Quemliclustat, also known by its development code AB-680, is an investigational, first-in-class, small-molecule therapeutic agent designed as a potent, selective, and reversible inhibitor of the ectoenzyme CD73 (cluster of differentiation 73).[1] Developed by Arcus Biosciences in collaboration with Gilead Sciences and Taiho Pharmaceutical, Quemliclustat represents a strategic intervention in the field of immuno-oncology, targeting the immunosuppressive adenosine axis within the tumor microenvironment (TME).[4] The fundamental mechanism of action involves the direct inhibition of CD73-mediated conversion of adenosine monophosphate (AMP) to adenosine, a nucleoside that has emerged as a critical mediator of immune evasion in a wide range of malignancies.[1] By blocking this final and rate-limiting step in extracellular adenosine production, Quemliclustat is designed to reverse adenosine-mediated lymphocyte suppression, thereby restoring and enhancing the anti-tumor activity of cytotoxic T-lymphocytes, natural killer (NK) cells, and other key immune effectors.[1]
The clinical development program for Quemliclustat is most advanced in metastatic pancreatic ductal adenocarcinoma (mPDAC), a disease with a historically poor prognosis and a significant unmet medical need.[9] The Phase 1b ARC-8 trial (NCT04104672) evaluated Quemliclustat in combination with standard-of-care chemotherapy (gemcitabine and nab-paclitaxel) in treatment-naive mPDAC patients. The study yielded promising results, with a pooled analysis of all patients receiving a 100 mg Quemliclustat-based regimen (n=122) demonstrating a median overall survival (OS) of 15.7 months, a figure that substantially exceeds historical benchmarks of approximately 9 months for chemotherapy alone.[11] Notably, the cohort receiving Quemliclustat plus chemotherapy without an anti-PD-1 antibody showed a median OS of 19.4 months.[11] These encouraging findings, achieved without significant added toxicity, supported the U.S. Food and Drug Administration (FDA) granting Orphan Drug Designation to Quemliclustat for pancreatic cancer in July 2025.[4]
Based on the strength of the ARC-8 data, Arcus Biosciences has initiated the global, registrational Phase 3 PRISM-1 trial (NCT06608927).[5] This pivotal study is designed to definitively assess the efficacy and safety of Quemliclustat plus gemcitabine/nab-paclitaxel versus placebo plus the same chemotherapy backbone in approximately 610 patients with treatment-naive mPDAC.[14] The successful outcome of this trial could establish Quemliclustat as a new standard of care, representing a significant therapeutic advancement for this challenging disease. This report provides a comprehensive analysis of Quemliclustat, detailing its molecular characteristics, the scientific rationale for its development, a thorough review of its preclinical and clinical data, and its current regulatory and commercial landscape.
A precise understanding of a drug's chemical identity is foundational to its pharmacological evaluation. Quemliclustat is a well-characterized small molecule with a distinct set of identifiers and physicochemical properties that define its structure and classification.
Quemliclustat is identified across scientific literature, clinical trial registries, and chemical databases by a variety of names and codes. Establishing these identifiers is crucial for accurate data aggregation and cross-referencing.
The chemical structure of Quemliclustat dictates its biological activity, solubility, and metabolic profile.
Quemliclustat is categorized based on its structural features, therapeutic use, and regulatory status.
Table 1: Key Identifiers and Physicochemical Properties of Quemliclustat
Property | Value | Source(s) |
---|---|---|
Generic Name (INN) | Quemliclustat | |
Development Code | AB-680 | |
IUPAC Name | amino]pyrazolo[3,4-b]pyridin-1-yl]-3,4-dihydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]methylphosphonic acid | |
CAS Registry Number | 2105904-82-1 | |
Molecular Formula | C20H24ClFN4O9P2 | |
Molecular Weight | 580.8 g/mol | |
InChIKey | MFYLCAMJNGIULC-KCVUFLITSA-N | |
SMILES String | CNC2=CC(=NC3=C2C=NN3[C@H]4O)Cl |
The concept of the tumor microenvironment (TME) has revolutionized the understanding of cancer biology, shifting the focus from a purely cancer cell-centric view to a complex ecosystem where malignant cells interact with a diverse array of non-malignant cells, including immune cells, fibroblasts, and endothelial cells. A key hallmark of cancer is its ability to evade destruction by the host immune system. Tumors achieve this by co-opting various physiological pathways to create a profoundly immunosuppressive TME. One of the most potent and pervasive of these immunosuppressive mechanisms is the generation of extracellular adenosine. High expression of CD73, the principal enzyme responsible for adenosine production, has been correlated with significantly poorer prognosis in multiple tumor types, including pancreatic cancer, establishing it as a high-value therapeutic target.
The generation of immunosuppressive adenosine within the TME is a well-defined, two-step enzymatic cascade that degrades extracellular adenosine triphosphate (ATP). Stressed or dying cells, a common occurrence in the rapidly turning-over TME, release large quantities of ATP. While high concentrations of extracellular ATP can act as a "danger signal" to alert and activate the immune system, this pro-inflammatory potential is subverted by tumor cells and associated stromal cells.
The first step in this pathway is the conversion of ATP to adenosine monophosphate (AMP) by the ectonucleotidase CD39. The second, and critically important, step is the dephosphorylation of AMP to adenosine, catalyzed by the ecto-5'-nucleotidase, CD73. CD73 is the primary and rate-limiting enzymatic producer of extracellular adenosine in the TME, making it a strategic point of intervention. By targeting this final, committed step in the pathway, a therapeutic agent can effectively shut down the production of the key immunosuppressive molecule at its source. This approach offers a distinct advantage over targeting other nodes in the pathway. For instance, inhibiting the upstream enzyme CD39 could have more complex effects on ATP metabolism, while targeting downstream adenosine receptors would need to account for multiple receptor subtypes with varied expression and function. The direct inhibition of CD73 with a potent molecule like Quemliclustat provides a mechanistically clean and direct method to abrogate this immunosuppressive signaling cascade.
Once produced by CD73, extracellular adenosine exerts its powerful immunosuppressive effects by binding to specific G-protein coupled receptors, primarily the A2A receptor (A2AR), expressed on the surface of various immune cells. This signaling cascade results in a broad dampening of anti-tumor immunity through several coordinated mechanisms:
The collective result of these actions is the creation of an "adenosinergic halo" around the tumor, a localized zone of profound immunosuppression that protects cancer cells from immune-mediated destruction and promotes tumor progression. The therapeutic rationale for Quemliclustat is therefore clear: by inhibiting CD73, it aims to dismantle this halo, reduce local adenosine concentrations, and thereby restore the functionality of the anti-tumor immune response.
The preclinical evaluation of Quemliclustat provided a robust foundation of evidence supporting its mechanism of action and therapeutic potential, justifying its advancement into human clinical trials. These studies characterized its molecular interactions, its effects on immune cells in vitro, and its anti-tumor activity in vivo.
Quemliclustat is a highly potent, selective, and reversible small-molecule competitive inhibitor of the human CD73 enzyme. Its exceptional potency is quantified by a very low inhibition constant (
Ki) of 5 pmol/L, indicating an extremely high affinity for its target. A critical feature of its design is its ability to effectively inhibit both the membrane-bound and soluble forms of CD73, ensuring a comprehensive blockade of all adenosine-generating activity of the enzyme within the TME.
Beyond simple competitive inhibition of the enzyme's active site, Quemliclustat exhibits a more complex and potentially more durable mechanism. Upon administration, the binding of Quemliclustat to CD73 on the cell surface leads to the clustering and subsequent internalization of the enzyme. This dual action is significant. While a standard competitive inhibitor can be overcome by high concentrations of the substrate (AMP), the physical removal of the CD73 enzyme from the cell surface provides a more profound and lasting blockade. This internalization effectively reduces the total amount of functional enzyme available to produce adenosine, suggesting that the biological effect may persist even as local drug concentrations fluctuate. This robust mechanism could contribute to the drug's high potency and the feasibility of less frequent dosing schedules. Furthermore, this process of internalization has been associated with a secondary benefit: a decrease in the migration of cancer cells, which could potentially help prevent metastasis.
A series of preclinical experiments provided strong proof-of-concept for Quemliclustat's immunomodulatory and antineoplastic activities.
The pharmacokinetic properties of a drug—its absorption, distribution, metabolism, and excretion—are critical determinants of its dosing, efficacy, and safety. Preclinical studies in various animal species revealed that Quemliclustat possesses a favorable pharmacokinetic profile characterized by low plasma clearance and a long half-life. These characteristics make it well-suited for long-acting parenteral (intravenous) administration, allowing for sustained target engagement with less frequent dosing. Initial pharmacokinetic data from Phase 1 studies in humans confirmed these findings, showing that Quemliclustat is well-tolerated and exhibits a long half-life compatible with the intravenous dosing schedules employed in later-phase clinical trials.
The clinical development of Quemliclustat has progressed rapidly, driven by its strong preclinical rationale and encouraging early-phase data. The program has investigated the drug across a portfolio of solid tumors known for high CD73 expression and an immunosuppressive TME. The highest phase of development reached is Phase 3 for adenocarcinoma, specifically metastatic pancreatic ductal adenocarcinoma (mPDAC), which has become the lead indication.
Other investigational indications that have been explored in Phase 1 or 2 trials include cholangiocarcinoma (bile duct cancer), non-small cell lung cancer (NSCLC), and colorectal cancer. Notably, development for prostate cancer has been discontinued, likely due to a lack of sufficient efficacy signal in early studies. The maximum clinical trial phase across all indications is Phase 2, with the exception of the pivotal Phase 3 study in mPDAC.
The ARC-8 study has been the cornerstone of Quemliclustat's clinical program, providing the most mature and compelling evidence of its potential benefit in mPDAC, a disease with a dire prognosis where the median OS with standard chemotherapy has remained below one year for decades.
ARC-8 is a Phase 1b, open-label, dose-escalation and expansion platform study designed to evaluate the safety, tolerability, and clinical activity of Quemliclustat in patients with previously untreated mPDAC. Following the dose-escalation phase, a dose of 100 mg administered intravenously was selected as the recommended dose for expansion. The study evaluated Quemliclustat in combination with the standard-of-care chemotherapy backbone of gemcitabine and nab-paclitaxel (G/nP). To explore potential synergy with PD-1 blockade, some cohorts also included the anti-PD-1 antibody zimberelimab (Z).
The primary efficacy analyses focused on several key patient cohorts with data as of a June 19, 2023 cutoff :
The efficacy results from ARC-8 were highly encouraging and exceeded historical benchmarks.
This divergence in outcomes between the doublet and triplet regimens is a pivotal finding. Pancreatic cancer is often characterized as an immunologically "cold" tumor, with low T-cell infiltration and a highly immunosuppressive stroma. The mechanism of PD-1 inhibitors relies on reinvigorating pre-existing but exhausted T-cell responses. In a "cold" TME, such responses may be sparse. In contrast, standard chemotherapy with gemcitabine/nab-paclitaxel is known to induce immunogenic cell death, a process that releases tumor antigens and danger signals capable of initiating a de novo anti-tumor immune response. However, the concurrent release of ATP, which is rapidly converted to immunosuppressive adenosine, can neutralize this effect. The synergy observed with the Q+G/nP doublet suggests that its primary benefit stems from Quemliclustat's ability to protect the nascent, chemotherapy-induced immune response from being immediately extinguished by adenosine. In this scenario, the addition of a PD-1 inhibitor provides little to no extra benefit because the limiting factor is the initial generation of an immune response, not the exhaustion of an existing one. This has significant implications for the design of future trials in mPDAC and other "cold" tumors.
A key aspect of the ARC-8 results was that the substantial efficacy benefit was achieved without introducing significant additional toxicity. The safety profile of the Quemliclustat-containing regimens was manageable and generally consistent with the known adverse event profile of the G/nP chemotherapy backbone. The most common Grade 3 or higher treatment-related adverse events were hematologic, including neutropenia (31-38%) and anemia (24-28%). No new or unexpected safety signals were observed, and deaths on study were not considered by investigators to be related to Quemliclustat or zimberelimab. This favorable safety profile is critical for a combination therapy intended for a broad patient population.
The ARC-8 study incorporated a strong translational research component that provided direct evidence of Quemliclustat's mechanism of action in human patients. Transcriptomic analysis of matched pre- and post-treatment tumor biopsies from 37 patients revealed two key findings. First, treatment with Quemliclustat-containing regimens led to a significant reduction in the tumor expression levels of the NR4A family of transcription factors (NR4A1, NR4A2, and NR4A3). Cell culture experiments confirmed that these genes are upregulated by adenosine signaling, establishing them as pharmacodynamic biomarkers of target engagement. Second, the observed decrease in NR4A expression was accompanied by a significant increase in tumor inflammation, as measured by a T-cell activation gene signature. These data provide a clear mechanistic link between Quemliclustat's inhibition of the adenosine pathway and the desired downstream effect of enhancing anti-tumor immunity within the TME.
Table 2: Summary of Efficacy Results from the ARC-8 Trial vs. Historical and Synthetic Controls
Regimen | n | Median OS (months) (95% CI) | 12-month OS Rate (%) | Median PFS (months) (95% CI) | ORR (%) (95% CI) | Source(s) |
---|---|---|---|---|---|---|
Q + G/nP (ARC-8 Cohort A2) | 29 | 19.4 (12.1, 23.0) | 72.3% | 8.8 (6.4, 12.6) | 41 (24, 61) | |
QZ + G/nP (ARC-8 Cohort A1) | 61 | 14.6 (10.6, 21.5) | 60.9% | 4.9 (3.7, 6.0) | 34 (23, 48) | |
All Pooled Q100 (ARC-8) | 122 | 15.7 (12.4, 20.9) | 62.7% | 6.3 (5.4, 7.7) | 39 (29.9, 47.8) | |
Synthetic Control Arm (SCA) | 122 | 9.8 (7.8, 11.4) | 41.1% | 5.5 (4.4, 6.6) | 41 (32.2, 50.3) | |
G/nP (MPACT Trial - Historical) | 431 | 8.5 (7.9, 9.5) | 35% | 5.5 (4.5, 5.8) | 23 (N/A) | |
NALIRIFOX (NAPOLI-3 Trial - Historical) | 383 | 11.1 (10.0, 12.1) | N/A | 7.4 (6.0, 7.7) | 42 (36.8, 47.5) |
Q=Quemliclustat; G/nP=gemcitabine/nab-paclitaxel; Z=zimberelimab; OS=Overall Survival; PFS=Progression-Free Survival; ORR=Objective Response Rate; N/A=Not Available
Table 3: Summary of Grade ≥3 Treatment-Related Adverse Events in ARC-8 (Pooled Cohorts)
Adverse Event | Frequency (%) | Source(s) |
---|---|---|
Neutropenia / Neutrophil Count Dec. | 31% - 38.7% | |
Anemia | 23.7% - 27.6% | |
Any Grade ≥3 TRAE | 73.0% - 85% | |
Serious TRAEs | 27.9% | |
AEs leading to Discontinuation | 23.0% |
TRAE = Treatment-Related Adverse Event
The promising results from the ARC-8 study, particularly from the doublet cohort, provided a clear path forward for a pivotal, registrational trial. Arcus initiated the PRISM-1 study in late 2024 to definitively establish the clinical benefit of Quemliclustat in first-line mPDAC.
Quemliclustat remains an investigational agent, but it has achieved a significant regulatory milestone that underscores its potential to address a major unmet medical need.
The development and potential commercialization of Quemliclustat are supported by a network of corporate partnerships, which is a common and effective strategy for advancing high-cost, late-stage oncology assets.
This strategic partnership structure is a significant asset for the Quemliclustat program. For a clinical-stage company like Arcus, the financial and operational demands of conducting a large, global Phase 3 trial and preparing for a multi-regional commercial launch are substantial. The collaboration with Gilead provides not only significant financial resources, which de-risks the late-stage development, but also access to Gilead's extensive global clinical operations infrastructure and established commercial footprint. Similarly, the partnership with Taiho leverages a regional expert's deep understanding of the regulatory and market access landscape in Asia. This tripartite alliance creates a robust global framework that significantly increases the probability of successfully bringing Quemliclustat to patients worldwide, should the PRISM-1 trial prove successful.
Quemliclustat has emerged as a highly promising investigational agent in oncology, distinguished by a clear and compelling mechanism of action that addresses a key pathway of immune evasion. By potently and selectively inhibiting CD73, the final enzyme in the immunosuppressive ATP-adenosine cascade, Quemliclustat is designed to dismantle the "adenosinergic halo" that protects tumors from immune attack. This mechanism is supported by a robust body of preclinical evidence demonstrating the drug's ability to restore the function of cytotoxic immune cells and synergize with other anti-cancer therapies, particularly chemotherapy.
The clinical data from the Phase 1b ARC-8 trial in first-line metastatic pancreatic ductal adenocarcinoma (mPDAC) represent a potential breakthrough. The median overall survival of 15.7 months in the pooled patient group, and particularly the 19.4 months observed in the doublet arm (Quemliclustat plus gemcitabine/nab-paclitaxel), far exceeds the historical benchmarks of 8.5-11.1 months for standard-of-care chemotherapy regimens. This substantial clinical benefit was achieved without adding significant toxicity, a critical factor for combination therapies in this patient population. The translational data from the trial, showing on-target modulation of the adenosine-regulated NR4A gene family and a corresponding increase in tumor inflammation, provide direct evidence that the drug is working in patients as intended.
From an expert perspective, the data package for Quemliclustat in mPDAC is exceptionally strong for a drug at this stage of development. The magnitude of the survival benefit seen in ARC-8, if confirmed, would be practice-changing. However, several critical points warrant consideration. The standout 19.4-month median OS was observed in a relatively small, non-randomized cohort (n=29), and such results from early-phase trials can sometimes be attenuated in larger, more heterogeneous Phase 3 populations. Therefore, the ongoing, randomized, placebo-controlled PRISM-1 trial is of paramount importance; its results will be the ultimate determinant of Quemliclustat's role in treating mPDAC.
The most intriguing and perhaps most important scientific question arising from the ARC-8 trial is the superior performance of the doublet regimen over the triplet regimen that included an anti-PD-1 antibody. This finding challenges the prevailing immuno-oncology paradigm of combining multiple checkpoint inhibitors. It suggests that for immunologically "cold" tumors like pancreatic cancer, the most effective strategy may not be to "release the brakes" on a non-existent T-cell response, but rather to use an agent like Quemliclustat to enable and protect a de novo immune response initiated by an inflammatory stimulus, such as immunogenic cell death induced by chemotherapy. This hypothesis, supported by the ARC-8 data, has significant implications for future combination strategies not only for Quemliclustat but for the field of immuno-oncology as a whole.
The immediate future of Quemliclustat is squarely focused on the execution and readout of the PRISM-1 trial. A positive result from this study would likely lead to regulatory submissions in the U.S., Europe, and Japan, positioning the combination of Quemliclustat with gemcitabine/nab-paclitaxel as a new global standard of care for the first-line treatment of mPDAC.
Beyond pancreatic cancer, the strong mechanistic rationale for CD73 inhibition suggests broad potential for Quemliclustat in other solid tumors. Future research will likely focus on identifying other tumor types where the adenosine axis is a dominant mechanism of immune suppression. Clinical trials are already underway in indications such as biliary tract cancer and NSCLC. The translational findings from ARC-8 also open the door for biomarker development. The expression of the NR4A gene family in baseline tumor tissue could be explored as a potential predictive biomarker to select patients most likely to benefit from Quemliclustat, paving the way for a more personalized therapeutic approach. In conclusion, Quemliclustat stands as a leading example of a rationally designed immuno-oncology agent that has translated a deep understanding of tumor biology into highly promising clinical activity, offering the potential for a significant improvement in outcomes for patients with one of the most difficult-to-treat cancers.
Published at: September 10, 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.