Lenalidomide: A Comprehensive Monograph on its Pharmacology, Clinical Utility, and Evolving Therapeutic Landscape
Introduction: The Evolution and Clinical Significance of Lenalidomide
Lenalidomide stands as a cornerstone in the modern treatment of several hematologic malignancies, most notably multiple myeloma (MM). Its development represents a pivotal chapter in the history of pharmacology, born from the effort to refine the therapeutic potential of its predecessor, thalidomide, while mitigating its infamous toxicity profile.[1] The journey of thalidomide, from a sedative linked to devastating teratogenicity in the 1950s and 1960s to a repurposed anti-cancer agent, set the stage for the rational design of its analogues.[3] This historical context is not merely a footnote; it directly dictates the stringent regulatory framework and risk management protocols that govern the use of lenalidomide today.
Lenalidomide, along with thalidomide and the more recent pomalidomide, constitutes a distinct class of agents known as Immunomodulatory Imide Drugs (IMiDs) or, more mechanistically, Cereblon (CRBN) E3 Ligase Modulators (CELMoDs).[1] By structurally modifying thalidomide—specifically, by adding an amino group at the 4-position of the phthaloyl ring and removing a carbonyl group—scientists created a compound with enhanced antineoplastic and immunomodulatory activity and a reduced incidence of certain side effects, such as somnolence and peripheral neuropathy.[1] However, the inherent risk of teratogenicity, as suggested by preclinical animal studies, remained a paramount concern.[3] Consequently, regulatory agencies, including the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA), mandated a strict Risk Evaluation and Mitigation Strategy (REMS) program as a condition of its approval, a direct legacy of the thalidomide tragedy that continues to shape every aspect of its clinical use.[11]
First approved by the FDA in December 2005 for myelodysplastic syndromes (MDS) and shortly thereafter in 2006 for multiple myeloma, lenalidomide rapidly transformed the treatment paradigms for these diseases.[2] Its therapeutic utility is rooted in a remarkable array of pleiotropic effects, encompassing potent antineoplastic, anti-angiogenic, anti-inflammatory, and immunomodulatory properties.[1] Since its initial market entry, its clinical applications have expanded dramatically, now covering frontline, maintenance, and relapsed/refractory settings for multiple myeloma, as well as approved indications in MDS and various B-cell lymphomas, including mantle cell lymphoma (MCL), follicular lymphoma (FL), and marginal zone lymphoma (MZL).[3] The most recent clinical data from 2024 and 2025 continue to cement its role as an indispensable backbone component in highly effective quadruplet regimens for newly diagnosed multiple myeloma, further solidifying its status as a transformative agent in hematologic oncology.[31]
Chemical Profile and Synthesis
A thorough understanding of lenalidomide begins with its fundamental chemical and physical characteristics, which dictate its formulation, stability, and biological behavior.
Chemical Structure and Physicochemical Properties
Lenalidomide is a synthetic compound with a well-defined chemical identity. Its formal chemical name is 3-(4-amino-1,3-dihydro-1-oxo-2H-isoindol-2-yl)-2,6-piperidinedione.[3] The molecule possesses both a piperidinedione and an indoline moiety, with a critical amino function on its aromatic ring system that contributes to its distinct properties compared to thalidomide.[37]
Lenalidomide contains a single chiral center, and as such, it is manufactured and administered as a racemic (1:1) mixture of its S(-) and R(+) enantiomers. Despite the presence of these two optically active forms, the compound has a net optical rotation of zero.[1] Physically, it presents as an off-white to pale-yellow crystalline solid or powder.[2] Its solubility profile is a key characteristic; it is poorly soluble in water but demonstrates greater solubility in organic solvents such as dimethyl sulfoxide (DMSO) and under acidic conditions, such as in 0.1N HCl buffer.[2] This solubility profile influences its oral absorption and formulation. The key physicochemical properties are summarized in Table 1.
Property | Value | Source(s) |
---|
Chemical Name | 3-(4-amino-1,3-dihydro-1-oxo-2H-isoindol-2-yl)-2,6-piperidinedione | 3 |
Synonyms | Revlimid, CC-5013, (3S)-3-(4-Amino-1-oxo-1,3-dihydro-2H-isoindol-2-yl)piperidine-2,6-dione | 3 |
CAS Number | 191732-72-6 | 3 |
DrugBank ID | DB00480 | 3 |
Molecular Formula | C13H13N3O3 | 3 |
Molecular Weight | 259.26 g/mol | 41 |
Appearance | Off-white to pale-yellow solid/powder | 2 |
Melting Point | 265–271 °C | 2 |
Solubility | Insoluble in water; Soluble in DMSO; Greatest solubility in 0.1N HCl buffer | 2 |
Chirality | Racemic mixture of S(-) and R(+) enantiomers | 1 |
Review of Synthetic Pathways
The chemical synthesis of lenalidomide has evolved, with various methods developed to improve efficiency, scalability, and environmental impact. The initial synthesis, disclosed in patents by Celgene, established a foundational route that has since been refined.[3]
A commonly described pathway begins with the starting material methyl 2-methyl-3-nitrobenzoate. This compound undergoes a bromination reaction, typically using N-bromosuccinimide (NBS) as the brominating agent, to introduce a bromine atom to the methyl group, forming methyl 2-(bromomethyl)-3-nitrobenzoate.[3] This activated intermediate is then reacted with 3-aminopiperidine-2,6-dione, a derivative of glutamine. This step involves a cyclization reaction to form the nitro-precursor of lenalidomide, where the piperidinedione ring is attached to the isoindolinone backbone.[3]
The final and critical step in the synthesis is the reduction of the nitro group on the phthaloyl ring to the primary amine. This transformation can be achieved through several methods. The classical approach involves catalytic hydrogenation using a palladium on carbon (Pd/C) catalyst.[3] More recent developments have focused on "greener" and more cost-effective methods, such as using iron powder in the presence of ammonium chloride, which avoids the use of precious metal catalysts.[51]
The key structural modifications that differentiate lenalidomide from its parent compound, thalidomide, are the addition of the amino group at the 4-position of the isoindolinone ring and the removal of one of the carbonyl groups from the same ring.[9] These precise changes are responsible for its altered biological activity and improved toxicity profile. Once synthesized, the active pharmaceutical ingredient is formulated into oral capsules for clinical use, combined with various excipients such as lactose anhydrous, microcrystalline cellulose, croscarmellose sodium, and magnesium stearate to ensure proper stability and delivery.[37]
Core Pharmacology: A Multi-faceted Mechanism of Action
Lenalidomide's therapeutic efficacy stems from a unique and complex mechanism of action that was elucidated years after its clinical benefits were first observed. It operates not as a simple enzyme inhibitor or receptor antagonist but as a "molecular glue," a novel pharmacological principle that involves actively reprogramming a key component of the cell's protein degradation machinery.[3] This results in a cascade of downstream effects that are simultaneously cytotoxic to cancer cells and stimulating to the host immune system.
The Cereblon (CRBN) E3 Ligase Complex: The Primary Molecular Target
The primary molecular target of lenalidomide is Cereblon (CRBN), a protein that functions as a substrate receptor for the Cullin 4-RING E3 ubiquitin ligase complex (CRL4^CRBN^).[3] This complex, which also includes DNA damage-binding protein 1 (DDB1), Cullin 4A (CUL4A), and Regulator of Cullins 1 (ROC1), is a central part of the ubiquitin-proteasome system, responsible for tagging proteins for destruction.[8] Lenalidomide binds directly to a shallow hydrophobic pocket within CRBN.[8] This binding event does not inhibit the ligase; instead, it fundamentally alters its surface, creating a neomorphic interface that changes its substrate specificity. This allows the CRL4^CRBN^ complex to recognize and bind to a new set of proteins, known as "neo-substrates," which it would not normally target for degradation.[3]
Targeted Degradation of IKZF1 and IKZF3 Transcription Factors
In the context of multiple myeloma and other B-cell malignancies, the most critical neo-substrates recruited by the lenalidomide-bound CRBN complex are the lymphoid transcription factors Ikaros (encoded by the IKZF1 gene) and Aiolos (encoded by the IKZF3 gene).[3] Once these transcription factors are brought into proximity with the E3 ligase, they are rapidly polyubiquitinated and subsequently targeted for destruction by the 26S proteasome.[3] This degradation is highly selective, as other members of the Ikaros protein family, such as IKZF2 and IKZF5, are not affected.[5]
The degradation of IKZF1 and IKZF3 is the linchpin of lenalidomide's activity. These proteins are essential for the survival and proliferation of myeloma cells. Experimental evidence confirms that the forced depletion of either IKZF1 or IKZF3 is sufficient to induce apoptosis in myeloma cell lines, and mutations that prevent their degradation render the cells resistant to lenalidomide's cytotoxic effects.[56]
Downstream Anti-Myeloma Effects: Disruption of the IRF4-MYC Axis
The reason IKZF1 and IKZF3 are so critical to myeloma cells is that they are master regulators of a key oncogenic pathway. Their degradation leads to the subsequent downregulation of Interferon Regulatory Factor 4 (IRF4), a transcription factor that is absolutely required for the survival of plasma cells, both normal and malignant. The reduction in IRF4 levels, in turn, leads to the downregulation of the potent oncogene MYC.[5] The disruption of this critical IKZF1/3-IRF4-MYC signaling network is the primary mechanism behind the direct anti-tumor and pro-apoptotic effects of lenalidomide on myeloma cells.
Pleiotropic Immunomodulatory Effects: T-Cell and NK-Cell Activation
The same core mechanism—degradation of IKZF1 and IKZF3—also explains lenalidomide's powerful immunomodulatory properties. In T-lymphocytes, IKZF3 functions as a transcriptional repressor of the gene encoding Interleukin-2 (IL-2), a potent cytokine that promotes immune cell activation and proliferation.[5] When lenalidomide triggers the degradation of IKZF3 in T-cells, this repression is lifted, leading to a surge in IL-2 production and secretion. This T-cell co-stimulation results in the proliferation of T-cells and enhances the cytotoxic activity of Natural Killer (NK) cells and NKT cells.[5]
This immunomodulatory effect is significantly more potent with lenalidomide than with thalidomide, by a factor of 100 to 1,000, and it underpins the drug's ability to enhance antibody-dependent cell-mediated cytotoxicity (ADCC), which provides a strong rationale for its synergistic use with monoclonal antibodies like rituximab and daratumumab.[18]
Anti-Angiogenic and Microenvironment-Modulating Properties
Beyond its direct effects on tumor cells and immune cells, lenalidomide also modulates the tumor microenvironment. It exerts anti-angiogenic effects by inhibiting the secretion of crucial growth factors, including Vascular Endothelial Growth Factor (VEGF) and basic Fibroblast Growth Factor (bFGF), from both tumor cells and the surrounding bone marrow stromal cells (BMSCs).[9] This disrupts the formation of new blood vessels necessary to sustain tumor growth.
Furthermore, lenalidomide inhibits the adhesion of myeloma cells to BMSCs, a process that is known to confer cell adhesion-mediated drug resistance (CAM-DR).[9] It also reduces the secretion of pro-inflammatory and pro-survival cytokines within the bone marrow, such as Tumor Necrosis Factor-alpha (TNF-α), IL-1, and IL-6, further disrupting the supportive niche that myeloma cells depend on.[9]
The discovery of this "molecular glue" mechanism has had implications far beyond lenalidomide itself. It established a new pharmacological principle: that a small molecule could be used to hijack the cell's own protein disposal system to eliminate disease-causing proteins. This insight has directly fueled the development of an entire new class of therapeutics, including Proteolysis-Targeting Chimeras (PROTACs) and next-generation CELMoDs, which aim to apply this principle to a wide range of proteins previously considered "undruggable," such as transcription factors and scaffolding proteins.[52] Lenalidomide, therefore, serves as the foundational clinical proof-of-concept for the entire field of targeted protein degradation, one of the most active areas of modern drug discovery.
Clinical Pharmacokinetics (ADME)
The clinical behavior of lenalidomide is governed by a relatively straightforward pharmacokinetic profile, characterized by rapid oral absorption, limited metabolism, and primary renal excretion. This profile is consistent across different patient populations but is critically dependent on renal function, a factor that dictates dosing strategy and toxicity management.
Absorption, Bioavailability, and Food Effects
Following oral administration, lenalidomide is rapidly and extensively absorbed, with an estimated absorption of over 90% of the administered dose under fasting conditions.[9] In healthy volunteers, peak plasma concentrations (Cmax) are typically achieved quickly, within 0.6 to 1.5 hours after dosing.[38]
The presence of food, particularly a high-fat meal, has a clinically significant impact on absorption. Food can reduce the Cmax by approximately 50% and the total drug exposure, as measured by the area under the concentration-time curve (AUC), by about 20%.[9] For this reason, while the drug can be taken with or without food, patients are advised to take it in a consistent manner relative to meals to ensure predictable exposure.
Pharmacokinetic studies have demonstrated that both Cmax and AUC increase in a dose-proportional manner. Importantly, there is no evidence of significant drug accumulation with repeated daily dosing, which is consistent with its relatively short half-life.[38]
Distribution, Protein Binding, and Metabolism
Lenalidomide exhibits wide distribution throughout the body. Plasma protein binding is low, ranging from 19% to 29%, indicating that a large fraction of the drug is free and available to distribute into tissues.[3] A clinically important aspect of its distribution is its ability to pass into semen. In a study of healthy males, lenalidomide concentrations in semen were found to be higher than in plasma at 2 and 24 hours after the last dose. However, the drug became undetectable in semen by 3 days after treatment cessation.[64] This finding is a critical piece of information for counseling male patients on the necessity of contraception to prevent fetal exposure.
The metabolism of lenalidomide in humans is limited. The main biotransformation pathways include chiral inversion between the S(-) and R(+) enantiomers, minor hydroxylation, and slow, non-enzymatic hydrolysis of the glutarimide ring.[64] Lenalidomide does not significantly inhibit or induce major cytochrome P450 (CYP) isoenzymes, which minimizes the potential for many common drug-drug interactions mediated by the CYP system.[40]
Renal Excretion and Elimination Half-Life
The defining characteristic of lenalidomide's elimination is its reliance on the kidneys. The primary route of excretion is renal, with approximately 82% of an oral dose being eliminated unchanged in the urine within 24 hours.[9] The drug's elimination half-life in individuals with normal renal function is correspondingly short, typically around 3 to 4 hours.[9]
This near-total dependence on renal clearance makes kidney function the single most important patient-specific factor influencing drug exposure. The total body clearance of lenalidomide is directly and linearly correlated with creatinine clearance (CrCl).[38] Consequently, any degree of renal impairment leads to reduced drug clearance, a prolonged elimination half-life (which can exceed 9 hours in patients with a CrCl < 50 mL/min), and a proportional increase in plasma AUC.[38] This direct causal link between renal function, drug exposure, and the risk of toxicity (particularly myelosuppression) is the scientific basis for the mandatory, evidence-based dose adjustments required for patients with renal insufficiency.[64]
Clinical Applications and Efficacy in Approved Indications
Lenalidomide has secured a central role in the treatment of several hematologic malignancies, with its efficacy established through numerous large-scale clinical trials. Its approvals span multiple diseases and treatment settings, from frontline therapy to the management of relapsed and refractory disease.
Multiple Myeloma (MM)
Lenalidomide's most significant impact has been in the treatment of multiple myeloma.
- Newly Diagnosed Multiple Myeloma (NDMM): Lenalidomide, in combination with dexamethasone, was established as a standard of care for transplant-ineligible patients based on pivotal trials like the FIRST study (NCT00689936), which demonstrated its superiority over older regimens such as melphalan, prednisone, and thalidomide (MPT).[67] It is also approved as a maintenance therapy following autologous stem cell transplantation (auto-HSCT), where it has been shown to significantly prolong progression-free survival.[3]
- Relapsed/Refractory Multiple Myeloma (RRMM): The initial approval for lenalidomide in MM was for patients who had received at least one prior therapy, based on studies showing high response rates when combined with dexamethasone.[3]
- The Era of Quadruplet Therapy (2024-2025): The therapeutic landscape for NDMM has been revolutionized by recent data from landmark clinical trials. Studies such as PERSEUS, ADVANCE, GMMG-HD7, and IsKia have consistently demonstrated that adding an anti-CD38 monoclonal antibody (daratumumab or isatuximab) to a lenalidomide-based backbone (e.g., lenalidomide, bortezomib, and dexamethasone or lenalidomide, carfilzomib, and dexamethasone) results in significantly higher rates of minimal residual disease (MRD) negativity and improved progression-free survival (PFS).[31] These findings have established quadruplet regimens as the new standard of care for both transplant-eligible and transplant-ineligible patients.
Myelodysplastic Syndromes (MDS) with Deletion 5q
Lenalidomide is approved as a monotherapy for the treatment of transfusion-dependent anemia in patients with low- or intermediate-1-risk MDS who have a specific chromosomal abnormality known as deletion 5q (del(5q)), with or without other cytogenetic abnormalities.[3] In this specific patient population, lenalidomide is highly effective, leading to transfusion independence in a majority of patients and thereby significantly improving their quality of life.[28]
Relapsed or Refractory Mantle Cell Lymphoma (MCL)
For mantle cell lymphoma, lenalidomide is approved as a monotherapy for patients whose disease has relapsed or progressed after at least two prior lines of therapy, one of which must have included the proteasome inhibitor bortezomib.[3]
Relapsed or Refractory Follicular and Marginal Zone Lymphoma (FL/MZL)
Lenalidomide is approved for use in combination with the anti-CD20 monoclonal antibody rituximab for patients with previously treated follicular lymphoma and marginal zone lymphoma.[3] This combination, often referred to as R², was approved by the EMA in late 2019 based on the results of the AUGMENT and MAGNIFY trials, which showed superior efficacy compared to rituximab alone.[74] More recent FDA approvals in 2025 have further expanded the therapeutic options in this space, incorporating agents like brentuximab vedotin or tafasitamab into lenalidomide-rituximab-based combinations for certain lymphoma subtypes.[75]
Off-Label and Investigational Uses
While its approved indications are broad, lenalidomide has also been explored in other conditions. It is sometimes used off-label, often with dexamethasone, for the treatment of AL amyloidosis.[3] A notable area where lenalidomide is
not recommended is in the treatment of chronic lymphocytic leukemia (CLL) outside of a controlled clinical trial. This is due to findings from a frontline study that showed an increased risk of death in the lenalidomide arm compared to standard chlorambucil, driven by a higher rate of serious cardiovascular adverse events.[11]
The table below summarizes pivotal clinical trials that have defined the role of lenalidomide across its major indications.
Trial Name / Identifier | Indication | Patient Population | Treatment Arms | Primary Endpoint | Key Result | Source(s) |
---|
FIRST (MM-020) | NDMM | Transplant-ineligible | Lenalidomide + low-dose Dex (Rd) continuous vs. Rd for 18 cycles vs. MPT | Progression-Free Survival (PFS) | Continuous Rd significantly improved PFS compared to MPT (25.5 vs. 21.2 months; HR 0.72). | 67 |
MAIA | NDMM | Transplant-ineligible | Daratumumab + Rd (D-Rd) vs. Rd | Progression-Free Survival (PFS) | D-Rd significantly improved PFS vs. Rd alone. | 78 |
ADVANCE | NDMM | Transplant-eligible and ineligible | Daratumumab + KRd (D-KRd) vs. KRd | MRD Negativity Rate | D-KRd resulted in a significantly higher MRD negativity rate (59% vs. 36%). | 31 |
PERSEUS | NDMM | Transplant-eligible | Daratumumab + VRd (D-VRd) vs. VRd | Progression-Free Survival (PFS) | D-VRd significantly improved PFS and doubled sustained MRD negativity rates. | 33 |
GMMG-HD7 | NDMM | Transplant-eligible | Isatuximab + VRd (Isa-VRd) vs. VRd | MRD Negativity Rate | Isa-VRd achieved superior MRD negativity rates (66% vs. 48%) and 3-year PFS. | 33 |
AUGMENT | R/R FL/MZL | Previously treated | Lenalidomide + Rituximab (R²) vs. Placebo + Rituximab | Progression-Free Survival (PFS) | R² significantly prolonged PFS compared to rituximab alone. | 74 |
MDS-003 | MDS with del(5q) | Transfusion-dependent, low/int-1 risk | Lenalidomide vs. Placebo | Transfusion Independence ≥ 26 weeks | 67% of patients on lenalidomide achieved transfusion independence vs. 6% on placebo. | 28 |
Dosing, Administration, and Management in Special Populations
The safe and effective use of lenalidomide requires strict adherence to indication-specific dosing regimens, coupled with proactive dose modifications for toxicity and in special populations, particularly those with renal impairment.
Recommended Dosing Regimens by Indication
Lenalidomide is administered orally via capsules that must be swallowed whole with water. The capsules should never be broken, opened, or chewed to avoid exposure to the powder.[12] It can be taken with or without food, but should be taken at approximately the same time each day to maintain consistent plasma levels.[15]
- Multiple Myeloma (Combination Therapy): The standard starting dose is 25 mg once daily, administered on Days 1 through 21 of a repeating 28-day cycle. It is typically given in combination with dexamethasone and/or other agents like proteasome inhibitors.[15]
- Multiple Myeloma (Maintenance Post-auto-HSCT): Treatment is initiated after adequate hematologic recovery. The recommended starting dose is 10 mg once daily, taken continuously on Days 1 through 28 of each cycle. The dose may be increased to 15 mg daily after 3 cycles if the initial dose is well tolerated.[15]
- Myelodysplastic Syndromes (MDS): The recommended starting dose is 10 mg once daily.[15]
- Mantle Cell Lymphoma (MCL): The starting dose is 25 mg once daily on Days 1 through 21 of a 28-day cycle.[15]
- Follicular/Marginal Zone Lymphoma (FL/MZL): The starting dose is 20 mg once daily on Days 1 through 21 of a 28-day cycle, typically for up to 12 cycles, in combination with rituximab.[15]
Dose Adjustments for Renal Impairment
Given that lenalidomide is almost exclusively cleared by the kidneys, dose adjustments for patients with renal impairment are mandatory to avoid excessive drug exposure and toxicity. The following table summarizes the recommended starting doses based on creatinine clearance (CrCl).
Indication | Standard Starting Dose | CrCl 30–60 mL/min | CrCl <30 mL/min (No Dialysis) | CrCl <30 mL/min (Dialysis) | Source(s) |
---|
MM (Combination) | 25 mg daily (Days 1-21) | 10 mg daily | 15 mg every 48 hours OR 5 mg daily | 5 mg daily, post-dialysis | 17 |
MCL / FL / MZL | 25 mg / 20 mg daily (Days 1-21) | 10 mg daily | 5 mg daily | 5 mg daily, post-dialysis | 24 |
MM (Maintenance) | 10 mg daily (continuous) | 5 mg daily | 2.5 mg daily | 2.5 mg daily, post-dialysis | 24 |
MDS | 10 mg daily | 5 mg daily | 2.5 mg daily | 2.5 mg daily, post-dialysis | 24 |
Dose Modifications for Hematologic Toxicity
Myelosuppression is the most common dose-limiting toxicity of lenalidomide. Proactive management through dose interruption and reduction is essential for patient safety and to allow for continued treatment. The following table provides a general guide for dose adjustments, though specific protocols may vary slightly.
Toxicity | Threshold for Action | Recommended Action | Dose Upon Resumption | Source(s) |
---|
Thrombocytopenia (MM/MCL/FL) | Platelets fall to < 30,000/mcL | Interrupt treatment. Monitor CBC weekly. | Resume at next lower dose level (e.g., 25 mg → 15 mg). Do not dose below 5 mg/day. | 17 |
Neutropenia (MM/MCL/FL) | ANC falls to < 1,000/mcL | Interrupt treatment. Consider G-CSF support. Monitor CBC weekly. | If neutropenia is only toxicity, resume at starting dose. If other toxicities, resume at next lower dose level. | 17 |
Thrombocytopenia (MDS) | Platelets fall to < 30,000/mcL (or < 50,000/mcL with transfusions) | Interrupt treatment. | Resume at next lower dose level (e.g., 10 mg → 5 mg). | 17 |
Neutropenia (MDS) | ANC falls to < 500/mcL for ≥ 7 days (or with fever) | Interrupt treatment. | Resume at next lower dose level (e.g., 10 mg → 5 mg). | 17 |
Safety Profile, Risk Management, and Adverse Events
The potent therapeutic activity of lenalidomide is accompanied by a significant and predictable toxicity profile. Safe use of the drug is contingent upon a thorough understanding of these risks, strict adherence to risk management protocols, and proactive management of adverse events.
Boxed Warnings: Embryo-Fetal Toxicity, Hematologic Toxicity, and Thromboembolism
The prescribing information for lenalidomide carries prominent boxed warnings for three major risks:
- Embryo-Fetal Toxicity (Teratogenicity): As a thalidomide analogue, lenalidomide is contraindicated for use during pregnancy. Based on its mechanism and findings in animal studies, it is presumed to be teratogenic in humans, capable of causing severe, life-threatening birth defects or embryo-fetal death. This risk is absolute and is the primary justification for the mandatory REMS program.[3]
- Hematologic Toxicity: Lenalidomide causes significant, dose-dependent myelosuppression. Severe neutropenia (low neutrophils) and thrombocytopenia (low platelets) are very common and can lead to an increased risk of serious infection and bleeding. In some cases, trilineage bone marrow hypoplasia or aplasia leading to pancytopenia has been reported. Frequent monitoring of complete blood counts (CBC) is required, especially during the initial cycles of therapy, to guide necessary dose interruptions and reductions.[3]
- Venous and Arterial Thromboembolism: Patients treated with lenalidomide have a significantly increased risk of developing blood clots, including deep vein thrombosis (DVT) and pulmonary embolism (PE), as well as arterial thromboses such as myocardial infarction (MI) and stroke. The risk is particularly elevated when lenalidomide is used in combination with dexamethasone or other agents. Prophylactic anticoagulation or antiplatelet therapy is recommended for most patients with multiple myeloma receiving lenalidomide.[3]
The Lenalidomide REMS Program: A Mandate for Safe Use
Due to the profound teratogenic risk, lenalidomide is subject to a highly restrictive distribution program known as the Lenalidomide REMS (Risk Evaluation and Mitigation Strategy) in the United States and similar pregnancy prevention programs in other regions.[1] The program's goal is to prevent fetal exposure to the drug. Key requirements include:
- Certified Prescribers and Pharmacies: Only healthcare providers and pharmacies certified with the REMS program can prescribe and dispense lenalidomide.
- Patient Enrollment: All patients must be counseled on the risks and enrolled in the program, signing a Patient-Physician Agreement Form.
- Requirements for Females of Reproductive Potential: These patients must have two negative pregnancy tests before initiating therapy and undergo regular pregnancy testing thereafter. They must commit to using two different forms of effective contraception simultaneously, beginning four weeks before treatment and continuing for at least four weeks after the final dose.
- Requirements for Males: Male patients must agree to use a condom during any sexual contact with a pregnant female or a female of reproductive potential, both during treatment and for four weeks after. They are prohibited from donating sperm during this period.
- Blood Donation Prohibition: All patients are prohibited from donating blood during therapy and for four weeks following discontinuation to prevent accidental administration to a pregnant individual.[11]
Comprehensive Review of Adverse Reactions
Beyond the boxed warnings, lenalidomide is associated with a wide range of adverse effects.
- Very Common Adverse Reactions (occurring in ≥20% of patients): In addition to the hematologic toxicities, other frequently reported side effects include fatigue, asthenia, diarrhea, constipation, muscle cramps/spasms, rash, fever, cough, peripheral edema, decreased appetite, and various infections (e.g., upper respiratory tract infection, bronchitis).[12]
- Serious Adverse Reactions:
- Second Primary Malignancies (SPM): An increased risk of developing new cancers, particularly hematologic malignancies like acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS), has been observed in patients treated with lenalidomide, especially in the multiple myeloma setting.[3]
- Hepatotoxicity: Severe, and in some cases fatal, liver injury has been reported. Periodic monitoring of liver function tests is recommended.[11]
- Severe Cutaneous Reactions: Rare but life-threatening skin reactions, including Stevens-Johnson Syndrome (SJS), Toxic Epidermal Necrolysis (TEN), and Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS), have been reported. Lenalidomide must be permanently discontinued if these occur.[3]
- Tumor Lysis Syndrome (TLS): In patients with a large tumor burden, the rapid killing of cancer cells can lead to TLS. Prophylactic measures and close monitoring of electrolytes and renal function are necessary for at-risk patients.[11]
- Tumor Flare Reaction: This reaction, more common in patients with lymphoma, involves tender lymph node swelling, low-grade fever, pain, and rash. It can typically be managed with NSAIDs or corticosteroids without discontinuing therapy.[11]
- Thyroid Dysfunction: Both hypothyroidism and hyperthyroidism have been reported, necessitating monitoring of thyroid function.[15]
The safety profile of lenalidomide necessitates a clinical management approach where supportive care is not merely an option but an integral part of the treatment plan. The high probability of VTE, myelosuppression, and the absolute risk of teratogenicity mean that a clinician prescribing lenalidomide is initiating a comprehensive care strategy that includes mandatory thromboprophylaxis, vigilant hematologic monitoring with dose adjustments and potential growth factor support, and strict adherence to the REMS program. The drug's clinical success is therefore a testament not only to its efficacy but also to the development of robust systems designed to manage its inherent and predictable risks.
Clinically Significant Drug-Drug Interactions
The potential for drug-drug interactions with lenalidomide is an important consideration for safe prescribing, primarily concerning agents that can potentiate its known toxicities or have a narrow therapeutic index.
Agents Increasing Thrombosis Risk
The baseline risk of venous and arterial thromboembolism associated with lenalidomide is exacerbated by concomitant use of other drugs that also promote clotting.
- Erythropoietin-Stimulating Agents (ESAs): These agents, used to treat anemia, are known to increase the risk of thrombosis. Their use with lenalidomide requires a careful benefit-risk assessment, and they should be discontinued if hemoglobin levels exceed 12 g/dL.[27]
- Estrogen-Containing Therapies: This includes hormonal contraceptives and hormone replacement therapy. As estrogens can be prothrombotic, their concurrent use with lenalidomide further elevates the risk of VTE. This is particularly relevant given the need for effective contraception in females of reproductive potential; non-hormonal methods should be strongly considered.[82]
Interactions with Narrow Therapeutic Index Drugs
- Digoxin: Co-administration of lenalidomide can increase the plasma concentrations of digoxin, a cardiac glycoside with a narrow therapeutic window. Clinical monitoring of digoxin levels is recommended to avoid toxicity.[15]
- Warfarin: The interaction between lenalidomide and warfarin is not fully characterized and may be influenced by concomitant dexamethasone. Therefore, close monitoring of the international normalized ratio (INR) is advised for patients receiving this combination to ensure appropriate anticoagulation.[27]
Other Significant Interactions
- Live Vaccines: Due to its immunosuppressive and immunomodulatory effects, lenalidomide can diminish the therapeutic effect of live vaccines and increase the risk of vaccine-induced infection. Live vaccines should be avoided during and for a period after lenalidomide therapy.[15]
- Myelosuppressive Agents: When used with other chemotherapeutic agents or drugs that suppress bone marrow function, lenalidomide can cause additive hematologic toxicity. This necessitates even more vigilant monitoring of blood counts and may require more frequent dose adjustments.[87]
- P-glycoprotein (P-gp) Interactions: While in vitro studies show lenalidomide is a weak substrate for the P-gp efflux pump, clinical studies have demonstrated that co-administration with strong P-gp inhibitors (e.g., quinidine) does not have a clinically meaningful effect on lenalidomide's pharmacokinetics. Therefore, dose adjustments are generally not required with P-gp inhibitors.[27]
The following table summarizes the most clinically important drug-drug interactions.
Interacting Drug/Class | Potential Effect | Clinical Management/Recommendation | Source(s) |
---|
Erythropoietin-Stimulating Agents (ESAs) | Increased risk of venous thromboembolism (VTE) | Use with caution based on benefit-risk assessment. Discontinue if hemoglobin > 12 g/dL. | 27 |
Estrogen-Containing Therapies | Increased risk of VTE | Use with caution. Consider alternative, non-hormonal methods of contraception. | 82 |
Digoxin | Increased plasma concentration of digoxin | Monitor digoxin levels and adjust digoxin dose as needed. | 27 |
Warfarin | Unpredictable effect on INR, potentially altered by concomitant dexamethasone | Monitor INR closely and adjust warfarin dose as needed. | 27 |
Live Vaccines | Diminished vaccine efficacy and increased risk of infection | Avoid co-administration. | 15 |
Other Myelosuppressive Agents | Additive hematologic toxicity (neutropenia, thrombocytopenia) | Monitor blood counts closely. Be prepared for more frequent dose modifications of lenalidomide and/or the concomitant agent. | 87 |
Mechanisms of Lenalidomide Resistance
Despite its profound efficacy, acquired resistance to lenalidomide is an inevitable event in the treatment of multiple myeloma, representing a major clinical challenge. Research into the mechanisms of resistance has revealed a complex picture involving both alterations in the drug's primary target and the activation of bypass pathways that allow cancer cells to survive.
CRBN-Dependent Resistance Mechanisms
The most direct and well-understood mechanisms of resistance involve the drug's molecular target, Cereblon (CRBN). For lenalidomide to work, it must bind to CRBN to induce the degradation of IKZF1 and IKZF3. Any alteration that disrupts this initial step can lead to resistance. These alterations include:
- Downregulation of CRBN Expression: Reduced transcription or translation of the CRBN gene leads to lower levels of the CRBN protein, providing fewer targets for lenalidomide to bind. This is one of the most common resistance mechanisms observed in patients.[7]
- Mutations in the CRBN Gene: Specific mutations within the lenalidomide-binding pocket of CRBN can prevent the drug from docking effectively, thereby abrogating its ability to recruit the neo-substrates.[91]
- Copy Number Loss or Deletion of the CRBN Gene: Loss of one or both copies of the CRBN gene also results in insufficient protein levels for a therapeutic effect.[93]
While these CRBN-centric mechanisms are clearly important, they are found in only about 20-30% of patients with acquired resistance to lenalidomide.[92] This significant gap has driven research into CRBN-independent pathways.
Emerging CRBN-Independent Pathways and Novel Drivers of Resistance
Recent investigations, particularly from 2024-2025, have uncovered sophisticated adaptive strategies that myeloma cells employ to evade lenalidomide's effects, even when CRBN is present and functional.
- ADAR1-Regulated dsRNA Sensing Pathway: A groundbreaking discovery has identified the RNA editing enzyme ADAR1 (adenosine deaminase acting on RNA 1) as a novel driver of lenalidomide resistance.[62] This mechanism reframes our understanding of lenalidomide's action. It is proposed that lenalidomide treatment leads to the accumulation of endogenous double-stranded RNA (dsRNA) within myeloma cells. In sensitive cells, this dsRNA acts as a "viral mimicry" signal, triggering an innate immune response through the sensor protein MDA5, leading to a type I interferon (IFN) response that contributes to apoptosis. However, in resistant cells, high expression of the ADAR1 enzyme edits this dsRNA, preventing it from being recognized by MDA5. This effectively short-circuits the anti-tumor immune response, allowing the cell to survive despite the presence of lenalidomide. Overexpression of ADAR1 has been shown to confer resistance, while its inhibition can re-sensitize myeloma cells to the drug.[62]
- Metabolic Reprogramming and Post-Translational Modifications (PTMs): Research published in 2025 has linked metabolic changes to resistance through the lens of PTMs. Lenalidomide-resistant myeloma cells exhibit increased glycolysis, which in turn drives a novel PTM called lysine lactylation (Kla) on various proteins, including those involved in chemoresistance. This suggests that the cancer cell reprograms its metabolism to epigenetically or post-translationally alter key proteins to promote survival. Importantly, inhibiting glycolysis was shown to reverse this resistance phenotype, pointing to a new therapeutic vulnerability.[95]
- Transcriptional Rewiring: Myeloma cells can develop resistance by rewiring their transcriptional networks to bypass their dependency on the IKZF1/3-IRF4-MYC axis. Even if lenalidomide successfully degrades IKZF1 and IKZF3, the cell can activate alternative pathways to maintain the expression of critical survival factors like MYC, rendering the drug's primary action less impactful.[61]
- Genetic and Microenvironmental Factors: Pre-existing high-risk genetic features, such as the gain or amplification of chromosome 1q (gain/amp1q) and deletion of chromosome 17p (which contains the TP53 tumor suppressor gene), are strongly associated with intrinsic and acquired resistance to therapy.[92] Furthermore, the bone marrow microenvironment can provide protective signals to myeloma cells, shielding them from the effects of therapy.
The elucidation of these complex, systems-level adaptations marks a significant evolution in our understanding of drug resistance. It moves beyond simple target mutations to encompass immune evasion, metabolic reprogramming, and epigenetic plasticity. This new knowledge is not merely academic; it provides a roadmap for the rational design of next-generation therapies. The discovery of the ADAR1 pathway, for instance, immediately suggests a new combination strategy: pairing lenalidomide with an ADAR1 inhibitor to prevent this adaptive resistance and restore the drug's efficacy. Similarly, targeting glycolysis could become a synergistic approach in lenalidomide-resistant disease. These insights are poised to directly influence the design of future clinical trials for this challenging patient population.
The Evolving Therapeutic Landscape: Recent and Future Developments (2024-2025)
The treatment paradigm for multiple myeloma is in a state of rapid evolution, with lenalidomide being repositioned from a key agent to the foundational backbone upon which more potent regimens are built. Data from major hematology conferences in 2024 and 2025 have solidified new standards of care and clarified the path forward for patients who become refractory to lenalidomide.
Novel Combination Regimens: The Era of Quadruplet Therapy
The most significant recent development in frontline myeloma therapy is the definitive establishment of four-drug, or quadruplet, regimens as the new standard of care for most patients with NDMM. A wealth of data from large, randomized phase 3 trials presented at the American Society of Clinical Oncology (ASCO), European Hematology Association (EHA), and American Society of Hematology (ASH) meetings has demonstrated the superiority of this approach.
- The PERSEUS and ADVANCE trials showed that adding the anti-CD38 monoclonal antibody daratumumab to lenalidomide-based triplets (VRd or KRd) leads to significantly higher rates of MRD negativity and prolonged PFS in both transplant-eligible and transplant-ineligible patients.[31]
- Similarly, the GMMG-HD7 and IsKia trials confirmed the benefit of adding a different anti-CD38 antibody, isatuximab, to lenalidomide-based backbones, with similar improvements in depth of response and survival outcomes.[33]
Collectively, these trials establish that for the majority of newly diagnosed patients, initial therapy should consist of a quadruplet regimen comprising a proteasome inhibitor (bortezomib or carfilzomib), an IMiD (lenalidomide), a steroid (dexamethasone), and an anti-CD38 monoclonal antibody.[34]
The Role of Minimal Residual Disease (MRD) in Guiding Lenalidomide Therapy
The goal of modern frontline therapy has shifted from achieving a clinical remission to achieving deep, sustained MRD negativity. MRD status, typically assessed by next-generation sequencing or flow cytometry in the bone marrow, has emerged as one of the most powerful prognostic factors and a key surrogate endpoint for long-term outcomes like PFS and OS in clinical trials.[33]
This is now translating into MRD-guided therapeutic strategies. For instance, a study presented at ASH 2024 explored the possibility of discontinuing lenalidomide maintenance therapy in patients who achieve sustained MRD negativity for at least three years post-transplant. The early results suggest that this may be a safe approach, with most patients remaining MRD-negative after stopping treatment. This strategy aims to reduce the long-term toxicity and financial burden of continuous therapy without compromising outcomes.[98] Conversely, the failure to achieve MRD negativity with frontline quadruplet therapy may identify patients who require alternative consolidation or maintenance strategies, such as the early introduction of cellular therapies.
Positioning Lenalidomide in the Age of Cellular Therapies and Bispecific Antibodies
The success of lenalidomide in the frontline setting has paradoxically created the next major clinical challenge: managing the growing population of patients whose disease is "lenalidomide-refractory".[91] For these individuals, novel immunotherapies are showing remarkable promise.
- CAR T-cell therapies, such as ciltacabtagene autoleucel (cilta-cel) and idecabtagene vicleucel (ide-cel), and bispecific antibodies that target BCMA or GPRC5D (e.g., teclistamab, talquetamab, linvoseltamab) are demonstrating high and durable response rates in heavily pretreated patients, the majority of whom are refractory to lenalidomide.[36] The CARTITUDE-4 trial, for example, directly compared cilta-cel to standard-of-care regimens in lenalidomide-refractory patients and showed vastly superior outcomes for the CAR T-cell therapy.[102]
The future role of lenalidomide will involve its integration with these powerful new modalities. Clinical trials are currently underway to explore lenalidomide as a maintenance therapy following CAR T-cell infusion to deepen or prolong responses (e.g., the CARTITUDE-2 study) or in combination with bispecific antibodies in earlier lines of therapy.[33] This strategic repositioning highlights lenalidomide's evolution: from a salvage agent to the cornerstone of induction therapy, with its failure now defining the critical juncture where next-generation immunotherapies are deployed.
Conclusion and Expert Recommendations
Lenalidomide is an oral immunomodulatory agent that has fundamentally reshaped the treatment of multiple myeloma and other hematologic cancers. Its development from thalidomide is a landmark example of rational drug design, and the elucidation of its novel "molecular glue" mechanism of action—reprogramming the CRL4^CRBN^ E3 ubiquitin ligase to degrade the transcription factors IKZF1 and IKZF3—has opened an entire new field of targeted protein degradation in pharmacology. This core mechanism elegantly explains its pleiotropic effects, which combine direct tumor cytotoxicity, potent T-cell and NK-cell activation, and modulation of the tumor microenvironment.
The clinical use of lenalidomide is a constant exercise in balancing its profound efficacy against a significant and predictable toxicity profile. The successful administration of lenalidomide is therefore inseparable from the proactive management of its side effects. This includes mandatory adherence to the REMS program to prevent embryo-fetal exposure, routine thromboprophylaxis to mitigate the high risk of VTE in myeloma patients, and vigilant monitoring of blood counts with guideline-based dose modifications for cytopenias and renal impairment.
In 2025, the therapeutic landscape positions lenalidomide as the indispensable backbone of first-line quadruplet regimens for nearly all patients with newly diagnosed multiple myeloma. The primary goal of these intensive induction therapies is to achieve deep and sustained MRD negativity. Consequently, the state of being "lenalidomide-refractory" has become the new, critical determinant for the use of next-generation immunotherapies, such as CAR T-cells and bispecific antibodies, in the relapsed setting.
Based on this comprehensive analysis, the following recommendations are proposed:
- Adherence to Safety Mandates: Strict, unwavering adherence to the Lenalidomide REMS program is non-negotiable.
- Proactive Toxicity Management: Clinicians must implement prophylactic and supportive care as an integral part of the treatment plan. This includes risk-stratified thromboprophylaxis for all myeloma patients, frequent hematologic monitoring with prompt, guideline-based dose adjustments, and consideration of G-CSF for neutropenia.
- Adoption of Quadruplet Regimens: For transplant-eligible and most transplant-ineligible patients with NDMM, an anti-CD38 antibody-based quadruplet regimen containing lenalidomide should be considered the standard of care to maximize the depth and duration of response.
- Overcoming Resistance: The highest priority for future research must be the development of strategies to overcome or prevent lenalidomide resistance. This should include the clinical investigation of agents targeting novel, CRBN-independent resistance pathways, such as ADAR1 inhibitors and glycolysis inhibitors, in combination with lenalidomide.
- MRD-Guided Therapy: Prospective, randomized clinical trials are urgently needed to validate MRD-guided treatment de-escalation strategies. Determining whether patients with sustained MRD negativity can safely discontinue lenalidomide maintenance is a critical question that could reduce long-term toxicity, improve quality of life, and decrease the financial burden of care.
- Optimal Sequencing and Combination: As the therapeutic armamentarium expands, research must focus on the optimal sequencing and combination of lenalidomide-based regimens with cellular therapies and bispecific antibodies to define the most effective, long-term treatment pathways for patients with multiple myeloma.
Works cited
- Lenalidomide hydrochloride monohydrate | DrugBank Online, accessed July 8, 2025, https://go.drugbank.com/salts/DBSALT003154
- Lenalidomide | 191732-72-6 - ChemicalBook, accessed July 8, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB3855432.htm
- Lenalidomide - Wikipedia, accessed July 8, 2025, https://en.wikipedia.org/wiki/Lenalidomide
- Biological Activity of Lenalidomide and Its Underlying Therapeutic Effects in Multiple Myeloma - PMC - PubMed Central, accessed July 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3417169/
- Lenalidomide induces degradation of IKZF1 and IKZF3 - PMC - PubMed Central, accessed July 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4292522/
- Synthesis and Pharmacology of Clinical Drugs Containing Isoindoline Heterocycle Core - Preprints.org, accessed July 8, 2025, https://www.preprints.org/manuscript/202411.1645/v1/download
- Novel insights into the mechanism of action of lenalidomide - Taylor & Francis Online, accessed July 8, 2025, https://www.tandfonline.com/doi/full/10.4161/onci.28386
- Cereblon-Targeting Ligase Degraders in Myeloma - BINASSS, accessed July 8, 2025, https://www.binasss.sa.cr/abr24/50.pdf
- Lenalidomide use in multiple myeloma (Review) - Spandidos Publications, accessed July 8, 2025, https://www.spandidos-publications.com/10.3892/mco.2023.2705
- 204026Orig1s000 - accessdata.fda.gov, accessed July 8, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2013/204026Orig1s000PharmR.pdf
- REVLIMID® (lenalidomide) | Indications and Important Safety Information, accessed July 8, 2025, https://www.bmsaccesssupport.com/revlimid/indications-important-safety-information.html
- REVLIMID® (lenalidomide) - Official Patient Website, accessed July 8, 2025, https://www.revlimid.com/
- Multiple Myeloma Treatment Option | REVLIMID® (lenalidomide), accessed July 8, 2025, https://www.revlimidhcp.com/mm
- Patient Guide to Risk Evaluation and Mitigation Strategy (REMS), accessed July 8, 2025, https://www.lenalidomiderems.com/media/wp-content/uploads/LEN_Patient_Guide.pdf
- Lenalidomide (oral route) - Mayo Clinic, accessed July 8, 2025, https://www.mayoclinic.org/drugs-supplements/lenalidomide-oral-route/description/drg-20068352
- Revlimid (Lenalidomide) - International Myeloma Foundation, accessed July 8, 2025, https://www.myeloma.org/revlimid-lenalidomide
- Revlimid (lenalidomide) capsules label - accessdata.fda.gov, accessed July 8, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/021880s028lbl.pdf
- Lenalidomide: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed July 8, 2025, https://go.drugbank.com/drugs/DB00480
- Lenalidomide – Knowledge and References - Taylor & Francis, accessed July 8, 2025, https://taylorandfrancis.com/knowledge/Medicine_and_healthcare/Pharmaceutical_medicine/Lenalidomide/
- Full article: A review of lenalidomide in combination with dexamethasone for the treatment of multiple myeloma - Taylor & Francis Online, accessed July 8, 2025, https://www.tandfonline.com/doi/full/10.2147/tcrm.s1445
- A review of lenalidomide in combination with dexamethasone for the treatment of multiple myeloma - PMC, accessed July 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2503648/
- go.drugbank.com, accessed July 8, 2025, https://go.drugbank.com/drugs/DB00480#:~:text=Lenalidomide%20is%20indicated%20for%20the,transplantation%20(auto%2DHSCT).
- Lenalidomide | Leukemia and Lymphoma Society, accessed July 8, 2025, https://www.lls.org/drug/lenalidomide
- Revlimid (lenalidomide) dosing, indications, interactions, adverse effects, and more, accessed July 8, 2025, https://reference.medscape.com/drug/revlimid-lenalidomide-342200
- Lenalidomide Accord | European Medicines Agency (EMA), accessed July 8, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/lenalidomide-accord
- Revlimid® (lenalidomide) Information for Healthcare Professionals Guide - Malta Medicines Authority, accessed July 8, 2025, https://medicinesauthority.gov.mt/file.aspx?f=5083
- Revlimid, INN-lenalidomide - EMA, accessed July 8, 2025, https://www.ema.europa.eu/en/documents/product-information/revlimid-epar-product-information_en.pdf
- Revlimid: Dosage, side effects, uses, and more - Medical News Today, accessed July 8, 2025, https://www.medicalnewstoday.com/articles/326466
- Lenalidomide (Revlimid) | General cancer information - Cancer Research UK, accessed July 8, 2025, https://www.cancerresearchuk.org/about-cancer/treatment/drugs/lenalidomide
- Annotation of FDA Label for lenalidomide - PharmGKB, accessed July 8, 2025, https://www.pharmgkb.org/labelAnnotation/PA166104834
- New Standard-of-Care Emerges for Multiple Myeloma - InventUM, accessed July 8, 2025, https://news.med.miami.edu/new-standard-of-care-emerges-for-multiple-myeloma/
- New four drug combination sets new standard in myeloma care - News-Medical.net, accessed July 8, 2025, https://www.news-medical.net/news/20250529/New-four-drug-combination-sets-new-standard-in-myeloma-care.aspx
- Novelties on Multiple Myeloma from the Main 2024 Hematology Conferences - PMC, accessed July 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11906126/
- Evolving Paradigms in Newly Diagnosed and R/R MM: Highlights from ASH 2024, accessed July 8, 2025, https://www.cancernetwork.com/around-the-practice/evolving-paradigms-in-newly-diagnosed-and-r-r-mm-highlights-from-ash-2024
- ASCO 2025: New standard of care emerges for multiple myeloma - ecancer, accessed July 8, 2025, https://ecancer.org/en/news/26524-asco-2025-new-standard-of-care-emerges-for-multiple-myeloma
- Top Myeloma Research Presented at ASCO, EHA & IMWG 2025 Webinar: The Patient Perspective, accessed July 8, 2025, https://www.myeloma.org/videos/top-myeloma-research-presented-asco-eha-imwg-2025-webinar-patient-perspective
- Chemistry Review(s) - accessdata.fda.gov, accessed July 8, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2005/021880s000_Revlimid_ChemR.pdf
- Attachment 1: Product information for AusPAR Lenalidomide (Revlimid) Celgene Pty Ltd PM, accessed July 8, 2025, https://www.tga.gov.au/sites/default/files/auspar-lenalidomide-160205-pi.pdf
- REVLIMID (lenalidomide) - Medsafe, accessed July 8, 2025, https://www.medsafe.govt.nz/profs/datasheet/r/revlimidcap.pdf
- Pharmacology Review(s) - accessdata.fda.gov, accessed July 8, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2005/021880s000_Revlimid_PharmR.pdf
- Lenalidomide | CAS 191732-72-6 | SCBT - Santa Cruz Biotechnology, accessed July 8, 2025, https://www.scbt.com/p/lenalidomide-191732-72-6
- www.tcichemicals.com, accessed July 8, 2025, https://www.tcichemicals.com/JP/en/p/L0332
- Lenalidomide (LENA) Hybrid Gold Complex Nanoparticles: Synthesis, Physicochemical Evaluation, and Perspectives in Nanomedicine | ACS Omega - ACS Publications, accessed July 8, 2025, https://pubs.acs.org/doi/10.1021/acsomega.0c02644
- 191732-72-6 Lenalidomide AKSci J90127, accessed July 8, 2025, https://aksci.com/item_detail.php?cat=J90127
- Lenalidomide | CC-5013 | CAS#191732-72-6 | TNF-alpha - MedKoo Biosciences, accessed July 8, 2025, https://www.medkoo.com/products/4689
- Lenalidomide (CC-5013, CAS Number: 191732-72-6) | Cayman Chemical, accessed July 8, 2025, https://www.caymanchem.com/product/14643/lenalidomide
- Lenalidomide | CAS 191732-72-6 - LGC Standards, accessed July 8, 2025, https://www.lgcstandards.com/PL/en/Lenalidomide/p/MM3641.00-0250
- Lenalidomide Unknown Status Phase 1 Trials for Lymphoma / Multiple Myeloma (MM) Treatment | DrugBank Online, accessed July 8, 2025, https://go.drugbank.com/drugs/DB00480/clinical_trials?conditions=DBCOND0006647%2CDBCOND0040908&phase=1&purpose=treatment&status=unknown_status
- Database of Drug Development for Rare Diseases - DDrare, accessed July 8, 2025, https://ddrare.nibn.go.jp/ddrare_Mar2023/ddrare_Mar2022/cgi-bin/drug_who_e.cgi?query=Lenalidomide&disease_id=16%2C%2026%2C%2028%2C%2034%2C%2049%2C%20284%2C%20300%2C%20331
- Lenalidomide = 98 HPLC 191732-72-6 - Sigma-Aldrich, accessed July 8, 2025, https://www.sigmaaldrich.com/US/en/product/sigma/sml2283
- Scalable and green process for the synthesis of anticancer drug lenalidomide | Request PDF - ResearchGate, accessed July 8, 2025, https://www.researchgate.net/publication/277625617_Scalable_and_green_process_for_the_synthesis_of_anticancer_drug_lenalidomide
- The novel mechanism of lenalidomide activity - PMC, accessed July 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4653765/
- Lenalidomide Stabilizes Protein–Protein Complexes by Turning Labile Intermolecular H-Bonds into Robust Interactions | Journal of Medicinal Chemistry - ACS Publications, accessed July 8, 2025, https://pubs.acs.org/doi/10.1021/acs.jmedchem.2c01692
- Direct Binding with Cereblon Mediates the Antiproliferative and Immunomodulatory Action of Lenalidomide and Pomalidomide | Blood | American Society of Hematology, accessed July 8, 2025, https://ashpublications.org/blood/article/118/21/738/83093/Direct-Binding-with-Cereblon-Mediates-the
- Mezigdomide—A Novel Cereblon E3 Ligase Modulator under Investigation in Relapsed/Refractory Multiple Myeloma - MDPI, accessed July 8, 2025, https://www.mdpi.com/2072-6694/16/6/1166
- Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells., accessed July 8, 2025, https://www.broadinstitute.org/publications/broad5496
- Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells, accessed July 8, 2025, https://pubmed.ncbi.nlm.nih.gov/24292625/
- Lenalidomide Causes Selective Degradation of IKZF1 and IKZF3 in Multiple Myeloma Cells, accessed July 8, 2025, https://beyondspringpharma.com/wp-content/uploads/2020/08/Kronke-2014.pdf
- Lenalidomide Causes Selective Degradation of IKZF1 and IKZF3 in Multiple Myeloma Cells, accessed July 8, 2025, https://www.researchgate.net/publication/259110094_Lenalidomide_Causes_Selective_Degradation_of_IKZF1_and_IKZF3_in_Multiple_Myeloma_Cells
- Lenalidomide promotes CRBN-mediated ubiquitination and degradation of IKZF1 and IKZF3, accessed July 8, 2025, https://ecancer.org/en/video/2538-lenalidomide-promotes-crbn-mediated-ubiquitination-and-degradation-of-ikzf1-and-ikzf3
- IMiD resistance in multiple myeloma: current understanding of the underpinning biology and clinical impact | Blood, accessed July 8, 2025, https://ashpublications.org/blood/article/142/2/131/494963/IMiD-resistance-in-multiple-myeloma-current
- ADAR1 in lenalidomide resistance: still immunomodulation? | Blood - ASH Publications, accessed July 8, 2025, https://ashpublications.org/blood/article/145/11/1104/535952/ADAR1-in-lenalidomide-resistance-still
- E3 Ligase Ligands in Successful PROTACs: An Overview of Syntheses and Linker Attachment Points - Frontiers, accessed July 8, 2025, https://www.frontiersin.org/journals/chemistry/articles/10.3389/fchem.2021.707317/full
- Clinical Pharmacokinetics and Pharmacodynamics of Lenalidomide - PubMed Central, accessed July 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5247551/
- Clinical Pharmacokinetics and Pharmacodynamics of Lenalidomide - ResearchGate, accessed July 8, 2025, https://www.researchgate.net/publication/304582549_Clinical_Pharmacokinetics_and_Pharmacodynamics_of_Lenalidomide
- Lenalidomide and dexamethasone in patients with relapsed multiple myeloma and impaired renal function: PrE1003, a PrECOG study, accessed July 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6127323/
- Study to Determine Efficacy and Safety of Lenalidomide Plus Low-dose Dexamethasone Versus Melphalan, Prednisone, Thalidomide in Patients With Previously Untreated Multiple Myeloma | ClinicalTrials.gov, accessed July 8, 2025, https://www.clinicaltrials.gov/study/NCT00689936
- REVLIMID U.S. Prescribing Information - Bristol Myers Squibb, accessed July 8, 2025, https://packageinserts.bms.com/pi/pi_revlimid.pdf
- Lenalidomide - Myeloma Patients Europe, accessed July 8, 2025, https://www.mpeurope.org/what-we-do/educational-resources/factsheets/lenalidomide/
- Key Trials From ASH 2024 Impact Treatment for Plasma Cell Disorders Going Forward, accessed July 8, 2025, https://www.targetedonc.com/view/key-trials-from-ash-2024-impact-treatment-for-plasma-cell-disorders-going-forward
- isatuximab, lenalidomide, bortezomib and dexamethasone for newly-diagnosed, transplant-eligible multiple myeloma: post transplantation interim analysis of the randomized phase iii gmmg-hd7 trial - EHA Library - The official digital education library of European Hematology Association (EHA), accessed July 8, 2025, https://library.ehaweb.org/eha/2024/eha2024-congress/422306/marc.s.raab.isatuximab.lenalidomide.bortezomib.and.dexamethasone.for.html?f=menu%3D6%2Abrowseby%3D8%2Asortby%3D2%2Ace_id%3D2552%2Aot_id%3D29201%2Amarker%3D5099%2Afeatured%3D18527
- Practice-changing updates on multiple myeloma: highlights from the 2024 ASH annual meeting - PMC, accessed July 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12077000/
- A review of the history, properties, and use of the immunomodulatory compound lenalidomide - PubMed, accessed July 8, 2025, https://pubmed.ncbi.nlm.nih.gov/21434945/
- EMA approval of lenalidomide plus rituximab for adult patients with R/R FL - Lymphoma Hub, accessed July 8, 2025, https://lymphomahub.com/medical-information/ema-approval-of-lenalidomide-plus-rituximab-for-adult-patients-with-rr-fl
- FDA approves brentuximab vedotin with lenalidomide and rituximab for relapsed or refractory large B-cell lymphoma, accessed July 8, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-brentuximab-vedotin-lenalidomide-and-rituximab-relapsed-or-refractory-large-b-cell
- Dr Mehta on the FDA Approval of Tafasitamab Plus Rituximab/Lenalidomide in R/R Follicular Lymphoma - OncLive, accessed July 8, 2025, https://www.onclive.com/view/dr-mehta-on-the-fda-approval-of-tafasitamab-plus-rituximab-lenalidomide-r-r-follicular-lymphoma
- FDA Approves Tafasitamab Plus Lenalidomide and Rituximab for R/R Follicular Lymphoma, accessed July 8, 2025, https://www.onclive.com/view/fda-approves-tafasitamab-plus-lenalidomide-and-rituximab-for-r-r-follicular-lymphoma
- FINAL SURVIVAL ANALYSIS OF DARATUMUMAB PLUS LENALIDOMIDE AND... by Thierry Facon - EHA Library, accessed July 8, 2025, https://library.ehaweb.org/eha/2024/eha2024-congress/421032/thierry.facon.final.survival.analysis.of.daratumumab.plus.lenalidomide.and.html
- Lenalidomide (Revlimid®) - Macmillan Cancer Support, accessed July 8, 2025, https://www.macmillan.org.uk/cancer-information-and-support/treatments-and-drugs/lenalidomide
- Lenalidomide and dexamethasone - Quick Reference Guide - MRSA Topical Eradication, accessed July 8, 2025, https://www.england.nhs.uk/south/wp-content/uploads/sites/6/2018/11/Lenalidomide-and-dexamethasonev2.pdf
- Lenalidomide Dosage Guide + Max Dose, Adjustments - Drugs.com, accessed July 8, 2025, https://www.drugs.com/dosage/lenalidomide.html
- Lenalidomide/Dasatinib Receive FDA Approval in Hematologic Malignancies, accessed July 8, 2025, https://www.cancernetwork.com/view/lenalidomide-dasatinib-receive-fda-approval-in-hematologic-malignancies
- REVLIMID (lenalidomide) - accessdata.fda.gov, accessed July 8, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/021880s057lbl.pdf
- Revlimid® (Lenalidomide) Capsules (2.5, 5, 10, 15, 20 and 25 mg), accessed July 8, 2025, https://packageinserts.bms.com/sds/sds_revlimid.pdf
- Amneal Launches Mesalamine and Receives U.S. FDA Approval for Lenalidomide, accessed July 8, 2025, https://investors.amneal.com/news/press-releases/press-release-details/2025/Amneal-Launches-Mesalamine-and-Receives-U.S.-FDA-Approval-for-Lenalidomide/default.aspx
- Lenalidomide (Revlimid): Uses, Side Effects, Interactions, Pictures, Warnings & Dosing, accessed July 8, 2025, https://www.webmd.com/drugs/2/drug-94831/revlimid-oral/details
- Lenalidomide Interactions Checker - Drugs.com, accessed July 8, 2025, https://www.drugs.com/drug-interactions/lenalidomide.html
- Lenalidomide: Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed July 8, 2025, https://www.rxlist.com/lenalidomide/generic-drug.htm
- www.medicalnewstoday.com, accessed July 8, 2025, https://www.medicalnewstoday.com/articles/drugs-revlimid-interactions#:~:text=Revlimid%20(lenalidomide)%20may%20interact%20with,blood%20clots%20or%20side%20effects.
- Revlimid interactions: Alcohol, medications, and other factors, accessed July 8, 2025, https://www.medicalnewstoday.com/articles/drugs-revlimid-interactions
- Lenalidomide in Multiple Myeloma: Review of Resistance Mechanisms, Current Treatment Strategies and Future Perspectives - PubMed Central, accessed July 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9913106/
- How Is Lenalidomide Resistance Developed? - HealthTree for Multiple Myeloma, accessed July 8, 2025, https://healthtree.org/myeloma/community/articles/how-is-lenalidomide-resistance-developed
- Resistance to immunomodulatory drugs in multiple myeloma: the cereblon pathway and beyond | Haematologica, accessed July 8, 2025, https://haematologica.org/article/view/11833
- New breakthrough combats lenalidomide resistance in multiple myeloma - ecancer, accessed July 8, 2025, https://ecancer.org/en/news/26305-new-breakthrough-combats-lenalidomide-resistance-in-multiple-myeloma
- Quantitative Proteomics Reveals the Role of Lysine Lactylation in Lenalidomide-Resistance in Multiple Myeloma Cells - ACS Publications - American Chemical Society, accessed July 8, 2025, https://pubs.acs.org/doi/10.1021/acschembio.5c00270
- Adaptive Biotechnologies Highlights New Data at 2025 ASCO Annual Meeting and EHA 2025 Congress Demonstrating How clonoSEQ® MRD Assessment is Optimizing Patient Care and Drug Development in Lymphoid Cancers | Morningstar, accessed July 8, 2025, https://www.morningstar.com/news/globe-newswire/9460513/adaptive-biotechnologies-highlights-new-data-at-2025-asco-annual-meeting-and-eha-2025-congress-demonstrating-how-clonoseq-mrd-assessment-is-optimizing-patient-care-and-drug-development-in-lymphoid-cancers
- EHA 2025 | Top abstracts in MM and other plasma cell dyscrasias - Multiple Myeloma Hub, accessed July 8, 2025, https://multiplemyelomahub.com/medical-information/eha-2025-top-abstracts-in-mm-and-other-plasma-cell-dyscrasias
- Sustained MRD Negativity for Three Years Can Guide Discontinuation of Lenalidomide Maintenance after ASCT in Multiple Myeloma: Results from a Prospective Cohort Study, accessed July 8, 2025, https://ashpublications.org/blood/article/144/Supplement%201/361/531161/Sustained-MRD-Negativity-for-Three-Years-Can-Guide
- Lenalidomide in Multiple Myeloma: Review of Resistance Mechanisms, Current Treatment Strategies and Future Perspectives - PubMed, accessed July 8, 2025, https://pubmed.ncbi.nlm.nih.gov/36765919/
- Forcing Ahead: Second-Line Treatment Options for Lenalidomide-Refractory Multiple Myeloma - PMC, accessed July 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11987876/
- A REAL-WORLD STUDY OF LENALIDOMIDE REFRACTORY MULTIPLE MYELOMA... by Marika Waltari - EHA Library, accessed July 8, 2025, https://library.ehaweb.org/eha/2025/eha2025-congress/4160853/marika.waltari.a.real-world.study.of.lenalidomide.refractory.multiple.myeloma.html?f=listing%3D0%2Abrowseby%3D8%2Asortby%3D2%2Amedia%3D3%2Asearch%3Disatuximab
- Ciltacabtagene autoleucel vs standard of care in lenalidomide-refractory multiple myeloma: Phase 3 CARTITUDE-4 subgroup analysis by cytogenetic risk - EHA Library, accessed July 8, 2025, https://library.ehaweb.org/eha/2024/eha2024-congress/421042/roberto.mina.ciltacabtagene.autoleucel.vs.standard.of.care.in.html
- May 2025 — Myeloma News from the Journals, accessed July 8, 2025, https://www.myeloma.org/news-events/multiple-myeloma-news/may-2025-myeloma-news-journals
- Highlights from ASCO June 2, 2025 - Society for Immunotherapy of Cancer (SITC), accessed July 8, 2025, https://www.sitcancer.org/blogs/thomas-martin/2025/06/03/highlights-from-asco-june-2-2025
- 25 Posts Not to Miss from EHA 2025 - Oncodaily, accessed July 8, 2025, https://oncodaily.com/societies/eha-2025-312591
- Linvoseltamab Receives EU Approval for Relapsed/Refractory Multiple Myeloma, accessed July 8, 2025, https://www.cancernetwork.com/view/linvoseltamab-receives-eu-approval-for-relapsed-refractory-multiple-myeloma
- January 2025 – Myeloma News from the Journals, accessed July 8, 2025, https://www.myeloma.org/news-events/multiple-myeloma-news/january-2025-myeloma-news-journals
- Clinical Trials Using Lenalidomide - NCI, accessed July 8, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/lenalidomide?pn=1
- ciltacabtagene autoleucel ± lenalidomide maintenance in newly diagnosed multiple myeloma with suboptimal response to frontline autologous stem cell transplant: cartitude-2 cohort d - EHA Library - European Hematology Association, accessed July 8, 2025, https://library.ehaweb.org/eha/2024/eha2024-congress/422309/wilfried.roeloffzen.ciltacabtagene.autoleucel.lenalidomide.maintenance.in.html?f=menu%3D6%2Abrowseby%3D8%2Asortby%3D2%2Ace_id%3D2552%2Aot_id%3D29201%2Amarker%3D5099%2Afeatured%3D18527
- UCSF Multiple Myeloma Clinical Trials — San Francisco Bay Area, accessed July 8, 2025, https://clinicaltrials.ucsf.edu/multiple-myeloma