MedPath

FGI-101-1A6 Advanced Drug Monograph

Published:Jun 14, 2025

Generic Name

FGI-101-1A6

A Comprehensive Analysis of Emerging Therapeutic Agents: Hetrombopag, CD7 CAR-T Cells, SAR-446523, and DOC-1021

Part I: Hetrombopag (DB16184) – A Thrombopoietin Receptor Agonist for Hematologic Support

1.1. Executive Summary & Introduction

Hetrombopag, also identified by the development code SHR-8735 and marketed in China as Hengqu®, is an orally bioavailable, nonpeptide small molecule thrombopoietin receptor (TPO-R) agonist.[1] Developed by the Chinese pharmaceutical company Jiangsu Hengrui Medicine Co., Ltd., Hetrombopag is designed to address thrombocytopenia, a condition characterized by a dangerously low platelet count, by stimulating endogenous platelet production.[1]

The compound's mechanism of action is analogous to that of endogenous thrombopoietin, the primary physiological regulator of platelet production. Hetrombopag binds to and activates the TPO receptor (also known as c-Mpl) on the surface of megakaryocyte precursors in the bone marrow. This agonistic activity initiates intracellular signaling cascades, including the JAK/STAT pathway, which promotes the proliferation and differentiation of these precursors into mature, platelet-producing megakaryocytes, ultimately leading to an increase in circulating platelet counts.[1] Although its mechanism is similar to that of the approved TPO-RA eltrombopag, Hetrombopag was structurally modified with the goal of enhancing its platelet-raising potency while mitigating the potential for hepatic toxicity, a known concern with some agents in this class.[3]

Hetrombopag has achieved regulatory approval and is currently in clinical development for several indications. On June 16, 2021, it received its first approval from the China National Medical Products Administration (NMPA) for the treatment of primary immune thrombocytopenia (ITP) and severe aplastic anemia (SAA) in adults who have had an insufficient response to prior therapies.[3] Concurrently, it is under investigation for chemotherapy-induced thrombocytopenia (CIT), a significant unmet need in oncology, for which it has been granted Orphan Drug Designation by the U.S. Food and Drug Administration (FDA).[5]

1.2. Clinical Development Program: A Dual Focus on Chronic and Acute Thrombocytopenia

The clinical development of Hetrombopag has strategically targeted both chronic and acute forms of thrombocytopenia, establishing its efficacy in established markets while pursuing indications with high unmet need globally.

1.2.1. Chemotherapy-Induced Thrombocytopenia (CIT)

CIT represents a major dose-limiting toxicity of many myelosuppressive chemotherapy regimens. It significantly increases the risk of bleeding complications, frequently necessitates chemotherapy dose reductions or delays, and can consequently compromise the efficacy of cancer treatment and worsen patient prognosis.[6] The development of a safe and effective oral agent to manage CIT is therefore a critical priority in supportive cancer care.

A pivotal multicenter, randomized, double-blind, placebo-controlled Phase II/III study (NCT03976882) was conducted to evaluate the efficacy and safety of Hetrombopag for the management of CIT in patients with advanced solid tumors.[6]

  • Trial Design: The study enrolled 129 adult patients (18-75 years) with a confirmed diagnosis of a solid tumor who were receiving chemotherapy and had previously experienced thrombocytopenia that resulted in a delay of their subsequent chemotherapy cycle. Eligible patients had an ECOG performance status of 0-1 and adequate organ function. Key exclusion criteria included a platelet count below 30×109/L at screening, a history of hematologic diseases other than CIT, and serious bleeding symptoms.[8] Patients were randomized to receive either Hetrombopag or a matching placebo.[6]
  • Efficacy Endpoints and Results: The primary efficacy endpoint was the proportion of treatment responders, defined as patients who successfully avoided a delay or dose reduction in their next chemotherapy cycle due to thrombocytopenia. The results from the Phase II portion of the study, involving 59 treated patients, demonstrated a statistically significant and clinically meaningful benefit for Hetrombopag. The proportion of responders was 60.7% (17 of 28 patients) in the Hetrombopag arm, compared to just 12.9% (4 of 31 patients) in the placebo arm. This represents a 47.6% difference in proportion, with an odds ratio of 10.44 (95% CI: 2.82–38.65; p < 0.001), unequivocally establishing the superiority of Hetrombopag in this setting.[6]
  • Safety Profile: During the double-blind treatment period, treatment-related adverse events (TRAEs) were reported in 25.0% of patients receiving Hetrombopag, compared to 9.7% in the placebo group. The safety profile was deemed acceptable and manageable within the context of patients receiving chemotherapy.[6]

1.2.2. Primary Immune Thrombocytopenia (ITP) and Severe Aplastic Anemia (SAA)

Hetrombopag's initial regulatory success was achieved in chronic hematologic conditions. Its approval by the NMPA in June 2021 for adult patients with ITP and SAA was based on robust clinical trial data establishing its efficacy and safety as a second-line treatment.[3]

A key aspect of its clinical profile is its potential for an improved therapeutic window compared to existing TPO-RAs. A post-hoc analysis of a Phase III ITP trial provided compelling evidence in this regard. In this trial, patients who were initially randomized to placebo were subsequently switched to eltrombopag. Those who completed the 14-week eltrombopag treatment were then eligible to switch to a 24-week course of Hetrombopag. The analysis showed that Hetrombopag treatment led to high response rates, even in patients who had demonstrated a limited response to eltrombopag. Furthermore, the safety profile appeared more favorable, with TRAEs reported in 38.1% of patients during the Hetrombopag period compared to 50.8% during the eltrombopag period. No severe adverse events were attributed to Hetrombopag in this analysis.[3] While these findings are from a post-hoc analysis and require confirmation in prospective, head-to-head trials, they suggest that Hetrombopag may offer a meaningful clinical advantage over the established standard of care.

1.3. Regulatory and Corporate Landscape

The development and commercialization of Hetrombopag are being driven by Jiangsu Hengrui Medicine Co., Ltd., a prominent pharmaceutical company in China with a growing global presence.[1] The company has navigated a sophisticated regulatory strategy, balancing domestic market approval with a clear path toward international expansion.

  • China (NMPA): Hetrombopag was first approved in China on June 16, 2021, for use in adult patients with ITP who have shown an insufficient response to corticosteroids or immunoglobulins, and for adult patients with SAA who have had a poor response to immunosuppressive therapy.[3] This approval provides a foundational revenue stream in a large domestic market and validates the drug's clinical utility in established indications for the TPO-RA class.
  • United States (FDA): On June 13, 2022, the FDA granted Orphan Drug Designation to Hetrombopag for the treatment of chemotherapy-induced thrombocytopenia.[5] This designation is a critical milestone for its development in the U.S. market. It is granted to therapies for rare diseases affecting fewer than 200,000 people and provides significant incentives, including tax credits for clinical trials, exemption from user fees, and the potential for seven years of market exclusivity upon approval.

The pursuit of these dual regulatory pathways illustrates a well-considered corporate strategy. By first securing approval in its home market for indications where the therapeutic class is well-established (ITP and SAA), Jiangsu Hengrui has established a commercial foothold and generated real-world evidence for Hetrombopag. Simultaneously, the company has targeted a high-unmet-need indication, CIT, for global development. The compelling efficacy data from the NCT03976882 trial, combined with the strategic acquisition of FDA Orphan Drug Designation, provides a strong foundation for this global ambition and positions Hetrombopag as a potentially best-in-class agent.

1.4. Conclusion and Future Outlook

Hetrombopag has emerged as a potent and effective TPO-R agonist with a promising safety profile. Its demonstrated efficacy in CIT, a condition with few effective treatments, and its regulatory approval in China for ITP and SAA underscore its clinical value. The data suggesting a potentially superior efficacy and safety profile compared to eltrombopag, if borne out in further studies, could position Hetrombopag as a leading agent in its class. Future development will be focused on completing the pivotal trials in CIT to support regulatory submissions in Western markets and potentially expanding into other thrombocytopenic conditions.

Table 1.1: Summary of Efficacy and Safety from the Hetrombopag Phase II CIT Trial (NCT03976882) [6]

EndpointHetrombopag (n=28)Placebo (n=31)Odds Ratio (95% CI)p-value
Primary Endpoint: Treatment Responders (%)60.7%12.9%10.44 (2.82–38.65)<0.001
Key Safety: Any Treatment-Related Adverse Event (%)25.0%9.7%Not ApplicableNot Applicable

Part II: CD7-Targeted CAR T-Cell Therapy – A New Frontier in T-Cell Malignancies

2.1. Executive Summary & Therapeutic Rationale

Chimeric Antigen Receptor (CAR) T-cell therapy targeting the CD7 antigen is an innovative immunotherapeutic strategy poised to address the significant unmet medical need in T-cell malignancies, including T-cell acute lymphoblastic leukemia (T-ALL) and T-cell lymphoblastic lymphoma (T-LBL).[9] These diseases, particularly in the relapsed/refractory (R/R) setting, have historically poor prognoses with limited effective treatment options,.

The therapeutic target, CD7, is a 40 kDa transmembrane glycoprotein of the immunoglobulin superfamily. It is highly and consistently expressed on the surface of malignant T-cells and their precursors, as well as on natural killer (NK) cells.[9] This high level of expression, which is often maintained at relapse, makes CD7 an ideal candidate for targeted immunotherapy.

However, the development of CD7-targeted CAR T-cell therapy has been fundamentally challenged by the shared expression of CD7 on both malignant cells and the normal T-cells used to manufacture the CAR-T product. This shared antigenicity gives rise to two primary obstacles:

  1. Fratricide: The phenomenon where CD7-targeting CAR T-cells recognize the CD7 antigen on each other's surface and engage in self-killing. This severely limits the ability to expand the cells during manufacturing and compromises their persistence and efficacy in vivo.[11]
  2. T-cell Aplasia: The on-target, off-tumor killing of the patient's healthy, non-malignant T-cells, leading to profound and prolonged immunosuppression and a high risk of life-threatening opportunistic infections,,.

Addressing these challenges has been the central focus of research and development in this field, leading to several innovative engineering and clinical strategies.

2.2. Engineering Solutions to Overcome Fratricide

Fratricide is the primary technical barrier to the successful production and function of CD7 CAR T-cells. Several sophisticated strategies have been developed to circumvent this issue.

  • Gene Editing with CRISPR/Cas9: This is currently the leading strategy to prevent fratricide. It involves using the CRISPR/Cas9 gene-editing system to precisely disrupt or knock out the CD7 gene in the T-cells that will be engineered into CAR T-cells.[12] By deleting the CD7 gene, the resulting CAR T-cells no longer express the target antigen on their surface and are thus "invisible" to each other, preventing fratricide and allowing for robust expansion. Wugen's allogeneic product, WU-CART-007, employs this technology. In addition to deleting CD7, Wugen also knocks out the T-cell receptor alpha constant (TRAC) gene. This second edit is crucial for an allogeneic ("off-the-shelf") product as it prevents the CAR T-cells from recognizing the recipient's tissues as foreign, thereby mitigating the risk of Graft-versus-Host-Disease (GvHD) [12],.
  • Protein Expression Blockers (PEBL): An alternative to permanent gene deletion is the use of a protein expression blocker. In this approach, T-cells are engineered to co-express the anti-CD7 CAR along with a small protein designed to trap newly synthesized CD7 molecules within the endoplasmic reticulum. This prevents the CD7 protein from ever reaching the cell surface, effectively rendering the CAR T-cells fratricide-resistant without altering their genome.
  • Natural Selection and Antigen Masking: Other methods have also been explored. One approach involves isolating and expanding the small, naturally occurring population of CD7-negative T-cells from a donor's blood to use for manufacturing,. Another strategy, termed "naturally selected" CAR T-cells, relies on the observation that the CAR construct itself can sometimes bind to and sequester the CD7 antigen intracellularly, effectively masking it from recognition by other CAR T-cells. This allows for the preferential survival and expansion of a fratricide-resistant population without additional genetic manipulation.

2.3. The Allogeneic Approach and Managing T-Cell Aplasia

The source of T-cells and the management of on-target toxicities are critical considerations for the clinical application of CD7 CAR-T therapy.

  • Autologous vs. Allogeneic Products: Autologous CAR T-cell therapies, manufactured from the patient's own T-cells, are a standard approach in B-cell malignancies. However, in T-cell malignancies, this approach carries a significant risk of the starting material (apheresis product) being contaminated with circulating malignant T-cells. These malignant cells could potentially be transduced with the CAR construct, leading to a CAR-positive relapse that is resistant to the therapy itself.[11] Allogeneic CAR T-cell therapies, such as WU-CART-007, are manufactured from T-cells collected from healthy donors.[12] This "off-the-shelf" approach eliminates the risk of tumor contamination and provides immediate availability of the therapy, which is a major advantage for patients with aggressive, rapidly progressing diseases who cannot wait for a personalized product to be manufactured.
  • T-Cell Aplasia and the Role of Transplant: The successful killing of CD7-positive tumor cells invariably leads to the depletion of normal CD7-positive T-cells and NK cells, resulting in profound and prolonged T-cell aplasia,,. This severe immunosuppression leaves patients highly vulnerable to opportunistic viral and fungal infections. Unlike B-cell aplasia, which can be managed with intravenous immunoglobulin (IVIG) replacement, there is no simple replacement therapy for a depleted T-cell compartment,.

This clinical reality has established a clear therapeutic paradigm for CD7 CAR-T therapy: its primary role is to serve as a highly effective "bridge-to-transplant". The therapy is exceptionally potent at inducing deep remissions, even in patients who are refractory to multiple lines of chemotherapy.[12] This remission provides a critical window of opportunity for the patient to proceed to a potentially curative allogeneic hematopoietic stem cell transplantation (allo-HSCT). The transplant then serves a dual purpose: it provides a definitive anti-leukemic therapy while simultaneously reconstituting the patient's entire immune system, including the T-cell population, thereby resolving the CAR T-cell-induced aplasia,,.

2.4. Clinical Landscape: Focus on Wugen's WU-CART-007

Wugen, Inc., a clinical-stage US biotechnology company, is a key developer in this space with its allogeneic candidate, WU-CART-007.[12]

  • Phase 1/2 Trial (NCT04984356): This global study has produced highly encouraging results in heavily pretreated patients with R/R T-ALL/LBL.
  • Efficacy: At the recommended Phase 2 dose (RP2D), WU-CART-007 achieved an overall response rate (ORR) of 91% and a composite complete remission (CRc) rate of 73%.[12] These response rates are remarkable in a patient population with very limited options.
  • Safety Profile: Data presented at the EHA 2024 Congress from the Phase 2 portion of the study (26 patients) showed that the therapy was associated with a manageable safety profile. Cytokine Release Syndrome (CRS) was observed in 88.5% of patients, with 19.2% experiencing Grade 3 or 4 events. Immune Effector Cell-Associated Neurotoxicity Syndrome (ICANS) was infrequent and low-grade, with only Grade 1 events reported in 7.7% of patients.[18] These toxicities were manageable with standard interventions.[17]
  • Pivotal Phase 2 Trial (T-RRex, NCT06514794): Based on the strength of the Phase 1/2 data, Wugen has initiated a global, single-arm, pivotal Phase 2 study. This trial will evaluate the efficacy and safety of WU-CART-007 in both pediatric and adult patients with R/R T-ALL/LBL. The dosing of the first patients in this pivotal trial was announced in March 2025, marking a significant step toward potential regulatory approval [[16]],, [[12]].
  • Regulatory Status: The potential of WU-CART-007 has been recognized by global regulatory agencies. It has received multiple designations from the US FDA intended to accelerate development, including Regenerative Medicine Advanced Therapy (RMAT), Fast Track, Orphan Drug, and Rare Pediatric Disease designations. In the European Union, it has been granted Priority Medicines (PRIME) designation by the EMA.[12]

Table 2.1: Comparative Overview of Key CD7 CAR-T Therapies

Therapy Name (Developer)TargetCell SourceFratricide Mitigation StrategyKey IndicationHighest PhaseReported ORR/CRc (%)Reported Grade ≥3 CRS/ICANS (%)
WU-CART-007 (Wugen) 12CD7AllogeneicCRISPR/Cas9 knockout of CD7 and TRACR/R T-ALL/LBLPhase 2 (Pivotal)91 / 73CRS: 19.2% / ICANS: 0%
RD13-01 (Zhejiang University)CD7AllogeneicGenetic modifications (unspecified)R/R T-cell malignanciesPhase 181.8 / 63.6CRS: 0% / ICANS: 0%
CRIMSON (Texas Children's) 10CD7AutologousCD28 co-stimulation (fratricide not explicitly addressed)R/R T-ALL/LBLPhase 1/2Not ReportedNot Reported
NS7CAR (Hebei Senlang Biotech)CD7Autologous/AllogeneicNatural Selection / Antigen MaskingR/R T-ALL/LBLPhase 190 / 90CRS: 25% / ICANS: 0%

Part III: SAR-446523 – An ADCC-Enhanced Monoclonal Antibody for Multiple Myeloma

3.1. Executive Summary & Therapeutic Rationale

SAR-446523 is an investigational, subcutaneously administered therapeutic being developed by Sanofi for patients with relapsed/refractory multiple myeloma (RRMM).[19] It is a monoclonal antibody designed to leverage a distinct immunological mechanism to target and eliminate malignant plasma cells.

The therapeutic target for SAR-446523 is the G-protein coupled receptor, class C, group 5, member D (GPRC5D). GPRC5D is an orphan receptor that has garnered significant interest as a target for MM immunotherapy due to its highly favorable expression profile. It is highly and selectively expressed on the surface of malignant plasma cells while its expression in normal tissues is largely restricted to hard keratinized tissues, such as hair follicles, with minimal to no expression on healthy hematopoietic cells [19],,,,,. This tumor-restricted expression provides a compelling therapeutic window, allowing for targeted destruction of myeloma cells while potentially sparing healthy tissues and minimizing off-target toxicities.

The mechanism of action (MOA) of SAR-446523 is based on enhanced Antibody-Dependent Cellular Cytotoxicity (ADCC). It is an IgG1-based monoclonal antibody that has been engineered in its Fc (fragment crystallizable) domain to increase its binding affinity to Fc receptors on immune effector cells.[19] The intended mechanism involves SAR-446523 binding to GPRC5D on the surface of myeloma cells. The engineered Fc region then acts as a beacon, recruiting and potently activating immune effector cells—primarily Natural Killer (NK) cells—which subsequently release cytotoxic granules to induce lysis and death of the myeloma cell [19],,,,,,,,.

3.2. Preclinical Profile and Differentiated Mechanism

The preclinical data for SAR-446523, presented at the American Association for Cancer Research (AACR) Annual Meeting in 2024, provide a strong rationale for its clinical development and highlight its differentiation from other GPRC5D-targeting agents.[25]

  • High Affinity and Potent ADCC: The AACR abstract (2727) reported that SAR-446523 demonstrated high affinity for the GPRC5D target. The engineered Fc domain also conferred increased affinity for both high and low affinity variants of the CD16a receptor (FCGR3A-158V and 158F), which is the key activating receptor on NK cells. This enhanced binding translated into potent ADCC activity in vitro. SAR-446523 induced strong cytotoxic effects in the picomolar range against a panel of MM cell lines, and this potency was observed regardless of whether the cell lines had low, medium, or high levels of GPRC5D expression. The cytotoxic activity was also reported to be higher than that of a clinical benchmark compound [25],.
  • Favorable Cytokine Release Profile: A crucial finding from the preclinical studies was the very low level of pro-inflammatory cytokine release (IL-6, TNFα, and IFNγ) associated with SAR-446523's anti-tumor activity. This was significantly lower than the cytokine release observed with a CD3xGPRC5D T-cell engager in a comparable in vitro assay.[25]
  • In Vivo Efficacy: The in vivo anti-tumor activity was confirmed in a humanized mouse model bearing disseminated MM cells, where SAR-446523 led to robust anti-tumoral efficacy and significantly improved mouse survival.[25]

Sanofi's decision to develop an ADCC-enhanced antibody against GPRC5D, rather than a T-cell engaging bispecific antibody like the approved drug talquetamab, appears to be a deliberate strategic choice rooted in a differentiated risk-benefit profile. The GPRC5D target, while promising, is not entirely tumor-specific, with on-target expression in keratinized tissues leading to notable skin, nail, and oral toxicities with T-cell engagers,,. Furthermore, T-cell redirecting therapies are commonly associated with high rates of Cytokine Release Syndrome (CRS). The preclinical data for SAR-446523 directly address this liability, demonstrating potent tumor killing with a minimal cytokine release signature. By relying on NK cell-mediated ADCC, a mechanism known to be less inflammatory than broad T-cell activation, Sanofi is positioning SAR-446523 as a potentially safer and more tolerable agent. This improved safety profile could be a significant competitive advantage, potentially allowing for use in combination regimens or in patient populations where the toxicities of T-cell engagers might be prohibitive. The clinical trial's exclusion of patients with prior NK-cell engaging therapy further emphasizes this distinct mechanistic focus.[22]

3.3. Clinical Development Program: A First-in-Human Study (NCT06630806)

SAR-446523 is currently being evaluated in a Phase 1/2, open-label, first-in-human clinical trial (NCT06630806). The study is actively recruiting adult participants with RRMM across multiple sites in the United States, Canada, Australia, and Italy [23],,, [[22]],,, [[16],,,,,,,.

The trial employs a two-part design to systematically evaluate the drug's safety and determine its optimal dose for future studies:

  • Part A (Dose Escalation): This non-randomized part of the study is designed to establish the safety profile of SAR-446523 and to determine the maximum administered dose (MAD) and maximum tolerated dose (MTD). Up to six escalating dose levels will be explored. The inclusion criteria for this part are designed to enroll a heavily pre-treated population. Patients must have received at least three prior lines of antimyeloma therapy, including an immunomodulator (IMiD), a proteasome inhibitor (PI), and an anti-CD38 monoclonal antibody. Importantly, in this initial safety-finding stage, prior exposure to both anti-GPRC5D and anti-BCMA therapies is permitted.[22]
  • Part B (Dose Optimization): Following the determination of a safe dose range from Part A, this part of the trial will randomize participants in a 1:1 ratio to one of two chosen dose regimens. The goal of Part B is to determine the optimal recommended Phase 2 dose (RP2D). The inclusion criteria for Part B are more stringent to ensure a cleaner efficacy signal. Patients must have received at least three prior lines of therapy, including an IMiD, a PI, an anti-CD38 mAb, and, critically, an anti-B Cell Maturation Antigen (anti-BCMA) agent. A key distinction from Part A is that patients with prior exposure to any anti-GPRC5D therapy are excluded from this part of the study.[22]

This staggered trial design reflects a sophisticated and risk-mitigating approach to early-phase clinical development. Allowing patients with prior GPRC5D therapy into the initial dose-escalation phase enables Sanofi to gather safety data in the most advanced and refractory patient population, where the unmet need is highest. However, for the crucial dose-optimization phase, these patients are excluded. This is a methodologically sound decision, as prior targeting of an antigen can lead to its downregulation or loss on tumor cells, which would confound the assessment of dose-response and could lead to the selection of a suboptimal dose for the intended GPRC5D-naive patient population. This design maximizes the chances of identifying a truly effective and safe dose for future pivotal trials.

3.4. Regulatory and Corporate Landscape

  • Developer: SAR-446523 is being developed by Sanofi, a global pharmaceutical company with a significant presence in oncology.[23]
  • Regulatory Status: The therapeutic potential of SAR-446523 has been recognized by the U.S. FDA, which has granted it Orphan Drug Designation for the treatment of multiple myeloma,. This designation provides incentives to support the development of drugs for rare diseases and underscores the unmet need in RRMM.

Part IV: DOC-1021 (Dubodencel) – A Personalized Dendritic Cell Vaccine for Solid Tumors

4.1. Executive Summary & Therapeutic Concept

DOC-1021, also known as Dubodencel, is a first-in-class, personalized, autologous dendritic cell (DC) vaccine platform under development by Diakonos Oncology Corp [27], [[29]],. The therapy is being investigated for the treatment of highly aggressive solid tumors, with glioblastoma (GBM) serving as the lead indication [[38]],.

The core innovation of DOC-1021 lies in its unique "double-loading" mechanism of action, which is designed to mimic a viral infection to elicit a powerful and comprehensive anti-tumor immune response [27],. The manufacturing process is entirely autologous and involves several steps:

  1. A patient's peripheral blood mononuclear cells (PBMCs) are collected via leukapheresis and are matured ex vivo into dendritic cells, the most potent antigen-presenting cells of the immune system.[28]
  2. A sample of the patient's own tumor is resected and used to create both a tumor lysate and amplified tumor-derived mRNA.[27]
  3. The dendritic cells are then "double-loaded" with both the tumor lysate (which contains a broad array of protein antigens for presentation on MHC Class II molecules) and the tumor mRNA (which is translated within the DC to provide endogenous antigens for presentation on MHC Class I molecules).[27]

This dual presentation of the complete cancer antigen repertoire is hypothesized to trigger a synergistic and exceptionally powerful, TH1-polarized cytotoxic T-cell response. This approach avoids the need for genetic modification of immune cells or the administration of high-dose interleukin-2 (IL-2).[27] To maximize the immune response, the vaccine is administered via ultrasound-guided injection near the tumor-draining lymph nodes (e.g., the deep cervical lymph nodes for GBM), which is intended to facilitate the trafficking of activated T-cells to the tumor site.[28]

4.2. Clinical Program in Glioblastoma (GBM)

The clinical development of DOC-1021 has focused initially on GBM, one of the most challenging solid tumors to treat.

4.2.1. Phase 1 Trial (NCT04552886)

A single-arm, open-label, first-in-human trial was conducted to evaluate the safety and feasibility of DOC-1021 in both newly diagnosed and recurrent GBM patients following standard of care (SOC) treatment [[38]], [[39]], [[30]], [[29]], [[39]], [[35]], [[16], [[30]], [[30].

  • Efficacy Results: Data presented at the 2025 ASCO Annual Meeting showed highly encouraging survival outcomes. Among 16 newly diagnosed patients, the 12-month overall survival (OS) rate was 88%. This result is particularly striking given that 94% of these patients had tumors with an unmethylated MGMT promoter, a biomarker associated with poor prognosis and resistance to standard temozolomide chemotherapy. The expected 12-month OS for this patient population with SOC is approximately 60%. The Kaplan-Meier projected median OS for the treated group is 19.7 months, compared to a historical control of 12.7 months for unmethylated GBM.[30]
  • Safety Profile: The therapy demonstrated an excellent safety profile. The most common adverse events were mild flu-like symptoms and local injection-site reactions. Importantly, no dose-limiting toxicities were reported across the four dose levels tested.[30]

4.2.2. Immunological Correlates and the Insight of Pseudo-progression

The Phase 1 trial provided crucial data linking the vaccine's mechanism to clinical outcomes.

  • Immune Response: Post-vaccination analyses confirmed the intended biological activity. There was a significant expansion of both CD4+ and CD8+ central memory T-cell compartments in the peripheral blood of treated patients. Furthermore, spatial transcriptomics analysis of post-treatment tumor tissue from three patients revealed the formation of "immune triads"—dense clusters of activated CD4+ T-cells, CD8+ T-cells, and migratory microglial cells—within the tumor microenvironment. These inflammatory foci were not present in pre-treatment samples, and regulatory T-cells (Tregs) appeared to be excluded from these areas post-treatment, suggesting a potent and targeted anti-tumor immune response.[30]
  • Pseudo-progression as a Biomarker of Response: A critical observation from the trial was the phenomenon of pseudo-progression. In immunotherapy, an influx of immune cells into the tumor can cause inflammation and edema that mimics tumor growth on standard MRI scans. In the DOC-1021 trial, eight patients showed this pattern of increased T1-weighted contrast enhancement on MRI early after treatment, despite being clinically stable. A striking divergence in outcomes was observed based on how this was managed. The median OS for the eight patients who were re-operated upon based on these imaging findings was 15.1 months. In contrast, for the eight patients who were observed without surgical intervention, the median OS has not yet been reached, and three of these patients remain alive and clinically well with no tumor visible on imaging at 22-33 months of follow-up.[30] This dramatic difference in survival provides strong clinical validation that this imaging finding represents a beneficial, immune-reactive microenvironment rather than true tumor progression. This insight is so significant that it has directly informed the protocol for the subsequent Phase 2 trial, which now includes criteria to avoid premature re-operation based on imaging findings that may represent pseudo-progression.[30]

4.2.3. Phase 2 Trial (NCT06805305)

Building on the promising Phase 1 data, Diakonos has initiated a randomized Phase 2 trial. This study will formally compare the efficacy of DOC-1021 combined with SOC versus SOC alone in 135 patients with newly diagnosed GBM [[38]],,,,,,,,.

4.3. Pipeline Expansion and Platform Potential

Diakonos Oncology is leveraging the DOC1021 platform technology across multiple difficult-to-treat solid tumors.

  • Pancreatic Cancer: A Phase 1 trial (NCT04157127) is currently enrolling patients with pancreatic ductal adenocarcinoma (PDAC) at Baylor College of Medicine. Early clinical results from this study have reportedly been encouraging and exceeded expectations,,,,, [[32]], [[31]].
  • Refractory Melanoma: The company also plans to initiate a study in refractory melanoma, a setting where immunotherapy has a more established role but where unmet need remains for patients who do not respond to checkpoint inhibitors,,,, [[34]].
  • Manufacturing: As an autologous cell therapy, manufacturing is a key consideration. Diakonos is actively engaged in process development with a contract development and manufacturing organization (CDMO) in Houston to enhance the efficiency of the manufacturing process, aiming to reduce the required amount of tumor tissue and leukocytes from the patient's apheresis, [[22],.

4.4. Regulatory and Corporate Landscape

  • Developer: Diakonos Oncology Corp. is a privately held, clinical-stage biotechnology company headquartered in Houston, Texas [[27]],, [[33]].
  • Regulatory Status: The DOC1021 platform has received significant validation and support from the FDA. It has been granted Fast Track Designation for both its glioblastoma and pancreatic cancer programs, as well as Orphan Drug Designation for malignant glioma, which includes GBM.[35] These designations are intended to facilitate and expedite the development and review of drugs that address serious conditions and fill an unmet medical need.

Part V: SAR-446523 – An ADCC-Enhanced Monoclonal Antibody for Multiple Myeloma

5.1. Executive Summary & Therapeutic Rationale

SAR-446523 is an investigational, subcutaneously administered therapeutic being developed by Sanofi for patients with relapsed/refractory multiple myeloma (RRMM).[19] It is a monoclonal antibody designed to leverage a distinct immunological mechanism to target and eliminate malignant plasma cells.

The therapeutic target for SAR-446523 is the G-protein coupled receptor, class C, group 5, member D (GPRC5D). GPRC5D is an orphan receptor that has garnered significant interest as a target for MM immunotherapy due to its highly favorable expression profile. It is highly and selectively expressed on the surface of malignant plasma cells while its expression in normal tissues is largely restricted to hard keratinized tissues, such as hair follicles, with minimal to no expression on healthy hematopoietic cells [19],,,,,. This tumor-restricted expression provides a compelling therapeutic window, allowing for targeted destruction of myeloma cells while potentially sparing healthy tissues and minimizing off-target toxicities.

The mechanism of action (MOA) of SAR-446523 is based on enhanced Antibody-Dependent Cellular Cytotoxicity (ADCC). It is an IgG1-based monoclonal antibody that has been engineered in its Fc (fragment crystallizable) domain to increase its binding affinity to Fc receptors on immune effector cells.[19] The intended mechanism involves SAR-446523 binding to GPRC5D on the surface of myeloma cells. The engineered Fc region then acts as a beacon, recruiting and potently activating immune effector cells—primarily Natural Killer (NK) cells—which subsequently release cytotoxic granules to induce lysis and death of the myeloma cell [19],,,,,,,,.

5.2. Preclinical Profile and Differentiated Mechanism

The preclinical data for SAR-446523, presented at the American Association for Cancer Research (AACR) Annual Meeting in 2024, provide a strong rationale for its clinical development and highlight its differentiation from other GPRC5D-targeting agents.[25]

  • High Affinity and Potent ADCC: The AACR abstract (2727) reported that SAR-446523 demonstrated high affinity for the GPRC5D target. The engineered Fc domain also conferred increased affinity for both high and low affinity variants of the CD16a receptor (FCGR3A-158V and 158F), which is the key activating receptor on NK cells. This enhanced binding translated into potent ADCC activity in vitro. SAR-446523 induced strong cytotoxic effects in the picomolar range against a panel of MM cell lines, and this potency was observed regardless of whether the cell lines had low, medium, or high levels of GPRC5D expression. The cytotoxic activity was also reported to be higher than that of a clinical benchmark compound [25],.
  • Favorable Cytokine Release Profile: A crucial finding from the preclinical studies was the very low level of pro-inflammatory cytokine release (IL-6, TNFα, and IFNγ) associated with SAR-446523's anti-tumor activity. This was significantly lower than the cytokine release observed with a CD3xGPRC5D T-cell engager in a comparable in vitro assay.[25]
  • In Vivo Efficacy: The in vivo anti-tumor activity was confirmed in a humanized mouse model bearing disseminated MM cells, where SAR-446523 led to robust anti-tumoral efficacy and significantly improved mouse survival.[25]

Sanofi's decision to develop an ADCC-enhanced antibody against GPRC5D, rather than a T-cell engaging bispecific antibody like the approved drug talquetamab, appears to be a deliberate strategic choice rooted in a differentiated risk-benefit profile. The GPRC5D target, while promising, is not entirely tumor-specific, with on-target expression in keratinized tissues leading to notable skin, nail, and oral toxicities with T-cell engagers,,. Furthermore, T-cell redirecting therapies are commonly associated with high rates of Cytokine Release Syndrome (CRS). The preclinical data for SAR-446523 directly address this liability, demonstrating potent tumor killing with a minimal cytokine release signature. By relying on NK cell-mediated ADCC, a mechanism known to be less inflammatory than broad T-cell activation, Sanofi is positioning SAR-446523 as a potentially safer and more tolerable agent. This improved safety profile could be a significant competitive advantage, potentially allowing for use in combination regimens or in patient populations where the toxicities of T-cell engagers might be prohibitive. The clinical trial's exclusion of patients with prior NK-cell engaging therapy further emphasizes this distinct mechanistic focus.[22]

5.3. Clinical Development Program: A First-in-Human Study (NCT06630806)

SAR-446523 is currently being evaluated in a Phase 1/2, open-label, first-in-human clinical trial (NCT06630806). The study is actively recruiting adult participants with RRMM across multiple sites in the United States, Canada, Australia, and Italy [23],,, [[22]],,, [[16],,,,,,,.

The trial employs a two-part design to systematically evaluate the drug's safety and determine its optimal dose for future studies:

  • Part A (Dose Escalation): This non-randomized part of the study is designed to establish the safety profile of SAR-446523 and to determine the maximum administered dose (MAD) and maximum tolerated dose (MTD). Up to six escalating dose levels will be explored. The inclusion criteria for this part are designed to enroll a heavily pre-treated population. Patients must have received at least three prior lines of antimyeloma therapy, including an immunomodulator (IMiD), a proteasome inhibitor (PI), and an anti-CD38 monoclonal antibody. Importantly, in this initial safety-finding stage, prior exposure to both anti-GPRC5D and anti-BCMA therapies is permitted.[22]
  • Part B (Dose Optimization): Following the determination of a safe dose range from Part A, this part of the trial will randomize participants in a 1:1 ratio to one of two chosen dose regimens. The goal of Part B is to determine the optimal recommended Phase 2 dose (RP2D). The inclusion criteria for Part B are more stringent to ensure a cleaner efficacy signal. Patients must have received at least three prior lines of therapy, including an IMiD, a PI, an anti-CD38 mAb, and, critically, an anti-B Cell Maturation Antigen (anti-BCMA) agent. A key distinction from Part A is that patients with prior exposure to any anti-GPRC5D therapy are excluded from this part of the study.[22]

This staggered trial design reflects a sophisticated and risk-mitigating approach to early-phase clinical development. Allowing patients with prior GPRC5D therapy into the initial dose-escalation phase enables Sanofi to gather safety data in the most advanced and refractory patient population, where the unmet need is highest. However, for the crucial dose-optimization phase, these patients are excluded. This is a methodologically sound decision, as prior targeting of an antigen can lead to its downregulation or loss on tumor cells, which would confound the assessment of dose-response and could lead to the selection of a suboptimal dose for the intended GPRC5D-naive patient population. This design maximizes the chances of identifying a truly effective and safe dose for future pivotal trials.

5.4. Regulatory and Corporate Landscape

  • Developer: SAR-446523 is being developed by Sanofi, a global pharmaceutical company with a significant presence in oncology.[23]
  • Regulatory Status: The therapeutic potential of SAR-446523 has been recognized by the U.S. FDA, which has granted it Orphan Drug Designation for the treatment of multiple myeloma,. This designation provides incentives to support the development of drugs for rare diseases and underscores the unmet need in RRMM.

Table 5.1: Key Preclinical Characteristics of SAR-446523 (from AACR 2024 Abstract) [25]

CharacteristicFinding
Target Binding AffinityHigh affinity for human GPRC5D and increased affinity for both high and low affinity variants of the CD16a receptor.
ADCC PotencyStrong cytotoxic potency (picomolar range) against MM cell lines, regardless of GPRC5D expression level.
Comparative CytotoxicityInduced higher cytotoxic activity compared to a clinical benchmark compound.
In Vitro Cytokine ReleaseVery low release of IL-6, TNFα, and IFNγ; much lower compared to a CD3xGPRC5D T-cell engager.
In Vivo Antitumor EfficacyLed to robust antitumoral efficacy and significantly improved survival in a NK humanized mouse model of disseminated MM.

Part VI: DOC-1021 (Dubodencel) – A Personalized Dendritic Cell Vaccine for Solid Tumors

6.1. Executive Summary & Therapeutic Concept

DOC-1021, also known as Dubodencel, is a first-in-class, personalized, autologous dendritic cell (DC) vaccine platform under development by Diakonos Oncology Corp [27], [[29]],. The therapy is being investigated for the treatment of highly aggressive solid tumors, with glioblastoma (GBM) serving as the lead indication [[38]],.

The core innovation of DOC-1021 lies in its unique "double-loading" mechanism of action, which is designed to mimic a viral infection to elicit a powerful and comprehensive anti-tumor immune response [27],. The manufacturing process is entirely autologous and involves several steps:

  1. A patient's peripheral blood mononuclear cells (PBMCs) are collected via leukapheresis and are matured ex vivo into dendritic cells, the most potent antigen-presenting cells of the immune system.[28]
  2. A sample of the patient's own tumor is resected and used to create both a tumor lysate and amplified tumor-derived mRNA.[27]
  3. The dendritic cells are then "double-loaded" with both the tumor lysate (which contains a broad array of protein antigens for presentation on MHC Class II molecules) and the tumor mRNA (which is translated within the DC to provide endogenous antigens for presentation on MHC Class I molecules).[27]

This dual presentation of the complete cancer antigen repertoire is hypothesized to trigger a synergistic and exceptionally powerful, TH1-polarized cytotoxic T-cell response. This approach avoids the need for genetic modification of immune cells or the administration of high-dose interleukin-2 (IL-2).[27] To maximize the immune response, the vaccine is administered via ultrasound-guided injection near the tumor-draining lymph nodes (e.g., the deep cervical lymph nodes for GBM), which is intended to facilitate the trafficking of activated T-cells to the tumor site.[28]

6.2. Clinical Program in Glioblastoma (GBM)

The clinical development of DOC-1021 has focused initially on GBM, one of the most challenging solid tumors to treat.

6.2.1. Phase 1 Trial (NCT04552886)

A single-arm, open-label, first-in-human trial was conducted to evaluate the safety and feasibility of DOC-1021 in both newly diagnosed and recurrent GBM patients following standard of care (SOC) treatment [[38]], [[39]], [[30]], [[29]], [[39]], [[35]], [[16], [[30]], [[30].

  • Efficacy Results: Data presented at the 2025 ASCO Annual Meeting showed highly encouraging survival outcomes. Among 16 newly diagnosed patients, the 12-month overall survival (OS) rate was 88%. This result is particularly striking given that 94% of these patients had tumors with an unmethylated MGMT promoter, a biomarker associated with poor prognosis and resistance to standard temozolomide chemotherapy. The expected 12-month OS for this patient population with SOC is approximately 60%. The Kaplan-Meier projected median OS for the treated group is 19.7 months, compared to a historical control of 12.7 months for unmethylated GBM.[30]
  • Safety Profile: The therapy demonstrated an excellent safety profile. The most common adverse events were mild flu-like symptoms and local injection-site reactions. Importantly, no dose-limiting toxicities were reported across the four dose levels tested.[30]

6.2.2. Immunological Correlates and the Insight of Pseudo-progression

The Phase 1 trial provided crucial data linking the vaccine's mechanism to clinical outcomes.

  • Immune Response: Post-vaccination analyses confirmed the intended biological activity. There was a significant expansion of both CD4+ and CD8+ central memory T-cell compartments in the peripheral blood of treated patients. Furthermore, spatial transcriptomics analysis of post-treatment tumor tissue from three patients revealed the formation of "immune triads"—dense clusters of activated CD4+ T-cells, CD8+ T-cells, and migratory microglial cells—within the tumor microenvironment. These inflammatory foci were not present in pre-treatment samples, and regulatory T-cells (Tregs) appeared to be excluded from these areas post-treatment, suggesting a potent and targeted anti-tumor immune response.[30]
  • Pseudo-progression as a Biomarker of Response: A critical observation from the trial was the phenomenon of pseudo-progression. In immunotherapy, an influx of immune cells into the tumor can cause inflammation and edema that mimics tumor growth on standard MRI scans. In the DOC-1021 trial, eight patients showed this pattern of increased T1-weighted contrast enhancement on MRI early after treatment, despite being clinically stable. A striking divergence in outcomes was observed based on how this was managed. The median OS for the eight patients who were re-operated upon based on these imaging findings was 15.1 months. In contrast, for the eight patients who were observed without surgical intervention, the median OS has not yet been reached, and three of these patients remain alive and clinically well with no tumor visible on imaging at 22-33 months of follow-up.[30] This dramatic difference in survival provides strong clinical validation that this imaging finding represents a beneficial, immune-reactive microenvironment rather than true tumor progression. This insight is so significant that it has directly informed the protocol for the subsequent Phase 2 trial, which now includes criteria to avoid premature re-operation based on imaging findings that may represent pseudo-progression.[30]

6.2.3. Phase 2 Trial (NCT06805305)

Building on the promising Phase 1 data, Diakonos has initiated a randomized Phase 2 trial. This study will formally compare the efficacy of DOC-1021 combined with SOC versus SOC alone in 135 patients with newly diagnosed GBM [[38]],,,,,,,,.

6.3. Pipeline Expansion and Platform Potential

Diakonos Oncology is leveraging the DOC1021 platform technology across multiple difficult-to-treat solid tumors.

  • Pancreatic Cancer: A Phase 1 trial (NCT04157127) is currently enrolling patients with pancreatic ductal adenocarcinoma (PDAC) at Baylor College of Medicine. Early clinical results from this study have reportedly been encouraging and exceeded expectations,,,,, [[32]], [[31]].
  • Refractory Melanoma: The company also plans to initiate a study in refractory melanoma, a setting where immunotherapy has a more established role but where unmet need remains for patients who do not respond to checkpoint inhibitors,,,, [[34]].
  • Manufacturing: As an autologous cell therapy, manufacturing is a key consideration. Diakonos is actively engaged in process development with a contract development and manufacturing organization (CDMO) in Houston to enhance the efficiency of the manufacturing process, aiming to reduce the required amount of tumor tissue and leukocytes from the patient's apheresis, [[22],.

6.4. Regulatory and Corporate Landscape

  • Developer: Diakonos Oncology Corp. is a privately held, clinical-stage biotechnology company headquartered in Houston, Texas [[27]],, [[33]].
  • Regulatory Status: The DOC1021 platform has received significant validation and support from the FDA. It has been granted Fast Track Designation for both its glioblastoma and pancreatic cancer programs, as well as Orphan Drug Designation for malignant glioma, which includes GBM.[35] These designations are intended to facilitate and expedite the development and review of drugs that address serious conditions and fill an unmet medical need.

Table 6.1: Summary of Key Outcomes from the DOC-1021 Phase 1 GBM Trial (NCT04552886) [30]

EndpointResultComparison/Context
Patient Population (newly diagnosed)n=16 (94% MGMT unmethylated)High-risk, poor-prognosis population
12-Month Overall Survival (OS) Rate88%Expected ~60% with standard of care
KM-Projected Median OS19.7 months12.7 months for historical unmethylated controls
Immune ResponseSignificant expansion of CD4+/CD8+ central memory T-cellsConfirms intended biological mechanism
SafetyNo dose-limiting toxicities reportedExcellent safety profile
Pseudo-progression SurvivalmOS not reached (Observed) vs. 15.1 months (Re-operated)Suggests imaging changes are a positive prognostic marker

Works cited

  1. Introduction to Hetrombopag Medication - Ontosight.ai, accessed June 14, 2025, https://ontosight.ai/glossary/term/introduction-to-hetrombopag-medication--67a24cc2c445bf945aeeb13d
  2. Hetrombopag: A promising thrombopoietin receptor agonist for the treatment of primary and secondary immune thrombocytopenia - ResearchGate, accessed June 14, 2025, https://www.researchgate.net/publication/389770635_Hetrombopag_A_promising_thrombopoietin_receptor_agonist_for_the_treatment_of_primary_and_secondary_immune_thrombocytopenia
  3. Hetrombopag: First Approval | Request PDF - ResearchGate, accessed June 14, 2025, https://www.researchgate.net/publication/353740159_Hetrombopag_First_Approval
  4. Eltrombopag: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed June 14, 2025, https://go.drugbank.com/drugs/DB06210
  5. Search Orphan Drug Designations and Approvals - FDA, accessed June 14, 2025, https://www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=882322
  6. Hetrombopag for the management of chemotherapy ... - ResearchGate, accessed June 14, 2025, https://www.researchgate.net/publication/381439193_Hetrombopag_for_the_management_of_chemotherapy-induced_thrombocytopenia_in_patients_with_advanced_solid_tumors_a_multicenter_randomized_double-blind_placebo-controlled_phase_II_study
  7. Hetrombopag Completed Phase 3 Trials for Chemotherapy-Induced Thrombocytopenia Treatment | DrugBank Online, accessed June 14, 2025, https://go.drugbank.com/drugs/DB16184/clinical_trials?conditions=DBCOND0033989&phase=3&purpose=treatment&status=completed
  8. Hetrombopag for the Treatment of Chemotherapy-Induced Thrombocytopenia in Subjects With Malignancy. | Clinical Research Trial Listing - CenterWatch, accessed June 14, 2025, https://www.centerwatch.com/clinical-trials/listings/NCT03976882/hetrombopag-for-the-treatment-of-chemotherapy-induced-thrombocytopenia-in-subjects-with-malignancy
  9. CAR-T Cell Therapy for T-Cell Malignancies - PMC - PubMed Central, accessed June 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10927222/
  10. Cancer and Hematology Center Cell Therapy for High Risk T-Cell Malignancies Using CD7-Specific CAR Expressed On Autologous T Cells (CRIMSON) | Texas Children's, accessed June 14, 2025, https://www.texaschildrens.org/departments/cancer-and-hematology-center/clinical-trials-and-novel-therapies/cell-therapy-for-high
  11. CAR T-cell therapies for T-cell malignancies: does cellular immunotherapy represent the best chance of cure? | Blood Advances | American Society of Hematology, accessed June 14, 2025, https://ashpublications.org/bloodadvances/article/9/4/913/534813/CAR-T-cell-therapies-for-T-cell-malignancies-does
  12. Wugen Announces Dosing of First Patients in Pivotal Trial of Off-the ..., accessed June 14, 2025, https://wugen.com/wugen-announces-dosing-of-first-patients-in-pivotal-trial-of-off-the-shelf-allogeneic-cd7-targeted-car-t-cell-therapy-wu-cart-007/
  13. CAR T-Cell Immunotherapy Treating T-ALL: Challenges and Opportunities - MDPI, accessed June 14, 2025, https://www.mdpi.com/2076-393X/11/1/165
  14. Three major challenges of CAR T therapy. (A) Fratricide: CAR T cells - ResearchGate, accessed June 14, 2025, https://www.researchgate.net/figure/Three-major-challenges-of-CAR-T-therapy-A-Fratricide-CAR-T-cells-as-they-have-target_fig1_367102243
  15. CD7-edited T cells expressing a CD7-specific CAR for the therapy of ..., accessed June 14, 2025, https://ashpublications.org/blood/article/130/3/285/36917/CD7-edited-T-cells-expressing-a-CD7-specific-CAR
  16. Wugen Announces Dosing of First Patients in Pivotal Trial of Off-the-Shelf, Allogeneic, CD7-Targeted CAR-T Cell Therapy WU-CART-007 - GlobeNewswire, accessed June 14, 2025, https://www.globenewswire.com/news-release/2025/03/20/3046089/0/en/Wugen-Announces-Dosing-of-First-Patients-in-Pivotal-Trial-of-Off-the-Shelf-Allogeneic-CD7-Targeted-CAR-T-Cell-Therapy-WU-CART-007.html
  17. WU-CART-007: A Breakthrough Allogeneic CAR-T Therapy, accessed June 14, 2025, https://www.delveinsight.com/eha-conference/eha-2024/wugen-phase-ii-wu-cart-007-study-results
  18. Anti-CD7 allogeneic WU-CART-007 in patients with relapsed ..., accessed June 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11326362/
  19. What GPRC5D inhibitors are in clinical trials currently? - Patsnap Synapse, accessed June 14, 2025, https://synapse.patsnap.com/article/what-gprc5d-inhibitors-are-in-clinical-trials-currently
  20. NCI Drug Dictionary, accessed June 14, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/
  21. ANZCTR search results | Australian Clinical Trials, accessed June 14, 2025, https://www.australianclinicaltrials.gov.au/anzctr-search-results?search_text=&condition_category=all&purpose_of_study=all&recruitment_status=Recruiting&recruitment_sites=all&phase=all&gender=all&healthy_volunteers=allðics_approval=Yes&condition_code=all&page=484
  22. A Study to Investigate the Safety and Efficacy of SAR446523 ..., accessed June 14, 2025, https://www.careacross.com/clinical-trials/trial/NCT06630806
  23. SAR-446523 - Drug Targets, Indications, Patents - Patsnap Synapse, accessed June 14, 2025, https://synapse.patsnap.com/drug/f9348f9905b74c889b803da8b130deba
  24. Application of GPRC5D Targeting Therapy in Relapsed Refractory Multiple Myeloma - PMC, accessed June 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11912436/
  25. Abstract 2727: Pre-clinical development of a novel anti-GPRC5D ..., accessed June 14, 2025, https://aacrjournals.org/cancerres/article/84/6_Supplement/2727/738783/Abstract-2727-Pre-clinical-development-of-a-novel
  26. Abstract 2727: Pre-clinical development of a novel anti-GPRC5D inducing potent anti-tumor effect through enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) for multiple myeloma - ResearchGate, accessed June 14, 2025, https://www.researchgate.net/publication/379210956_Abstract_2727_Pre-clinical_development_of_a_novel_anti-GPRC5D_inducing_potent_anti-tumor_effect_through_enhanced_antibody-dependent_cell-mediated_cytotoxicity_ADCC_for_multiple_myeloma
  27. Diakonos Oncology to Present Phase I Results of Dubodencel (DOC1021), a Double-Loaded Dendritic Cell Therapy for Glioblastoma, at the ASCO 2025 Annual Meeting - BioSpace, accessed June 14, 2025, https://www.biospace.com/press-releases/diakonos-oncology-to-present-phase-i-results-of-dubodencel-doc1021-a-double-loaded-dendritic-cell-therapy-for-glioblastoma-at-the-asco-2025-annual-meeting
  28. Dubodencel - Drug Targets, Indications, Patents - Patsnap Synapse, accessed June 14, 2025, https://synapse.patsnap.com/drug/89d87c4207814803a932681b432e1aff
  29. FDA Grants Fast Track Designation to DOC1021 for Glioblastoma - OncLive, accessed June 14, 2025, https://www.onclive.com/view/fda-grants-fast-track-designation-to-doc1021-for-glioblastoma
  30. Vaccination by homologous antigenic loading with DOC1021 as ..., accessed June 14, 2025, https://ascopubs.org/doi/10.1200/JCO.2025.43.16_suppl.2014
  31. CNS 2024 Conference Presentation: Diakonos Oncology's ..., accessed June 14, 2025, https://www.biospace.com/press-releases/cns-2024-conference-presentation-diakonos-oncologys-autologous-dendritic-cell-vaccine-doc1021-yields-93-12-month-survival-for-glioblastoma-patients
  32. Diakonos Oncology Presents Promising Phase I Results of Dubodencel (DOC1021) for the Treatment of Glioblastoma at the ASCO 2025 Annual Meeting - BioSpace, accessed June 14, 2025, https://www.biospace.com/press-releases/diakonos-oncology-presents-promising-phase-i-results-of-dubodencel-doc1021-for-the-treatment-of-glioblastoma-at-the-asco-2025-annual-meeting
  33. Diakonos Oncology Presents Promising Phase I Results of Dubodencel (DOC1021) for the Treatment of Glioblastoma at the ASCO 2025 Annual Meeting - PR Newswire, accessed June 14, 2025, https://www.prnewswire.com/news-releases/diakonos-oncology-presents-promising-phase-i-results-of-dubodencel-doc1021-for-the-treatment-of-glioblastoma-at-the-asco-2025-annual-meeting-302469896.html
  34. Evaluating Immunotherapy DOC1021 for Glioblastoma - CGTLive®, accessed June 14, 2025, https://www.cgtlive.com/view/evaluating-immunotherapy-doc1021-glioblastoma
  35. Diakonos' DOC1021 Phase I Results | AACR 2024 - DelveInsight, accessed June 14, 2025, https://www.delveinsight.com/blog/doc1021-phase-i-results
  36. Diakonos' brain tumour vaccine wins FDA's orphan drug designation, accessed June 14, 2025, https://www.pharmaceutical-technology.com/news/diakonos-brain-tumour-vaccine-wins-fdas-orphan-drug-designation/
  37. FDA Gives Dendritic Cell Vaccine Fast Track Status for Pancreatic Cancer - CancerNetwork, accessed June 14, 2025, https://www.cancernetwork.com/view/fda-gives-dendritic-cell-vaccine-fast-track-status-for-pancreatic-cancer
  38. Dendritic Cell Immunotherapy for Treatment of Adult GBM - ClinicalTrials.Veeva, accessed June 14, 2025, https://ctv.veeva.com/study/dendritic-cell-immunotherapy-for-treatment-of-adult-gbm
  39. FDA Grants Orphan Drug Designation to DOC1021 for Malignant Glioma, accessed June 14, 2025, https://www.targetedonc.com/view/fda-grants-orphan-drug-designation-to-doc1021-for-malignant-glioma

Published at: June 14, 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.