Fact-checked by Grok 2 weeks ago

Cereblon E3 ligase modulator

Cereblon E3 ligase modulators are small molecules that bind directly to the (CRBN) protein, serving as the substrate receptor in the Cullin 4-RING (CRL4CRBN) complex, to reprogram its substrate specificity and thereby promote the ubiquitination and subsequent proteasomal of select neo-substrate proteins. These modulators, exemplified by and its analogs and —collectively known as immunomodulatory drugs (IMiDs)—induce the of transcription factors such as () and IKZF3 (Aiolos), which underlies their efficacy in treating and other hematologic malignancies. Originally identified through the teratogenic and therapeutic effects of , this mechanism has spurred the development of next-generation compounds termed Cereblon modulators (CELMoDs), which exhibit enhanced binding affinity and broader substrate profiles for potential applications in and beyond. The paradigm has also foundational role in proximity-inducing targeted protein technologies, including proteolysis-targeting chimeras (PROTACs), by leveraging CRBN recruitment to eliminate disease-associated proteins.

Discovery and Historical Development

Thalidomide's initial applications and withdrawal

Thalidomide was first synthesized in 1954 by chemists at in , following its discovery in 1953, initially developed as a non-barbiturate with purported properties. Marketed under names like Contergan starting in , it gained rapid approval in over 40 countries due to claims of low toxicity, including no lethal dose observed in , and was widely prescribed for , anxiety, and especially morning sickness in pregnant women despite limited preclinical testing in pregnant models. By the early , annual sales exceeded 10 million tablets globally, with distribution in at least 46 countries, as its perceived safety profile—bolstered by over-the-counter availability in some markets—drove adoption even amid emerging reports of from prolonged use, which manufacturers and regulators largely dismissed as unrelated or reversible. The drug's teratogenic effects became evident in 1961 when Australian obstetrician William McBride and German pediatrician Widukind Lenz independently linked it to a surge in and other limb malformations, with causal exposure confined to days 20-36 post-fertilization during embryonic limb bud development. This period aligned with vulnerability, resulting in an estimated 10,000 to 12,000 affected births worldwide by 1962, primarily in , as retrospective epidemiological data confirmed dose-dependent correlations absent in control populations. Chemie withdrew on November 26, 1961, followed by most distributors by 1962, exposing deficiencies in regulatory reliance on rodent models where teratogenicity was not detected, underscoring the limitations of interspecies without broader empirical validation in or humans. Post-withdrawal, thalidomide's utility reemerged in 1964 when Israeli physician Jacob Sheskin observed rapid remission of leprosum (ENL) symptoms in a patient treated for , prompting controlled trials that established its efficacy in modulating inflammatory responses without the overdose risks of prior uses. The U.S. approved it in July 1998 specifically for acute ENL treatment and maintenance in , under a restricted distribution program (later formalized as iPLEDGE/REMS) mandating pregnancy testing and contraception to mitigate teratogenic risks, reflecting data-driven risk-benefit reassessment from observational and clinical evidence in non-pregnant populations.

Identification of cereblon as the primary target

In 2010, researchers led by Takumi Ito identified cereblon (CRBN), a substrate receptor in the Cullin-Ring E3 ubiquitin ligase complex CRL4^{DDB1}, as the primary cellular target of thalidomide through affinity purification using thalidomide-immobilized beads, which specifically pulled down CRBN from rabbit embryo extracts. This discovery built on prior observations of CRBN gene mutations associated with autosomal recessive nonsyndromic intellectual disability, providing a functional context for CRBN's role in developmental processes. Biochemical assays confirmed direct binding of thalidomide to CRBN with a dissociation constant in the micromolar range, and binding was competed by thalidomide analogs, establishing specificity over nonspecific interactions. To link CRBN binding to thalidomide's teratogenic effects, Ito et al. employed chick embryo models, where thalidomide exposure suppressed expression of fibroblast growth factors Fgf8 and Fgf10 in developing limb buds, critical for proximal-distal patterning; this suppression was absent in embryos expressing a thalidomide-resistant CRBN mutant (R419X, derived from human intellectual disability cases), which reduced drug affinity and blocked limb defects. Complementary experiments in zebrafish demonstrated that CRBN ortholog knockdown via morpholinos phenocopied thalidomide-induced reductions in caudal fin development, while overexpression of wild-type CRBN but not the resistant mutant restored normal morphogenesis in the presence of the drug, providing causal evidence that CRBN modulation drives teratogenicity rather than indirect pathways like anti-angiogenesis. Parallel investigations into 's therapeutic efficacy in revealed that CRBN engagement promotes ubiquitination and proteasomal degradation of transcription factors () and IKZF3 (Aiolos), contradicting earlier cytokine-centric hypotheses by demonstrating direct control; , a thalidomide analog, induced selective IKZF1/3 degradation in myeloma cell lines within hours, an effect abolished in CRBN-knockdown cells or with proteasome inhibitors. confirmed IKZF1/3 as primary neo-substrates, with degradation correlating to antiproliferative effects independent of . Crystal structures published in 2014 by Eric S. Fischer et al. resolved the DDB1-CRBN complex bound to at 2.0 Å resolution, visualizing how the drug occupies a conserved pocket on CRBN's , inducing a conformational change that exposes a novel β-sheet surface for neo-substrate recruitment, thus mechanistically validating the "molecular glue" hypothesis and enabling structure-based design of targeted degraders. These findings shifted paradigms from correlative binding to causal degradation mechanisms, underscoring CRBN's role in both and .

Evolution from IMiDs to targeted degraders

, approved by the FDA on December 27, 2005, for use in combination with dexamethasone in patients who had received at least one prior therapy, represented a significant advancement over as an immunomodulatory drug (IMiD) with enhanced potency in inducing degradation of transcription factors and IKZF3, estimated at 100- to 1,000-fold greater in cellular assays compared to due to improved binding affinity. , approved on February 8, 2013, for relapsed or refractory after at least two prior therapies including and , further optimized this profile by promoting more rapid and complete ubiquitination of IKZF substrates than or in preclinical models. Post-2010 rational efforts shifted toward E3 ligase modulators (CELMoDs), exemplified by iberdomide (CC-220) and mezigdomide (CC-92480), which exhibit higher -binding affinity than IMiDs to enable selective neo-substrate recruitment and immune modulation while addressing resistance. In phase 1/2 trials for relapsed/ , iberdomide plus dexamethasone yielded overall response rates exceeding 30% in heavily pretreated patients to multiple therapies, including prior IMiDs. Mezigdomide demonstrated enhanced tumoricidal activity against both IMiD-sensitive and resistant cells in early clinical data from the CC-92480-MM-002 study, supporting its advancement in combination regimens. This evolution extended to proteolysis-targeting chimeras (PROTACs) incorporating CRBN-recruiting moieties, which by 2025 had been developed against over 60 protein targets, allowing of substrates beyond the limited neo-substrates of IMiDs and CELMoDs by linking diverse ligands to CRBN binders like derivatives. A primary empirical driver was overcoming IMiD conferred by CRBN , which disrupt IMiD-induced neo-substrate ; preclinical studies indicate CELMoDs retain activity against certain CRBN mutants due to their superior , though varies by type.

Molecular Mechanism of Action

Binding and modulation of cereblon-CRL4 E3 ligase

(CRBN) serves as the recognition receptor within the cullin-RING ubiquitin ligase complex CRL4^{CRBN}, which comprises CRBN, damaged DNA-binding protein 1 (DDB1), cullin-4A/B (CUL4), and RING-box protein 1 (RBX1). In its physiological state, CRBN recruits specific , such as or MEIS2 protein, to the complex for polyubiquitination by an E2-conjugating enzyme, marking them for proteasomal degradation. This process regulates diverse cellular functions, including amino acid and developmental signaling. CELMoDs, including classical immunomodulatory drugs (IMiDs) like thalidomide, lenalidomide, and pomalidomide, bind within a conserved pocket on CRBN's helical domain, often termed the thalidomide-binding pocket, characterized by three tryptophan residues (TRIP12 motif). Binding affinities vary, with thalidomide exhibiting a dissociation constant (K_d) of approximately 250 nM, while pomalidomide achieves tighter binding in the 200-500 nM range; these interactions induce a conformational shift in CRBN, exposing a cryptic neo-surface that alters substrate specificity. Crystal structures resolved in 2014 (e.g., PDB: 4CI1) and subsequent cryo-EM analyses (resolutions up to 3.4 Å from 2020-2025) reveal how this binding rigidifies ternary complexes between CRBN, the modulator, and neo-substrates, positioning lysine residues proximal to the E2 active site for efficient ubiquitination. IMiDs function primarily as molecular glues, small monovalent molecules (<500 Da) that stabilize otherwise weak or transient CRBN-neo-substrate interactions without requiring a flexible linker, enabling degradation at DC_{50} values often in the 10-100 nM range for neo-substrates like IKZF1/3. In contrast, bifunctional proteolysis-targeting chimeras (PROTACs) incorporate a CRBN-recruiting warhead (typically an IMiD derivative) linked to a target-binding moiety, forming extended ternary complexes that tether distant proteins for ubiquitination; this mechanism accommodates larger architectures but demands optimized linker lengths (10-20 atoms) for proximity-induced degradation kinetics. Both modalities hijack without altering the core ubiquitination machinery, though glue-induced complexes exhibit higher rigidity and potentially faster on/off kinetics compared to PROTAC-mediated ones. Empirical evidence from structural biology underscores causality in substrate redirection: thalidomide binding to CRBN provokes degradation of transcription factors like SALL4, disrupting mesenchymal-to-epithelial transition in limb bud development, as evidenced by impaired angiogenesis and outgrowth in CRBN-dependent models. Degradation efficiency correlates with ternary complex stability, with DC_{50} values reflecting not just binding affinity but also ubiquitination rates (half-life reductions from hours to minutes for neo-substrates). These biophysical insights, derived from unbiased screens and high-resolution structures spanning 2014-2025, highlight how pocket occupancy modulates CRL4^{CRBN} without intrinsic enzymatic activation.

Neo-substrate recruitment and ubiquitination

Cereblon E3 ligase modulators, such as immunomodulatory drugs (IMiDs), induce the recruitment of non-native substrates, termed neo-substrates, to the CRL4CRBN ubiquitin ligase complex by binding to (CRBN) and altering its substrate-binding pocket. This binding promotes the formation of a stable ternary complex consisting of the modulator, CRBN, and neo-substrates like the zinc-finger transcription factors and IKZF3, facilitating their polyubiquitination on lysine residues and subsequent proteasomal degradation. studies using in IMiD-treated cells have empirically confirmed the selective ubiquitination and depletion of IKZF1 and IKZF3, with degradation levels correlating directly with drug exposure. The efficiency of neo-substrate ubiquitination depends on the stability of the ternary complex rather than simple binary binding affinities, as structural analyses reveal that modulator-induced conformational changes in CRBN expose a neo-substrate recognition interface essential for activity. In cells, degradation exhibits dose-dependence: low concentrations achieve partial /3 depletion sufficient for modulating cellular , while higher doses drive near-complete degradation, triggering particularly in cells reliant on expression regulated by these transcription factors. Unbiased global screens have identified additional neo-substrates beyond /3, comprising a degradome of dozens of proteins, with off-target effects accounting for unintended ubiquitination events that contribute to both therapeutic toxicities and ancillary benefits such as anti-angiogenic activity observed in preclinical models.

Downstream effects on transcription factors and immunity

Degradation of the transcription factors (Ikaros) and IKZF3 (Aiolos) by modulators, such as and , disrupts their repressive function on . In human T cells, this loss derepresses production of interleukin-2 (IL-2) and interferon-gamma (IFN-γ), promoting a Th1-skewed response. assays of human peripheral blood mononuclear cells (PBMCs) demonstrate enhanced proliferation and secretion from CD4+ and CD8+ T cells upon co-stimulation with these agents at concentrations of 0.03–1 μM. This /3 degradation also augments natural killer () and NKT cell activation, increasing their cytotoxic potential against myeloma cells through elevated IFN-γ and IL-2 output. In models, the resulting downregulation—mediated indirectly via IKZF loss—correlates with direct tumor cell and improved (PFS) in lenalidomide-treated patients, as observed in clinical trials where responders showed rapid IRF4 reduction within 8 hours of exposure. These effects underscore as a primary , rather than secondary , challenging earlier emphases on broad anti-inflammatory actions like TNF-α suppression, which predated substrate identification and apply more to than next-generation IMiDs. Empirical data reveal species-specific variations in immunomodulatory potency; human exhibits stronger IKZF recruitment than rodent orthologs, explaining weaker T-cell and responses in murine models and historical gaps in preclinical predictions. In human PBMC assays, enhances + T-cell antitumor functions via ERK signaling and IFN-γ secretion, independent of direct modulation in . Clinical correlations, such as PFS extension in del(5q) myeloma subsets unresponsive to TNF-α pathways, further prioritize degradation-driven immunity over inhibition narratives.

Chemical Classes and Structure-Activity Relationships

Classical immunomodulatory drugs (IMiDs)


Classical immunomodulatory drugs (IMiDs), comprising , , and , share a or isoindolinone moiety fused to a glutarimide ring, with the latter serving as the primary for (CRBN) engagement. binds CRBN weakly, with dissociation constants (K_d) reported in the range of 0.6–30 μM depending on the protein construct and assay conditions, limiting its efficiency in neo-substrate recruitment. Introduction of an amino group at the 4-position of the aromatic ring in and forms hydrogen bonds with and residues in CRBN's thalidomide-binding domain (TBD), boosting binding affinity 100- to 500-fold to sub-micromolar levels (K_d ≈ 0.16–0.5 μM). This enhancement translates to potent ubiquitination and proteasomal degradation of neo-substrates like and IKZF3, with half-maximal degradation concentrations (DC_{50}) reaching 5–50 nM, far surpassing thalidomide's micromolar thresholds.
Structural variations in the non-glutarimide portion influence ancillary activities. The isoindolinone scaffold in , versus the in and , reduces and attenuates sedation linked to off-target phosphodiesterase 4 (PDE4) inhibition, while maintaining CRBN-modulating glue functionality. Quantitative structure-activity relationship (QSAR) studies from Celgene's analog libraries reveal that moderate increases in —via alkyl or aryl substitutions—correlate with heightened efficiency for IKZF proteins, optimizing ternary complex stability without proportionally elevating binding affinity.
CompoundCRBN K_d (approximate)IKZF DC_{50} (approximate)Key Modification
0.6–30 μM>1 μMUnsubstituted phthalimide
~0.16 μM10–50 nM4-Amino-isoindolinone
~0.3 μM5–20 nM4-Amino-phthalimide
Potency gains incur selectivity trade-offs; steeper dose-response curves for neo-substrate degradation in more affine IMiDs align with clinical observations of dose-limiting , potentially stemming from PU.1 downregulation and myeloid maturation blockade in hematopoietic progenitors. , the most potent, exhibits higher grade 3–4 neutropenia incidence (up to 50% in trials) compared to (30–40%), underscoring a potency-toxicity continuum.

Next-generation CELMoDs

Iberdomide (CC-220) exemplifies SAR advancements through extensions of the ring, including fluorination at the 3-position, which enhance selectivity for family proteins (/3) degradation over classical IMiDs like . These modifications yield half-maximal degradation concentrations (DC50) for in the low nanomolar range (approximately 2.4 nM), surpassing 's potency by an in comparable assays, while data indicate fewer off-target substrates due to tighter CRBN engagement and reduced . This selectivity stems from stabilized ternary complex formation, promoting efficient ubiquitination kinetics without broadly altering CRL4CRBN substrate recognition. Mezigdomide (CC-92480) incorporates cyclic fusions to the glutarimide core, augmenting ternary complex stability with CRBN and neo-substrates via deeper pocket occupancy and conformational locking of CRBN in its closed state. These structural iterations result in superior , enabling mezigdomide to elicit in iberdomide-resistant myeloma cell lines , where IMiDs fail, through prolonged /3 depletion and downstream apoptotic signaling. Crystal structures confirm enhanced hydrophobic interactions in the CRBN thalidomide-binding domain, correlating with higher binding affinity (Kd values sub-nanomolar) relative to predecessors. Broader principles guiding CELMoD optimization involve halogen substitutions and ring appendages that fine-tune CRBN pocket filling, directly linking occupancy enhancements to rates in primary myeloma samples; for instance, fluoro-additions increase , boosting neo-substrate recruitment efficiency by 5-10 fold in assays. These evolutions prioritize causal potency over mere , yielding compounds with faster on-rates for ubiquitination compared to IMiDs, as quantified by time-resolved .

Cereblon-recruiting PROTACs and molecular glues

Cereblon-recruiting proteolysis-targeting chimeras (PROTACs) are heterobifunctional small molecules comprising a for a (), a chemical linker, and a cereblon-binding warhead, such as or analogs, designed to induce ubiquitination and proteasomal degradation of the POI via the CRL4CRBN complex. Unlike classical immunomodulatory drugs (IMiDs), which act as molecular glues to degrade endogenous neo-substrates like /3, PROTACs enable target-specific degradation by incorporating POI-selective binders, expanding the addressable beyond natural CRBN interactors. Structure-activity relationship () studies for PROTACs emphasize warhead potency and linker optimization to favor ternary complex (POI-PROTAC-CRBN) formation over complexes. High-affinity CRBN warheads, often featuring piperidine-2,6-dione scaffolds with aryl substitutions, ensure robust recruitment, while POI ligands (e.g., for CDK4/6) are tethered via variable linkers. Empirical data indicate that PEG-based linkers spanning 15-20 Å—corresponding to 4-6 units—optimize spatial geometry for CDK4/6 degraders, yielding degradation maxima (Dmax) >90% and half-maximal degradation concentrations (DC50) in the low nanomolar range, surpassing the stoichiometric limitations of glue-induced neo-substrate degradation. Longer or rigid linkers may disrupt proximity, reducing efficiency, whereas suboptimal lengths promote non-productive binding. Molecular glues, in contrast, lack dedicated POI ligands and instead stabilize aberrant CRBN-neo-substrate interfaces through conformational modulation, with guided by binder affinity and induced-fit dynamics rather than linker design. Non-bifunctional "mini-glues" target novel CRBN pockets with compact scaffolds, prioritizing rigidity (e.g., fused rings or constrained conformations) to drive transient, event-specific degradation events that minimize off-target effects. For instance, indazole-based CRBN ligands exhibit favoring planarity for enhanced ternary cooperativity in cyclin K degradation. A hallmark empirical for both modalities is the hook effect, wherein excessive concentrations saturate binary complexes (e.g., POI-PROTAC or CRBN-PROTAC), inhibiting assembly and ; this bell-shaped dose-response is pronounced in PROTACs due to their extended structures. strategies include linker tuning for higher cooperativity or multivalent designs, such as trivalent PROTACs incorporating additional motifs to sustain productive complexes at elevated doses. Glues, being smaller, often evade severe hooking but remain constrained by endogenous substrate competition.

Clinical Applications

Lenalidomide combined with dexamethasone demonstrated significant efficacy in relapsed or refractory in the phase III MM-009 and MM-010 trials conducted in 2005-2006, achieving an overall response rate (ORR) of 60.6% compared to 21.9% with dexamethasone alone, with median overall survival (OS) prolonged to approximately 38 months versus 31.6 months in long-term follow-up. This regimen effectively doubled (PFS) relative to historical dexamethasone monotherapy benchmarks of around 18 months OS in relapsed settings, establishing as a standard second-line option post-bortezomib failure. However, these gains reflect median outcomes in a heterogeneous , with actual long-term survival limited by inevitable relapse and without curative intent. Pomalidomide, approved for heavily pretreated or refractory , yields ORR around 30% when combined with dexamethasone in trials such as MM-003 (2012-2013), particularly in lenalidomide-refractory cases, with median PFS of 4 months and OS of 12 months. In French Intergroupe Francophone du Myelome (IFM) studies from 2013 onward, pomalidomide-based regimens in triple-class refractory disease show comparable response durability, though efficacy diminishes in patients with prior immunomodulatory drug exposure. These agents extend remission modestly but do not overcome intrinsic refractoriness, underscoring their role as bridging therapies rather than definitive cures. Next-generation E3 ligase modulators (CELMoDs), such as mezigdomide, exhibit enhanced activity in pomalidomide-refractory , with phase 1/2 trials (CC-92480-MM-002, 2022-2024) reporting ORR of 40-50% in combination with dexamethasone among heavily pretreated patients, including those with extramedullary disease. Median PFS reaches 6-8 months in these cohorts, surpassing benchmarks, though myelosuppression limits dosing intensity. Ongoing data from 2024-2025 emphasize CELMoDs' potential in overcoming partial IMiD cross-resistance via tighter binding, yet responses remain transient without multi-agent integration. Combination regimens incorporating IMiDs or CELMoDs with proteasome inhibitors like or monoclonal antibodies such as further prolong PFS and OS; for instance, -pomalidomide-dexamethasone achieves ORR over 60% and median PFS of 8-12 months in relapsed settings per APOLLO and OPTIMIS trials. Similarly, lenalidomide--dexamethasone extends remission post-induction, with 3-year OS rates exceeding 70% in transplant-eligible patients, though benefits attenuate in non-transplant candidates where median survival gains are modest (6-12 months). Empirical evidence highlights CRBN pathway aberrations, including mutations or deletions in 12-30% of cases, as predictors of non-response, occurring more frequently post-IMiD exposure and rendering cells insensitive to substrate degradation. Despite these advances, IMiDs and CELMoDs do not achieve cure in , with median survival extensions often confined to 1-2 years in relapsed disease absent autologous transplantation, which amplifies frontline gains but fails to prevent eventual progression in over 80% of cases. Claims of transformative efficacy must account for selection biases in trials excluding high-risk , where resistance emerges rapidly via CRBN-independent pathways like /3 dysregulation. Real-world outcomes lag trial data due to comorbidities and sequential therapy exhaustion, reinforcing the need for resistance biomarkers over optimistic narratives.

Applications in autoimmune and inflammatory diseases

, the prototypical cereblon E3 ligase modulator, was reintroduced in the 1960s for managing leprosum (ENL), an immune-mediated inflammatory reaction in patients refractory to corticosteroids and dapsone. Early clinical trials reported rapid symptom resolution, including reduced skin lesions and fever, in 70-80% of severe, recurrent ENL cases, with mechanisms involving TNF-α suppression via cereblon-mediated degradation of transcription factors like and Aiolos. However, neuropathy incidence reached 20-50% with prolonged use (>6 months), a often downplayed in initial efficacy-focused studies from leprosy-endemic regions, where short-term benefits outweighed documented sensory nerve damage in empirical follow-up data. In (GVHD), particularly chronic forms post-allogeneic , and have shown partial efficacy through , with response rates of 30-50% in steroid-refractory cutaneous and mucosal manifestations via targeted ubiquitination of immune transcription factors. , at doses of 5-10 mg daily, induced objective improvements in 40-60% of chronic GVHD cases in phase II trials, but empirical observations revealed paradoxical T-cell activation and flares exacerbating acute GVHD in up to 20% of patients, limiting its adoption beyond niche salvage settings. These risks, rooted in off-target immune stimulation rather than pure suppression, highlight causal discrepancies in narratives portraying IMiDs as broadly anti-inflammatory, as trial discontinuations due to progression underscore selective reporting biases in oncology-adjacent literature. Lenalidomide trials in systemic (SLE) and (RA) yielded mixed outcomes, with efficacy confined to refractory cutaneous (CLE) subsets, where 50-70% of patients achieved clearance at 5-10 mg doses over 6-12 months. In SLE, histologic improvements correlated with reduced B-cell activity, yet release syndromes and systemic flares occurred in 10-30% of cases, driven by empirical T-cell data that contradict generalized anti-inflammatory claims and prompted early trial halts. RA studies similarly reported insufficient joint response rates (<40%) alongside hematologic toxicities, supporting niche utility only where standard immunosuppressants fail, per causal analyses of immune pathway activation over suppression. Emerging cereblon modulators like iberdomide, a next-generation agent, demonstrated in a 2022 phase II SLE trial (NCT03081791) that 0.45 mg daily dosing achieved Systemic Lupus Erythematosus Responder Index-4 (SRI-4) responses in 45% of patients versus 20% placebo at 24 weeks, with reductions in autoantibodies via enhanced IKZF1/3 degradation. However, cytopenias led to dose reductions in 15-25% and dropouts in 10%, reflecting persistent class effects underemphasized in preliminary efficacy summaries from sponsor-driven reports, where long-term autoimmune applicability remains constrained by these empirical safety signals absent in optimistic projections.

Emerging uses in other conditions

Cereblon-recruiting PROTACs have entered early clinical testing for solid tumors, including prostate cancer, where they target proteins like the androgen receptor (AR) for degradation. ARV-110, a bifunctional degrader linking AR to cereblon, achieved prostate-specific antigen (PSA) reductions in phase 1/2 trials for metastatic castration-resistant prostate cancer, with 34% of full-dose patients (23/68) reaching PSA30 and 55% (11/20) at the 900 mg twice-daily dose, indicating preliminary antitumor activity as of May 2025. Similarly, ARV-766, another cereblon-based AR degrader, has progressed to phase 2 evaluation in advanced prostate cancer, building on phase 1 safety data without exceeding predefined toxicity thresholds. These applications extend beyond hematologic malignancies, leveraging cereblon's ubiquitination machinery to address drivers in epithelial-derived cancers. In neurological disorders, IMiDs and related cereblon modulators show preclinical potential for neurodegeneration by modulating neuroinflammation and protein aggregation, as explored in models of conditions like amyotrophic lateral sclerosis (ALS) through enhanced clearance of aberrant proteins. However, human evidence remains limited, with no advanced trials reported for ALS or similar diseases by 2025, and repurposing efforts confined to exploratory in vitro and animal studies assessing immunomodulatory effects on neuronal survival. Translational challenges persist, including unpredictable off-target degradation of neo-substrates, which complicates selectivity and contributes to high attrition; over 20 cereblon-based PROTACs have reached phase 1/2 by 2025, but few demonstrate sufficient efficacy-toxicity ratios for broader advancement, underscoring empirical hurdles in non-hematologic contexts.

Adverse Effects and Safety Profile

Teratogenic risks and reproductive toxicity

Thalidomide exerts teratogenic effects primarily through binding to , a substrate receptor in the Cullin-Ring E3 ubiquitin ligase complex, which redirects its activity to ubiquitinate and degrade specific transcription factors such as . This degradation disrupts downstream signaling pathways, including those involving essential for limb bud formation, leading to phocomelia and other malformations in developing embryos. The mechanism has been replicated in vertebrate models, including zebrafish exhibiting pectoral fin defects upon thalidomide exposure, and human induced pluripotent stem cell (iPSC)-derived models demonstrating similar disruptions in mesodermal patterning and organogenesis. Empirical evidence confirms causality, as CRBN mutations resistant to thalidomide binding abolish these effects, while overexpression of wild-type CRBN sensitizes non-responsive species like rodents. Critical exposure occurs between 20 and 37 days post-conception, corresponding to the embryonic period of limb, ear, and cardiac development, with even single doses sufficient to induce defects. Historical data from the 1950s-1960s indicate that exposure during this window resulted in birth defects in approximately 20-30% of affected pregnancies, far exceeding the 3-5% background rate, though overall incidence varied with dosing and timing. In the modern era, U.S. reintroduction of in 1998 under the System for Thalidomide Education and Prescribing Safety (S.T.E.P.S.) program, followed by integration into iPLEDGE-like risk evaluation and mitigation strategies (REMS) for IMiDs, has reduced but not eliminated fetal exposures; post-1998 reports document over 200 pregnancies among treated patients, underscoring persistent compliance failures despite mandatory contraception, pregnancy testing, and prescriber registration. Cereblon E3 ligase modulators structurally analogous to , such as and , retain high teratogenic potency due to shared CRBN-binding and neo-substrate degradation profiles, inducing similar embryonic toxicities in preclinical models. Both carry FDA boxed warnings contraindicating use in pregnancy, with evidence of embryo-fetal lethality and malformations in rats and rabbits at doses below human equivalents, necessitating identical REMS protocols including negative pregnancy tests and abstinence requirements for patients of reproductive potential. Semen from treated males contains these agents, prompting recommendations for condom use to prevent indirect fetal exposure.

Hematologic and immunologic toxicities

Hematologic toxicities associated with cereblon E3 ligase modulators, particularly immunomodulatory drugs (IMiDs) such as lenalidomide and pomalidomide, primarily manifest as dose-limiting cytopenias, including neutropenia, thrombocytopenia, and anemia. In clinical trials for relapsed/refractory multiple myeloma, grade 3/4 neutropenia occurs in approximately 25-50% of patients treated with lenalidomide, with rates reaching up to 41.5% in some cohorts, while thrombocytopenia affects 11-15% at grade 3/4. For pomalidomide plus low-dose dexamethasone, grade 3/4 neutropenia is reported in 49.7% of cases, thrombocytopenia in 24.1%, and anemia in 33%. These events are often the leading causes of dose interruptions or reductions, with neutropenia and thrombocytopenia prompting adjustments in over 50% of lenalidomide-treated patients in certain regimens. The causal mechanism involves IMiD-induced ubiquitination and proteasomal degradation of transcription factors IKZF1 and IKZF3 via the cereblon E3 ligase complex, which disrupts normal hematopoiesis in CD34+ progenitor cells. This degradation shifts differentiation toward immature myeloid precursors and inhibits apoptosis in hematopoietic progenitors, exacerbating myelosuppression independently of anti-myeloma efficacy. Severe cytopenias are dose-dependent and more pronounced after multiple cycles, though they rarely lead to permanent discontinuation in less than 5% of cases when managed proactively. Immunologic toxicities stem from secondary immunosuppression, increasing susceptibility to infections such as herpes zoster reactivation, with elevated incidence reported in lenalidomide-treated multiple myeloma patients due to altered T-cell function and cytokine profiles. This risk is compounded by neutropenia-related immune compromise, prompting routine antiviral prophylaxis in clinical practice. Other opportunistic infections, including pneumonia, occur at rates up to 17-29% for grade 3/4 events in IMiD regimens. Empirical management through dose reductions or delays effectively mitigates these toxicities while maintaining therapeutic efficacy; for instance, reductions after 12 months of therapy preserve progression-free survival without compromising outcomes. In real-world data, such adjustments occur in 35-62% of patients and correlate with prolonged treatment duration and reduced severe event rates. Growth factor support, such as granulocyte colony-stimulating factor for neutropenia, further aids in sustaining dosing intensity.

Long-term and off-target effects

Long-term use of thalidomide, a prototypical , is associated with peripheral neuropathy in 20-50% of patients, with incidence rising cumulatively over time and reaching up to 73% between 6 and 12 months in some cohorts based on actuarial analyses. Sensory nerve conduction studies indicate that 10-20% of these cases prove irreversible, particularly after exposure exceeding 12 months, limiting sustained therapy and necessitating dose reductions or discontinuation. While lenalidomide and pomalidomide exhibit lower neuropathy rates than thalidomide, primarily due to reduced cumulative dosing in maintenance regimens, vigilance remains essential in multiple myeloma patients receiving prolonged treatment. Lenalidomide maintenance therapy in multiple myeloma has been linked to a modest increase in secondary primary malignancies, including myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML), with incidences of 3.1 per 100 patient-years versus 1.2 in placebo controls, equating to roughly a 1-2% excess risk over several years. This association persists after adjusting for prior melphalan exposure, though debate centers on whether it reflects direct drug genotoxicity or selection effects from underlying disease biology and extended survival enabling malignancy emergence. Longitudinal data from trials like CALGB 100104 underscore the need to weigh this against survival benefits, as median survival post-MDS/AML diagnosis remains poor at around 2.4 months. Off-target degradation poses risks in next-generation cereblon modulators, including and , where proteomics profiling of degradomes reveals unintended ubiquitination and proteasomal breakdown of neo-substrates beyond intended targets like . Such promiscuity, stemming from cereblon pocket conformational changes, may contribute to chronic symptoms like fatigue and gastrointestinal disturbances through dysregulated pathways, though causality requires further longitudinal validation. In IMiD-based systems, retention of parent compound liabilities exacerbates these effects, highlighting the imperative for degradome mapping in preclinical optimization to mitigate long-term toxicities.

Pharmacokinetics and Drug Metabolism

Absorption, distribution, and elimination of key agents

Thalidomide is rapidly absorbed after oral administration, achieving peak plasma concentrations within 2-4 hours, with an estimated bioavailability approaching 90% due to minimal first-pass metabolism. Its elimination half-life averages 5-7 hours, reflecting a flip-flop phenomenon where absorption rate exceeds elimination, leading to hepatic metabolism via non-enzymatic hydrolysis and cytochrome P450 pathways; both enantiomers undergo rapid racemization in vivo, complicating stereoselective disposition. Less than 5% of the dose is excreted unchanged in urine, with the remainder transformed into metabolites that are primarily renally eliminated. Lenalidomide demonstrates rapid oral absorption independent of dose, reaching maximum concentrations in approximately 1 hour, though absolute bioavailability remains undetermined due to lack of intravenous data. The terminal elimination half-life is 3-4 hours in patients with normal renal function, with approximately 82% of the administered dose excreted unchanged in urine via glomerular filtration and active tubular secretion, underscoring dominant renal clearance. In individuals with creatinine clearance below 60 mL/min, half-life extends to 9-16 hours, requiring dosage adjustments to mitigate accumulation. Minimal hepatic metabolism occurs, primarily via CYP-mediated oxidation to inactive hydroxy metabolites. Pomalidomide exhibits rapid absorption post-oral dosing, with bioavailability of at least 73% and time to peak concentration around 2-3 hours. Its elimination half-life ranges from 6.5-8 hours in healthy subjects, extending slightly in multiple myeloma patients to about 7.5 hours, driven by hepatic metabolism through , , and to hydroxylated and hydrolyzed products. Approximately 73% of the dose appears in urine and 15% in feces, with only 2-8% as unchanged parent compound, indicating extensive biotransformation prior to elimination. Next-generation cereblon E3 ligase modulators (CELMoDs), such as iberdomide, show enhanced oral pharmacokinetics with dose-proportional increases in Cmax and AUC following single doses from 0.1-1 mg, supporting improved systemic exposure over earlier IMiDs. Absorption is rapid, with half-lives typically in the 6-10 hour range, though specific elimination pathways involve hepatic metabolism modulated by physicochemistry; distribution varies, with lipophilicity influencing blood-brain barrier penetration—more polar CELMoDs exhibit limited central nervous system entry, while optimized analogs achieve therapeutic brain concentrations in preclinical models. Renal and fecal elimination contribute, but data remain preliminary from phase 1 studies emphasizing tolerability over detailed mass balance.

Drug interactions and resistance factors

Pomalidomide, a cereblon E3 ligase modulator, is primarily metabolized via (major pathway) and (minor pathway), rendering it susceptible to interactions with modulators. Strong inhibitors, such as fluvoxamine, should be avoided due to potential increases in pomalidomide exposure and toxicity risk. inducers like rifampin can reduce systemic exposure to immunomodulatory imide drugs () by 20-50%, necessitating dose adjustments or alternative therapies to maintain efficacy. In contrast, exhibits minimal cytochrome P450 involvement, with primary renal elimination, resulting in fewer enzymatic interactions but potential additive risks with other renally cleared agents. Resistance to cereblon modulators in relapsed multiple myeloma often arises from genetic alterations in the CRBN-CRL4 complex, including deletions or mutations in CRBN and CUL4 genes observed in approximately 20% of refractory cases. These changes disrupt the molecular glue activity of IMiDs, which relies on CRBN binding to recruit substrates like IKZF1/3 for ubiquitination and degradation, thereby empirically bypassing therapeutic proteolysis without fully abolishing ligase function. Point mutations, such as the YW/AA variant in CRBN's thalidomide-binding pocket, confer resistance by impairing IMiD docking, though next-generation cereblon E3 ligase modulators (CELMoDs) demonstrate partial efficacy against select deleterious mutations via enhanced affinity or altered binding modes. Pharmacodynamic factors influencing response include baseline IKZF1 and IKZF3 expression levels, where higher pre-dose IKZF abundance correlates with greater degradation efficiency and improved clinical outcomes in IMiD-treated patients, serving as a predictive biomarker for sensitivity. Low IKZF levels prior to treatment may signal intrinsic resistance, independent of CRBN integrity, highlighting the need for IKZF profiling in therapeutic decision-making.

Synthesis and Chemical Production

Routes for thalidomide and early IMiDs

The synthesis of thalidomide typically commences with the condensation of phthalic anhydride and L-glutamic acid to yield N-phthaloyl-DL-glutamic acid, followed by cyclization to form the glutarimide ring, often employing acetic anhydride or urea as dehydrating agents. This multi-step process achieves overall yields of approximately 70%, though it is susceptible to impurities arising from side reactions during anhydride formation and cyclization. Thalidomide possesses a chiral center at the 3-position of the glutarimide ring, and while starting from enantiopure L-glutamic acid, the molecule undergoes rapid racemization in vivo due to enolization facilitated by the adjacent carbonyl groups, complicating stereospecific attributions of activity. Early production of thalidomide encountered challenges with batch-to-batch variability, including residual solvents and incomplete cyclization products, which heightened scrutiny on manufacturing consistency amid emerging toxicity reports. Lenalidomide, an early derivative, is synthesized via alkylation of or its precursors with derivatives of pipecolic acid to construct the piperidine-2,6-dione moiety, followed by nitro group reduction if employed in intermediate steps. Optimized routes prioritize enantiopure (S)-pipecolic acid to maintain chiral integrity at the piperidine C-3 position, yielding the active enantiomer with purities exceeding 99% and overall process efficiencies around 60%. Unlike thalidomide, lenalidomide exhibits slower epimerization rates due to the cyclic structure stabilizing the chiral center, enabling better control over stereochemistry in pharmaceutical formulations. Pomalidomide follows a analogous path to thalidomide, substituting 3-nitrophthalic anhydride for phthalic anhydride in the glutamic acid condensation, with subsequent nitro reduction to the 4-amino substituent post-cyclization, addressing stereochemical instability through refined purification to minimize racemic impurities. These early IMiD syntheses underscored the need for rigorous chiral resolution and impurity profiling to mitigate empirical risks observed in initial scalability efforts.

Modern synthesis of CELMoDs and PROTACs

The synthesis of modern cereblon E3 ligase modulators (CELMoDs), such as iberdomide (CC-220), emphasizes kilogram-scale processes for single-enantiomer intermediates, enabling progression to clinical trials. Key routes involve the preparation of chiral glutarimide-phthalimide scaffolds through optimized coupling of advanced intermediates, including boronic acid derivatives (e.g., Iberdomide·BSA) for subsequent aryl attachments, with focus on stereocontrol and impurity management. These convergent strategies reduce overall steps compared to early IMiDs, incorporating palladium-catalyzed couplings like for modular assembly of piperidine-containing side chains on the phthalimide ring, achieving high efficiency in late-stage transformations suitable for good manufacturing practice (GMP) conditions. For analogous CELMoDs like CC-90009, process development features mild transformations such as phthalide chlorination to benzoate esters, followed by nitrile and amide coupling, yielding purified product for early clinical supply with enhanced purity and reduced waste. Such optimizations highlight efficiency gains, including >80% yields in optimized steps, scalable to multi-kilogram batches as a foundation for commercial routes. PROTAC synthesis targeting leverages , particularly copper-catalyzed azide-alkyne (CuAAC), to conjugate variable linkers to warheads like , enabling rapid iteration of hundreds of analogues for structure-activity relationship studies. This approach streamlines linker diversification, from PEG-based flexible chains to rigid or motifs, minimizing synthetic iterations for potency tuning. Scalability advancements in the include solid-phase methods, where ligands are resin-bound for sequential attachment of linkers and target protein binders, reducing solution-phase steps by up to 50% and supporting parallel library production per recent protocols and patents.

Current Research and Challenges

Clinical trials of novel modulators

In phase 1/2 trials of mezigdomide (CC-92480), a potent CELMoD, combined with dexamethasone in heavily pretreated relapsed/ patients, overall response rates (ORR) reached 43.1% at the 160 mg dose, with updated data from 2024 showing durable responses and median (PFS) exceeding 12 months in select cohorts. Combinations with bortezomib-dexamethasone or carfilzomib-dexamethasone in the CC-92480-MM-002 trial (NCT03989414) yielded ORR above 40% and PFS over one year as of mid-2025 updates, particularly in patients to prior immunomodulatory drugs. These results highlight mezigdomide's activity in high-risk populations, though and infections remain key adverse events. Iberdomide (CC-220), another CELMoD with immunostimulatory properties, advanced to phase 3 evaluation in the EXCALIBER-RRMM trial (NCT04975997), where it plus daratumumab-dexamethasone significantly improved minimal residual disease negativity rates compared to standard therapies in relapsed/refractory multiple myeloma, with topline efficacy data released in September 2025. Ongoing phase 3 studies, including frontline combinations initiated in 2025, target ORR and PFS endpoints in newly diagnosed patients, building on phase 1/2 data showing ORR of 30-50% in refractory settings. Safety profiles align with class effects, emphasizing thrombocytopenia management. Cereblon-recruiting PROTACs have entered early-to-late-stage trials outside myeloma, focusing on solid tumors. ARV-471 (vepdegestrant), an oral degrader, achieved a 40% clinical benefit rate (complete/partial response or stable disease ≥24 weeks) in a phase 2 expansion cohort for ER+/HER2- advanced , with >90% target degradation observed in tumor biopsies. The phase 3 VERITAC-2 trial reported a 2.9-month PFS improvement versus in May 2025, confirming >50% engagement across doses. Other CRBN-PROTACs, such as those targeting or , remain in phase 1/2 for and hematologic cancers, with degradation efficiencies informing dose escalation. These trials underscore PROTACs' potential for overcoming resistance via deep , though challenges persist.

Mechanisms of resistance and optimization strategies

Resistance to E3 ligase modulators, such as and , often arises from genomic alterations in the CRBN gene itself, disrupting the binding pocket required for modulator-induced substrate recruitment. Missense mutations in CRBN, identified in patients refractory to these agents, have been modeled in cellular assays to assess functional impact; for instance, certain mutations (e.g., H397Y) confer resistance to by impairing of neo-substrates like /3 while preserving baseline CRBN ligase activity. These changes, including point mutations clustered in the thalidomide-binding domain, appear in up to 20.7% of lenalidomide-refractory cases and are associated with inferior outcomes to subsequent immunomodulatory drugs. In vitro selection of resistant cell lines frequently yields CRBN mutations alongside alterations in the CRL4^CRBN complex, underscoring a direct causal role in evading drug-dependent ubiquitination. Epigenetic and transcriptional adaptations further contribute to resistance by compensating for the loss of key transcription factors targeted by these modulators. Overexpression of , a downstream effector normally downregulated via /3 degradation, enables bypass of the apoptotic effects in cells; this occurs through mechanisms like BATF heterodimerization, which sustains activity despite IKZF depletion. Such adaptations highlight how resistant clones maintain oncogenic signaling independent of CRBN-modulator axis disruption, as evidenced in IMiD-refractory models where levels correlate with sustained viability. Optimization strategies focus on designing next-generation agents that circumvent these resistance pathways via enhanced binding kinetics or alternative recruitment modes. modulators (CELMoDs), such as iberdomide, exhibit higher CRBN affinity and allosteric modulation, retaining efficacy against IMiD-resistant mutants that abolish traditional molecular glue activity; structural studies confirm CELMoDs engage distinct conformational states, evading pocket mutations. Dual recruitment approaches, explored in monovalent degraders, switch specificity (e.g., from DCAF16 to FBXO22) with minimal structural tweaks, enabling degradation in CRBN-mutated contexts by leveraging redundant networks. These tactics, validated in preclinical resistant lines, prioritize empirical potency over single-ligase dependency to restore therapeutic windows.

Potential expansions beyond oncology

Preclinical investigations have explored cereblon-based PROTACs for degrading immune signaling proteins in autoimmune conditions, such as IRAK4 degraders that reduce production in models of relevant to diseases like . However, these approaches face limitations from potential cytokine storms induced by off-target degradation of neosubstrates, as observed with CRBN modulators altering transcription factors like /3, which can dysregulate immune responses. STAT3-targeted PROTACs, often leveraging CRBN recruitment, have demonstrated selective degradation in cellular assays, offering theoretical benefits for STAT3-driven , but clinical translation remains hindered by toxicity profiles and incomplete pathway suppression. In neurodegeneration, efforts to develop CRBN-dependent tau PROTACs aim to clear pathogenic aggregates in tauopathies like , with in vitro validation showing ternary complex formation and proteasomal . Progress has stalled due to poor blood- barrier , as most PROTACs exhibit limited CNS exposure owing to their large molecular weight and , alongside risks of unintended within the degradome. Additional challenges include optimizing linker for bioavailability without compromising E3 ligase engagement. Broader empirical data indicate that while over 100 PROTAC , including non-oncologic ones, have shown efficacy via CRBN , clinical advancement lags at under 10% success rate, predominantly attributable to pharmacokinetic barriers like poor oral , rapid clearance, and tissue distribution issues. These hurdles underscore the need for refined molecular properties to enable non-oncology applications, tempered by inherent toxicities from ubiquitin-proteasome pathway perturbations.