Biotech
1864871-20-4
Cusatuzumab is an investigational, first-in-class, glyco-engineered humanized IgG1 monoclonal antibody targeting the CD70 antigen, a member of the tumor necrosis factor (TNF) superfamily. Developed initially by argenx, cusatuzumab is being evaluated primarily for the treatment of Acute Myeloid Leukemia (AML), a hematological malignancy characterized by poor outcomes, particularly in elderly patients unfit for intensive chemotherapy. The therapeutic rationale for targeting CD70 is compelling; while its expression is transient and restricted in normal tissues, it is aberrantly overexpressed in a multitude of cancers, including over 95% of AML blasts. In AML, the interaction of CD70 with its receptor, CD27, promotes a cell-autonomous signaling cascade that is critical for the proliferation and survival of Leukemia Stem Cells (LSCs)—the chemotherapy-resistant cell population responsible for disease initiation and relapse.
The mechanism of action of cusatuzumab is dual-pronged, combining direct antagonism with potent immune-mediated killing. By binding to CD70, it blocks the pro-survival CD70/CD27 signaling pathway. Concurrently, its Fc region, which has been defucosylated via Potelligent® technology, exhibits dramatically enhanced binding to Fcγ receptors on immune effector cells, leading to powerful Antibody-Dependent Cellular Cytotoxicity (ADCC), Complement-Dependent Cytotoxicity (CDC), and Antibody-Dependent Cellular Phagocytosis (ADCP). This dual mechanism is designed to eliminate both bulk tumor cells and the underlying LSC reservoir.
The clinical development of cusatuzumab has been marked by both significant promise and substantial setbacks. Initial Phase I/II studies in combination with azacitidine demonstrated a favorable safety profile and remarkable preliminary efficacy, which culminated in a major global collaboration and licensing agreement with Janssen, an affiliate of Johnson & Johnson, valued at up to $1.6 billion. However, subsequent results from the randomized Phase II CULMINATE trial, while confirming a dose-response relationship favoring the 20 mg/kg dose, showed a complete remission rate that was not competitive with the newly established standard of care, venetoclax plus azacitidine. This shift in the therapeutic landscape led Janssen to terminate the collaboration in 2021.
Despite this, the unique LSC-targeting hypothesis and promising data from a Phase Ib trial (ELEVATE) evaluating a triplet combination with venetoclax and azacitidine supported the drug's continued development. Global rights were returned to argenx and subsequently licensed to OncoVerity, an asset-centric spin-off company. OncoVerity has initiated a pivotal, randomized Phase II trial (OV-AML-1231) comparing the triplet regimen against the standard-of-care doublet. This trial features a risk-stratified design and a primary endpoint of Overall Survival, representing a mature strategic pivot that directly addresses the questions necessary to establish clinical value in the modern AML treatment paradigm. The future of cusatuzumab now hinges on the outcome of this study, which will determine if its unique biological mechanism can translate into a meaningful survival benefit for high-risk AML patients.
Cusatuzumab is a biologic therapeutic agent classified as a humanized monoclonal antibody.[1] As a complex protein-based therapy, it is defined by a specific set of identifiers used across chemical, regulatory, and clinical databases to ensure precise tracking and characterization. The International Nonproprietary Name (INN) assigned to the molecule is Cusatuzumab.[2] It is also widely known by its development code names, primarily ARGX-110, which was used by its originator company, argenx, and later as JNJ 74494550 during its co-development with Janssen.[3]
The molecule is a humanized IgG1 isotype antibody, signifying that its constant regions are derived from human immunoglobulin G1, while its antigen-binding variable regions originate from a non-human source—in this case, a llama—and have been engineered to reduce immunogenicity.[2] A key structural feature of cusatuzumab is its defucosylation, a specific form of glyco-engineering designed to enhance its biological activity.[2] The antibody is produced using recombinant DNA technology in a mammalian cell expression system, specifically Chinese Hamster Ovary (CHO) cells.[6] The approximate molecular weight of the final glycoprotein is 144.36 kDa.[8]
Table 1: Core Identifiers and Properties of Cusatuzumab
| Property | Identifier / Value | Source(s) |
|---|---|---|
| International Nonproprietary Name (INN) | Cusatuzumab | 2 |
| DrugBank ID | DB15100 | 2 |
| CAS Number | 1864871-20-4 | 2 |
| UNII (Unique Ingredient Identifier) | RR5U4UFJ8L | 2 |
| NCI Thesaurus Code | C106116 | 2 |
| Synonyms / Code Names | ARGX-110, JNJ 74494550, Anti-CD70 Monoclonal Antibody ARGX-110 | 2 |
| Molecular Class | Biotech; Humanized, Defucosylated IgG1 Monoclonal Antibody | 2 |
| Molecular Weight (Approx.) | 144.36 kDa | 8 |
| Host Cell | Chinese Hamster Ovary (CHO) Cells | 6 |
As a canonical IgG1 antibody, cusatuzumab possesses a symmetric, Y-shaped structure composed of four polypeptide chains: two identical heavy chains and two identical light chains, linked by disulfide bonds. Each heavy chain consists of a variable domain ($V_H$) and three constant domains ($C_H1$, $C_H2$, $C_H3$). Each light chain comprises one variable domain ($V_L$) and one constant domain ($C_L$). The pairing of the $V_H$ and $V_L$ domains at the tips of the "Y" arms forms the two identical antigen-binding fragments (Fab), which are responsible for the antibody's high-affinity recognition of the CD70 antigen. The stem of the "Y" is formed by the paired $C_H2$ and $C_H3$ domains, creating the fragment crystallizable (Fc) region, which mediates the antibody's interaction with components of the immune system, such as Fc receptors on effector cells and the C1q component of the complement system.
The primary structure, or amino acid sequence, is the fundamental blueprint that dictates the antibody's folding, antigen specificity, and effector functions. The complete amino acid sequences for the heavy and light chains of cusatuzumab have been validated and are publicly available through the FDA Global Substance Registration System (GSRS).[10]
Table 2: Amino Acid Sequence of Cusatuzumab Heavy and Light Chains
| Chain | Amino Acid Sequence | Source |
|---|---|---|
| Heavy Chain (2 identical chains) | EVQLVESGGGLVQPGGSLRLSCAASGFTFSVYYMNWVRQAPGKGLEWVSDINNEGGTTYYADSVKGRFTISRDNSKNSLYLQMNSLRAEDTAVYYCARDAGYSNHVPIFDSWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK | 10 |
| Light Chain (2 identical chains) | QAVVTQEPSLTVSPGGTVTLTCGLKSGSVTSDNFPTWYQQTPGQAPRLLIYNTNTRHSGVPDRFSGSILGNKAALTITGAQADDEAEYFCALFISNPSVEFGGGTQLTVLGQPKAAPSVTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSKQSNNKYAASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPTECS | 10 |
Cusatuzumab has received significant regulatory acknowledgments for its potential to treat a rare and serious disease. On April 22, 2020, the European Medicines Agency (EMA) granted a positive orphan designation to cusatuzumab for the "Treatment of acute myeloid leukaemia".[2] Similarly, the United States Food and Drug Administration (FDA) has also granted orphan drug designation for the treatment of AML.[11]
The acquisition of orphan drug status is a critical strategic asset in the development of a new therapeutic. This designation is granted to drugs intended for the treatment, prevention, or diagnosis of a life-threatening or chronically debilitating rare disease (affecting fewer than 200,000 people in the U.S. or not more than 5 in 10,000 in the E.U.). The status provides substantial development incentives, including protocol assistance from regulatory agencies, tax credits for clinical trial costs, waived application fees, and, most importantly, a period of market exclusivity upon regulatory approval. In the U.S., this exclusivity period is seven years, while in the E.U., it is ten years. This protected market period is a powerful commercial advantage, designed to encourage investment in therapies for rare diseases that might otherwise be commercially unviable. For a drug like cusatuzumab, with a complex and capital-intensive development history, this designation underscores the recognized high unmet medical need in AML and provides a crucial framework for its potential commercialization by its current developer, OncoVerity.
The therapeutic targeting of cusatuzumab is rooted in the distinct biological roles of the CD70/CD27 signaling axis in normal versus malignant tissues. CD70 (also known as TNFSF7) is a type II transmembrane protein and a member of the tumor necrosis factor (TNF) ligand superfamily.[5] Its sole known receptor is CD27 (TNFRSF7), a type I transmembrane protein belonging to the TNF receptor superfamily.[1]
Under normal physiological conditions, the expression of CD70 is tightly regulated and transient. It is primarily found on the surface of highly activated T-cells, B-cells, and mature dendritic cells.[1] The interaction between CD70 on these activated immune cells and CD27, which is constitutively expressed on lymphocytes, provides a critical co-stimulatory signal. This signal is essential for the clonal expansion, differentiation, and long-term survival of T-cells and B-cells during an adaptive immune response.[1] Following the resolution of an immune challenge, CD70 expression is downregulated, thereby helping to control the magnitude and duration of the lymphocyte response. This transient and restricted expression pattern in healthy tissues is a key characteristic that makes CD70 an attractive target for cancer therapy, as it minimizes the potential for on-target, off-tumor toxicity.
In stark contrast to its controlled expression in the immune system, CD70 is aberrantly and constitutively overexpressed in a wide range of both hematological and solid tumors.[2] In the context of AML, studies have documented CD70 expression on the surface of blasts from over 95% of newly diagnosed patients.[13]
This overexpression is not a passive marker but an active driver of malignant cell survival and proliferation. When CD70 on a cancer cell binds to CD27, which is often co-expressed on the same or adjacent cancer cells, it triggers a downstream signaling cascade that culminates in the activation of the Nuclear Factor-kappa B (NF-κB) pathway.[1] NF-κB is a master regulator of genes involved in cell survival, proliferation, and inflammation, and its constitutive activation is a hallmark of many cancers, including AML. This CD70-driven signaling thus creates a positive feedback loop that promotes the growth of the malignancy.[1]
Furthermore, the engagement of the CD70/CD27 axis leads to the proteolytic cleavage and shedding of the CD27 ectodomain from the cell surface, releasing a soluble form of the receptor (sCD27) into the circulation. Elevated urinary and serum concentrations of sCD27 have been identified as a negative prognostic biomarker, correlating with poorer outcomes in patients with various malignancies.[1] CD70 may also contribute to immune evasion by creating a tumor microenvironment that recruits and supports immunosuppressive regulatory T-cells (Tregs).[1]
The scientific rationale for cusatuzumab is most sharply focused on its potential to eradicate Leukemia Stem Cells (LSCs). AML is understood to be a hierarchical disease, initiated and sustained by a small, resilient subpopulation of LSCs.[12] These cells possess properties of self-renewal and are largely resistant to conventional chemotherapy, allowing them to persist after treatment and drive disease relapse, which is the primary cause of mortality in AML.[12] Therefore, therapies capable of specifically targeting and eliminating the LSC population are considered essential for achieving durable remissions and potential cures.
Crucially, LSCs have been shown to consistently express both CD70 and its receptor CD27. This co-expression establishes a cell-autonomous signaling loop that is vital for LSC propagation and survival.[12] By directly promoting the self-renewal of the cells at the apex of the leukemic hierarchy, the CD70/CD27 axis represents a fundamental dependency of the disease.
A key discovery that formed the basis for the combination therapy strategy with cusatuzumab involves the effect of hypomethylating agents (HMAs) like azacitidine, a standard therapy for AML. Preclinical studies demonstrated that treatment with HMAs leads to the demethylation of the CD70 gene promoter.[17] This epigenetic modification results in a paradoxical upregulation of CD70 expression on the surface of LSCs.[12] This phenomenon creates a unique therapeutic opportunity. While HMAs have their own anti-leukemic activity, they are generally insufficient to eliminate the LSC pool on their own. However, by increasing the density of the CD70 target on the most critical and dangerous cell population, HMAs effectively "paint a target" on the LSCs, priming them for enhanced recognition and destruction by a CD70-directed antibody like cusatuzumab. This provides a strong mechanistic basis for a synergistic interaction, where the HMA not only treats the disease but also renders the root of the disease—the LSC—more vulnerable to the targeted immunotherapy.
Cusatuzumab employs a sophisticated, dual mechanism of action that combines the direct interruption of oncogenic signaling with the potent recruitment of the host immune system to actively destroy malignant cells. This two-pronged approach is designed to provide a more comprehensive and durable anti-leukemic effect than either mechanism could achieve alone.[12]
The first component of cusatuzumab's activity is its function as a direct antagonist of the CD70/CD27 pathway. As a high-affinity monoclonal antibody, cusatuzumab binds selectively to the extracellular domain of the human CD70 molecule.[2] This binding physically obstructs the interaction between CD70 and its receptor, CD27, effectively neutralizing the pathway's activity.[1]
By blocking this interaction, cusatuzumab interrupts the downstream pro-survival signaling cascade mediated by NF-κB that drives the proliferation of malignant cells and, most critically, Leukemia Stem Cells (LSCs).[1] This leads to an inhibition of tumor cell growth and can induce myeloid differentiation, pushing the immature blasts toward a more mature, non-proliferative state.[16] In addition to inhibiting proliferation, this blockade also prevents the proteolytic shedding of the CD27 receptor, thereby reducing the levels of soluble CD27 (sCD27), a marker associated with poor prognosis.[1] This mechanism directly targets the cell-autonomous growth advantage conferred by aberrant CD70 expression.
The second, and arguably more powerful, mechanism of action involves the active elimination of CD70-expressing tumor cells through Fc-mediated effector functions.[19] The Fc region of the cusatuzumab IgG1 antibody acts as a bridge, linking the CD70-positive cancer cell to various components of the innate immune system.
A defining feature of cusatuzumab is that it has been specifically glyco-engineered through defucosylation, a process utilizing Potelligent® technology.[22] In a standard IgG1 antibody, the N-glycans attached to the Fc region contain fucose sugar residues. These fucose molecules sterically hinder the binding of the Fc region to the FcγRIIIa (CD16a) receptor, which is expressed at high levels on Natural Killer (NK) cells, the primary mediators of Antibody-Dependent Cellular Cytotoxicity (ADCC). By manufacturing cusatuzumab in a cell line that lacks the enzyme responsible for fucosylation, the resulting antibody has a significantly higher affinity for FcγRIIIa. This enhanced binding leads to a much more efficient engagement of NK cells, which, upon activation, release cytotoxic granules (containing perforin and granzymes) that induce apoptosis in the targeted cancer cell. This "enhanced ADCC" is a core design feature of cusatuzumab, intended to maximize its cell-killing potential.[2]
In addition to ADCC, the Fc region of cusatuzumab can also initiate two other killing mechanisms:
This suite of cytotoxic mechanisms ensures that cusatuzumab does not merely suppress tumor growth but actively works to eliminate CD70-positive cells from the body.
The biological effects of cusatuzumab's dual mechanism of action have been validated in both preclinical models and clinical trial settings. In vitro and xenotransplantation experiments provided the foundational proof-of-concept. These studies demonstrated that cusatuzumab was significantly more effective at eliminating AML LSCs than a functionally equivalent antibody that could only block CD70/CD27 signaling but lacked enhanced ADCC capabilities. The superiority of cusatuzumab was particularly evident in the presence of human NK cells, confirming the critical contribution of the enhanced ADCC mechanism.[12]
These preclinical findings were subsequently corroborated by pharmacodynamic data from early-phase human trials. In the initial Phase I/II study (NCT03030612), patients treated with even a single dose of cusatuzumab monotherapy exhibited a significant reduction in bone marrow blasts, with an average decrease of approximately 30-32% within two weeks.[12] More importantly, the treatment led to a significant reduction in the frequency of LSCs in the bone marrow of all analyzed patients, directly confirming the drug's activity against its primary target population.[12]
To understand the molecular underpinnings of this effect, single-cell RNA sequencing was performed on samples from treated patients. This advanced analysis revealed that cusatuzumab monotherapy induced profound changes in the gene expression profiles of LSCs. Specifically, it triggered the upregulation of gene signatures associated with apoptosis (programmed cell death) and myeloid differentiation.[12] This provides strong molecular evidence that cusatuzumab not only kills LSCs but also pushes surviving cells out of their self-renewing, stem-like state toward a more mature, non-malignant phenotype. These pharmacodynamic results provide a clear biological rationale for the clinical responses observed in AML patients.
The characterization of a drug's pharmacokinetic (PK) profile—its absorption, distribution, metabolism, and excretion—is fundamental to establishing a rational and effective dosing regimen. For cusatuzumab, PK parameters were assessed as a secondary objective in its clinical trials to understand its behavior in the human body and ensure that the chosen dose and schedule would result in sustained exposure sufficient for therapeutic activity.[12]
Detailed pharmacokinetic data for cusatuzumab comes from a Phase 1 study conducted in Japanese patients with newly diagnosed AML who were ineligible for intensive chemotherapy. In this study, participants received cusatuzumab at a dose of 20 mg/kg.[23] The PK analysis from this cohort provides key insights into the drug's concentration-time profile.
Following a single intravenous infusion, cusatuzumab exhibited PK properties typical of a monoclonal antibody. The key parameters observed were [23]:
The pharmacokinetic profile of cusatuzumab provides a strong justification for the dosing schedule used in its major clinical trials. The primary goal of antibody therapy is to maintain serum concentrations consistently above the level required for target saturation and biological effect throughout the entire treatment cycle.
The observed terminal half-life of approximately 9.3 days is a critical parameter. A long half-life is characteristic of IgG antibodies and allows for less frequent dosing. The biweekly (every two weeks) administration schedule—specifically, on Days 3 and 17 of a 28-day cycle—used in the CULMINATE and ELEVATE trials is well-supported by this PK profile.[3] A 14-day dosing interval is short enough relative to the 9.3-day half-life to prevent drug concentrations from falling to sub-therapeutic levels. The substantial trough concentrations of over 100 µg/mL confirm that this schedule maintains high levels of circulating antibody. This sustained exposure is essential for ensuring continuous pressure on CD70-expressing cells, allowing both the signal blockade and the Fc-mediated killing mechanisms to remain active throughout the treatment period. The dose-escalation studies (1, 3, 10, and 20 mg/kg) also established a predictable, dose-proportional increase in exposure, which informed the selection of the 10 mg/kg and 20 mg/kg doses for later-stage evaluation.[26]
The clinical development of cusatuzumab has been strategically focused on addressing a significant unmet medical need: the treatment of newly diagnosed Acute Myeloid Leukemia (AML) in patients who are considered ineligible for intensive induction chemotherapy due to age or comorbidities.[3] This population has historically had a very poor prognosis, creating a clear opportunity for novel, less toxic therapies. The clinical program has evolved over time, reflecting both the growing understanding of the drug's properties and the dynamic changes in the standard of care for AML. The trajectory can be traced through a series of key clinical trials, each designed to answer a specific question, from initial safety and dose-finding to randomized comparisons against the contemporary standard of care.
Table 3: Summary of Key Clinical Trials for Cusatuzumab in AML
| Trial ID (NCT) | Phase | Official Title / Name | Combination Agents | Patient Population | Primary Endpoint(s) | Status |
|---|---|---|---|---|---|---|
| NCT03030612 | I/II | A Study of Cusatuzumab Plus Azacitidine in Participants With Newly Diagnosed AML or High-risk MDS | Azacitidine | ND AML/HR-MDS, unfit for intensive chemo | Phase I: RP2D; Phase II: ORR | Completed |
| NCT04023526 | II | A Phase 2 Study of Cusatuzumab Plus Azacitidine in Patients With Newly Diagnosed AML Who Are Not Candidates for Intensive Chemotherapy (CULMINATE) | Azacitidine | ND AML, unfit for intensive chemo | Complete Remission (CR) Rate | Active, Not Recruiting |
| NCT04150887 | Ib | A Study of Cusatuzumab in Combination With Background Therapy for the Treatment of Subjects With AML (ELEVATE) | Azacitidine, Venetoclax | ND AML, unfit for intensive chemo | Safety and Tolerability | Active, Not Recruiting |
| NCT06384261 | II | A Study Comparing Venetoclax and Azacitidine Plus Cusatuzumab to Venetoclax and Azacitidine in Newly Diagnosed AML Ineligible for Intensive Therapy (OV-AML-1231) | Azacitidine, Venetoclax | ND AML, unfit for intensive chemo | Overall Survival (OS) | Recruiting |
ND: Newly Diagnosed; HR-MDS: High-Risk Myelodysplastic Syndrome; RP2D: Recommended Phase 2 Dose; ORR: Overall Response Rate.
The clinical journey of cusatuzumab began with a two-part, open-label, multicenter Phase I/II trial (NCT03030612) designed to establish the safety, tolerability, and preliminary efficacy of cusatuzumab in combination with the hypomethylating agent azacitidine.[15] The Phase I dose-escalation portion enrolled patients into four sequential cohorts, testing intravenous cusatuzumab at doses of 1, 3, 10, and 20 mg/kg.[26]
The study demonstrated a very favorable safety profile. No dose-limiting toxicities (DLTs) were observed across the entire dose range, and a maximum tolerated dose (MTD) was not reached.[12] This indicated that the drug was well-tolerated even at the highest dose tested.
The preliminary efficacy signals from this initial study were exceptionally strong and generated significant excitement. An early report on the first 12 enrolled patients revealed a 92% overall response rate (ORR), with 11 of 12 patients responding to the therapy. This included 9 patients (82%) who achieved a complete response with or without complete hematologic recovery (CR/CRi).[24] Furthermore, 42% of these patients achieved minimal residual disease (MRD) negativity, suggesting a deep level of response.[24] More mature data from the full cohort of 38 patients (12 in Phase I, 26 in Phase II) later confirmed this activity, showing an objective response in 19 of 38 patients (50%), with 14 of 38 (37%) achieving a complete remission.[29] It was these highly promising initial results that served as the primary catalyst for the multi-billion dollar partnership with Janssen.[15]
Following the encouraging Phase I/II results, the CULMINATE trial (NCT04023526) was initiated. This was a larger, randomized, open-label Phase II study designed to formally compare two dose levels of cusatuzumab—10 mg/kg versus 20 mg/kg—when combined with azacitidine, to determine the optimal dose for future development.[3] The primary efficacy endpoint was the rate of complete remission (CR).[3]
The trial enrolled 103 patients, who were randomized 1:1 to the two dose arms.[3] The results demonstrated a clear dose-response relationship in favor of the higher dose.
Table 4: Efficacy Outcomes from the CULMINATE (NCT04023526) Trial
| Efficacy Endpoint | Cusatuzumab 10 mg/kg Cohort (n=51) | Cusatuzumab 20 mg/kg Cohort (n=52) |
|---|---|---|
| Complete Remission (CR) Rate | 12% (95% CI: 6-23) | 27% (95% CI: 17-40) |
| Composite Complete Remission (CRc) Rate | Not explicitly reported, but lower than 20mg/kg arm | 40% |
Data sourced from.[3] CRc includes CR and CR with incomplete hematologic recovery.
The totality of the clinical data from the study led to the conclusion that 20 mg/kg was the optimal dose of cusatuzumab for further investigation.[27] However, the 27% CR rate observed with the optimal dose, while demonstrating clear biological activity, was viewed unfavorably in the context of a rapidly evolving therapeutic landscape. During the course of the CULMINATE trial, results from the pivotal VIALE-A study established the combination of venetoclax and azacitidine, which demonstrated a CR/CRi rate of approximately 66%, as the new global standard of care for this patient population. The efficacy bar had been significantly raised. Consequently, Part 2 of the CULMINATE trial (a single-arm expansion) was cancelled, and this reassessment of the competitive landscape was the primary driver behind Janssen's decision to terminate its collaboration with argenx.[3]
Recognizing the shift in the standard of care, the development strategy for cusatuzumab pivoted to investigate its potential as part of a triplet combination. The ELEVATE trial (NCT04150887) is a Phase 1b study designed to evaluate the safety, tolerability, and preliminary activity of adding cusatuzumab to the new backbone of venetoclax and azacitidine (a regimen sometimes abbreviated as CVA or VAC).[33]
Interim data from this single-arm study were highly encouraging. In an evaluable population of 42 patients, the triplet combination demonstrated very high response rates [33]:
These numerically impressive response rates suggested that adding cusatuzumab to the venetoclax/azacitidine backbone could be beneficial and was well-tolerated.[33] However, a critical limitation of these findings is the single-arm, non-randomized design of a Phase 1b study. The venetoclax/azacitidine doublet is itself a highly active regimen, making it difficult to definitively attribute the incremental efficacy to cusatuzumab without a direct comparator arm. The key unanswered questions were whether the addition of cusatuzumab could lead to deeper responses (i.e., higher rates of MRD negativity) and, most importantly, more durable responses that translate into improved long-term survival. Answering these questions required a randomized, controlled trial.
Following the reacquisition of rights and the formation of OncoVerity, a new, well-designed clinical trial was initiated to definitively assess the value of cusatuzumab. The OV-AML-1231 study (NCT06384261) is a randomized, open-label, multicenter Phase II trial that represents a mature and necessary strategic pivot for the program.[19]
The trial design directly addresses the limitations of the previous studies. It will randomize approximately 120 newly diagnosed, unfit AML patients in a 2:1 ratio to receive either [37]:
This randomized design will provide the high-quality, comparative data needed to isolate the specific contribution of cusatuzumab. Crucially, the primary endpoint of the study is Overall Survival (OS), the gold standard for demonstrating clinical benefit in oncology and the most meaningful outcome for patients.[37]
A key feature of this new trial is its focus on a precision-medicine approach. The study protocol specifies that the trial population will be enriched for patients with adverse cytogenetic and molecular risk features.[37] OncoVerity is leveraging an advanced bioinformatics platform to guide this development, based on the hypothesis that cusatuzumab's unique LSC-targeting mechanism will be most impactful in these high-risk patients, who often have poor responses or early relapse on standard therapies.[19] This targeted, randomized, OS-focused trial design gives cusatuzumab its best and most rational opportunity to demonstrate its value and secure a place in the modern AML treatment armamentarium.
Across its clinical development program, which has included treatment of over 100 patients with AML, cusatuzumab has demonstrated a generally manageable and predictable safety profile.[19] In the foundational Phase I/II dose-escalation study (NCT03030612), no dose-limiting toxicities were identified at doses up to and including 20 mg/kg, and a maximum tolerated dose was not reached.[12] This early finding established a favorable therapeutic window for the drug.
The overall safety profile observed in combination studies is largely consistent with the known toxicities of the background therapeutic agents (azacitidine and/or venetoclax) and the complications associated with the underlying disease in a typically frail, elderly patient population with compromised bone marrow function.[23] The addition of cusatuzumab does not appear to introduce novel, unexpected toxicities but rather contributes a specific and manageable set of adverse events, primarily infusion-related reactions.
The most frequently reported treatment-emergent adverse events (TEAEs) are hematologic in nature, which is expected in a population with AML undergoing treatment that affects the bone marrow. Data from the randomized CULMINATE trial provides a clear picture of the most common Grade 3 or worse TEAEs.
Table 5: Summary of Common Treatment-Emergent Adverse Events (Grade ≥3) in the CULMINATE Trial
| Adverse Event (Grade ≥3) | Cusatuzumab 10 mg/kg + Azacitidine (n=51) | Cusatuzumab 20 mg/kg + Azacitidine (n=51) |
|---|---|---|
| Thrombocytopenia | 47% (24 patients) | 57% (29 patients) |
| Anemia | 47% (24 patients) | 33% (17 patients) |
| Neutropenia | 39% (20 patients) | 39% (20 patients) |
| Leukopenia | Not specified in this source | Not specified in this source |
| Pneumonia | Not specified in this source | Not specified in this source |
| Serious TEAEs (Any) | 86% (44 patients) | 78% (40 patients) |
Data sourced from.[3] Note: Some sources report slightly different aggregated numbers based on the full study population.[31]
Infections are also a common and serious complication in this patient population. Across the full cohort of the Phase I/II study (NCT03030612), infections of any kind were reported in 84.2% of patients.[29] Treatment-related deaths have occurred in the trials, with causes including pneumonia and septic shock, which are known risks for patients with AML and profound neutropenia.[3]
Infusion-related reactions (IRRs) are a known class effect for monoclonal antibody therapies. In the Phase I/II study, IRRs were reported in 18.4% of patients, including two Grade 3 events.[29] These reactions are generally manageable through the standard practice of premedication with acetaminophen, an antihistamine, and a corticosteroid prior to infusion.[23]
The randomized design of the CULMINATE trial allowed for a direct comparison of the safety profiles of the 10 mg/kg and 20 mg/kg doses of cusatuzumab. The data indicated that the higher, more efficacious dose of 20 mg/kg did not come with a significant safety penalty. The rates of Grade 3 or worse TEAEs, including key hematologic toxicities like neutropenia, were similar between the two arms. The rate of serious TEAEs was also comparable, and was numerically slightly lower in the 20 mg/kg arm (78%) compared to the 10 mg/kg arm (86%).[3] This finding was crucial in supporting the selection of the 20 mg/kg dose for all subsequent clinical development.
In the ELEVATE trial, which evaluated the triplet combination of cusatuzumab with venetoclax and azacitidine, the regimen was reported to be well-tolerated, with a safety profile consistent with prior studies of cusatuzumab and the known toxicities of the venetoclax/azacitidine backbone.[33] This suggests that cusatuzumab can be safely integrated into the current standard-of-care regimen without introducing overlapping or synergistic toxicities that would make the combination unfeasible.
Cusatuzumab, originally designated ARGX-110, was discovered and developed by argenx, a clinical-stage biotechnology company based in the Netherlands and Belgium.[11] The antibody was generated using argenx's proprietary SIMPLE Antibody™ platform. This technology leverages the unique immune system of llamas (Camelidae), which produce, in addition to conventional antibodies, a class of simpler, heavy-chain-only antibodies. The platform allows for the rapid generation of a diverse repertoire of antibodies with high affinity and specificity against complex disease targets. The variable domains from these llama-derived antibodies are then "humanized" and grafted onto a human IgG1 Fc backbone to create therapeutic candidates like cusatuzumab, which are designed for optimal activity and reduced immunogenicity in patients.[11]
Based on the compelling preclinical rationale targeting LSCs and the exceptionally promising early clinical data from the Phase I/II study (NCT03030612), cusatuzumab attracted major pharmaceutical interest. In December 2018, argenx entered into an exclusive, worldwide collaboration and license agreement with Cilag GmbH International, an affiliate of the Janssen Pharmaceutical Companies of Johnson & Johnson, to co-develop and commercialize cusatuzumab.[21]
The financial terms of the agreement were substantial, reflecting the high perceived potential of the asset. The deal included [11]:
This agreement, with a total potential value exceeding $1.6 billion, provided a massive infusion of capital and validation for argenx and placed the development of cusatuzumab on a global stage with a major oncology partner.
However, in June 2021, Janssen announced its decision to terminate the collaboration and return all rights to cusatuzumab to argenx.[32] The decision was not precipitated by any new or unexpected safety concerns. Instead, Janssen's official statement cited a "review of all available cusatuzumab data and in consideration of the evolving standard of care for the treatment of acute myeloid leukemia (AML)".[30] This was a direct reference to the emergence of the venetoclax plus azacitidine regimen as the new, highly effective standard of care following the publication of the VIALE-A trial results. The 27% complete remission rate observed for the cusatuzumab/azacitidine doublet in the CULMINATE trial, while clinically meaningful, was not commercially competitive against the approximately 66% CR/CRi rate of the new standard. Janssen's decision was therefore a strategic and financial one, based on a reassessment of the commercial landscape and the increased risk and investment that would be required to prove the value of a triplet combination in a randomized trial.
Following the termination of the Janssen partnership, argenx regained full global rights to cusatuzumab in June 2021.[33] Rather than abandoning the program, argenx sought a new path forward. In March 2023, this new path was revealed with the announcement that worldwide licensing rights for cusatuzumab had been acquired by OncoVerity, Inc..[30]
OncoVerity is an "asset-centric spin-off" company specifically created to optimize the development of cusatuzumab. The company represents a strategic fusion of argenx's deep expertise in the CD70/CD27 pathway with the translational biology and clinical expertise in AML of Dr. Clayton Smith from the University of Colorado.[30] The new venture was launched with a $30 million Series A financing round, with participation from argenx itself, signaling its continued belief in the asset.[30]
This transition marks a significant strategic pivot for the cusatuzumab program. Instead of pursuing a broad development strategy, OncoVerity is employing a focused, precision-medicine approach. This is embodied in the design of its new randomized Phase II trial (NCT06384261), which is specifically enriching for high-risk patient populations and leveraging advanced bioinformatics to identify those most likely to benefit from cusatuzumab's unique LSC-targeting mechanism.[19] This revival under a new, specialized entity represents a rational and data-driven effort to find the right patient population where cusatuzumab's distinct biological activity can provide a clear and substantial clinical benefit over the current standard of care.
The therapeutic landscape for newly diagnosed AML in patients unfit for intensive chemotherapy has been transformed by the approval of the BCL-2 inhibitor venetoclax in combination with a hypomethylating agent (HMA). This regimen has established a new benchmark for efficacy, with high rates of complete remission and a significant improvement in overall survival compared to HMA monotherapy. Consequently, any new therapeutic agent entering this space must demonstrate a clear and substantial benefit on top of this potent backbone.
Cusatuzumab is positioned to address the primary limitation of the current standard of care: relapse. Despite high initial response rates with venetoclax-based therapy, many patients, particularly those with high-risk molecular features, eventually relapse. This is believed to be driven by the persistence of drug-resistant Leukemia Stem Cells (LSCs). The central therapeutic hypothesis for cusatuzumab is that its unique ability to target and eliminate the CD70-positive LSC population can lead to deeper, more durable remissions and, ultimately, improved long-term survival. Its role is not to replace the current standard of care, but to augment it, specifically targeting the root cause of relapse in a way that venetoclax and azacitidine may not.
The clinical data for cusatuzumab tells a story of a drug with clear biological activity but one whose development has been challenged by a moving goalpost. The initial Phase I/II data were highly impressive, suggesting a best-in-class profile for an HMA-doublet and justifying a landmark partnership. However, the more robust, randomized data from the CULMINATE trial, while confirming a dose-response, yielded an efficacy level that was quickly superseded by the venetoclax combination. The promising, albeit uncontrolled, high response rates from the ELEVATE triplet study provided the rationale to continue, but underscored the need for definitive, comparative data.
The future of cusatuzumab now rests almost entirely on the outcome of the ongoing, randomized Phase II OV-AML-1231 trial (NCT06384261). This study is well-designed to provide a definitive answer to the most critical question: does the addition of cusatuzumab to venetoclax and azacitidine improve overall survival compared to the standard of care alone? A positive result, particularly if driven by benefit in the pre-specified high-risk subgroups, would validate the LSC-targeting hypothesis, vindicate the precision-medicine strategy of OncoVerity, and could establish a new standard of care for a defined patient population. A negative result, conversely, would likely signal the end of its development in AML.
To maximize the potential for success and fully leverage the ongoing clinical investment, the following strategic recommendations are proposed:
Published at: October 23, 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.