generic formulations has marked a significant inflection point, beginning to dismantle the cost barriers that previously restricted its use and shifting clinical practice towards more liberal, upfront mobilization strategies.[6] This report provides an exhaustive analysis of plerixafor, covering its chemical nature, detailed pharmacology, pharmacokinetic profile, clinical efficacy, safety considerations, and its evolving position within the therapeutic and commercial landscape, including its investigational horizons.

Chemical Profile and Physicochemical Properties

A comprehensive understanding of plerixafor's clinical behavior begins with its fundamental chemical and physical characteristics. Its unique molecular architecture is directly responsible for its specific pharmacological activity and its pharmacokinetic disposition.

Chemical Structure and Nomenclature

Plerixafor is classified as a small molecule and is a member of the azamacrocycle chemical class.[1] Structurally, it is a highly specific and symmetrical bicyclam derivative. The molecule is composed of two identical 1,4,8,11-tetraazacyclotetradecane rings, commonly known as cyclam rings.[1] These two macrocyclic rings are covalently joined by a 1,4-phenylenebis(methylene) linker, which attaches to one of the amine nitrogens on each cyclam ring.[1]

The systematic International Union of Pure and Applied Chemistry (IUPAC) name for plerixafor is 1,1'-[1,4-phenylenebis(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane.[1] Its molecular formula is

C28​H54​N8​, corresponding to a molecular weight of approximately 502.8 g/mol.[1]

Figure 1: 2D Chemical Structure of Plerixafor

!(httpse://pubchem.ncbi.nlm.nih.gov/image/imgsrv.fcgi?cid=65015&t=l)

Source: PubChem CID 65015 1

Physicochemical Properties

Plerixafor exists as a solid at room temperature and has a reported melting point of 131.5 °C.[1] It is characterized as being slightly soluble in water.[1] A defining feature of its chemistry is its basicity. The molecule contains eight nitrogen atoms, all of which can readily accept protons.[13] At physiological pH, these nitrogen atoms are protonated, giving the entire molecule a strong net positive charge, estimated to be +4.[4] This polycationic nature is a critical determinant of its mechanism of action. The dissociation constants (pKa) have been reported as being in the range of 8.5-11.5 for the secondary amines and <2.4 for the tertiary amines, confirming its character as a strong base.[1]

The molecule's structure also allows the two cyclam rings to act as chelating agents for divalent metal ions, with a particular affinity for zinc (Zn2+), copper (Cu2+), and nickel (Ni2+).[13] The biologically active form of plerixafor is believed to be its zinc complex, which further influences its interaction with its biological target.[13]

The structure of plerixafor is not merely a chemical curiosity; it is the blueprint for its function and fate within the body. The symmetrical bicyclam architecture, with its eight basic nitrogen atoms, results in a molecule that is highly charged and water-soluble at physiological pH.[4] This high positive charge is the key that unlocks its pharmacological effect, enabling strong electrostatic interactions with complementary negatively charged amino acid residues within the binding pocket of the CXCR4 receptor.[4] This same feature—high charge and hydrophilicity—profoundly influences its pharmacokinetic profile. Charged molecules are generally poor substrates for the lipophilic enzymes of the cytochrome P450 system, explaining why plerixafor is not significantly metabolized.[13] Consequently, the body relies on the kidneys to eliminate the unchanged, water-soluble drug, making renal function the critical determinant of its clearance.[13] This direct line from molecular structure to mechanism of action and route of elimination creates a cohesive understanding of the drug's benefits and risks.

Table 2.1: Key Identifiers and Properties of Plerixafor

Identifier TypeValueSource Snippet(s)
DrugBank IDDB068091
CAS Number110078-46-11
UNIIS915P5499N1
Molecular FormulaC28​H54​N8​16
Molecular Weight502.8 g/mol1
IUPAC Name1,1'-[1,4-phenylenebis(methylene)]-bis-1,4,8,11-tetraazacyclotetradecane1
Physical FormSolid1
Melting Point131.5 °C1
SolubilitySlightly soluble1
pKa8.5-11.5 and <2.41

Pharmacology: Mechanism of Action and Pharmacodynamic Effects

Plerixafor's therapeutic effect is derived from its precise and potent modulation of a fundamental biological pathway governing cell trafficking. Its journey from a failed antiviral candidate to a cornerstone of transplant medicine is a testament to the importance of understanding a drug's mechanism and observing its pharmacodynamic effects.

The CXCR4/SDF-1α Axis: The "Anchor and Cable" of HSC Retention

The retention of hematopoietic stem cells (HSCs) within the protective microenvironment of the bone marrow is not a passive process. It is actively maintained by a complex network of cellular interactions and signaling pathways. Central to this process is the interaction between the C-X-C chemokine receptor type 4 (CXCR4) and its sole cognate ligand, stromal cell-derived factor-1-alpha (SDF-1α), also known as CXCL12.[1]

CXCR4 is a seven-transmembrane G-protein-coupled receptor (GPCR) ubiquitously expressed on the surface of many cell types, including hematopoietic cells, most notably CD34+ HSCs.[4] Its ligand, SDF-1α, is constitutively expressed and secreted by bone marrow stromal cells.[2] The binding of SDF-1α to CXCR4 on HSCs initiates a signaling cascade that promotes cell adhesion and retention. This interaction functions as a biological "anchor and cable," tethering the stem cells to the marrow niche and regulating their homing and egress.[1] As long as this axis is intact, HSCs remain quiescent and sequestered within the bone marrow, with only a small number circulating in the peripheral blood.

Selective and Reversible Antagonism of CXCR4 by Plerixafor

Plerixafor functions as a highly selective, potent, and reversible antagonist of the CXCR4 receptor.[1] It physically occupies the ligand-binding pocket on the receptor, thereby competitively blocking the binding of SDF-1α.[4] By severing this "anchor," plerixafor disrupts the primary retention signal that holds HSCs in the marrow.[2]

The molecular basis for this interaction is a unique binding mode characterized by strong charge-charge interactions. The highly positive (+4) charge of the protonated plerixafor molecule forms electrostatic bonds with three key negatively charged acidic amino acid residues within the CXCR4 binding site: Asp171, Asp262, and Glu288.[4] This tight-binding interaction explains its potency, with an IC50 (half maximal inhibitory concentration) for CXCR4 reported to be 44 nM.[16] Plerixafor's selectivity is a key attribute; it has been tested against a wide panel of other chemokine receptors (including CXCR1, CXCR2, CXCR3, CXCR7, and multiple CCRs) and has shown no significant inhibitory activity, confirming its specificity for CXCR4.[4]

While plerixafor is overwhelmingly classified as a pure antagonist, with no ability to elicit downstream signaling on its own, some reports suggest it may exhibit weak partial agonist activity at very high concentrations or with mutated forms of the receptor.[4] Additionally, it has been described as an allosteric agonist of CXCR7, another receptor for SDF-1α, although the clinical relevance of this secondary interaction remains less defined.[13]

The origin story of plerixafor provides a compelling narrative of drug repurposing and the power of clinical observation. The molecule was not initially designed for hematology. It was first synthesized in 1987 and later developed by AnorMED under the code name AMD3100 as a potential anti-HIV drug.[4] The therapeutic hypothesis was sound: certain strains of HIV (T-tropic or X4-using) utilize the CXCR4 receptor, in addition to CD4, to gain entry into T-cells.[4] By blocking this co-receptor, plerixafor was intended to prevent viral infection. During Phase I clinical trials in HIV-infected individuals, however, investigators noted a consistent and unexpected "side effect": a rapid and significant increase in the number of circulating white blood cells.[4] Rather than dismissing this as a simple demargination effect, researchers, notably led by Hal Broxmeyer, correctly deduced that this leukocytosis was the result of active mobilization of progenitor cells from the bone marrow.[4] This serendipitous pharmacodynamic finding was a turning point. While its development as an anti-HIV agent was ultimately not pursued, its profound ability to mobilize HSCs was recognized as a far more promising therapeutic application, leading to a complete pivot in its clinical development path and its eventual success as a hematologic drug.[4] This history underscores how a drug engineered for high potency and specificity against a target can reveal new therapeutic avenues when its effects are rigorously observed in humans.

Pharmacodynamic Outcomes: Rapid and Predictable Mobilization

The direct pharmacodynamic consequence of CXCR4 antagonism by plerixafor is the rapid, transient, and dose-dependent mobilization of HSCs from the bone marrow into the peripheral circulation.[4] This manifests as a measurable increase in total circulating leukocytes (leukocytosis) and, critically for its therapeutic purpose, a substantial elevation in the number of CD34+ stem cells in the peripheral blood.[2]

The kinetics of this effect are remarkably swift and predictable. Following a single subcutaneous injection, the concentration of circulating CD34+ cells begins to rise, reaching a peak approximately 6 to 9 hours post-administration.[2] This predictable peak is the basis for the clinical recommendation to perform apheresis (the process of collecting the stem cells from the blood) within this time window to maximize the harvest.[11] This kinetic profile stands in stark contrast to that of G-CSF, the other agent used for mobilization. G-CSF works through a more indirect mechanism, requiring multiple days of administration to achieve its peak effect, which is often less predictable from patient to patient.[4] The rapid and reliable pharmacodynamic effect of plerixafor is one of its primary clinical advantages.

Pharmacokinetics: Absorption, Distribution, Metabolism, and Excretion (ADME)

The pharmacokinetic profile of plerixafor is characterized by its simplicity and predictability, which are direct consequences of its chemical properties. This profile underpins its dosing regimen and highlights its key safety considerations, particularly regarding renal function.

Absorption

Following subcutaneous (SC) administration, plerixafor is absorbed rapidly into the systemic circulation. Peak plasma concentrations (Tmax​) are consistently achieved within 30 to 60 minutes after injection.[11] Studies comparing SC to intravenous administration suggest that its bioavailability is very high, approaching 100%.[21] This rapid and complete absorption ensures a reliable onset of action, which is crucial for timing apheresis procedures.

Distribution

Once in the bloodstream, plerixafor exhibits moderate binding to human plasma proteins, with approximately 58% of the drug being bound.[11] Its apparent volume of distribution (

Vd​) in humans is reported to be 0.3 L/kg.[11] This value indicates that while the drug distributes out of the plasma into the extravascular fluid space, it does not extensively accumulate in tissues, consistent with its hydrophilic nature.

Metabolism

A defining feature of plerixafor's pharmacokinetics is its lack of significant metabolism.[13] Its highly charged, water-soluble structure makes it a poor substrate for the hepatic cytochrome P450 (CYP450) enzyme system. In vitro studies using human liver microsomes and primary hepatocytes have confirmed that plerixafor is not metabolized by these enzymes.[18] Furthermore, plerixafor does not inhibit or induce the major CYP450 isoenzymes (including CYP1A2, CYP2B6, and CYP3A4) or the drug transporter P-glycoprotein.[13]

Excretion

Given its lack of metabolism, plerixafor is eliminated from the body primarily through renal excretion of the unchanged drug.[13] In individuals with normal renal function, approximately 70% of an administered dose is recovered unchanged in the urine within the first 24 hours.[13] The elimination from plasma follows a biphasic pattern with a terminal half-life (

t1/2​) of 3 to 5 hours.[11] This relatively short half-life means the drug is cleared from the system quickly, minimizing the risk of prolonged effects.

The pharmacokinetic profile of plerixafor can be described as remarkably "clean," but with a single, critical vulnerability. Its near-total lack of hepatic metabolism is a significant clinical advantage, especially in oncology patients who are often on complex, multi-drug regimens.[18] This "clean" profile minimizes the risk of pharmacokinetic drug-drug interactions mediated by the CYP450 system, which are a common source of toxicity and therapeutic failure.[18] However, this metabolic inertness comes at a cost: a heavy reliance on a single organ system for elimination. With approximately 70% of the drug cleared unchanged by the kidneys, renal function becomes the sole determinant of its systemic exposure.[13] This creates a single point of failure. In patients with compromised renal function, the drug cannot be cleared effectively, leading to accumulation, prolonged exposure, and an increased risk of toxicity. This pharmacokinetic reality is the direct reason for the most important safety precaution associated with the drug: the mandatory dose adjustment in patients with renal impairment.[18]

Table 4.1: Summary of Plerixafor Pharmacokinetic Parameters

ParameterValueClinical ImplicationSource Snippet(s)
Route of AdministrationSubcutaneous (SC)Standard clinical route.11
Peak Plasma Time (Tmax​)30–60 minutesAllows for precise timing of apheresis 6-11 hours post-dose to coincide with peak HSC mobilization.11
Protein Binding~58%Moderate binding; a significant fraction of the drug is free and active.11
Volume of Distribution (Vd​)0.3 L/kgDrug is primarily in the extravascular fluid space, not sequestered in tissues.11
MetabolismNegligibleVery low potential for drug-drug interactions with agents metabolized by CYP450 enzymes.13
Primary Route of ExcretionRenal (~70% unchanged)Dose reduction is mandatory in patients with moderate-to-severe renal impairment.13
Elimination Half-life (t1/2​)3–5 hoursRapid clearance allows for daily dosing without significant accumulation in patients with normal renal function.11

Clinical Application and Efficacy

Plerixafor has established itself as an indispensable tool in the setting of autologous stem cell transplantation, providing a reliable method to ensure adequate stem cell collection, particularly in challenging patient populations.

Approved Indications and Therapeutic Role

Plerixafor is approved by the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for use in combination with a granulocyte-colony stimulating factor (G-CSF), such as filgrastim.[1] Its specific indication is to mobilize hematopoietic stem cells to the peripheral blood for collection (via apheresis) and subsequent autologous transplantation in adult patients with non-Hodgkin's lymphoma (NHL) and multiple myeloma (MM).[1] The EMA has also approved a pediatric indication for mobilizing HSCs in children with lymphoma or malignant solid tumors.[1]

The primary therapeutic role of plerixafor is to augment the mobilization effect of G-CSF. While G-CSF alone is effective for many, a significant proportion of patients (estimated at 5-30%) fail to mobilize a sufficient number of CD34+ cells to proceed with transplantation.[2] This is particularly common in patients with risk factors for poor mobilization, such as advanced age, extensive prior chemotherapy or radiation, or treatment with certain agents like lenalidomide.[2] Plerixafor is used either "upfront" in patients predicted to be poor mobilizers or as a "just-in-time" or "rescue" therapy for those who have a suboptimal response to G-CSF alone.[6]

Pivotal Clinical Trials and Efficacy Data

The regulatory approvals for plerixafor were based on the strength of two pivotal, multicenter, randomized, double-blind, placebo-controlled Phase III studies.[6] These trials definitively established its superiority when added to G-CSF.

  • NHL Study: In the trial involving 298 adult patients with NHL, the addition of plerixafor to G-CSF resulted in a significantly higher rate of successful mobilization. 59% of patients in the plerixafor arm achieved the target collection of ≥5×106 CD34+ cells/kg within four or fewer apheresis days, compared to only 20% of patients in the placebo plus G-CSF arm.[7]
  • MM Study: In the trial of 302 adult patients with MM, the results were similarly compelling. 72% of patients receiving plerixafor plus G-CSF met the collection target of ≥6×106 CD34+ cells/kg within four or fewer apheresis days, versus just 34% in the placebo group.[7]

Subsequent systematic reviews and meta-analyses have reinforced these findings. One large meta-analysis calculated that patients receiving the plerixafor/G-CSF combination had an odds ratio of 5.33 for achieving the predetermined apheresis yield compared to those receiving G-CSF alone.[8] This robust effect was observed across patient populations with MM, NHL, and Hodgkin's lymphoma (HL).[8] A key clinical benefit highlighted in these trials is the ability to reach the target cell dose in fewer apheresis sessions, which reduces the physical burden on the patient, minimizes procedural risks, and can lead to significant cost savings for the healthcare system.[1]

Dosing and Administration

The administration of plerixafor follows a specific and timed protocol to maximize its efficacy.

  • Standard Dose: The recommended dose is a subcutaneous injection of 0.24 mg/kg, calculated based on the patient's actual body weight.[11]
  • Timing: Treatment with plerixafor is initiated only after the patient has received four consecutive daily morning doses of G-CSF (typically 10 µg/kg).[2] The plerixafor injection is administered in the evening, approximately 6 to 11 hours prior to the planned start of the apheresis procedure the next morning.[11]
  • Dose Capping and Duration: Due to increasing systemic exposure with higher body weights, the total daily dose of plerixafor should not exceed 40 mg.[11] The optimal dosing for patients weighing more than 175% of their ideal body weight has not been formally investigated.[18] Treatment can be continued daily for up to four consecutive days, with some clinical trial experience extending to seven days if necessary to reach the collection goal.[11]

Use in Special Populations

  • Renal Impairment: As plerixafor is primarily cleared by the kidneys, dose adjustment is critical in patients with renal dysfunction. For patients with moderate to severe renal impairment, defined as a creatinine clearance (Clcr​) of ≤50 mL/min, the dose must be reduced by one-third to 0.16 mg/kg/day. The maximum daily dose in this population should not exceed 27 mg.[18]
  • Pregnancy and Lactation: Plerixafor is classified as US Pregnancy Category D. It has demonstrated teratogenicity in animal studies and can cause harm to a developing fetus.[10] Therefore, it is essential to verify pregnancy status before initiation. Females of reproductive potential must use an effective form of contraception during treatment and for one week following the final dose.[10] It is unknown if plerixafor is excreted in human milk, but due to the potential for serious adverse reactions in the infant, breastfeeding should be discontinued during therapy and for one week after.[18]

The high cost of brand-name Mozobil historically created a significant barrier to its use, forcing a clinical and economic debate between two main strategies: "just-in-time" (JIT) versus "upfront" administration. The JIT, or rescue, approach was born from economic necessity; institutions developed risk-adapted protocols where plerixafor was reserved only for patients who were actively failing to mobilize after an initial attempt with G-CSF alone.[6] This conserved resources but risked delayed or failed collections. In contrast, an "upfront" strategy involves administering plerixafor from the start of mobilization to all patients, or at least to those with known risk factors for poor mobilization (e.g., MM patients requiring high cell yields for tandem transplants).[6] Studies began to show that for these high-risk groups, the upfront approach could actually be more cost-effective by reliably reducing the number of apheresis days and preventing outright mobilization failure.[6] The recent market entry of multiple, lower-cost generic versions of plerixafor is a transformative event.[12] This development is poised to resolve the debate by diminishing the economic driver behind the restrictive JIT strategy. As the cost barrier lowers, clinical practice is likely to shift towards more widespread upfront use, particularly in patients with any risk factors, with the goal of making stem cell collection more predictable, efficient, and successful for a broader patient population.

Safety and Tolerability Profile

Plerixafor has a well-established safety profile derived from robust clinical trials and over a decade of post-marketing experience. While generally well-tolerated, it is associated with a predictable set of adverse effects and requires adherence to specific warnings and precautions.

Common and Serious Adverse Events

The adverse reactions associated with plerixafor are typically mild to moderate in severity and transient in nature.

  • Common Adverse Reactions (incidence ≥10%): The most frequently reported side effects in pivotal clinical trials were predominantly gastrointestinal and constitutional. These include diarrhea (37%), nausea (34%), injection site reactions (such as pain, erythema, and swelling; 34%), fatigue (27%), headache (22%), arthralgia (13%), dizziness (11%), and vomiting (10%).[10] The majority of these events were Grade 1 or 2.[10]
  • Serious Adverse Reactions: Although uncommon, serious events have been reported. The most significant of these are serious hypersensitivity reactions, including life-threatening anaphylactic shock, which have occurred in less than 1% of patients.[10] For this reason, patients should be observed for at least 30 minutes after administration in a setting where treatment for anaphylaxis is immediately available.[10] Vasovagal reactions, orthostatic hypotension, and syncope have also been reported in less than 1% of subjects following subcutaneous injection.[32]

Table 6.1: Incidence of Common Adverse Reactions (≥10%) in Mobilization Clinical Trials

Adverse ReactionPlerixafor + G-CSF (%)Placebo + G-CSF (%)Source Snippet(s)
Diarrhea37Not specified10
Nausea34Not specified10
Injection Site Reactions34Not specified10
Fatigue27Not specified10
Headache22Not specified10
Arthralgia13Not specified10
Dizziness11Not specified10
Vomiting10Not specified10
Note: Placebo arm data was not consistently available in the provided sources for a direct comparison.

Warnings, Precautions, and Contraindications

There are no FDA-issued "black box" warnings for plerixafor. The only formal contraindication is a history of a serious hypersensitivity reaction to the drug.[11] However, several important warnings and precautions must be observed.

  • Splenic Enlargement and Rupture: Cases of splenic enlargement and, rarely, splenic rupture have been reported following the co-administration of plerixafor and G-CSF. This is a known risk of G-CSF therapy that may be exacerbated. Patients who report left upper abdominal pain, scapular pain, or shoulder pain should be promptly evaluated for splenic integrity.[13]
  • Tumor Cell Mobilization: A significant mechanistic concern is the potential for plerixafor to mobilize malignant cells from the bone marrow niche along with normal HSCs. This is because many hematologic malignancies, including lymphoma and myeloma, express CXCR4 and utilize the SDF-1α axis for homing and survival.[2] Due to this risk, plerixafor is not intended for HSC mobilization in patients with leukemia, as it may contaminate the apheresis product with leukemic cells.[10] For NHL and MM, the potential for tumor cell collection and subsequent reinfusion exists. The long-term clinical consequence of this phenomenon has not been well-studied and remains a key area of post-marketing evaluation.[10]
  • Hematologic Effects:
  • Leukocytosis: The combination of plerixafor and G-CSF induces a marked increase in circulating leukocytes. White blood cell counts must be monitored during therapy.[10]
  • Thrombocytopenia: A decrease in platelet counts has been observed in patients receiving plerixafor. Platelet counts should be monitored in all patients, particularly before and after apheresis procedures, which can also lower platelet levels.[10]
  • Embryo-Fetal Toxicity: Plerixafor has demonstrated teratogenic effects in animal models and is presumed to carry a risk of fetal harm in humans. Strict contraception measures are required for both male and female patients during treatment and for one week after the final dose.[10]

The most significant clinical uncertainty surrounding the long-term use of plerixafor is the "known unknown" of tumor cell mobilization. The biological principle is straightforward: the drug works by disrupting the CXCR4/SDF-1α anchor that holds cells in the bone marrow.[4] Since many lymphoma and myeloma cells also express CXCR4 and use this same anchor for survival and drug resistance, it is biologically inevitable that plerixafor will dislodge them along with the desired HSCs.[2] These mobilized malignant cells are then collected during apheresis and, critically, are re-infused back into the patient following high-dose, myeloablative chemotherapy.[10] The central question, which is repeatedly highlighted as "not well-studied," is whether this re-infusion of viable tumor cells increases the long-term risk of disease relapse.[10] While the potent chemotherapy is intended to eradicate any residual disease, the practice of re-seeding the patient with their own cancer cells is a source of clinical concern. This issue represents a major gap in the current understanding of plerixafor's risk-benefit profile and underscores the need for long-term follow-up studies and large-scale registry analyses to determine if there is any impact on overall survival or progression-free survival.

Drug-Drug Interactions

Plerixafor has a very favorable drug-drug interaction profile due to its pharmacokinetic properties.

  • Pharmacokinetic Interactions: As it is not a substrate, inhibitor, or inducer of the CYP450 enzyme system or P-glycoprotein, the potential for metabolic drug interactions is extremely low.[13] This is a major advantage in cancer patients who are often receiving multiple medications. The only theoretical pharmacokinetic interaction risk would be with other drugs that are actively secreted by the kidneys or that reduce renal function, as this could lead to increased plerixafor concentrations. However, no specific severe interactions have been formally identified or reported.[11]
  • Pharmacodynamic Interactions: In clinical studies, the addition of the monoclonal antibody rituximab to a mobilization regimen of plerixafor and G-CSF did not have a negative impact on patient safety or CD34+ cell yield.[18]

The Evolving Therapeutic Landscape

After more than a decade as a unique therapeutic agent, plerixafor is now situated within a rapidly evolving clinical and commercial landscape. Its established mechanism has inspired research into new indications, while the arrival of both generic versions and a new mechanistic competitor is reshaping its clinical use and economic considerations.

Investigational and Off-Label Uses

The targeted mechanism of CXCR4 antagonism has prompted investigation of plerixafor in a variety of conditions beyond its approved indications.

  • WHIM Syndrome: This rare, congenital primary immunodeficiency is caused by an autosomal dominant gain-of-function mutation in the CXCR4 gene, leading to hyper-retention of neutrophils and other leukocytes in the bone marrow. Plerixafor represents a direct, mechanism-based therapy. Phase 1 clinical trials using long-term, low-dose subcutaneous plerixafor have shown promise in correcting the chronic neutropenia and other cytopenias associated with the syndrome, providing preliminary evidence for its efficacy in this orphan disease.[35]
  • Solid Tumors: The CXCR4/SDF-1α axis is implicated in tumor metastasis and angiogenesis. Preclinical studies in mouse models of glioblastoma have shown that plerixafor can reduce tumor recurrence after radiotherapy by blocking the recruitment of bone marrow-derived pro-vasculogenic cells to the tumor site.[13] It has also been investigated in pancreatic cancer, where blocking the CXCR4 pathway was shown to increase the efficacy of chemotherapy.[2]
  • Expanded Hematologic Use: While not formally approved for these uses, National Comprehensive Cancer Network (NCCN) guidelines support the use of plerixafor for HSC mobilization in patients with Hodgkin lymphoma and for allogeneic (donor) transplants where the donor has an insufficient collection with G-CSF alone.[36]
  • Gene Therapy: Plerixafor is utilized as a mobilization agent to harvest autologous HSCs for the manufacturing of ex vivo gene therapies, such as Zynteglo (betibeglogene autotemcel), which is approved for the treatment of beta-thalassemia.[36]
  • Other Research Areas: Plerixafor has been explored in diverse preclinical and early clinical settings, including for its potential to counteract opioid-induced hyperalgesia (in animal models) and to accelerate wound healing in diabetic mice, though a 2020 study in humans found no benefit for diabetic arterial insufficiency ulcers.[13]

Comparative Efficacy and Positioning

Plerixafor's position in the therapeutic armamentarium is now defined not only by its own merits but also by its comparison to alternatives and competitors.

  • Brand (Mozobil) vs. Generic Plerixafor: The recent availability of generic plerixafor in the US and Europe is a major development.[12] Retrospective clinical studies have been conducted to ensure therapeutic equivalence. A 2024 study comparing brand-name Mozobil to a generic formulation in multiple myeloma patients found no significant difference in the primary endpoints of cumulative CD34+ cell yield or post-transplant engraftment times.[12] Notably, the study reported that the generic cohort required significantly fewer plerixafor doses and fewer apheresis days to reach their collection goal, suggesting at least equivalent, and possibly more efficient, mobilization.[12] These findings provide clinical reassurance that lower-cost generics can be used interchangeably with the brand-name product.
  • Plerixafor vs. Other Mobilization Agents:
  • G-CSF Alone: The combination of plerixafor and G-CSF is unequivocally superior to G-CSF alone in achieving target stem cell yields, particularly in patients who mobilize poorly.[8] A meta-analysis showed the combination was over five times more likely to result in a successful collection.[8]
  • Chemomobilization: The use of cytotoxic chemotherapy (e.g., cyclophosphamide) followed by G-CSF is another effective mobilization strategy that can yield high cell counts.[38] However, it is associated with greater toxicity, including neutropenic fever, and less predictable timing for apheresis compared to the plerixafor/G-CSF regimen.[38]
  • Motixafortide (Aphexda): Approved by the FDA in September 2023, motixafortide is a second-generation CXCR4 antagonist and the first direct competitor to plerixafor.[40] It is a cyclic peptide, distinguishing it structurally from the bicyclam plerixafor.[42] Indicated for HSC mobilization in MM patients, it shares the same mechanism of action.[44] In its pivotal GENESIS trial, motixafortide plus filgrastim enabled 67.5% of patients to achieve the primary collection goal, compared to 9.5% for placebo plus filgrastim.[46] The approval of motixafortide validates CXCR4 as a key mobilization target and introduces market competition for the first time.

The choice of mobilization strategy is a complex decision based on patient diagnosis, prior treatments, institutional protocols, and cost. The introduction of generic plerixafor and a new brand-name competitor, motixafortide, has expanded the options and necessitates a nuanced understanding of each approach.

Table 7.1: Comparative Overview of HSC Mobilization Agents

Agent/RegimenMechanism of ActionTypical Use CaseKey AdvantagesKey Disadvantages
G-CSF AloneInduces HSC proliferation and egress via indirect mechanisms.Standard mobilization for healthy donors and many first-line patients.Well-established, relatively low cost.5-30% failure rate, multi-day administration, unpredictable kinetics, side effects (e.g., bone pain). 2
Chemomobilization + G-CSFCytotoxic agents damage the marrow niche, followed by G-CSF-driven rebound hematopoiesis.Often used in lymphoma as part of salvage therapy.Can yield very high cell counts; provides anti-tumor effect.High toxicity (e.g., neutropenic fever), unpredictable timing of collection, hospitalization often required. 38
Plerixafor + G-CSFSelective, reversible CXCR4 antagonism, blocking HSC retention.Upfront for poor mobilizers; rescue for G-CSF failures. Approved for NHL and MM.Rapid, predictable kinetics; high success rate; well-tolerated.High cost (brand); risk of tumor cell mobilization; requires renal dose adjustment. 4
Motixafortide + G-CSFSelective CXCR4 antagonism, blocking HSC retention.Approved for HSC mobilization in MM.High success rate in pivotal trial; introduces competition.High cost (brand); requires premedication for hypersensitivity; limited post-marketing data. 40

Regulatory and Commercial History

The trajectory of plerixafor from a repurposed molecule to a blockbuster drug, and now to a competitive generic market, provides a compelling case study in the pharmaceutical lifecycle.

Development and Approval Timeline

Plerixafor's journey to market was unconventional and spanned several decades and corporate entities.

  • Discovery and Early Development: The molecule was first synthesized in 1987 as part of basic chemical research.[13] It was later identified as a potent anti-HIV agent and developed under the code name AMD3100 by the biopharmaceutical company AnorMED.[5]
  • Acquisition and Strategic Pivot: In 2006, AnorMED was acquired by Genzyme Corporation.[2] Following the observation of its potent HSC-mobilizing effects in early trials, Genzyme made the pivotal decision to deprioritize the anti-HIV indication and focus development on its use in autologous transplantation.[4]
  • Regulatory Submission and Approval: Genzyme filed New Drug Applications (NDAs) with both the FDA and EMA in June 2008.[48] The FDA granted the application Priority Review status, recognizing its potential to address an unmet medical need.[48] On December 15, 2008, the FDA approved plerixafor injection, under the brand name Mozobil.[1] The drug was also granted orphan drug designation in both the United States and the European Union, a status given to therapies for rare diseases.[1] Genzyme was subsequently acquired by Sanofi, which managed the brand for most of its patent life.

Market Dynamics: The Impact of Generic Entry

For over a decade, the commercial landscape for plerixafor was defined by brand-name exclusivity.

  • Era of Brand Exclusivity (2008-2023): As the sole plerixafor product, Mozobil (Sanofi) commanded a premium price, with reports of a single vial costing over $8,000 to $10,000 in the United States.[28] This high cost was a significant factor limiting its routine use and led many transplant centers to develop restrictive, risk-adapted protocols to manage expenditures.[12] Despite these limitations, Mozobil became a successful product, with global sales reaching approximately $300 million in 2022.[50]
  • The Generic Wave (2023-Present): The patent exclusivity for plerixafor ended in 2023, triggering a paradigm shift. In July 2023, the FDA approved the first generic versions of plerixafor injection.[29] Multiple manufacturers, including Amneal Pharmaceuticals, Dr. Reddy's Laboratories, Teva Pharmaceuticals, and Meitheal Pharmaceuticals, have since entered the market.[29]
  • Economic and Clinical Implications: The introduction of generic competition has led to a significant reduction in the price of plerixafor.[6] This is expected to have profound effects on clinical practice by democratizing access to the drug. The economic pressures that favored "just-in-time" strategies are diminishing, likely leading to more widespread "upfront" use in patients with risk factors for poor mobilization. This shift has the potential to improve the efficiency and success rate of autologous transplants across the board.

Plerixafor's commercial journey represents the full circle of the pharmaceutical market lifecycle. It began as a high-value, first-in-class, innovative orphan drug whose high price directly influenced and constrained its clinical application.[28] Its market matured with the arrival of the first direct mechanistic competitor, motixafortide, in 2023, which validated the drug class but also signaled the end of its monopoly.[41] The nearly simultaneous loss of patent exclusivity and the flood of generic entrants has rapidly transformed plerixafor from a high-margin specialty product into a more commoditized therapeutic agent where price and access, rather than just efficacy, are key drivers of use.[6] Consequently, the clinical conversation is shifting. For a decade, the debate was "Plerixafor vs. G-CSF alone," a question of efficacy versus cost. The new debate will be "Plerixafor vs. motixafortide," a comparison of two active agents, while the cost of plerixafor itself becomes a less prohibitive factor in the decision-making process.

Conclusion and Future Directions

Plerixafor has secured its place in the history of hematology and oncology as a transformative therapeutic agent. As a selective CXCR4 antagonist, its introduction provided a novel, potent, and predictable mechanism for mobilizing hematopoietic stem cells, fundamentally improving the feasibility and success of autologous transplantation for patients with non-Hodgkin's lymphoma and multiple myeloma. Its well-characterized pharmacokinetic profile, manageable safety, and robust evidence base have made the combination of plerixafor and G-CSF the undisputed standard of care for enhancing HSC mobilization, especially in patients who are difficult to mobilize.

The therapeutic landscape for plerixafor is currently in a state of dynamic flux. The recent and concurrent arrival of multiple generic formulations and a new brand-name competitor, motixafortide, marks the end of its era as a niche, high-cost innovator. The availability of lower-cost generics is the single most significant recent development, poised to democratize access, reshape clinical protocols away from cost-based restrictions, and make successful mobilization achievable for a wider patient population.

Despite its success, critical questions and opportunities for future research remain. These will define the next chapter in the story of plerixafor.

  • Defining Long-Term Safety: The most pressing unanswered question is the long-term clinical impact of re-infusing tumor cells that are inevitably mobilized along with HSCs. While no definitive signal of harm has emerged, this remains a theoretical risk that requires rigorous investigation through long-term follow-up of patients from pivotal trials and large-scale, real-world registry analyses to assess any potential impact on relapse rates and overall survival.
  • Optimizing Use in the Modern Era: The treatment paradigms for lymphoma and myeloma continue to evolve with the introduction of new classes of drugs (e.g., CD38-targeted antibodies, novel immunomodulators) that can themselves impact a patient's ability to mobilize stem cells. Future research must focus on defining the optimal timing and use of plerixafor within these modern, multi-agent induction regimens to ensure transplant remains a viable option.
  • Expanding Therapeutic Horizons: The promise of plerixafor in treating the rare genetic disorder WHIM syndrome warrants further, more definitive clinical trials. Its potential as an anti-metastatic or chemosensitizing agent in solid tumors, while still in early stages, represents an exciting avenue for exploration. Formalizing its role in allogeneic donation and other hematologic malignancies through prospective trials is also a logical next step.
  • Navigating a Competitive Market: With the entry of motixafortide, the era of CXCR4 antagonist competition has begun. Head-to-head clinical trials comparing the efficacy, safety, and overall cost-effectiveness of plerixafor-based regimens versus motixafortide-based regimens will be essential to guide clinical decision-making and formulary choices in the years to come.

In conclusion, plerixafor has transitioned from a serendipitous discovery to an essential component of cancer care. Its future will be shaped by ongoing efforts to refine its use, answer critical long-term safety questions, and explore its full therapeutic potential in a newly competitive and accessible market.

Works cited

  1. Plerixafor | C28H54N8 | CID 65015 - PubChem, accessed July 29, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Plerixafor
  2. Plerixafor – Knowledge and References - Taylor & Francis, accessed July 29, 2025, https://taylorandfrancis.com/knowledge/Medicine_and_healthcare/Pharmaceutical_medicine/Plerixafor/
  3. Plerixafor | Leukemia and Lymphoma Society, accessed July 29, 2025, https://www.lls.org/drug/plerixafor
  4. Physiology and Pharmacology of Plerixafor - PMC, accessed July 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3776399/
  5. Mozobil® (Plerixafor, AMD3100), 10 years after its approval by the US Food and Drug Administration, accessed July 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6379795/
  6. Comparison of the efficacy of a generic plerixafor versus Mozobil as adjunct peripheral blood stem cell mobilization agents in multiple myeloma patients - PMC, accessed July 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11637246/
  7. Mozobil | European Medicines Agency (EMA), accessed July 29, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/mozobil
  8. G-CSF+ plerixafor versus G-CSF alone mobilized hematopoietic ..., accessed July 29, 2025, https://www.tandfonline.com/doi/full/10.1080/07853890.2024.2329140
  9. Efficacy and safety of plerixafor for hematopoietic stem cell mobilization for autologous transplantation in patients with non-Hodgkin lymphoma and multiple myeloma: A systematic review and meta-analysis - PMC - PubMed Central, accessed July 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6614714/
  10. Mozobil® (plerixafor) injection | HCP - Sanofi campus, accessed July 29, 2025, https://pro.campus.sanofi/us/products/mozobil
  11. Mozobil (plerixafor) dosing, indications, interactions, adverse effects, and more, accessed July 29, 2025, https://reference.medscape.com/drug/mozobil-plerixafor-342168
  12. Comparison of the Efficacy of Generic Plerixafor Versus Mozobil as Adjunct Peripheral Blood Stem-Cell Mobilization Agents in Patients With Multiple Myeloma - Journal of Hematology Oncology Pharmacy, accessed July 29, 2025, https://www.jhoponline.com/issue-archive/2024-issues/march-2024-vol-14-special-feature/comparison-of-the-efficacy-of-generic-plerixafor-versus-mozobil-as-adjunct-peripheral-blood-stem-cell-mobilization-agents-in-patients-with-multiple-myeloma
  13. Plerixafor - Wikipedia, accessed July 29, 2025, https://en.wikipedia.org/wiki/Plerixafor
  14. Chemical structure of plerixafor. | Download Scientific Diagram - ResearchGate, accessed July 29, 2025, https://www.researchgate.net/figure/Chemical-structure-of-plerixafor_fig1_49603394
  15. Definition of plerixafor - NCI Drug Dictionary, accessed July 29, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/plerixafor
  16. Plerixafor free base | CAS#110078-46-1 | immunostimulant | MedKoo, accessed July 29, 2025, https://www.medkoo.com/products/20129
  17. Plerixafor: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed July 29, 2025, https://go.drugbank.com/drugs/DB06809
  18. Mozobil, INN-plerixafor - EMA, accessed July 29, 2025, https://www.ema.europa.eu/en/documents/product-information/mozobil-epar-product-information_en.pdf
  19. Plerixafor - brand name list from Drugs.com, accessed July 29, 2025, https://www.drugs.com/ingredient/plerixafor.html
  20. Plerixafor is a Selective CXCR4 Antagonist and Mobilizes Hematopoietic Stem Cells in Cancer, accessed July 29, 2025, https://www.cancer-research-network.com/2024/01/15/plerixafor-is-a-selective-cxcr4-antagonist-and-mobilizes-hematopoietic-stem-cells-in-cancer/
  21. Fig. 1. Pharmacokinetics and pharmacodynamics of plerixafor. A : plasma... - ResearchGate, accessed July 29, 2025, https://www.researchgate.net/figure/Pharmacokinetics-and-pharmacodynamics-of-plerixafor-A-plasma-clearance-of-14_fig1_264390786
  22. Plerixafor Monograph for Professionals - Drugs.com, accessed July 29, 2025, https://www.drugs.com/monograph/plerixafor.html
  23. Plerixafor - NCI, accessed July 29, 2025, https://www.cancer.gov/about-cancer/treatment/drugs/plerixafor
  24. Stem Cell Mobilization with and without Plerixafor: A Comparative Analysis, accessed July 29, 2025, https://www.heraldopenaccess.us/openaccess/stem-cell-mobilization-with-and-without-plerixafor-a-comparative-analysis
  25. G-CSF + plerixafor versus G-CSF alone mobilized hematopoietic stem cells in patients with multiple myeloma and lymphoma: a systematic review and meta-analysis - Taylor & Francis Online, accessed July 29, 2025, https://www.tandfonline.com/doi/abs/10.1080/07853890.2024.2329140
  26. Plerixafor: Side Effects, Uses, Dosage, Interactions, Warnings - Rx List, accessed July 29, 2025, https://www.rxlist.com/plerixafor/generic-drug.htm
  27. plerixafor - Wikidata, accessed July 29, 2025, https://www.wikidata.org/wiki/Q905835
  28. Efficacy and Cost Analysis of a Plerixafor Protocol for Peripheral Blood Stem-Cell Mobilization in Patients with Multiple Myeloma or Non-Hodgkin Lymphoma - Journal of Hematology Oncology Pharmacy, accessed July 29, 2025, https://www.jhoponline.com/issue-archive/2016-issues/jhop-december-2016-vol-6-no-4/efficacy-and-cost-analysis-of-a-plerixafor-protocol-for-peripheral-blood-stem-cell-mobilization-in-patients-with-multiple-myeloma-or-non-hodgkin-lymphoma
  29. Generic Mozobil Availability - Drugs.com, accessed July 29, 2025, https://www.drugs.com/availability/generic-mozobil.html
  30. Mozobil (plerixafor): Uses, Side Effects, Dosage & Reviews - GoodRx, accessed July 29, 2025, https://www.goodrx.com/mozobil/what-is
  31. Mozobil (Plerixafor Injection): Side Effects, Uses, Dosage, Interactions, Warnings - Rx List, accessed July 29, 2025, https://www.rxlist.com/mozobil-drug.htm
  32. MEDICAL REVIEW(S) - accessdata.fda.gov, accessed July 29, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2008/022311s000_MedR.pdf
  33. Plerixafor (Mozobil): Uses, Side Effects, Interactions, Pictures, Warnings & Dosing - WebMD, accessed July 29, 2025, https://www.webmd.com/drugs/2/drug-151672/mozobil-subcutaneous/details
  34. Plerixafor Uses, Side Effects & Warnings - Drugs.com, accessed July 29, 2025, https://www.drugs.com/mtm/plerixafor.html
  35. A phase 1 clinical trial of long-term, low-dose treatment of WHIM syndrome with the CXCR4 antagonist plerixafor | Blood | American Society of Hematology, accessed July 29, 2025, https://ashpublications.org/blood/article/123/15/2308/32255/A-phase-1-clinical-trial-of-long-term-low-dose
  36. Mozobil (plerixafor) - Medical Drug Clinical Criteria, accessed July 29, 2025, https://www.carelonrx.com/ms/pharmacyinformation/Mozobil.pdf
  37. Comparison of the efficacy of a generic plerixafor versus Mozobil as adjunct peripheral blood stem cell mobilization agents in multiple myeloma patients - PubMed, accessed July 29, 2025, https://pubmed.ncbi.nlm.nih.gov/39367642/
  38. New Strategies for Stem Cell Mobilization - PMC, accessed July 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3499993/
  39. Plerixafor and Filgrastim Following Cyclophosphamide for Stem Cell Mobilization in Patients With Multiple Myeloma | ClinicalTrials.gov, accessed July 29, 2025, https://clinicaltrials.gov/study/NCT01074060
  40. 217159Orig1s000 - accessdata.fda.gov, accessed July 29, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2023/217159Orig1s000IntegratedR.pdf
  41. Aphexda (motixafortide) FDA Approval History - Drugs.com, accessed July 29, 2025, https://www.drugs.com/history/aphexda.html
  42. go.drugbank.com, accessed July 29, 2025, https://go.drugbank.com/drugs/DB14939#:~:text=Similar%20in%20mechanism%20to%20the,cells%20to%20bone%20marrow%20matrix.
  43. Motixafortide | C97H144FN33O19S2 | CID 91865076 - PubChem, accessed July 29, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Motixafortide
  44. Motixafortide: Uses, Interactions, Mechanism of Action - DrugBank, accessed July 29, 2025, https://go.drugbank.com/drugs/DB14939
  45. What is the mechanism of Motixafortide? - Patsnap Synapse, accessed July 29, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-motixafortide
  46. FDA Approved Aphexda, in Combination With Filgrastim, to Mobilize Stem Cells for Transplant in Patients With Multiple Myeloma - American Health & Drug Benefits, accessed July 29, 2025, https://www.ahdbonline.com/web-exclusives/fda-approvals/fda-approved-aphexda-in-combination-with-filgrastim-to-mobilize-stem-cells-for-transplant-in-patients-with-multiple-myeloma
  47. FDA Approves Motixafortide Plus Filgrastim for Stem Cell Mobilization in Multiple Myeloma, accessed July 29, 2025, https://www.ajmc.com/view/fda-approves-motixafortide-plus-filgrastim-for-stem-cell-mobilization-in-multiple-myeloma
  48. Mozobil (plerixafor) FDA Approval History - Drugs.com, accessed July 29, 2025, https://www.drugs.com/history/mozobil.html
  49. Plerixafor Approved for Stem Cell Transplant - CancerNetwork, accessed July 29, 2025, https://www.cancernetwork.com/view/plerixafor-approved-stem-cell-transplant
  50. Generic PLERIXAFOR INN entry, pharmaceutical patent expiration information and freedom to operate - DrugPatentWatch, accessed July 29, 2025, https://www.drugpatentwatch.com/p/generic-api/PLERIXAFOR
  51. Mozobil Prices - U.S. & International | PharmacyChecker.com, accessed July 29, 2025, https://www.pharmacychecker.com/mozobil/
  52. Plerixafor 20 mg / mL Injection 1.2 mL - McKesson Medical-Surgical, accessed July 29, 2025, https://mms.mckesson.com/product/1236824/Amneal-Pharmaceuticals-LLC-70121169402
  53. Plerixafor Coupons 2025: Up to 80% Discount - SingleCare, accessed July 29, 2025, https://www.singlecare.com/prescription/plerixafor
  54. Plerixafor Prices, Coupons, Copay Cards & Patient Assistance - Drugs.com, accessed July 29, 2025, https://www.drugs.com/price-guide/plerixafor

Published at: July 29, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.