This report provides a comprehensive analysis of the investigational drug CDX-301, a recombinant human Fms-like tyrosine kinase-3 ligand (rhuFlt3L) developed by Celldex Therapeutics. The analysis synthesizes data from preclinical studies, foundational clinical trials, and multiple oncology combination studies to evaluate its mechanism of action, clinical efficacy, safety profile, and strategic positioning within the biopharmaceutical landscape.
A critical point of clarification is the existence of two distinct therapeutic candidates designated CDX-301. The subject of this report is Celldex Therapeutics' rhuFlt3L, a potent hematopoietic cytokine. This must be distinguished from an unrelated zeaxanthin-based pharmaceutical candidate from Cardax, Inc., also named CDX-301, which is under development for macular degeneration.
Celldex's CDX-301 functions by binding to the Flt3 receptor (CD135), stimulating the proliferation and mobilization of hematopoietic stem cells (HSCs) and, most notably, dendritic cells (DCs). This dual mechanism has supported two distinct development paths: hematopoietic stem cell transplantation (HSCT) and immuno-oncology. A foundational Phase 1 study in healthy volunteers (NCT01465139) established a favorable safety profile and demonstrated potent, duration-dependent expansion of CD34+ progenitor cells and key DC subsets, validating its biological activity.
While an initial pilot study was launched to explore its utility in allogeneic HSCT, the program appears to have been deprioritized in favor of its application in oncology. In this arena, CDX-301 has been strategically positioned as an immune-priming agent. Clinical trials in melanoma (NCT02129075) and non-small cell lung cancer (NSCLC) (NCT02839265) have successfully demonstrated that pre-treatment or concurrent administration of CDX-301 can significantly enhance the immunogenicity and efficacy of cancer vaccines and stereotactic radiotherapy, respectively. The observation of a systemic "abscopal effect" in the NSCLC trial provides compelling evidence for its ability to generate a broad anti-tumor immune response.
Despite these successes, corporate strategy at Celldex Therapeutics has evolved. CDX-301 is no longer featured as a lead asset in the company's pipeline, which now prioritizes novel antibody-based therapies for inflammatory diseases and other oncology targets. The development of CDX-301 continues primarily through investigator-sponsored trials and as an internal tool to potentiate other proprietary agents, such as the CD40 agonist CDX-1140. Its trajectory reflects a strategic repositioning from a potential standalone product to a valuable platform technology, a shift that mirrors the broader evolution of the immuno-oncology field toward complex, synergistic combination therapies.
A crucial initial step in the analysis of CDX-301 is to resolve a significant ambiguity arising from the use of the same designation by two separate companies for two fundamentally different drug candidates. The name "CDX-301" refers to both a protein-based immunotherapy agent from Celldex Therapeutics and a carotenoid-based pharmaceutical from Cardax, Inc..[1]
Celldex Therapeutics' CDX-301, the subject of this report, is a recombinant human Fms-like tyrosine kinase-3 ligand (rhuFlt3L). It is a potent hematopoietic cytokine investigated for its ability to mobilize stem cells and expand dendritic cell populations, primarily for applications in hematopoietic stem cell transplantation and as a combination agent in cancer immunotherapy.[1]
Cardax, Inc.'s CDX-301 is an entirely unrelated compound. It is a pharmaceutical candidate based on zeaxanthin, a xanthophyll carotenoid, being developed for the treatment of macular degeneration.[2] Its mechanism involves accumulating in the human eye to provide protection against oxidative damage and inflammation associated with retinal disorders, with a target initial indication of Stargardt disease.[2] Cardax has stated it may seek to monetize this asset through licensing or sale.[2]
To ensure clarity, this report will focus exclusively on the rhuFlt3L molecule developed by Celldex Therapeutics. All subsequent references to CDX-301 pertain to this specific agent.
CDX-301 is classified as a protein-based therapy, specifically a soluble, recombinant human protein.[3] Its therapeutic class is broad, encompassing roles as a cytokine, an immunotherapy agent, an antineoplastic, a haematopoiesis stimulant, and an adjuvant.[4] The drug is designated as a new molecular entity.[10]
From a manufacturing perspective, CDX-301 is produced using a modern bioprocess. This involves a proprietary Chinese hamster ovary (CHO) cell production line and a serum-free cell culture system.[1] This updated manufacturing process distinguishes it from earlier versions of rhuFlt3L and ensures a consistent, scalable supply for clinical development. The resulting protein has an identical amino acid sequence and comparable biological activity to the rhuFlt3L that was previously in clinical development decades ago.[1]
The clinical journey of rhuFlt3L began with Immunex Inc. (later acquired by Amgen), which first demonstrated its safety and biological activity in clinical studies involving over 150 healthy volunteers and 380 cancer patients.[3] Despite this initial work, clinical development was discontinued for over a decade.[1]
In 2009, Celldex Therapeutics in-licensed the program from Amgen and reinitiated its development under the new designation, CDX-301.[12] This decision to revive the program was not merely a continuation but a strategic repositioning. The original development of rhuFlt3L by Immunex occurred before the modern era of immuno-oncology. At that time, its primary value was perceived to be in stem cell mobilization. However, with the advent of checkpoint inhibitors and a deeper understanding of the central role of dendritic cells (DCs) in orchestrating anti-tumor immunity, the strategic value of a potent DC growth factor increased exponentially. Celldex's re-initiation of the program appears to have been a calculated move to leverage this new therapeutic context, betting that rhuFlt3L's true potential was not as a standalone agent but as a foundational priming agent capable of synergizing with the new wave of cancer immunotherapies. This hypothesis has been the guiding principle of its subsequent clinical development path.
The biological activity of CDX-301 is mediated through its specific interaction with a single receptor target: the Fms-like tyrosine kinase-3 (Flt3), also known by its cluster of differentiation designation, CD135.[1] This receptor is a hallmark of immature hematopoietic cells and is expressed on the surface of hematopoietic stem cells (HSCs), early hematopoietic progenitor cells (HPCs), immature thymocytes, and steady-state dendritic cells (DCs).[1]
The binding of CDX-301 (Flt3L) to the Flt3 receptor initiates a cascade of intracellular signaling events. This signaling directly promotes the proliferation, differentiation, and development of these Flt3-expressing cell populations within the bone marrow. A key consequence of this stimulation is the mobilization of these cells from the bone marrow into the peripheral circulation and their trafficking to lymphoid organs, thereby increasing their numbers systemically.[1] This targeted expansion of specific immune and progenitor cell populations forms the basis of CDX-301's therapeutic potential.
A primary and well-characterized effect of CDX-301 is the robust expansion and mobilization of CD34+ cells from the bone marrow into the peripheral blood.[1] The CD34+ cell population is a heterogeneous group that includes the multipotent HSCs and more committed HPCs responsible for generating all lineages of blood cells. This mechanism is the foundation for investigating CDX-301 as an agent for hematopoietic stem cell transplantation (HSCT).[3]
The goal in HSCT is to collect a sufficient number of HSCs from a donor (allogeneic) or patient (autologous) to reconstitute the entire hematopoietic system following high-dose chemotherapy or radiation. Early clinical studies with the original rhuFlt3L demonstrated that when administered in combination with standard mobilizing agents like granulocyte-colony stimulating factor (G-CSF) or granulocyte-macrophage colony-stimulating factor (GM-CSF), it resulted in higher and more sustained levels of circulating HPCs in the apheresis product compared to G-CSF alone.[13] This suggests that CDX-301 could improve the efficiency of stem cell collection, potentially reducing the number of apheresis sessions required and enabling successful collection in patients who mobilize poorly with standard agents.
While its effects on stem cells are significant, the most strategically important function of CDX-301 in the context of modern oncology is its "unique capacity" to dramatically increase the number of circulating dendritic cells.[4] DCs are the most potent antigen-presenting cells (APCs) in the immune system, responsible for capturing antigens (such as those from tumor cells), processing them, and presenting them to naive T cells to initiate an adaptive immune response.
CDX-301 administration leads to the effective peripheral expansion of multiple critical DC subsets. This includes key myeloid DC populations, such as BDCA-1+ (cDC2) and BDCA-3 high (cDC1) cells, as well as plasmacytoid DCs (pDCs), identified by the marker BDCA-2+.[1] The cDC1 subset, in particular, is considered crucial for cross-presentation of tumor antigens and the generation of effective cytotoxic CD8+ T cell responses.
This potent DC expansion is the central rationale for using CDX-301 in immuno-oncology combination therapies. The core hypothesis is that by administering CDX-301 prior to or concurrently with another therapy, the immune system can be "primed." This expansion creates a larger army of DCs ready to capture and present tumor antigens released by other treatments. This strategy is designed to amplify the therapeutic effect of:
Beyond its established roles in hematopoiesis and immunology, preclinical research has uncovered a novel mechanism of action for CDX-301 as a potential radioprotective and radiomitigatory agent. Studies in murine models demonstrated that administration of CDX-301 24 hours prior to total body irradiation could prevent radiation-induced injury and hematopoietic acute radiation syndrome.[22]
The underlying mechanism appears to involve the regulation of microRNAs (miRNAs), which are small non-coding RNAs that control gene expression. CDX-301 was found to prevent the widespread dysregulation of miRNA expression and biogenesis caused by radiation in both serum and spleen samples. Specifically, it inhibits the activation of the HOTAIR (HOX transcript antisense intergenic RNA) regulatory pathway, a key player in radiation-induced injury responses. Furthermore, CDX-301 attenuates radiation-induced inflammatory responses and restores the expression of genes within the MAP kinase and TGF-β signaling pathways that are typically altered by radiation exposure.[22]
This discovery suggests a potential dual benefit when CDX-301 is combined with radiotherapy in cancer treatment. The clinical trial in NSCLC (NCT02839265) was designed around the concept of an in situ vaccination, where CDX-301 enhances the immune response to radiation-killed tumor cells.[19] However, these preclinical findings imply a concurrent, complementary mechanism may be at play. In such a combination, CDX-301 could not only be making the radiation more immunogenically effective but also potentially safer by protecting surrounding healthy tissues from radiation-induced damage. If translatable to humans, this dual function—enhancing systemic tumor kill while mitigating local toxicity—would represent a significant therapeutic advantage.
The foundational human study for the modern CDX-301 program was NCT01465139, a Phase 1, open-label, non-randomized, single-group, dose-escalating trial conducted in 30 healthy volunteers.[1] The study was sponsored by Celldex Therapeutics and conducted at Rockefeller University.[3] Its primary objectives were to comprehensively evaluate the safety, tolerability, pharmacokinetic (PK), pharmacodynamic (PD), and immunologic profile of subcutaneously administered CDX-301.[1]
The study enrolled healthy male and female volunteers aged 18 to 55 and employed a standard 3+3 dose-escalation design across seven sequential cohorts. These cohorts were designed to explore various doses and schedules to characterize the biological response and identify optimal regimens for future trials. The dosing regimens tested were:
Participants were monitored for safety for 28 days following the last dose, with blood samples collected periodically to assess the drug's effects on circulating immune and hematopoietic cell populations.[23]
The study successfully demonstrated that CDX-301 induces a potent, dose- and duration-dependent expansion of its target cell populations in the peripheral blood.
A critical finding from the pharmacodynamic analysis was the superior efficacy of extended dosing duration compared to higher dose intensity. The cohort receiving 25 $μg/kg/day$ for 10 days produced significantly greater increases in both CD34+ cells and myeloid DCs than the cohort receiving a threefold higher dose (75 $μg/kg/day$) for only 5 days.[1] This observation indicated that sustained exposure to CDX-301, rather than a higher peak concentration, was the primary driver of maximal cell expansion. This finding was instrumental in guiding dose-schedule selection for subsequent clinical trials.
The table below summarizes the magnitude of cellular expansion observed with the optimal 10-day dosing regimen.
Table 1: Summary of Pharmacodynamic Effects of CDX-301 on Peripheral Blood Cell Counts (NCT01465139)
| Cell Type | Baseline Count (mean/µL) | Peak Count (mean/µL) | Fold Increase | Optimal Dosing Regimen |
|---|---|---|---|---|
| CD34+ HSC/HPC | 2.4 | 55.3 | 23-fold | 25 $μg/kg/day$ x 10 days |
| BDCA-1+ Myeloid DC | 10.2 | 1323 | 130-fold | 25 $μg/kg/day$ x 10 days |
| BDCA-3+ Myeloid DC | 0.99 | 47.5 | 48-fold | 25 $μg/kg/day$ x 10 days |
| Data compiled from Anandasabapathy et al., 2015.1 |
The pharmacokinetic profile of CDX-301 was evaluated through periodic measurement of its concentration in circulating blood, assessing parameters such as area under the curve (AUC), maximum concentration ($C_{max}$), time to maximum concentration ($T_{max}$), and half-life ($T_{1/2}$).[23] The terminal disposition half-life was found to be dose-dependent, ranging from approximately 12.3 hours at the 10 $μg/kg/day$ dose to 28 hours at the 75 $μg/kg/day$ dose.[13]
Across all seven cohorts and dosing regimens, CDX-301 was demonstrated to be safe and generally well-tolerated.[1] All 30 healthy volunteers completed the full dosing schedule and safety follow-up.[12]
The most frequently reported adverse events were mild and transient. These included Grade 1 lymphadenopathy, observed in six volunteers, all of whom were in the highest dose cohorts (25 $μg/kg$ and 75 $μg/kg$), and Grade 1 diarrhea, observed in two volunteers.[4]
There was a single serious adverse event (SAE) reported in the study: one volunteer with a remote history of community-acquired pneumonia (CAP) developed Grade 3 CAP on study day 12. The event was considered a possible dose-limiting toxicity (DLT), but the volunteer responded rapidly to antibiotics and recovered fully. No other infections, DLTs, or SAEs were observed throughout the trial.[1]
A key safety finding was the lack of immunogenicity. No anti-CDX-301 antibodies were detected in any of the volunteers through the end of the study follow-up period, mitigating concerns about the development of neutralizing antibodies that could compromise efficacy or cause adverse immune reactions upon repeated exposure.[12] The overall safety profile from this foundational study supported the continued development of CDX-301 in both transplantation and oncology settings.
The investigation of CDX-301 for use in HSCT is grounded in its potent and proven ability to mobilize large quantities of CD34+ hematopoietic stem and progenitor cells from the bone marrow into the peripheral blood.[1] The standard of care for mobilization has long been G-CSF, often with the addition of plerixafor for poor mobilizers.[26] The rationale for CDX-301 was that it could offer an alternative or complementary mechanism to enhance the yield and quality of the stem cell graft collected for transplantation.
This hypothesis was supported by preclinical data showing that CDX-301 could improve the reconstitution of blood cells post-transplant.[4] Furthermore, published studies with earlier forms of rhuFlt3L suggested additional benefits beyond simple cell numbers, including the potential to improve the speed of engraftment and, critically for allogeneic transplants, to reduce the risk of graft-versus-host disease (GvHD).[12] The mobilization of a broader range of immune progenitors, including dendritic cell precursors, was thought to contribute to a more balanced and rapid immune reconstitution, which could be particularly beneficial for older or more frail patients.[12]
Based on the strong rationale and positive Phase 1 data, Celldex initiated a pilot study (NCT02200380) in 2014 to evaluate CDX-301 in the clinical setting of allogeneic HSCT.[15] This was an open-label, multicenter, prospective study designed to assess CDX-301 as a mobilizing agent in healthy adult sibling donors who were HLA-matched to recipients with various hematological malignancies, including acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), myelodysplastic syndrome (MDS), and various lymphomas.[15]
The study's design involved two experimental arms to evaluate CDX-301 both as a standalone agent and in combination with an established mobilizing drug:
The primary objective was to assess the safety and tolerability of these regimens in healthy donors. A key secondary endpoint was to determine the efficacy of mobilization, measured by the proportion of donors from whom a sufficient yield of CD34+ cells could be collected for transplantation.[15]
Despite the initiation of this trial, its development appears to have been halted or deprioritized. The public record for NCT02200380 on ClinicalTrials.gov was last updated in April 2017, and no study results have been posted or published in the provided materials.[15] The absence of any subsequent press releases or publications from Celldex regarding the outcomes of this study is conspicuous.
This halt in the HSCT program likely reflects a strategic shift within Celldex. During the period following the trial's initiation in 2014, the company was concurrently launching and advancing multiple combination studies in immuno-oncology (e.g., melanoma trial NCT02129075, NSCLC trial NCT02839265).[18] By 2018, a corporate update following a pipeline revamp clarified that CDX-301's development would continue primarily through investigator-sponsored trials and as an internal combination agent, with no mention of a company-led HSCT program.[29] This timeline strongly suggests that a corporate decision was made to allocate resources away from the competitive and well-established HSCT mobilization market, where CDX-301 would compete with G-CSF and plerixafor, and toward the more proprietary and potentially higher-value field of immuno-oncology, where it could be used to synergistically enhance Celldex's other pipeline assets.
The central and most enduring strategic application for CDX-301 in oncology has been its use as an immune-priming agent. It is consistently referred to as a "dendritic cell growth factor" whose primary role is to expand the population of DCs available to orchestrate an anti-tumor immune response.[16] The underlying principle is that by administering CDX-301 before or during another cancer therapy, the resulting surge in DCs creates a more favorable microenvironment for generating a robust, tumor-specific T-cell attack. This strategy has been explored across a diverse range of malignancies and in combination with various therapeutic modalities.
One of the first and most definitive tests of the immune-priming hypothesis was the NCT02129075 trial, a Phase II, randomized, multicenter study in 60 patients with fully resected stage IIB-IV melanoma.[9] The study was conducted by the Cancer Immunotherapy Trials Network (CITN) and sponsored by the National Cancer Institute (NCI).[18]
Building on the priming concept, this study explored the synergy between CDX-301 and stereotactic body radiotherapy (SBRT) to create an in situ vaccine.[19] The trial enrolled 29 patients with advanced NSCLC who had multifocal active disease and had failed at least one prior line of systemic therapy; a vast majority (90%) had previously received immune checkpoint inhibitors.[19]
This Phase I trial represents a more complex, multi-layered immunotherapeutic strategy in patients with stage III/IV HER2-negative breast cancer, including triple-negative breast cancer.[20]
This Phase II trial, sponsored by Washington University School of Medicine, was designed to investigate the immunologic effects of combining CDX-301 with CDX-1140 in patients with surgically resectable pancreatic ductal adenocarcinoma.[33]
The most recent and mechanistically ambitious strategy involving CDX-301 is a Phase 1 trial investigating a four-part in situ immunomodulation regimen in patients with unresectable and metastatic solid tumors, including melanoma, sarcoma, and breast cancer.[21]
The following table provides a consolidated overview of the key clinical trials that have defined the role of CDX-301 in oncology.
Table 2: Overview of Key Clinical Trials for CDX-301 in Oncology
| Indication | Trial ID | Phase | Combination Agents | Key Outcome/Finding | Status |
|---|---|---|---|---|---|
| Melanoma | NCT02129075 | II | CDX-1401 (vaccine), poly-ICLC | Pre-treatment with CDX-301 significantly enhanced vaccine-specific T-cell responses. | Completed |
| NSCLC | NCT02839265 | I/II | Stereotactic Body Radiotherapy (SBRT) | Well-tolerated with high PFS4 (60%) and OS (55%) rates; induced systemic abscopal effects in 14-31% of patients. | Completed |
| Breast Cancer | NCI-2022-08956 | I | CDX-1140 (CD40 agonist), Pegylated Liposomal Doxorubicin (chemo) | Triplet combination designed to orchestrate antigen release, DC expansion, and DC activation. | Active, not recruiting |
| Pancreatic Cancer | NCT04536077 | II | CDX-1140 (CD40 agonist) | Designed to assess immunologic effects of the combination in resectable disease. | Terminated |
| Solid Tumors | NCT05673876 | I | Radiation, poly-ICLC, CDX-1140 | In situ immunomodulation with intratumoral CDX-301 to generate a localized and systemic immune response. | Recruiting |
The development trajectory of CDX-301 cannot be fully understood without considering the broader corporate strategy of its developer, Celldex Therapeutics. The company has undergone significant strategic shifts, particularly following pipeline setbacks with other lead candidates. In 2018, after the failure of its antibody-drug conjugate glembatumumab vedotin, Celldex announced a major strategic prioritization of its pipeline.[29]
This corporate revamp had direct consequences for CDX-301. While the company chose to focus its internal resources on advancing assets like the CD40 agonist CDX-1140 and the ErbB3 inhibitor CDX-3379, the plan for CDX-301 was explicitly altered. The company stated that its development would "continue externally through investigator-sponsored initiatives and internally through inclusion in combination studies".[29] This statement marked a formal shift in strategy, moving CDX-301 away from the path of a company-sponsored, standalone development program.
This repositioning has been reinforced by subsequent corporate communications. Celldex's pipeline updates from 2020 and beyond have increasingly focused on a new set of lead assets, particularly barzolvolimab (CDX-0159) and the bispecific antibody CDX-622, which are targeted at inflammatory, allergic, and autoimmune disorders.[39] In these more recent presentations of the company's pipeline, CDX-301 is no longer featured as a primary or strategic asset. The termination of the Washington University-sponsored pancreatic cancer trial (NCT04536077) in early 2024 for "corporate reasons" further solidifies the conclusion that CDX-301, while still active in some trials, is not a central pillar of Celldex's forward-looking strategy.[38]
An analysis of Celldex's public financial filings, such as its 10-K annual reports, provides further context. The 2019 10-K filing lists CDX-301 as a key program being developed in combination with CDX-1140.[46] However, by the time of the 2021 10-K filing, while CDX-301 is mentioned as being manufactured in-house for ongoing clinical trials, the overall strategic narrative of the company had clearly pivoted to its mast cell-focused programs.[47]
The collective evidence points to a deliberate repositioning of CDX-301. It is no longer being pursued as a lead asset with a direct path to commercialization as a standalone product. Instead, it has transitioned into the role of a valuable mechanistic tool or a "platform technology" within the Celldex portfolio. Its proven ability to expand dendritic cell populations makes it a powerful agent to deploy in combination studies to enhance the efficacy of other, more proprietary assets like CDX-1140. This is a capital-efficient strategy: it allows Celldex to leverage a biologically active and clinically validated asset to de-risk and potentially improve the outcomes of its primary drug candidates, without incurring the substantial costs of late-stage Phase 3 trials and commercialization for what is essentially a supportive agent.
CDX-301 is a molecule with unequivocally demonstrated and potent biological activity. The Phase 1 trial in healthy volunteers provided a clear and compelling dataset showing that it can safely and effectively expand key hematopoietic and immune cell populations, most notably dendritic cells. Its clinical utility as an immune-priming agent has been validated in multiple immuno-oncology settings, where it has successfully enhanced the immune response to both a cancer vaccine in melanoma and to stereotactic radiotherapy in NSCLC.
However, the strategic value of CDX-301 to its parent company has evolved in parallel with the broader immuno-oncology landscape. A decade ago, a potent cytokine might have been a viable candidate for standalone development. In today's complex therapeutic environment, dominated by checkpoint inhibitors, bispecific antibodies, and cellular therapies, its greatest value lies in synergy. The story of CDX-301 is not one of failure, but of finding its optimal, albeit supportive, role.
Its future development will likely continue along its current path. It will serve as a key component in investigator-sponsored trials exploring novel immunotherapy combinations, providing valuable scientific insights into immune modulation. Internally at Celldex, it remains a strategic tool that can be combined with next-generation proprietary molecules to enhance their activity, particularly in immunologically "cold" tumors that are resistant to other forms of immunotherapy. The lack of an Orphan Drug Designation from either the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA) further supports its positioning for broader, combination use rather than for a niche indication.[10] Ultimately, CDX-301 has been successfully repositioned from a potential product into a valuable instrument for building more effective cancer treatments.
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.