Small Molecule
C26H34N8O
1628256-23-4
Lerociclib is an investigational, orally bioavailable, small molecule drug that functions as a potent and selective inhibitor of cyclin-dependent kinases 4 and 6 (CDK4/6). As a member of the third generation of CDK inhibitors, it represents a refined therapeutic agent designed to address the primary drivers of cell cycle progression in certain malignancies, most notably hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer. The mechanism of action is centered on the inhibition of the Cyclin D-CDK4/6-Retinoblastoma (Rb) protein pathway, a critical regulator of the G1-S phase transition in the cell cycle. By preventing the phosphorylation of Rb, lerociclib induces a durable G1 cell cycle arrest, thereby suppressing the proliferation of cancer cells dependent on this pathway.
The clinical development of lerociclib has been robust, culminating in two pivotal, randomized, double-blind, placebo-controlled Phase III trials—LEONARDA-1 and LEONARDA-2. These studies have successfully demonstrated statistically significant and clinically meaningful improvements in progression-free survival (PFS) in patients with HR+/HER2- advanced or metastatic breast cancer. LEONARDA-1 established its efficacy in combination with fulvestrant in an endocrine-resistant population, while LEONARDA-2 confirmed its benefit as a first-line therapy in combination with letrozole. The magnitude of benefit observed, with hazard ratios for disease progression or death consistently around 0.46, positions lerociclib's efficacy as highly competitive with established market leaders in the CDK4/6 inhibitor class.
A key distinguishing feature of lerociclib is its differentiated safety and tolerability profile. While retaining the class-defining efficacy, it has demonstrated a lower incidence of severe (Grade 3/4) neutropenia and gastrointestinal toxicity, particularly diarrhea, compared to other approved agents. This favorable profile enables a continuous, twice-daily oral dosing regimen without a mandatory treatment-free interval, or "drug holiday," which is required for some competitors to allow for bone marrow recovery. This continuous target inhibition, coupled with improved tolerability, forms the core of its clinical value proposition.
The development and commercialization trajectory of lerociclib has been characterized by a series of strategic global licensing agreements. Originally developed by G1 Therapeutics, the asset achieved its first global approval in May 2025 from China's National Medical Products Administration (NMPA) under the stewardship of its regional partner, Genor Biopharma. Following a period of corporate transition for its Western partner, the development and commercialization rights for the US, Europe, and other key markets are now held by Pepper Bio. Beyond its primary indication in breast cancer, preclinical evidence suggests promising future applications for lerociclib in other oncologic settings, such as pediatric sarcomas and in combination regimens to overcome resistance to targeted therapies in non-small cell lung cancer. This comprehensive monograph details the molecular, preclinical, clinical, and strategic landscape of lerociclib, positioning it as a significant and refined entrant into the therapeutic armamentarium for HR+ breast cancer and beyond.
Lerociclib is classified as a synthetic organic, small molecule drug, designed for oral administration.[1] Its chemical identity is well-defined through a comprehensive set of nomenclature and registry identifiers that facilitate its tracking across scientific literature, patent databases, and regulatory filings.
The drug is known by its generic name, Lerociclib, which is its United States Adopted Name (USAN).[4] Its International Nonproprietary Name (INN) is lerociclibum.[3] Throughout its development, it has been referred to by several codes, including G1T38, GB-491, and EQ132, reflecting its journey through different developing organizations.[1]
Its molecular formula is C26H34N8O, and its structure is characterized by a complex, multi-ring spirocyclic system.[4] This intricate architecture is precisely engineered to achieve high-affinity binding within the ATP-binding pocket of its target kinases. The systematic IUPAC name for this structure is 4-[[5-(4-propan-2-ylpiperazin-1-yl)pyridin-2-yl]amino]spiro[1,3,5,11-tetrazatricyclo[7.4.0.0$^{2,7}$]trideca-2,4,6,8-tetraene-13,1'-cyclohexane]-10-one.[3] The molecular structure is the fundamental determinant of its pharmacological activity. The core pyridine-amine-pyrimidine scaffold is a common motif among ATP-competitive kinase inhibitors, serving to anchor the molecule within the kinase "hinge" region through hydrogen bonding.[10] The specific arrangement of the propan-2-ylpiperazinyl and spiro[cyclohexane] moieties confers the high degree of potency and selectivity that characterizes lerociclib.
Physicochemical properties, which govern its absorption, distribution, and drug-like characteristics, have been computationally predicted. These properties are summarized in Table 1. The molecule adheres to Lipinski's Rule of Five, a widely used guideline to assess druglikeness and predict oral bioavailability, indicating a favorable profile for an oral therapeutic.[2]
Table 1: Identification and Physicochemical Properties of Lerociclib
Parameter | Value | Source(s) |
---|---|---|
Generic Name | Lerociclib | 2 |
Developmental Codes | G1T38, GB-491, EQ132 | 1 |
DrugBank ID | DB16218 | 2 |
CAS Number | 1628256-23-4 | 4 |
Molecular Formula | C26H34N8O | 4 |
Molecular Weight | 474.6 g/mol | 4 |
IUPAC Name | 4-[[5-(4-propan-2-ylpiperazin-1-yl)pyridin-2-yl]amino]spiro[1,3,5,11-tetrazatricyclo[7.4.0.0$^{2,7}$]trideca-2,4,6,8-tetraene-13,1'-cyclohexane]-10-one | 3 |
SMILES | CC(C)N1CCN(CC1)C2=CN=C(C=C2)NC3=NC=C4C=C5C(=O)NCC6(N5C4=N3)CCCCC6 | 4 |
InChIKey | YPJRHEKCFKOVRT-UHFFFAOYSA-N | 4 |
Water Solubility | 0.134 mg/mL (Predicted) | 2 |
logP | 3.49 (Predicted) | 2 |
pKa (Strongest Acidic) | 11.59 (Predicted) | 2 |
pKa (Strongest Basic) | 8.19 (Predicted) | 2 |
Rule of Five Compliance | Yes | 2 |
Lerociclib exerts its antineoplastic activity through the targeted inhibition of the cell cycle machinery.[4] It is an ATP-competitive inhibitor, meaning it binds to the ATP-binding pocket of Cyclin-Dependent Kinase 4 (CDK4) and Cyclin-Dependent Kinase 6 (CDK6), preventing these enzymes from carrying out their normal function.[5]
In normal and malignant cell physiology, the progression from the G1 (first gap) phase to the S (synthesis) phase of the cell cycle is a tightly regulated process. A key checkpoint is controlled by the complex formed between D-type cyclins and their kinase partners, CDK4 and CDK6. In response to mitogenic signals, this active complex phosphorylates the Retinoblastoma tumor suppressor protein (Rb).[2] Phosphorylation of Rb causes it to release the E2F family of transcription factors. Once liberated, E2F activates the transcription of a suite of genes essential for DNA replication and cell division, thereby committing the cell to enter the S phase and proliferate.[2]
In many cancers, particularly HR+ breast cancer, this pathway is frequently dysregulated, often through the overexpression of Cyclin D1, which is a direct transcriptional target of the estrogen receptor (ER).[10] This leads to constitutive CDK4/6 activity, uncontrolled Rb phosphorylation, and relentless cell proliferation.
Lerociclib directly intervenes in this pathological process. By selectively binding to and inhibiting the kinase activity of CDK4 and CDK6, it blocks the phosphorylation of Rb.[4] This action maintains Rb in its active, hypophosphorylated state, where it remains bound to E2F, effectively sequestering it and preventing the activation of downstream target genes.[2] The ultimate cellular consequence is a potent and sustained cell cycle arrest in the G1 phase.[4] This cytostatic effect suppresses tumor cell proliferation and forms the basis of its therapeutic efficacy.[4] A critical prerequisite for the activity of lerociclib and other drugs in its class is the presence of a functional, non-mutated Rb protein; tumors that have lost Rb function ("Rb-null") are inherently resistant to CDK4/6 inhibition.[5]
The pharmacological profile of lerociclib is defined by its high potency against its intended targets and its selectivity against other related kinases, which is a crucial determinant of its therapeutic window. Biochemical assays have quantified its inhibitory activity, revealing nanomolar potency. The half-maximal inhibitory concentration (IC50) of lerociclib has been measured at 1 nM for the CDK4/CyclinD1 complex and 2 nM for the CDK6/CyclinD3 complex.[5] These values establish it as one of the most potent CDK4/6 inhibitors developed.
While highly potent against CDK4 and CDK6, lerociclib was designed to be selective, minimizing activity against other kinases to reduce off-target effects. Its kinase selectivity profile has been characterized, showing significantly less activity against other members of the CDK family. The most closely related off-target with notable inhibition is CDK9, which is involved in transcriptional regulation. Lerociclib exhibits an IC50 of 28 nM against the CDK9/cyclin T complex.[9] This represents an approximately 30-fold selectivity window for CDK4 over CDK9, a feature that may contribute to its distinct tolerability profile. Inhibition of CDK9 is associated with different cellular effects and potential toxicities than CDK4/6 inhibition, and the relative sparing of this target is a key molecular attribute.
In cell-based assays, this biochemical potency translates to effective inhibition of cell proliferation. Lerociclib demonstrates a half-maximal effective concentration (EC50) of approximately 20-100 nM for inducing G1 arrest and inhibiting proliferation in Rb-competent cancer cell lines.[5] In stark contrast, its activity in Rb-null cell lines is negligible, with
EC50 values greater than 3 µM, confirming that its cellular effects are mediated through its intended on-target mechanism.[5]
The clinical development of lerociclib was underpinned by a robust preclinical data package that established its proof-of-concept as an antineoplastic agent. In vitro studies using a panel of cancer cell lines demonstrated its potent and selective activity. In multiple estrogen receptor-positive (ER+) breast cancer cell lines, including MCF7, BT474, and ZR-75-1, lerociclib effectively inhibited estrogen-driven proliferation. Its potency in these models was shown to be comparable to, and in some cases greater than, the selective estrogen receptor degrader (SERD) fulvestrant.[5] Critically, this growth-inhibitory effect was specific to the HR+ context, as no activity was observed in ER-negative cell lines such as MDA-MB-436, providing a clear rationale for its development in this specific breast cancer subtype.[5] The drug also showed broad antiproliferative activity against other tumor types, including melanoma, leukemia, and lymphoma cell lines, suggesting its potential utility beyond breast cancer.[6]
This promising in vitro activity was successfully translated into in vivo models. In xenograft studies using human breast cancer cells (e.g., MCF7) implanted in immunocompromised mice, daily oral administration of lerociclib led to significant, durable, and dose-dependent inhibition of tumor growth.[5] Similar potent antitumor effects were observed in a genetically engineered mouse model (GEMM) driven by the HER2/neu oncogene, further validating its efficacy in a more complex, immunologically intact tumor microenvironment.[5] These animal model studies were instrumental in confirming its oral bioavailability and establishing a clear relationship between drug exposure and antitumor response, providing the necessary evidence to proceed with clinical investigation.
From its early stages, the development of lerociclib was strategically focused on its potential as a combination therapy, a cornerstone of modern oncology. Preclinical studies systematically built the scientific rationale for combining lerociclib with other targeted agents to enhance efficacy and overcome resistance.
The most critical combination explored was with endocrine therapy for HR+ breast cancer. Studies pairing lerociclib with G1T48, a novel oral SERD, demonstrated synergistic activity. This combination was particularly effective in models of acquired endocrine resistance, including tamoxifen-resistant (TamR), long-term estrogen-deprived (LTED), and patient-derived xenograft (PDX) tumors.[5] These findings provided a strong preclinical mandate for the clinical strategy of combining lerociclib with agents that block the ER signaling pathway.
The utility of lerociclib was also investigated in the context of overcoming resistance to other targeted therapies. In models of non-small cell lung cancer (NSCLC) with activating EGFR mutations, resistance to EGFR tyrosine kinase inhibitors (TKIs) like osimertinib often arises through the activation of downstream or parallel "bypass" signaling pathways. Preclinical studies showed that adding lerociclib to osimertinib not only enhanced initial tumor killing but also significantly delayed the emergence of acquired resistance.[18] In xenograft models, this combination led to 100% complete responses and a 43% tumor cure rate, compared to 60% complete responses and 0% cures with osimertinib alone.[18] Mechanistic work suggested that lerociclib's inhibition of the cell cycle blocks the proliferative escape mediated by bypass pathways such as MET and AXL. Similar synergistic effects were observed when combining lerociclib with TKIs targeting ALK and RET fusions, highlighting a broad potential role in thwarting TKI resistance.[18]
Furthermore, in pancreatic ductal adenocarcinoma (PDAC), a cancer characterized by near-universal KRAS mutations and frequent dysregulation of the CDK4/6/Rb axis, lerociclib was tested in combination with inhibitors of KRAS effector pathways. Using a high-throughput ex vivo drug screening platform with 24 PDAC PDX models, lerociclib was found to significantly enhance the efficacy of inhibitors targeting PI3K (pictilisib) and ERK (ulixertinib).[19] This provided a strong rationale for investigating dual pathway inhibition in this notoriously difficult-to-treat malignancy. Collectively, these preclinical studies did not merely validate a single indication but strategically positioned lerociclib as a versatile and potent combination partner capable of addressing key clinical challenges like acquired resistance across multiple cancer types.
Lerociclib is formulated as an orally bioavailable small molecule, a key characteristic for convenient, long-term administration in the treatment of advanced cancer.[4] Clinical trial protocols for its pivotal studies specified that lerociclib should be administered with food, suggesting that food may enhance its absorption or, more commonly, reduce inter-patient variability in exposure, a strategy employed for other drugs in this class.[12]
Pharmacokinetic (PK) assessments were integral endpoints in the early-phase clinical trials designed to establish the drug's safety and optimal dosing regimen.[21] Data from the Phase 1b/2a dose-finding study (NCT02983071) demonstrated a dose-proportional increase in drug exposure (Area Under the Curve, AUC) with increasing doses, indicating predictable PK behavior within the therapeutic range investigated.[24] A key pharmacological attribute that distinguishes lerociclib is its relatively short plasma half-life compared to first-to-market CDK4/6 inhibitors. This property is foundational to its ability to be administered on a continuous daily schedule without accumulating to toxic levels, thereby avoiding the need for a treatment-free interval.[25]
To formally characterize the complete ADME profile of the drug in humans, a radiolabeled version, Carbon C-14 lerociclib ([14C]GB491), was synthesized.[28] The use of such a tracer in a human ADME study is the gold standard for definitively determining the routes and rates of metabolism and excretion, and for identifying and quantifying all circulating metabolites. While the development of this tool indicates that a formal ADME study was planned or conducted, the detailed results from such a study are not publicly available in the provided documentation.
Although a formal human ADME study report is not available, a reliable assessment of lerociclib's drug-drug interaction (DDI) potential can be inferred from the stringent exclusion criteria outlined in its clinical trial protocols. Multiple studies, including NCT05085002 and the planned NCT05712941, explicitly prohibited the concomitant use of known strong or moderate inducers or strong inhibitors of the Cytochrome P450 3A (CYP3A) enzyme subfamily.[29] This is a standard safety measure for investigational drugs whose metabolic clearance is predominantly dependent on the CYP3A4 isoenzyme. Co-administration with a strong CYP3A4 inhibitor (e.g., ketoconazole) would be expected to significantly increase lerociclib plasma concentrations, raising the risk of toxicity. Conversely, co-administration with a strong CYP3A4 inducer (e.g., rifampin) would likely decrease lerociclib exposure, potentially compromising its efficacy.
Furthermore, these trial protocols also excluded patients who were taking other medications that have a narrow therapeutic window and are themselves primarily metabolized by CYP3A4/5.[29] This suggests that lerociclib is likely a clinically relevant inhibitor of CYP3A4. By inhibiting this enzyme, lerociclib could increase the plasma concentrations of co-administered CYP3A4 substrates, which can be dangerous for drugs where a small increase in exposure can lead to significant toxicity. This metabolic and DDI profile is highly consistent with that of the approved CDK4/6 inhibitors palbociclib, ribociclib, and abemaciclib, all of which are substrates and inhibitors of CYP3A4, necessitating careful management of concomitant medications in clinical practice.[12] Therefore, despite the absence of a dedicated publication, the available evidence strongly indicates that CYP3A4-mediated interactions are a critical consideration for the safe and effective use of lerociclib.
The clinical efficacy of lerociclib in its primary indication of HR+, HER2- advanced breast cancer (ABC) has been systematically evaluated through a phased development program, culminating in two positive, large-scale Phase III trials.
The foundational clinical study for lerociclib was a Phase 1b/2a trial (NCT02983071) designed to establish the safety, tolerability, pharmacokinetics, and preliminary efficacy of lerociclib in combination with the SERD fulvestrant in patients with ER+, HER2- ABC who had progressed on prior therapies.[23] This study was crucial for identifying the optimal dose and schedule for subsequent pivotal trials.
The trial explored both once-daily (QD) and twice-daily (BID) continuous dosing regimens. The results clearly favored the BID schedule, which demonstrated an improved safety and tolerability profile, most notably with lower rates of gastrointestinal adverse events compared to QD dosing.[22] Based on the overall balance of safety and activity, the dose of 150 mg administered orally twice daily was selected as the recommended dose for further development.[22]
At this dose, the combination of lerociclib and fulvestrant showed compelling antitumor activity. Among 19 evaluable patients in the 150 mg BID cohort, the confirmed partial response (PR) rate was 32%, and 47% of patients achieved stable disease.[23] The clinical benefit rate (CBR), defined as the proportion of patients with a complete response, partial response, or stable disease lasting at least 24 weeks, was an impressive 74%.[23] The median progression-free survival (PFS) across all dose levels in the study was 15 months.[33] These efficacy data were considered highly consistent with the results observed in pivotal trials of other approved CDK4/6 inhibitors combined with fulvestrant, providing strong justification for advancing lerociclib into Phase III development.[23]
The efficacy of lerociclib was definitively established through the successful execution of the LEONARDA program, which comprised two large, randomized, placebo-controlled Phase III trials targeting the two primary settings for CDK4/6 inhibitor use in HR+ ABC.
The LEONARDA-1 study was designed to confirm the benefit of lerociclib in patients with HR+/HER2- locally advanced or metastatic breast cancer whose disease had relapsed or progressed on a prior endocrine therapy.[21] A total of 275 patients were randomized on a 1:1 basis to receive either lerociclib (150 mg BID continuously) plus fulvestrant or a matching placebo plus fulvestrant.[15]
The study successfully met its primary endpoint of investigator-assessed PFS. The addition of lerociclib to fulvestrant more than doubled the median PFS, from 5.49 months in the placebo arm to 11.07 months in the lerociclib arm. This represented a 54% reduction in the risk of disease progression or death, which was both statistically significant and clinically meaningful (Hazard Ratio: 0.458; 95% Confidence Interval [CI]: 0.317-0.661; p<0.001).[21] The benefit was further corroborated by a Blinded Independent Central Review (BICR), which reported an even more pronounced effect (HR: 0.353; 95% CI: 0.228-0.547; p<0.00001).[20]
Secondary endpoints also favored the lerociclib arm. The Objective Response Rate (ORR) was significantly higher with the lerociclib combination compared to fulvestrant alone (26.9% vs. 9.9%).[21] At the time of the primary analysis, overall survival (OS) data were immature and did not show a statistically significant difference.[20]
The LEONARDA-2 study evaluated lerociclib in the first-line setting for patients with HR+/HER2- advanced or metastatic breast cancer who had not received any prior systemic therapy for their advanced disease.[38] In this trial, 279 patients were randomized 1:1 to receive lerociclib (150 mg BID) plus the aromatase inhibitor letrozole, or placebo plus letrozole.[38]
This study also met its primary endpoint of investigator-assessed PFS at a planned interim analysis. The median PFS in the placebo plus letrozole arm was 16.56 months, while the median PFS in the lerociclib plus letrozole arm had not yet been reached, indicating a durable and prolonged benefit. The addition of lerociclib resulted in a 54% reduction in the risk of progression or death (HR: 0.464; 95% CI: 0.293-0.733; p=0.0004).[38] For patients with measurable disease at baseline, the ORR was numerically higher in the lerociclib group (62.3%) compared to the placebo group (48.5%).[38]
The successful outcomes of both LEONARDA trials validate lerociclib's efficacy across the continuum of care for HR+ advanced breast cancer, mirroring the development pathways of its competitors and establishing it as a potent therapeutic agent in both first-line and endocrine-resistant settings.
A critical aspect of the clinical data for lerociclib is the consistency of its treatment benefit across various clinically relevant patient subgroups. In both the LEONARDA-1 and LEONARDA-2 trials, subgroup analyses demonstrated that the PFS advantage conferred by lerociclib was maintained regardless of baseline characteristics.[37] This consistent benefit was observed in patients with visceral metastases (including liver metastases), those with a high burden of disease (multiple metastatic sites), patients with primary endocrine resistance (a historically poor-prognosis group), and across different menopausal statuses (pre/peri- and postmenopausal).[20] This broad and consistent efficacy underscores the robustness of lerociclib's clinical activity and supports its use in a wide range of patients with HR+ advanced breast cancer.
Table 2: Summary of Major Clinical Trials for Lerociclib
Trial ID (NCT) | Phase | Indication | Intervention Arms | Key Endpoints | Status | Sponsor(s) | Source(s) |
---|---|---|---|---|---|---|---|
NCT02983071 | 1b/2a | HR+/HER2- Advanced Breast Cancer | Lerociclib + Fulvestrant (Dose Escalation/Expansion) | Safety, Tolerability, PK, Preliminary Efficacy (ORR, CBR, PFS) | Active, not recruiting | G1 Therapeutics, Inc. | 23 |
NCT03455829 | 1/2 | EGFR-Mutant NSCLC | Lerociclib + Osimertinib | Safety, Tolerability, Preliminary Efficacy | Completed | G1 Therapeutics, Inc. | 32 |
LEONARDA-1 (NCT05054751) | 3 | HR+/HER2- Advanced Breast Cancer (Endocrine-Resistant) | Lerociclib + Fulvestrant vs. Placebo + Fulvestrant | Primary: PFS. Secondary: OS, ORR, Safety | Completed / Results Reported | Genor Biopharma | 21 |
LEONARDA-2 (NCT05851014) | 3 | HR+/HER2- Advanced Breast Cancer (First-Line) | Lerociclib + Letrozole vs. Placebo + Letrozole | Primary: PFS. Secondary: OS, ORR, Safety | Completed / Results Reported | Genor Biopharma | 38 |
NCT05085002 | 2 | HR+/HER2- Advanced Breast Cancer (1L & 2L) | Lerociclib + Endocrine Therapy (Letrozole or Fulvestrant) | Safety, Efficacy | Terminated | EQRx, Inc. | 29 |
NCT05712941 | 3 | Advanced/Recurrent Endometrial Cancer | Lerociclib + Letrozole vs. Placebo + Letrozole | PFS, Safety | Withdrawn | EQRx International, Inc. | 8 |
Table 3: Key Efficacy Results from the Phase III LEONARDA-1 and LEONARDA-2 Trials
Endpoint | Trial | Lerociclib Arm | Control Arm | Hazard Ratio (95% CI) / Rate | p-value | Source(s) |
---|---|---|---|---|---|---|
Median PFS (Investigator) | LEONARDA-1 | 11.07 months | 5.49 months | 0.458 (0.317, 0.661) | <0.001 | 21 |
Median PFS (Investigator) | LEONARDA-2 | Not Reached | 16.56 months | 0.464 (0.293, 0.733) | 0.0004 | 38 |
Median PFS (BICR) | LEONARDA-1 | Not Reported | Not Reported | 0.353 (0.228, 0.547) | 0.000002 | 20 |
Objective Response Rate (ORR) | LEONARDA-1 | 26.9% | 9.9% | N/A (Rate Difference) | Not Reported | 21 |
Objective Response Rate (ORR) | LEONARDA-2 | 62.3% | 48.5% | N/A (Rate Difference) | Not Reported | 38 |
The safety profile of lerociclib has been extensively characterized in its clinical development program, revealing a pattern of adverse events (AEs) that is consistent with the CDK4/6 inhibitor class but with notable differentiations in the frequency and severity of key toxicities. In the pivotal LEONARDA-1 trial, any-grade AEs were reported in 98.5% of patients receiving lerociclib plus fulvestrant, compared to 80.4% in the placebo plus fulvestrant arm.[20] Grade 3 or higher AEs occurred in 57.7% of patients in the lerociclib arm versus 15.2% in the control arm.[20]
The most frequently reported AEs were hematological in nature. In the LEONARDA-1 study, the most common AEs of any grade in the lerociclib arm were neutropenia (90.5%), leukopenia (86.9%), anemia (34.3%), and thrombocytopenia (19.7%).[21] Diarrhea was the most common non-hematologic AE, occurring in 19.7% of patients.[21]
Serious adverse events (SAEs) were reported in a minority of patients, with rates of 5.8% to 13.1% in the lerociclib arms across the LEONARDA studies, which were generally comparable to or only slightly higher than the rates in the placebo arms (7.7% to 8.0%).[21] A crucial indicator of the overall manageability of a drug's side effect profile is the rate of treatment discontinuation due to AEs. For lerociclib, this rate was exceptionally low, at approximately 0.7% in both large Phase III trials.[21] This suggests that while AEs are common, they are typically manageable with supportive care and dose modifications, allowing the vast majority of patients to remain on therapy.
The value of lerociclib is most apparent when its safety profile is analyzed in the context of other CDK4/6 inhibitors. It appears to strike a favorable balance, mitigating the most problematic toxicities associated with its competitors.
Hematologic Toxicity: Neutropenia is the hallmark dose-limiting toxicity for the CDK4/6 inhibitor class, resulting from the inhibition of CDK6 in hematopoietic progenitor cells. Lerociclib is no exception, with neutropenia being its most common AE. However, the severity appears to be attenuated compared to first-generation agents. In the LEONARDA-1 and -2 trials, the rate of Grade 3/4 neutropenia was consistently 46.7%, with Grade 4 neutropenia occurring in only 5.1% of patients.[21] These rates, particularly for Grade 4 neutropenia, are lower than those reported in the pivotal trials for palbociclib and ribociclib, which often exceed 20%.[8] This more manageable myelosuppression is a key feature that underpins the rationale for lerociclib's continuous dosing schedule, as it does not necessitate a one-week break for bone marrow recovery.[23] Febrile neutropenia, a serious complication of severe neutropenia, was reported in only one patient (5.0%) in the 150 mg BID cohort of the Phase 2 study.[22]
Gastrointestinal Toxicity: Diarrhea is the primary dose-limiting toxicity of abemaciclib, the other continuously dosed CDK4/6 inhibitor, with Grade 3 events occurring in up to 15% of patients in pivotal trials.[8] Lerociclib demonstrates a markedly different and more favorable gastrointestinal profile. While any-grade diarrhea was reported, severe (Grade 3 or higher) diarrhea was exceptionally rare. Across the two large Phase III studies involving over 270 patients on lerociclib, only a single case of Grade 3 diarrhea (0.7%) was reported, with no cases reported in the LEONARDA-1 trial.[21] This low rate of severe diarrhea is a major differentiating safety feature.
Other Toxicities: Lerociclib appears to have a benign profile with respect to other known class-associated toxicities. In the LEONARDA-2 study, the incidence of QTc prolongation was comparable to that of the placebo arm, and no cases of venous thromboembolism were reported.[38] Early clinical trials also noted low rates of other bothersome AEs such as stomatitis and alopecia.[15] This overall safety profile—characterized by manageable neutropenia, minimal severe diarrhea, and a low incidence of other significant toxicities—supports the conclusion that lerociclib offers a differentiated and favorable benefit-risk balance.
Formal contraindications for lerociclib are pending review and approval by Western regulatory agencies like the FDA and EMA. However, the exclusion criteria from its pivotal clinical trials provide a robust framework for identifying patient populations in whom the drug should be used with caution or avoided.
Key exclusion criteria across the clinical program included patients with clinically significant, uncontrolled heart disease or a history of cardiac repolarization abnormalities such as prolonged QT syndrome or Torsades de Pointes.[29] Patients with active or symptomatic central nervous system (CNS) metastases, severe impairment of lung function (e.g., interstitial pneumonia), or an ejection fraction of ≤45% were also excluded.[29] In most trials, patients with prior exposure to any CDK4/6 inhibitor were not eligible, although this is an eligibility criterion rather than a contraindication.[22]
As detailed in the pharmacokinetics section, the most critical contraindication relates to drug-drug interactions. The co-administration of strong or moderate CYP3A inducers or strong inhibitors of CYP3A was strictly forbidden in clinical trials and will almost certainly be a contraindication or carry a strong warning in the final product label.[29] This requires careful review of a patient's concomitant medications before initiating therapy.
The developmental and corporate history of lerociclib is a compelling case study in modern, partnership-driven pharmaceutical strategy. The molecule was originally discovered and advanced through early clinical development by G1 Therapeutics, a U.S.-based biopharmaceutical company co-founded by Dr. Norman Sharpless, the former Director of the National Cancer Institute.[43]
Recognizing the significant capital required for late-stage global development and commercialization, G1 Therapeutics pursued a strategy of regional licensing to maximize the asset's value. In June 2020, G1 entered into a pivotal agreement with Genor Biopharma, granting them the exclusive rights to develop and commercialize lerociclib in the Asia-Pacific region (excluding Japan).[43] This partnership proved highly successful, as Genor Biopharma efficiently conducted the pivotal LEONARDA-1 and LEONARDA-2 trials, ultimately securing the drug's first global regulatory approval in China.
Concurrently, in July 2020, G1 licensed the rights for the United States, Europe, Japan, and all other ex-Asia-Pacific markets to EQRx, a company founded with a mission to develop innovative medicines at lower prices.[43] EQRx initiated further clinical studies, including a Phase 2 trial in advanced breast cancer (NCT05085002).[32] However, the trajectory of lerociclib in the West was altered by corporate events. In August 2023, EQRx was acquired by Revolution Medicines. Following the acquisition, Revolution Medicines announced a strategic decision to focus on its internal RAS-inhibitor pipeline and to wind down the legacy EQRx programs, including the development of lerociclib.[45]
This development prompted G1 Therapeutics to re-secure the ex-Asia-Pacific rights and establish new partnerships. In May 2024, G1 announced two new licensing agreements. The rights for all oncology indications were licensed to Pepper Bio, a transomics drug discovery company tasked with advancing lerociclib through further clinical trials and regulatory submissions in the U.S. and Europe.[25] A separate, more niche agreement was made with Deimos Biosciences for the development of lerociclib for radioprotective uses, though this license was subsequently terminated in June 2025.[7] This complex history of licensing and re-licensing highlights a capital-efficient strategy that has allowed the asset to progress and achieve validation in a major market, thereby de-risking it for its new partners in the West.
As of the latest available information, the regulatory status of lerociclib varies significantly by region.
Lerociclib is entering a mature and competitive market dominated by three well-entrenched CDK4/6 inhibitors: palbociclib (Ibrance®), ribociclib (Kisqali®), and abemaciclib (Verzenio®). Its clinical and commercial success will depend on its ability to differentiate itself from these established agents. A comparative analysis across key attributes is presented in Table 4.
The efficacy of lerociclib, as measured by the PFS hazard ratios in the LEONARDA trials, is highly competitive and numerically similar to the pivotal trial results of the approved drugs, confirming its potent, class-level antitumor activity.[10] The most significant differentiation lies in its unique combination of a continuous dosing schedule and a favorable safety profile. Unlike palbociclib and ribociclib, it does not require a one-week treatment holiday, theoretically providing sustained target inhibition. Unlike abemaciclib, it is associated with a very low rate of severe diarrhea, a major quality-of-life concern and dose-limiting toxicity for that agent. This "best-of-both-worlds" profile—combining the convenience and sustained pressure of continuous dosing with a more manageable safety burden—represents its core competitive advantage.
Table 4: Comparative Profile of Lerociclib versus Approved CDK4/6 Inhibitors
Feature | Lerociclib | Palbociclib | Ribociclib | Abemaciclib |
---|---|---|---|---|
Primary Targets | CDK4, CDK6 | CDK4, CDK6 | CDK4, CDK6 | CDK4, CDK6 |
Key Off-Targets | CDK9 (moderate) | Highly selective | Highly selective | CDK1, CDK2, CDK9, GSK3β |
Recommended Dose | 150 mg BID | 125 mg QD | 600 mg QD | 150 mg BID |
Schedule | Continuous | 3 weeks on, 1 week off | 3 weeks on, 1 week off | Continuous |
PFS HR (1L vs. AI) | ~0.46 (LEONARDA-2) | ~0.58 (PALOMA-2) | ~0.57 (MONALEESA-2) | ~0.55 (MONARCH 3) |
PFS HR (2L vs. Fulvestrant) | ~0.46 (LEONARDA-1) | ~0.46 (PALOMA-3) | ~0.57 (MONALEESA-3) | ~0.55 (MONARCH 2) |
Grade 3/4 Neutropenia | ~47% | ~66% | ~60% | ~27% |
Grade 3/4 Diarrhea | <1% | <1% | <2% | ~15% |
Other Notable AEs | Manageable hematologic | Myelosuppression | QTc prolongation, LFT elevation | Diarrhea, VTE, Fatigue |
CNS Penetration | Not established | Poor | Poor | Yes |
FDA/EMA Status | Investigational | Approved | Approved | Approved |
First Approval | China (May 2025) | USA (Feb 2015) | USA (Mar 2017) | USA (Sep 2017) |
(Note: Efficacy and safety data are derived from respective pivotal trials and are not from head-to-head comparisons. Sources: 8) |
While the primary focus of lerociclib's development has been HR+ breast cancer, a body of preclinical evidence and early clinical exploration points to significant potential in other areas.
Pediatric Sarcomas: One of the most compelling areas for future investigation is in pediatric sarcomas. These are heterogeneous and often aggressive tumors for which new therapeutic options are urgently needed. Preclinical studies have demonstrated that the CDK4/6 pathway is often dysregulated in sarcomas. Lerociclib treatment of sarcoma cell lines in vitro and in advanced 3D bioprinted microtumor models resulted in decreased cell viability, reduced proliferation, G1 cell cycle arrest, and a significant reduction in cellular motility and stemness (tumor sphere formation).[1] These robust preclinical findings provide a strong scientific rationale for initiating clinical trials to evaluate lerociclib as a novel therapeutic strategy for this patient population.
Endometrial Cancer: The rationale for using CDK4/6 inhibitors extends to other hormone-driven cancers. Endometrial cancer, particularly low-grade endometrioid subtypes, often expresses hormone receptors and relies on pathways that converge on cell cycle activation. Recognizing this, EQRx had planned a Phase 3 trial (NCT05712941) to evaluate lerociclib in combination with letrozole for patients with advanced or recurrent, low-grade endometrial cancer.[8] Although this trial was withdrawn following the corporate acquisition of EQRx, the initial plan signifies a high level of interest and a strong biological rationale for exploring lerociclib in this indication.
Radioprotection: A novel and non-oncologic application for lerociclib was identified in the field of radioprotection. The transient cell cycle arrest induced by CDK4/6 inhibitors can protect normal, healthy cells—particularly hematopoietic stem and progenitor cells—from the damaging effects of cytotoxic therapies. This concept is clinically validated by trilaciclib (Cosela®), another G1 Therapeutics asset, which is approved for myelopreservation in patients receiving chemotherapy. Lerociclib was being explored for a similar protective effect against DNA damage induced by radiation therapy.[3] A licensing agreement was established with Deimos Biosciences specifically for this indication, and while it was later terminated, it highlights a scientifically plausible and potentially valuable application for lerociclib beyond direct cancer treatment.[7]
Lerociclib has emerged as a potent and highly effective third-generation CDK4/6 inhibitor, with a clinical profile that is both competitive with and differentiated from the established leaders in its class. The successful completion of the LEONARDA-1 and LEONARDA-2 Phase III trials has unequivocally demonstrated its ability to significantly prolong progression-free survival for patients with HR+/HER2- advanced breast cancer, both in the first-line and endocrine-resistant settings. The magnitude of clinical benefit is on par with that of palbociclib, ribociclib, and abemaciclib, firmly establishing its class-level efficacy.
The primary and most compelling attribute of lerociclib is its differentiated safety and tolerability profile. It has been engineered to strike an optimal balance between potent target inhibition and patient tolerability. The clinical data consistently show a lower incidence of severe (Grade 4) neutropenia compared to palbociclib and ribociclib, and a near-absence of the severe diarrhea that characterizes abemaciclib. This unique safety signature enables a continuous, twice-daily dosing regimen without a mandatory treatment holiday. This regimen offers the theoretical advantage of sustained pressure on the cell cycle, potentially preventing the tumor cell cycle re-entry that could occur during the off-treatment week of intermittent schedules, while simultaneously improving patient quality of life by minimizing severe gastrointestinal toxicity. This "best-of-both-worlds" profile represents its core value proposition and a clear rationale for its adoption in clinical practice.
The journey of lerociclib from discovery to market is a testament to a modern, capital-efficient, partnership-driven development model. Its first global approval by the NMPA in China not only provides a new therapeutic option for patients in that region but also serves as a critical regulatory validation of the asset, significantly de-risking its path forward in Western markets. Despite a complex history of licensing agreements and corporate strategy shifts, the strength of the underlying clinical data has ensured the drug's continued development.
Looking ahead, the future of lerociclib appears promising. Beyond its established role in breast cancer, compelling preclinical data support its investigation in new indications with high unmet need, such as pediatric sarcomas. Under the stewardship of its new development partners, the next critical steps will be to successfully navigate the regulatory pathways with the FDA and EMA and to strategically design trials that further explore its potential in other malignancies. In conclusion, lerociclib is not merely a "me-too" compound but a refined and optimized CDK4/6 inhibitor that offers a distinct and favorable balance of efficacy and tolerability, positioning it to become a valuable addition to the oncology armamentarium.
Published at: September 9, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.