MedPath

LM-108 Advanced Drug Monograph

Published:Sep 12, 2025

Generic Name

LM-108

Comprehensive Analysis of the Investigational Anti-CCR8 Monoclonal Antibody: Cafelkibart (LM-108)

Executive Summary

Cafelkibart (LM-108) is an investigational, first-in-class, Fc-optimized humanized monoclonal antibody targeting the C-C motif chemokine receptor 8 (CCR8). Developed by LaNova Medicines Ltd. and now under the strategic stewardship of Sino Biopharmaceutical Ltd., LM-108 represents a novel immuno-oncology strategy designed to address a critical unmet need: resistance to immune checkpoint inhibitors (ICIs).[1] The core therapeutic thesis of LM-108 is predicated on its ability to selectively deplete tumor-infiltrating regulatory T cells (Tregs), a key immunosuppressive cell population within the tumor microenvironment (TME), thereby reactivating a potent anti-tumor immune response.[4]

Clinical development has yielded compelling efficacy signals in heavily pre-treated patient populations, particularly in cancers where ICIs have shown limited utility or where resistance has developed. In a pooled analysis of Phase 1/2 studies in patients with advanced gastric cancer, the majority of whom had failed prior anti-PD-1 therapy, LM-108 in combination with an anti-PD-1 antibody demonstrated an Objective Response Rate (ORR) of 36.1%.[7] This efficacy was profoundly enriched in a biomarker-selected subgroup of patients with high CCR8 expression who had progressed on first-line therapy, where an unprecedented ORR of 87.5% was observed.[7] Similarly, in advanced pancreatic cancer, the combination therapy achieved an ORR of 20.3% and a median Overall Survival (OS) of 10.02 months, with response rates rising to 33.3% in patients with high CCR8 expression.[4]

The safety profile of LM-108, characterized by common immune-related adverse events such as elevated transaminases, rash, and cytopenias, has been described by investigators as manageable and consistent with its potent immunomodulatory mechanism.[4] This acceptable therapeutic window appears to be a direct result of the drug's high target specificity, which spares peripheral Tregs and minimizes systemic autoimmunity. Strategically, LM-108 is positioned as a leading agent in the CCR8-targeting class, with a clear development path focused on overcoming ICI resistance in biomarker-defined patient populations across a range of solid tumors.

Scientific Rationale and Mechanism of Action

The Immunosuppressive Role of Tumor-Infiltrating Regulatory T Cells (Tregs)

The efficacy of modern cancer immunotherapies, particularly those targeting the PD-1/PD-L1 axis, is fundamentally dependent on the host's ability to mount an effective anti-tumor T-cell response. However, a significant proportion of patients either do not respond to these therapies (primary resistance) or develop resistance after an initial response (acquired resistance). A primary driver of this resistance is the establishment of a profoundly immunosuppressive tumor microenvironment (TME). Central to this immunosuppressive network are regulatory T cells (Tregs), a specialized subset of CD4+ T cells characterized by the expression of the transcription factor Foxp3.[5]

Tumors actively recruit and expand Treg populations, which then function to suppress the activity of effector immune cells, most notably cytotoxic CD8+ T cells, through various mechanisms including the secretion of inhibitory cytokines and direct cell-cell contact.[9] The accumulation of these Tregs within the tumor creates a formidable barrier to effective anti-tumor immunity, thereby limiting the clinical benefit of ICIs.[4] Seminal preclinical work has established that the ratio of effector T cells (Teffs) to Tregs within the TME is a critical determinant of immune response and a strong correlate of ICI efficacy.[5] A high Teff-to-Treg ratio is associated with a "hot," immune-inflamed TME and favorable outcomes, whereas a low ratio signifies an immunosuppressed or "cold" TME and poor response to therapy. This understanding forms the basis of a compelling therapeutic hypothesis: selectively depleting Tregs within the tumor, thereby shifting the Teff-to-Treg ratio in favor of the effector response, could overcome a major mechanism of ICI resistance and unlock their therapeutic potential in a broader patient population.

C-C Motif Chemokine Receptor 8 (CCR8) as a Precision Target

The primary challenge in targeting Tregs for therapeutic benefit is achieving selectivity. Systemic, non-specific depletion of all Tregs would likely lead to severe, life-threatening autoimmune toxicities, as these cells are crucial for maintaining immune homeostasis in peripheral tissues. The ideal target would be a cell surface protein that is uniquely and highly expressed on the immunosuppressive Tregs within the TME, but largely absent from beneficial peripheral Tregs and other essential immune cells.

C-C motif chemokine receptor 8 (CCR8), a G protein-coupled receptor (GPCR) belonging to the chemokine receptor subfamily, has emerged as such a target.[8] Extensive analysis of single-cell sequencing data from various human cancers, including breast, colorectal, and lung cancer, has revealed a distinct expression pattern for CCR8. Its expression is specifically and highly upregulated on tumor-infiltrating Tregs, where it is co-expressed with the lineage marker Foxp3.[5] Conversely, CCR8 expression is low or absent in peripheral blood cells, normal tissues, and, critically, on peripheral Tregs.[8] This differential expression provides a unique therapeutic window, allowing for the precise targeting of the most pathogenic Treg population within the tumor while sparing the essential homeostatic Tregs circulating in the periphery. Furthermore, high expression of CCR8 in the TME has been correlated with poor patient prognosis, validating it as a clinically relevant and high-value therapeutic target.[11]

Cafelkibart (LM-108): Molecular Design and Depletive Mechanism

Cafelkibart (LM-108) is a novel, humanized monoclonal antibody of the immunoglobulin G1 (IgG1) subclass, engineered to specifically bind to human CCR8.[5] Patent filings reveal that LM-108 is a humanized version of the murine antibody 137D1H10, with specific complementarity-determining regions (CDRs) that confer its high-affinity binding to the CCR8 protein.[12]

A critical feature of LM-108's design is its "Fc-optimized" IgG1 backbone.[5] The selection of the IgG1 isotype is deliberate, as it is the most potent human isotype for engaging Fc gamma receptors (

FcγR) on the surface of effector immune cells, such as Natural Killer (NK) cells and macrophages. The Fc region of LM-108 has been further engineered to enhance this engagement, thereby maximizing its ability to mediate two powerful cytotoxic mechanisms: Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC) and Antibody-Dependent Cellular Phagocytosis (ADCP).[5] This design choice reflects a specific therapeutic strategy: it is not sufficient to simply block the signaling of the CCR8 receptor by its natural ligand, CCL1; the physical elimination of the immunosuppressive Treg cell itself is required to achieve a robust and durable reversal of TME-mediated immunosuppression.

The mechanism of action proceeds as follows: upon intravenous administration, LM-108 circulates and preferentially binds to CCR8-expressing cells within the TME. This binding achieves two primary effects. First, it blocks the CCL1-CCR8 signaling axis, which is involved in Treg migration and function.[11] Second, and more importantly, the Fc portion of the antibody acts as a beacon, "tagging" the CCR8-positive Treg for destruction. NK cells recognize the Fc region via their

FcγRs and release cytotoxic granules, inducing ADCC. Similarly, macrophages bind to the Fc region and engulf the targeted Treg via ADCP.[5] The net result is the selective and efficient depletion of tumor-infiltrating Tregs, removing a key source of local immunosuppression and thereby unleashing the latent cytotoxic potential of tumor-specific CD8+ T cells to attack and destroy cancer cells.[11]

Preclinical Evidence and Proof-of-Concept

The advancement of LM-108 into clinical trials was supported by a robust body of preclinical data that validated its mechanism of action and demonstrated its anti-tumor potential, both as a monotherapy and in combination with ICIs.

In Vitro Characterization

Initial laboratory studies focused on confirming the binding characteristics and functional activity of LM-108. Flow cytometry assays demonstrated that LM-108 binds with high affinity and specificity to human CCR8 in a dose-dependent manner.[5] This was confirmed using various cell lines engineered to express different levels of CCR8. For instance, in cell-based binding assays, LM-108 bound to human CCR8-overexpressing HEK293 cells with a half-maximal effective concentration (

EC50​) of 1.03 nM, and to endogenously expressing U2OS and Jurkat cells with EC50​ values of 0.25 nM and 0.21 nM, respectively. Crucially, no binding was observed in non-CCR8-expressing control cells, underscoring its high target specificity.[12]

Functional assays subsequently confirmed the potency of its Fc-optimized design. LM-108 was shown to induce powerful ADCC and ADCP effects against CCR8-expressing target cells when co-cultured with human effector cells.[5] Patent data quantifies this potency, showing that LM-108 mediated ADCC against CCR8-expressing cells with an

EC50​ as low as 0.002 nM when using primary human peripheral blood mononuclear cells (PBMCs) as effectors.[12] These in vitro results provided the foundational evidence that LM-108 could effectively bind its target and trigger the intended depletive mechanisms.

In Vivo Efficacy in Murine Models

The therapeutic hypothesis was then tested in vivo using syngeneic mouse tumor models, which allow for the study of a fully competent immune system. Using a murine surrogate antibody in models such as the CT26 colon carcinoma and MC38 colorectal adenocarcinoma, treatment with the anti-CCR8 antibody as a monotherapy resulted in significant inhibition of tumor growth.[5] Analysis of the TME in these treated animals confirmed the on-target biological effect: there was a marked and effective depletion of tumor-infiltrating Tregs, which was accompanied by a corresponding and therapeutically favorable increase in the CD8+ T cell to Treg ratio within the tumors.[5]

The most critical preclinical validation came from combination studies. In multiple mouse models, including those designed to be resistant to anti-PD-1 therapy, the combination of the anti-CCR8 antibody with a PD-1 inhibitor demonstrated potent synergistic efficacy, leading to significantly greater tumor growth inhibition than either agent alone.[5] This provided the definitive proof-of-concept for the primary clinical development strategy of using LM-108 to overcome ICI resistance.

Mechanistic Insights from Preclinical Studies

Deeper investigation into the TME of treated animals revealed a more nuanced mechanism underlying the observed synergy. The therapeutic depletion of CCR8+ Tregs was found to remodel the TME by disrupting a key immunosuppressive interaction between these Tregs and a population of C-C motif chemokine ligand 5 (CCL5)-positive dendritic cells (DCs).[9] Dendritic cells are the most potent antigen-presenting cells in the body, responsible for priming naive T cells and initiating adaptive immune responses.

By removing the suppressive influence of the Tregs, the combination of anti-CCR8 and anti-PD-1 therapy led to a dramatic increase in the secretion of interleukin-12 (IL-12) by these DCs, an effect mediated through the Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway.[9] IL-12 is a powerful pro-inflammatory cytokine that is critical for promoting the cytotoxic activity of CD8+ T cells and NK cells. This finding suggests that the efficacy of LM-108 is not merely a passive effect of "releasing the brakes" by removing Tregs. It also actively "steps on the gas" by enhancing the antigen-presenting and T-cell-priming function of dendritic cells. This dual action—removing suppression while simultaneously boosting activation—provides a strong molecular rationale for its potent effects, particularly in immunologically "cold" or ICI-resistant tumors that may lack a pre-existing robust immune infiltrate.

Clinical Development Program Overview

Global Clinical Strategy

The clinical development of LM-108 has been pursued with a rapid and global strategy. The program achieved key Investigational New Drug (IND) application approvals from major regulatory bodies in quick succession: Australia's Therapeutic Goods Administration (TGA) in November 2021, the U.S. Food and Drug Administration (FDA) in January 2022, and China's National Medical Products Administration (NMPA) in May 2022.[8] Notably, the NMPA approval marked LM-108 as the first CCR8-targeting antibody to enter the clinical stage in China, establishing an early lead in a key pharmaceutical market.[8]

The overarching clinical program consists of multiple Phase 1/2, open-label, multi-center studies designed to evaluate the safety, tolerability, pharmacokinetics, and preliminary efficacy of LM-108.[17] While monotherapy activity is being assessed, the primary strategic focus is on combination therapies, pairing LM-108 with established anti-PD-1 antibodies such as pembrolizumab, toripalimab, and sintilimab, and in some cases, with standard-of-care chemotherapy.[4] The program is currently in Phase 2, its highest stage of development, with plans announced for the initiation of pivotal Phase 3 registrational trials in both China and the United States during the second half of 2024.[9]

Targeted Indications and Trial Designs

The clinical strategy employs an initial broad evaluation followed by focused expansion in indications with strong biological rationale and high unmet need.

  • Advanced Solid Tumors (Basket Trials): The program was initiated with basket trials, such as NCT05255484 and NCT05518045, which enrolled patients with a variety of recurrent or refractory advanced solid tumors. These studies served to establish the initial safety profile, determine the recommended Phase 2 dose (RP2D), and identify early signals of efficacy across different cancer types.[17]
  • Pancreatic Cancer: Following promising early signals, a dedicated focus was placed on pancreatic cancer. Efficacy and safety have been evaluated in a pooled analysis of two Phase 1/2 trials (NCT05199753, NCT05518045), which enrolled patients with advanced disease who had progressed on at least one prior line of systemic therapy. The intervention in these trials is LM-108 combined with either pembrolizumab or toripalimab.[4]
  • Gastric Cancer: This indication has emerged as a cornerstone of the LM-108 program. A pooled analysis of three Phase 1/2 studies (NCT05199753, NCT05255484, NCT05518045) has evaluated LM-108 plus an anti-PD-1 antibody (pembrolizumab or toripalimab). A key feature of this cohort is the high proportion of patients (nearly 90%) who had previously been treated with and failed anti-PD-1 therapy, positioning the trial to directly test the drug's ability to overcome ICI resistance.[7]
  • Non-Small Cell Lung Cancer (NSCLC): The ACCLAIM study (NCT06479759) is a multi-cohort trial specifically designed for patients with locally advanced or metastatic NSCLC. It is evaluating LM-108 in combination with the anti-PD-1 antibody sintilimab, both with and without the addition of chemotherapy, in treatment-naïve and previously treated patient populations.[19]
  • Triple-Negative Breast Cancer (TNBC): A Phase 2, single-center trial (NCT06387628) is investigating LM-108 in a three-drug combination with toripalimab and a chemotherapeutic agent (either nab-paclitaxel or eribulin). The study is enrolling patients with recurrent or metastatic TNBC in both first-line and later-line settings.[20]

The breadth of this program, summarized in the table below, reflects a deliberate strategy to establish LM-108 as a broadly applicable therapy for overcoming immunotherapy resistance across multiple major solid tumor types.

Trial Identifier (NCT Number)PhaseIndication(s)Intervention(s)Patient PopulationStatus
NCT051997531/2Gastric Cancer, Pancreatic Cancer, Solid TumorsLM-108 + Pembrolizumab/ToripalimabAdvanced, RefractoryActive
NCT052554841/2Gastric Cancer, Solid TumorsLM-108 (monotherapy or + Pembrolizumab)Advanced, RefractoryActive
NCT055180451/2Gastric Cancer, Pancreatic Cancer, Solid TumorsLM-108 (monotherapy or + Toripalimab)Advanced, RefractoryActive
NCT06479759 (ACCLAIM)Not specifiedNon-Small Cell Lung Cancer (NSCLC)LM-108 + Sintilimab ± ChemotherapyLocally Advanced or MetastaticRecruiting
NCT063876282Triple-Negative Breast Cancer (TNBC)LM-108 + Toripalimab + ChemotherapyRecurrent or MetastaticActive
NCT068254941/2Solid TumorsLM-108 ± Penpulimab + ChemotherapyAdvancedRecruiting

Table 1: Summary of Active Clinical Trials for Cafelkibart (LM-108). Data compiled from.[4]

Clinical Efficacy in Advanced Solid Tumors

The clinical data reported to date from the LM-108 program, primarily from pooled analyses in pancreatic and gastric cancer, have provided strong validation for its therapeutic hypothesis, demonstrating encouraging anti-tumor activity in patient populations with high unmet needs and limited treatment options.

Pancreatic Cancer: A High Unmet Need Setting

Pancreatic cancer remains one of the most lethal malignancies, with notoriously poor responses to conventional chemotherapy and very limited success for immunotherapies to date. The data for LM-108 in this setting comes from a pooled analysis of two Phase 1/2 studies (NCT05199753, NCT05518045) that included 80 patients from China and Australia with advanced pancreatic cancer who had progressed on prior systemic therapy.[4] The patient population was characteristic of this difficult-to-treat disease, with 65.0% presenting with liver metastases and 22.5% having already failed prior anti-PD-1 therapy.[4]

The efficacy results, with a data cutoff of December 2, 2024, are summarized below.

Efficacy EndpointOverall Population (n=74)Patients with 1 Prior Line (n=45)Patients with High CCR8 Expression (n=9)
Objective Response Rate (ORR)20.3% (95% CI 11.8-31.2%)24.4% (95% CI 12.9-39.5%)33.3% (95% CI 7.5-70.1%)
Disease Control Rate (DCR)62.2% (95% CI 50.1-73.2%)71.1% (95% CI 55.7-83.6%)77.8% (95% CI 40.0-97.2%)
Median Duration of Response (DoR)5.49 months (95% CI 3.02-8.87)6.93 months (95% CI 3.02-NA)Not Reported
Median Progression-Free Survival (PFS)3.12 months (95% CI 1.61-4.86)4.86 months (95% CI 2.79-6.90)6.90 months (95% CI 1.22-NA)
Median Overall Survival (OS)10.02 months (95% CI 6.41-13.11)Not Reached9.15 months (95% CI 3.61-NA)

Table 2: Efficacy of Cafelkibart (LM-108) plus Anti-PD-1 Therapy in Pancreatic Cancer (Pooled Analysis). Data from.[4]

In the overall efficacy-evaluable cohort of 74 patients, the combination therapy achieved an ORR of 20.3% and a median OS of 10.02 months.[4] These results are encouraging in a historically chemo-refractory disease. The data becomes more compelling when examining the subgroup of 45 patients who had progressed on only one prior line of therapy. In this less heavily pre-treated population, the ORR increased to 24.4%, the median PFS improved to 4.86 months, and the median OS was not yet reached at the time of analysis, with a promising 12-month OS rate of 51.6%.[4] The most significant finding, however, is the emergence of a potential predictive biomarker. In a small subset of 9 patients from this cohort who had high CCR8 expression, the ORR was 33.3%, and the median PFS was 6.90 months, suggesting that patient selection based on CCR8 expression could substantially enrich for response.[4]

Gastric Cancer: Overcoming Immunotherapy Resistance

The most striking clinical results for LM-108 have emerged from the gastric cancer cohort. This analysis pooled data from three Phase 1/2 studies (NCT05199753, NCT05255484, NCT05518045), including 48 patients with advanced gastric cancer.[7] The defining characteristic of this cohort was its profound resistance to immunotherapy: 89.6% of patients had received prior anti-PD-1 therapy and progressed.[7] This population represents one of the most significant challenges in modern oncology, and any meaningful activity in this setting is of high clinical importance.

The efficacy data, with a cutoff of December 25, 2023, demonstrated a remarkable ability to re-sensitize these tumors to immunotherapy.

Efficacy EndpointOverall Efficacy-Evaluable (n=36)Progressed on 1st-Line Tx (n=11)Progressed on 1st-Line Tx with High CCR8 (n=8)
Objective Response Rate (ORR)36.1% (95% CI 20.8-53.8%)63.6% (95% CI 30.8-89.1%)87.5% (1 CR, 6 PR)
Disease Control Rate (DCR)72.2% (95% CI 54.8-85.8%)81.8% (95% CI 48.2-97.7%)100% (1 CR, 6 PR, 1 SD)
Median Progression-Free Survival (PFS)6.53 months (95% CI 2.96-NA)Not ReportedNot Reported

Table 3: Efficacy of Cafelkibart (LM-108) plus Anti-PD-1 Therapy in Gastric Cancer (Pooled Analysis). CR = Complete Response; PR = Partial Response; SD = Stable Disease. Data from.[7]

Among the 36 efficacy-evaluable patients, the combination achieved an impressive ORR of 36.1% and a median PFS of 6.53 months.[7] The results in the key subgroup of 11 patients whose disease had progressed on first-line treatment—the direct target population for an ICI-resistance-breaking agent—were exceptional, with an ORR of 63.6%.[7]

The data from the biomarker-defined subgroup is even more profound. Among 8 patients from the first-line progressor group who had high tumor CCR8 expression, the ORR was 87.5%, including one complete response and six partial responses, and the DCR was 100%.[7] An ORR of this magnitude in a PD-1 refractory population is nearly unprecedented and strongly validates the central therapeutic hypothesis. It suggests that for a significant subset of patients who fail first-line immunotherapy, the accumulation of CCR8+ Tregs is not merely one of several resistance mechanisms, but is likely the dominant, targetable driver of resistance. The ability of LM-108 to effectively eliminate this population appears to fundamentally restore sensitivity to PD-1 blockade, representing a potential paradigm shift in the treatment of ICI-resistant gastrointestinal cancers.

Emerging Indications (NSCLC and TNBC)

Building on the strong biological rationale and compelling clinical signals in GI cancers, the LM-108 development program is expanding into other solid tumors where Treg-mediated immunosuppression is a known resistance mechanism. The initiation of trials in NSCLC (NCT06479759) and TNBC (NCT06387628) is supported by preclinical data indicating that CCR8 is specifically upregulated on tumor-infiltrating Tregs in lung and breast cancer models.[10] While clinical efficacy data from these trials are not yet available, their designs—combining LM-108 with both immunotherapy and chemotherapy—reflect a sophisticated strategy to tackle these complex diseases from multiple angles.

Safety and Tolerability Profile

A comprehensive assessment of the safety and tolerability of LM-108 is critical to understanding its therapeutic index. The data from the pooled analyses in pancreatic and gastric cancer provide a clear picture of the adverse event profile associated with potent, targeted immunomodulation.

Pooled Safety Data Analysis

In the pancreatic cancer cohort of 80 treated patients, treatment-related adverse events (TRAEs) of any grade were reported in 95.0% of patients, a high frequency indicative of a biologically active combination.[4] The most common TRAEs (occurring in ≥25% of patients) included increased aspartate aminotransferase (AST), increased alanine aminotransferase (ALT), anemia, rash, pyrexia, decreased platelet count, and increased conjugated bilirubin. Grade ≥3 TRAEs occurred in a majority of patients (52.5%). The most frequent of these severe events (≥5%) were lipase elevation (7.5%), increased ALT (6.3%), increased AST (5.0%), immune-mediated enterocolitis (5.0%), hypokalemia (5.0%), and rash (5.0%).[4]

In the gastric cancer cohort of 48 treated patients, the safety profile was broadly similar, though with a slightly lower incidence of severe events. All-grade TRAEs were reported in 81.3% of patients.[7] The most common events (≥15%) were increased ALT, increased AST, decreased white blood cell count, and anemia. Grade ≥3 TRAEs occurred in 37.5% of patients. The most common severe events (≥4%) were anemia (8.3%), increased lipase (4.2%), rash (4.2%), and decreased lymphocyte count (4.2%).[7]

Interpretation of Safety Profile

The constellation of observed adverse events is consistent with the known toxicities of anti-PD-1 therapy combined with a potent immunomodulatory agent. The high rates of hepatic toxicity (elevated transaminases), hematologic events (anemia, cytopenias), and dermatologic toxicity (rash) are expected consequences of widespread immune activation. The occurrence of specific immune-mediated events, such as enterocolitis, provides direct clinical evidence of the on-target immunological effects of the combination therapy.

Despite the high frequency of TRAEs, particularly the Grade ≥3 events in the pancreatic cancer cohort, the investigators in both analyses concluded that the safety profile was "manageable" and "well tolerated".[4] This suggests that the adverse events, while common, are generally predictable, monitorable, and reversible with standard clinical management protocols, such as treatment interruption, dose reduction, and the use of corticosteroids for immune-mediated toxicities.

The ability to manage these toxicities is the critical counterpart to the drug's high efficacy. This favorable therapeutic window—the capacity to induce a powerful anti-tumor immune response without triggering uncontrollable systemic autoimmunity—is arguably the most crucial element for the drug's ultimate clinical and commercial viability. This balance is a direct consequence of the molecular design and target selectivity of LM-108. By specifically targeting CCR8, which is preferentially expressed on Tregs within the tumor, the therapy is designed to focus its potent depletive effects locally within the TME while largely sparing the peripheral Treg population that is essential for maintaining systemic immune tolerance.[4] This precision is what allows for potent efficacy with a safety profile that, while significant, remains within the realm of clinical manageability.

Strategic and Regulatory Analysis

The trajectory of LM-108 from an early-stage asset to a leading candidate in its class has been shaped by strategic corporate development and a successful global regulatory strategy, positioning it for late-stage trials and potential market entry.

Corporate Development and Commercial Outlook

LM-108 was developed in-house by LaNova Medicines Ltd., a Shanghai-based biotechnology company founded in 2019 with a focus on creating innovative biologics against challenging targets like GPCRs.[8] The company rapidly advanced a pipeline of novel agents, with LM-108 emerging as a key clinical-stage asset.

In a significant strategic move reported in July 2025, Sino Biopharmaceutical Ltd. agreed to fully acquire LaNova Medicines.[3] This acquisition is a major validation of the potential of LaNova's pipeline and, in particular, of LM-108. By bringing LaNova under its corporate umbrella, Sino Biopharm provides the substantial financial resources, global development infrastructure, and commercial expertise necessary to execute large-scale Phase 3 trials and navigate the complex path to market approval and launch. This transition significantly de-risks the late-stage development of LM-108 and signals strong confidence from a major pharmaceutical player in its potential to become a best-in-class therapy.[1]

Regulatory Trajectory and Designations

The regulatory pathway for LM-108 has been characterized by rapid progress and the attainment of multiple special designations that are intended to expedite its development and review. The program's global nature was established early with IND approvals in Australia, the United States, and China within a seven-month period from late 2021 to mid-2022.[8]

More importantly, LM-108 has been granted several prestigious designations from major global health authorities based on its promising preliminary data:

  • FDA Fast Track Designation: In April 2023, the U.S. FDA granted Fast Track Designation to LM-108 for the treatment of NSCLC.[10] This designation is intended for drugs that treat serious conditions and fill an unmet medical need. It allows for more frequent communication with the FDA and makes the drug eligible for Accelerated Approval and Priority Review, potentially shortening the time to market.
  • FDA Orphan Drug Designation: In December 2023, the FDA granted Orphan Drug Designation to LM-108 for the treatment of pancreatic cancer.[9] This status provides significant development incentives, including tax credits for clinical trials, waiver of prescription drug user fees, and, most importantly, the potential for seven years of market exclusivity in the U.S. upon approval for that indication.
  • China Breakthrough Therapy Designation: LM-108 has also received Breakthrough Therapy designation from China's NMPA.[9] Similar to the FDA's designation, this is designed to expedite the development and review of drugs that have shown encouraging preliminary evidence of a substantial improvement over available therapies for serious diseases.

The combination of these multiple, high-value regulatory designations from both the FDA and NMPA is a powerful external validation of the clinical data. It signals that regulatory agencies in the world's two largest pharmaceutical markets recognize the potential of LM-108 to offer a significant therapeutic advance for patients with cancer and are actively facilitating its development. This collection of designations is not routine and suggests a smoother and more rapid path to approval than is typical for a new oncology agent.

Competitive Landscape and Future Directions

The therapeutic potential of targeting CCR8 to deplete tumor-infiltrating Tregs has been recognized by several pharmaceutical companies, creating a competitive but still nascent landscape. LM-108 is positioned as one of the top three most advanced projects worldwide targeting CCR8, giving it a significant first-mover advantage.[14] Key competitors in clinical development include Bristol-Myers Squibb (BMS-986340), Gilead/Jounce (JTX-1811), Shionogi (S-531011), Bayer (BAY3375968), and Roche (RO7502175).[8] Additionally, small molecule CCR8 antagonists, such as IPG7236 from Immunophage, are also in early-stage development.[25]

Looking ahead, the clinical development path for LM-108 will almost certainly be guided by a biomarker-driven strategy. The stark difference in response rates between the overall population and the CCR8-high subgroups in both the gastric and pancreatic cancer trials provides a clear and compelling rationale for using CCR8 expression as a patient selection criterion in future pivotal trials.[4] The development and validation of a robust companion diagnostic assay to accurately identify patients with "high CCR8 expression" will be a critical parallel activity to ensure the success of this strategy. The ultimate goal for the LM-108 program is to establish it as a new standard of care for overcoming ICI resistance in biomarker-defined patient populations across a wide range of solid tumors.

Conclusions

Cafelkibart (LM-108) has emerged as a highly promising, next-generation immuno-oncology agent with a well-defined mechanism of action and a clear strategic path. Its design as an Fc-optimized, depletive anti-CCR8 antibody is strongly supported by a scientific rationale targeting a key mechanism of immunotherapy resistance—the accumulation of tumor-infiltrating Tregs.

The preclinical and, more importantly, the clinical data generated to date provide compelling evidence of its potential. The unprecedented Objective Response Rate of 87.5% observed in patients with PD-1 refractory, CCR8-high gastric cancer is a landmark finding that validates the therapeutic hypothesis in a setting of profound unmet medical need. The encouraging activity in advanced pancreatic cancer further broadens its potential impact. These efficacy results, paired with a safety profile deemed manageable by clinical investigators, establish a favorable therapeutic index that is underpinned by the drug's high target selectivity.

The strategic acquisition by Sino Biopharm and the attainment of multiple prestigious regulatory designations from both the U.S. FDA and China's NMPA de-risk its late-stage development and signal strong external confidence in its clinical promise. The clear biomarker signal points toward a precision-medicine approach that could maximize clinical benefit in future trials.

In conclusion, cafelkibart (LM-108) is a leading clinical-stage asset that has demonstrated the potential to fundamentally alter the treatment landscape for patients with solid tumors that are resistant to current immune checkpoint inhibitors. Further investigation in pivotal, biomarker-driven trials is strongly supported by the existing data and is essential to confirm its role as a new cornerstone of cancer immunotherapy.

Works cited

  1. cafelkibart (LM-108) / Sino Biopharm - LARVOL DELTA, accessed September 12, 2025, https://delta.larvol.com/Products/?ProductId=21ee6016-0b68-4808-b3d8-8534f5f71b84
  2. LaNova Medicines Announces Global Exclusive License Agreement with AstraZeneca for LM-305, a Novel GPRC5D-Targeting Antibody Drug Conjugate - PR Newswire, accessed September 12, 2025, https://www.prnewswire.com/news-releases/lanova-medicines-announces-global-exclusive-license-agreement-with-astrazeneca-for-lm-305-a-novel-gprc5d-targeting-antibody-drug-conjugate-301823364.html
  3. LM-108 - Page 1 - BioWorld, accessed September 12, 2025, https://www.bioworld.com/keywords/55732-lm-108
  4. Efficacy and safety of cafelkibart (LM-108), an anti-CCR8 ..., accessed September 12, 2025, https://ascopubs.org/doi/10.1200/JCO.2025.43.16_suppl.4010
  5. Effective depletion of tumor-infiltrating Tregs by a novel anti-CCR8 ..., accessed September 12, 2025, https://aacrjournals.org/cancerres/article/82/12_Supplement/6008/704499/Abstract-6008-Effective-depletion-of-tumor
  6. Efficacy and safety of cafelkibart (LM-108), an anti-CCR8 monoclonal antibody, in combination with anti-PD-1 therapy in patients with pancreatic cancer: Results from phase 1/2 studies. - ASCO, accessed September 12, 2025, https://www.asco.org/abstracts-presentations/ABSTRACT494890
  7. Efficacy and safety of LM-108, an anti-CCR8 monoclonal antibody ..., accessed September 12, 2025, https://ascopubs.org/doi/10.1200/JCO.2024.42.16_suppl.2504
  8. LaNova Medicines Announces the IND Approval of CCR8-targeting monoclonal antibody LM-108 by China NMPA, accessed September 12, 2025, http://www.lanovamedicines.com/en/news-center/press-release/56.html
  9. LM-108 - Drug Targets, Indications, Patents - Patsnap Synapse, accessed September 12, 2025, https://synapse.patsnap.com/drug/57fa4aaacbbc4d43ba30addfff73095f
  10. LM-108 Receives Fast Track Designations for Cancer Treatments from US FDA, accessed September 12, 2025, https://www.lanovamedicines.com/en/news-center/press-release/64.html
  11. Definition of cafelkibart - NCI Drug Dictionary, accessed September 12, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/cafelkibart
  12. AU2021277712B2 - Anti-CCR8 monoclonal antibodies and uses ..., accessed September 12, 2025, https://patents.google.com/patent/AU2021277712B2/en
  13. Efficacy and safety of LM-108, an anti-CCR8 monoclonal antibody, in combination with, accessed September 12, 2025, https://www.cabrini.com.au/app/uploads/4-Gary-Richardson-LM-108.pdf
  14. LaNova Medicines Completes USD 42 Million Series C1 Financing to Accelerate Clinical Progress and Innovation Platform Development, accessed September 12, 2025, https://www.lanovamedicines.com/en/news-center/press-release/66.html
  15. Efficacy and safety of LM-108, an anti-CCR8 monoclonal antibody, in combination with an anti-PD-1 antibody in patients with gastric cancer, accessed September 12, 2025, https://ascopubs.org/doi/pdf/10.1200/JCO.2024.42.16_suppl.2504
  16. LM-108 Price - AbMole BioScience, accessed September 12, 2025, https://www.abmole.com/products/LM-108.html
  17. Study of LM-108 as a Single Agent or in Combination With Pembrolizumab in Subjects With Advanced Solid Tumours - National Cancer Institute, accessed September 12, 2025, https://www.cancer.gov/research/participate/clinical-trials-search/v?id=NCI-2022-10238
  18. Study of LM-108 as a Single Agent or in Combination With ..., accessed September 12, 2025, https://trial.medpath.com/clinical-trial/2083b5b2928a1269/nct05518045-clinical-study-lm-108-toripalimab-solid-tumours
  19. Study Details | LM-108 Antibody Combination With Sintilimab for Locally Advanced or Metastatic Non-Small Cell Lung Cancer | ClinicalTrials.gov, accessed September 12, 2025, https://clinicaltrials.gov/study/NCT06479759
  20. Study Details | NCT06387628 | LM-108 in Combination With PD-1 Based Treatment for Patients With Recurrent or Metastatic Triple - Negative Breast Cancer | ClinicalTrials.gov, accessed September 12, 2025, https://clinicaltrials.gov/study/NCT06387628
  21. LaNova Medicines Announces Initiation of Phase 1 Clinical Trial of Anti-PD-1/VEGF Bispecific Antibody LM-299 and Completion of $42 Million Series C1 Financing - PR Newswire, accessed September 12, 2025, https://www.prnewswire.com/news-releases/lanova-medicines-announces-initiation-of-phase-1-clinical-trial-of-anti-pd-1vegf-bispecific-antibody-lm-299-and-completion-of-42-million-series-c1-financing-302280157.html
  22. FDA Granted LM-108 Orphan Drug Designations for Cancer Treatments, accessed September 12, 2025, https://www.lanovamedicines.com/en/news-center/press-release/65.html
  23. Selective depletion of tumor-infiltrating regulatory T cells with BAY ..., accessed September 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11164760/
  24. Preclinical and translational pharmacology of afucosylated anti ..., accessed September 12, 2025, https://pubmed.ncbi.nlm.nih.gov/38486310/
  25. Discovery of a Potent and Selective CCR8 Small Molecular ..., accessed September 12, 2025, https://pubs.acs.org/doi/abs/10.1021/acs.jmedchem.3c00030

Published at: September 12, 2025

This report is continuously updated as new research emerges.

MedPath

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

© 2025 MedPath, Inc. All rights reserved.