MedPath

DR30206 Advanced Drug Monograph

Published:Oct 31, 2025

Generic Name

DR30206

DR30206: A Comprehensive Analysis of a Novel Trispecific Antibody for the Treatment of Solid Tumors

1.0 Executive Summary

DR30206 is an investigational, first-in-class trispecific antibody fusion protein engineered to simultaneously target three critical pathways that govern the tumor microenvironment (TME): Programmed death-ligand 1 (PD-L1), vascular endothelial growth factor (VEGF), and transforming growth factor-beta (TGF-β).[1] Developed by Zhejiang Doer Biologics Co., Ltd., this agent is built upon the company's proprietary MultipleBody® platform and embodies a sophisticated therapeutic hypothesis: that comprehensive, simultaneous blockade of these interconnected pathways can synergistically dismantle tumor defenses, overcome resistance to existing immunotherapies, and elicit potent, durable anti-tumor responses.[1]

The molecule's innovative architecture features the well-established anti-VEGF antibody Bevacizumab as its structural backbone, with an anti-PD-L1 single-domain antibody (VHH) and a TGF-β "trap" fused to its termini.[3] Preclinical studies have provided a strong proof-of-concept, demonstrating that DR30206 possesses superior anti-tumor activity compared not only to monotherapies but also to the combination of separate anti-PD-L1 and anti-VEGF antibodies, suggesting a true synergistic effect conferred by the single-molecule format.[3]

The clinical development program for DR30206 is proceeding on an aggressive, parallel track. A Phase 1 monotherapy trial (NCT06132828) is evaluating its safety and preliminary efficacy in a broad population of patients with advanced solid tumors, while a Phase Ib/IIa trial (NCT07056777) is assessing its potential in combination with standard chemotherapy for gastrointestinal cancers.[5] This strategy is designed to rapidly define the drug's therapeutic window and identify its most promising clinical applications.

DR30206 enters a competitive landscape where the clinical and commercial success of the PD-1/VEGF bispecific antibody ivonescimab has set a high benchmark for efficacy.[7] The primary challenge for DR30206 will be to demonstrate that the addition of TGF-β inhibition provides a clear and clinically meaningful improvement in efficacy and/or durability over this established dual-blockade approach, while maintaining a manageable safety profile. Consequently, DR30206 represents a high-risk, high-reward program at the forefront of multi-specific antibody engineering, with the potential to significantly alter the treatment paradigm for solid tumors if its foundational scientific rationale is validated in human trials.

2.0 Scientific Rationale and Therapeutic Hypothesis

The development of DR30206 is predicated on a deep understanding of the complex, interconnected signaling networks that tumors exploit to evade immune destruction. By targeting three key regulators of the TME, the drug aims to orchestrate a multi-pronged attack that current therapies cannot achieve.

2.1 The Interconnected Immuno-Oncology Axis: PD-L1, VEGF, and TGF-β in the Tumor Microenvironment (TME)

Solid tumors are not merely masses of malignant cells but complex ecosystems involving stromal cells, blood vessels, and immune cells. Within this TME, three signaling pathways are particularly critical for tumor growth and immune evasion:

  • PD-L1: As a primary immune checkpoint ligand, PD-L1 is often overexpressed on tumor cells and other cells within the TME. Its binding to the PD-1 receptor on activated T-cells delivers an inhibitory signal that leads to T-cell exhaustion and anergy, effectively shutting down the anti-tumor immune response.[2]
  • VEGF: This is the principal driver of tumor angiogenesis, the formation of new blood vessels required to supply tumors with oxygen and nutrients. The resulting vasculature is often abnormal and leaky, leading to a hypoxic, acidic, and high-pressure TME that physically impedes the infiltration of cytotoxic T-lymphocytes (CTLs) and promotes an immunosuppressive milieu.[2]
  • TGF-β: This pleiotropic cytokine is a master regulator of immunosuppression within the TME. It inhibits the function of CTLs and Natural Killer (NK) cells, promotes the differentiation of regulatory T-cells (Tregs), and contributes to the formation of a dense, fibrotic stroma that acts as a physical barrier to immune cell entry. Crucially, elevated TGF-β signaling is a well-documented mechanism of resistance to anti-PD-(L)1 therapies.[2]

These pathways do not operate in isolation. They form a self-reinforcing network of immunosuppression. For example, hypoxia induced by VEGF can increase the expression of both PD-L1 and TGF-β. TGF-β, in turn, can stimulate VEGF production by cancer-associated fibroblasts (CAFs) and upregulate PD-L1 on tumor cells, creating a vicious cycle that makes the TME highly resistant to immune attack.[12]

2.2 The Rationale for Trispecific Blockade: A Strategy to Overcome Immunotherapy Resistance

The therapeutic hypothesis behind DR30206 is that a single molecule capable of simultaneously neutralizing all three of these pathways can achieve a synergistic effect that is greater than the sum of its parts. By co-localizing these three distinct inhibitory functions within the TME, DR30206 is designed to comprehensively remodel the tumor ecosystem from an immune-suppressive to an immune-supportive state.[3]

This approach is a direct and rational evolution based on the limitations of prior therapeutic strategies. Immune checkpoint inhibitor (ICI) monotherapies have revolutionized oncology, but their efficacy is limited in many "cold," non-immunogenic tumors.[3] Combination therapies, such as atezolizumab (anti-PD-L1) and bevacizumab (anti-VEGF), have demonstrated the value of dual blockade in indications like hepatocellular and renal cell carcinoma, but they do not address the potent immunosuppressive and fibrotic effects of TGF-β.[13]

The design of DR30206 can be viewed as an engineered response to past clinical failures and a logical progression in drug development. The field has witnessed the discontinuation of bintrafusp alfa, a bifunctional fusion protein targeting PD-L1 and TGF-β, in late-stage trials, suggesting that dual blockade of these two pathways alone may be insufficient to overcome entrenched resistance mechanisms in many tumors.[11] The central premise of DR30206 is that the addition of anti-VEGF activity is the critical third component. By inhibiting angiogenesis, normalizing tumor vasculature, and relieving hypoxia, the anti-VEGF component is expected to improve immune cell infiltration and create a TME in which the simultaneous blockade of PD-L1 and TGF-β can be maximally effective. This represents a sophisticated attempt to solve the well-defined clinical problem of ICI resistance by targeting the underlying biology in a more comprehensive manner.

3.0 Molecular Profile and Mechanism of Action of DR30206

The therapeutic potential of DR30206 is rooted in its unique and intelligently designed molecular structure, which combines validated and novel components to achieve its trifunctional activity.

3.1 Molecular Architecture: A Fusion Protein on the MultipleBody® Platform

DR30206 is a trispecific antibody fusion protein developed using Zhejiang Doer Biologics' proprietary MultipleBody® platform, which is designed to create multi-targeted biotherapeutics.[1] The molecule is composed of three distinct functional domains engineered into a single polypeptide construct [3]:

  1. Structural Backbone: The foundation of the molecule is the full-length humanized IgG1 monoclonal antibody Bevacizumab. This well-characterized, blockbuster drug specifically targets and neutralizes human vascular endothelial growth factor A (VEGF-A).[3]
  2. N-Terminus Fusion: Fused to the N-terminus of the Bevacizumab heavy chains is a variable domain of a heavy-chain-only antibody (VHH), also known as a nanobody, that targets human PD-L1.[3]
  3. C-Terminus Fusion: Fused to the C-terminus of the Bevacizumab heavy chains is the extracellular domain (ECD) of the human TGF-β receptor II (TGF-β RII). This component functions as a "ligand trap," sequestering active TGF-β isoforms in the TME.[3]

This molecular design reflects a pragmatic and sophisticated engineering strategy. The use of Bevacizumab as the backbone is a significant de-risking choice, leveraging decades of clinical data on its safety, pharmacokinetics, and anti-angiogenic activity. This allows development to focus on the novel functionalities of the fused domains. The selection of a VHH for PD-L1 targeting is another advanced choice, as these small, stable domains are well-suited for creating multi-specific constructs and can offer advantages in manufacturing and biophysical properties over larger antibody fragments. This fusion of a proven therapeutic with cutting-edge protein domains illustrates a strategy aimed at maximizing technical success while pursuing a highly innovative biological concept.

3.2 Multi-Modal Mechanism of Action

The unique structure of DR30206 enables it to engage its three targets simultaneously, leading to a multi-pronged attack on the tumor and its microenvironment [2]:

  • PD-L1 Blockade: The anti-PD-L1 VHH binds to PD-L1 expressed on tumor cells, preventing its interaction with the PD-1 receptor on T-cells. This action releases the "brakes" on the immune system, restoring the anti-tumor activity of CTLs and NK cells, which can then recognize and kill cancer cells.[2]
  • VEGF Neutralization: The Bevacizumab component binds to circulating VEGF-A, preventing it from activating its receptors (VEGFRs) on endothelial cells. This abrogates VEGF-mediated signaling, which in turn inhibits the formation of new tumor blood vessels (neovascularization). This anti-angiogenic effect can slow tumor growth and may also help "normalize" the existing tumor vasculature, reducing immunosuppression and improving the delivery of other therapeutic agents and immune cells.[1]
  • TGF-β Sequestration: The TGF-β RII ECD acts as a high-affinity decoy receptor, capturing and neutralizing TGF-β in the TME. This prevents TGF-β from signaling through its endogenous receptors, thereby blocking its potent immunosuppressive effects on T-cells and NK cells, as well as its pro-fibrotic and pro-metastatic activities.[2]

In addition to these primary mechanisms, preclinical data indicate that DR30206, by virtue of its IgG1 Fc region derived from Bevacizumab, may also be capable of eliciting antibody-dependent cellular cytotoxicity (ADCC), providing another potential mechanism for immune-mediated tumor cell killing.[3]

4.0 Preclinical Evidence and Proof-of-Concept

The scientific foundation for the clinical development of DR30206 is supported by a robust preclinical data package, with key findings published in a peer-reviewed journal.

4.1 Publication and Key Findings

The core preclinical validation for DR30206 was published in the journal International Immunopharmacology in March 2024.[3] This study detailed the design and characterization of the molecule. The data demonstrated that DR30206 exhibits high antigen-binding affinities to all three of its targets—PD-L1, VEGF, and TGF-β—and effectively blocks their respective signaling pathways. These are fundamental properties required for therapeutic efficacy.[3] The study also confirmed the molecule's ability to stimulate a mixed lymphocyte reaction (MLR), further supporting its immune-activating potential.[3]

4.2 Comparative Efficacy in In Vivo Models

The most significant finding from the preclinical research relates to the molecule's anti-tumor activity in animal models. The in vivo experiments revealed that the efficacy of DR30206 was superior to that of several important comparators:

  • Monoclonal antibodies targeting either PD-L1 or VEGF alone.
  • A bispecific antibody targeting both PD-L1 and TGF-β.
  • The combination of separate anti-PD-L1 and anti-VEGF monoclonal antibodies administered together.[3]

This latter finding is particularly crucial, as it forms the central pillar of the drug's value proposition. The demonstrated superiority over the co-administration of separate antibodies targeting two of its three targets strongly suggests that the single-molecule, trispecific format confers a true synergistic advantage. This is likely due to the ability of DR30206 to ensure the simultaneous, co-localized blockade of all three pathways within the same region of the TME, orchestrating a more coordinated and potent biological effect than is possible with separate antibodies that have independent pharmacokinetics and biodistribution. This preclinical evidence of synergy justifies the development of a complex trispecific molecule and sets a high but clear objective for the clinical trials: to replicate this superior efficacy in patients. Based on these promising results, the authors of the study suggested that DR30206 holds great therapeutic potential for multiple cancer types, with a particular emphasis on lung cancer, colon adenocarcinoma, and breast carcinoma.[3]

5.0 Clinical Development Program Assessment

Zhejiang Doer Biologics is advancing DR30206 through a sophisticated and aggressive clinical development program, characterized by two distinct trials running in parallel to rapidly evaluate the drug's potential as both a monotherapy and a combination agent.

The table below summarizes the key features of the two ongoing foundational studies for DR30206.

ParameterNCT06132828NCT07056777
TitleA Multicenter, Open-Label, Phase I Study to Evaluate the Safety, Tolerability, Pharmacokinetics of DR30206 in Patients With Advanced or Metastatic Solid TumorsA Phase Ib/IIa Clinical Trial Evaluating the Preliminary Efficacy and Safety of DR30206 in Combination With Standard Therapy in Patients With Gastrointestinal Tumors
PhasePhase 1Phase Ib/IIa
DesignOpen-Label, Non-Randomized, Single Group AssignmentOpen-Label, Non-Randomized, Single Group Assignment
InterventionDR30206 for Injection (intravenous monotherapy)DR30206 (intravenous) + Standard Chemotherapy (e.g., Oxaliplatin, Capecitabine, 5-FU, Irinotecan)
Patient PopulationAdvanced or metastatic solid tumors who have failed standard therapyUnresectable advanced or metastatic gastrointestinal tumors (e.g., colorectal, gastric, esophageal)
Primary ObjectivesPart A: DLTs, MTD, RDE. Part A+B: AEs, SAEs. Part B: RP2D.Phase Ib: DLTs, MTD, RP2D, TEAEs. Phase IIa: Objective Response Rate (ORR) per RECIST v1.1.
Status (as of latest data)RecruitingRecruiting
SponsorZhejiang Doer Biologics Co., Ltd.Zhejiang Doer Biologics Co., Ltd.
Est. Study CompletionJuly 30, 2026 [6]June 30, 2026 5

5.1 Phase 1 Monotherapy Study in Advanced Solid Tumors (NCT06132828)

This first-in-human study is designed to establish the foundational safety and activity profile of DR30206 as a single agent.

  • Design and Objectives: The trial follows a standard two-part, open-label design. Part A is a dose-escalation stage intended to characterize the drug's safety, tolerability, and pharmacokinetics (PK), and to determine the maximum tolerated dose (MTD) and the recommended dose for expansion (RDE). Part B is a dose-expansion stage designed to further evaluate the safety and gather preliminary data on the anti-tumor activity, measured by Objective Response Rate (ORR), at the identified optimal dose.[6]
  • Patient Population: The study enrolls patients aged 18 to 75 with histologically or cytologically confirmed advanced or metastatic solid tumors for whom standard therapy has failed or is not tolerated.[20] This broad "all-comers" approach in the initial phase is typical for novel oncology agents, aiming to identify early signals of efficacy across a range of tumor types.
  • Expansion Cohorts: The protocol specifies that once a recommended dose is established, the study may open expansion cohorts in specific tumor types. These include non-small cell lung cancer (NSCLC), cervical cancer, hepatocellular carcinoma (HCC), triple-negative breast cancer (TNBC), gastric cancer, ovarian cancer, and colorectal cancer. The selection of these cohorts will be guided by any efficacy signals (complete or partial responses) observed during the dose-escalation phase.[6]
  • Key Exclusion Criteria: The eligibility criteria are designed to protect patient safety and reflect the known risks of the targeted pathways. Key exclusions include patients with a history of severe allergies to monoclonal antibodies, active autoimmune diseases, known active central nervous system (CNS) metastases, severe infections, or clinically significant cardiovascular disease.[20]

5.2 Phase Ib/IIa Combination Therapy Study in Gastrointestinal Tumors (NCT07056777)

This trial explores a more pragmatic therapeutic strategy, testing DR30206's ability to enhance the efficacy of the standard-of-care backbone in a major oncology market.

  • Design and Rationale: This is a multicenter, open-label Phase Ib/IIa study being conducted in China. It evaluates DR30206 administered intravenously in combination with standard chemotherapy regimens (such as FOLFOX, FOLFIRI, or XELOX).[5] The rationale is to determine if DR30206 can synergize with chemotherapy to produce superior outcomes in patients with advanced gastrointestinal (GI) tumors.[23]
  • Patient Population: The study enrolls patients with unresectable, locally advanced, or metastatic GI tumors. This includes patients with colorectal cancer (either treatment-naïve or after progression on first-line therapy) and patients with HER2-negative gastric, esophageal, or gastroesophageal junction (GEJ) adenocarcinoma who have not received prior systemic chemotherapy for advanced disease.[5]
  • Objectives: The Phase Ib portion of the study focuses on the safety and tolerability of the combination, with the goal of identifying the recommended Phase 2 dose (RP2D). The primary objective of the subsequent Phase IIa portion is to evaluate the anti-tumor efficacy of the combination, with ORR (as assessed by RECIST v1.1) serving as the primary endpoint.[5]

The decision to run these two distinct trials concurrently represents a sophisticated, capital-intensive strategy that maximizes speed and optionality. This dual-track approach allows the company to simultaneously explore the drug's intrinsic, single-agent activity across multiple cancers while also testing a specific, commercially relevant hypothesis in combination with a standard therapy backbone. Positive data from either trial would provide a viable path toward registration, significantly de-risking the overall program and positioning DR30206 for multiple potential roles in the oncology armamentarium.

6.0 Competitive Landscape and Strategic Positioning

DR30206 is entering one of the most dynamic and competitive areas of oncology drug development: multi-specific antibodies. Its strategic position will be defined by its ability to differentiate itself from a growing field of bispecific and trispecific agents, a landscape largely driven by innovation from Chinese biopharmaceutical companies.[7]

The table below provides a comparative overview of DR30206 and selected competitors targeting related pathways.

Drug NameDeveloper(s)TargetsHighest Development PhaseKey Differentiator/Comment
DR30206Zhejiang Doer BiologicsPD-L1 x VEGF x TGF-βPhase 1/2aTrispecific; Bevacizumab backbone; VHH for PD-L1.
PM8003BiotheusPD-L1 x VEGF x TGF-βPhase 1/2Direct trispecific competitor to DR30206.14
IvonescimabAkeso / Summit TherapeuticsPD-1 x VEGFPhase 3 / Approved (China)Key clinical and commercial benchmark for dual blockade.7
Bintrafusp AlfaMerck KGaA / GSKPD-L1 x TGF-βDevelopment DiscontinuedCautionary tale for TGF-β targeting; its failure raises the bar for proving the benefit of this mechanism.11
LBL-015Leads BiolabsPD-1 x TGF-βRIIPhase 1Example of PD-1/TGF-β dual targeting approach.[24]
PM8002 (BNT327)Biotheus / BioNTechPD-L1 x VEGFPhase 1/2Example of PD-L1/VEGF dual targeting approach.7

6.1 Direct and Indirect Competitors in the Multi-Specific Antibody Space

The competitive environment for DR30206 is multifaceted:

  • Direct Trispecific Competitor: The most direct rival is PM8003, developed by Biotheus, which is also an anti-PD-L1/VEGF/TGF-β trispecific antibody currently in a Phase 1/2 trial in China.[14] The relative development timelines and emerging data from this program will be a critical factor in the competitive positioning of DR30206.
  • Key Bispecific Benchmark: The most important benchmark for DR30206 is ivonescimab, a PD-1/VEGF bispecific antibody developed by Akeso Biopharma and licensed by Summit Therapeutics. Ivonescimab's clinical success and recent approval in China have validated the concept of dual PD-1/VEGF blockade and established a high bar for both efficacy and safety that any new agent in this space must meet or exceed.[7]
  • Broader Landscape: The field also includes numerous other agents targeting subsets of these pathways, including PD-(L)1/TGF-β bispecifics (such as LBL-015 and SHR-1701) and other PD-(L)1/VEGF bispecifics (like PM8002/BNT327).[7] These programs contribute to a crowded and rapidly evolving therapeutic area.

6.2 Differentiation and Potential Advantages

DR30206's primary point of differentiation is its unique trispecificity. The central strategic question for the program is whether this "triple whammy" approach can translate into a clinically meaningful advantage over dual-blockade strategies.[14] This advantage would need to manifest as a higher objective response rate, more durable responses, or, critically, activity in tumor types or patient populations that are resistant to PD-(L)1/VEGF bispecifics.

The strategic positioning of DR30206 is therefore shaped by two pivotal precedents in the field. First, the high-profile failure of the PD-L1/TGF-β bispecific bintrafusp alfa has cast a shadow of skepticism over TGF-β as a therapeutic target, placing a significant burden of proof on Zhejiang Doer Biologics to demonstrate that its inhibition provides a tangible clinical benefit.[14] Second, the resounding success of ivonescimab has both validated the multi-specific antibody approach and created a formidable competitor.[7] Consequently, the clinical development of DR30206 must be strategically designed not merely to achieve regulatory approval, but to generate comparative data that unequivocally demonstrates a superior benefit-risk profile over the established dual-blockade standard.

7.0 Expert Analysis, Unanswered Questions, and Future Outlook

DR30206 is a scientifically compelling and rationally designed therapeutic candidate that represents the next wave of innovation in immuno-oncology. However, its path to clinical and commercial success is accompanied by significant questions and challenges.

7.1 SWOT Analysis

  • Strengths: The molecule is based on a strong scientific rationale for comprehensive TME remodeling. Its innovative trispecific design incorporates a de-risked and validated Bevacizumab backbone. The preclinical data package is highly promising, suggesting true synergy and superiority over combination therapies. The parallel clinical development strategy is aggressive and well-designed to maximize the speed of data generation.
  • Weaknesses: As an investigational agent, there is no published human data to confirm its safety or efficacy. There is a significant risk of cumulative or novel toxicities arising from the simultaneous blockade of three major biological pathways. The drug is being developed by a smaller biotechnology company, which may face challenges in executing large-scale global trials and commercialization without a major pharmaceutical partner.
  • Opportunities: DR30206 has the potential to be the first-in-class approved therapy targeting the PD-L1/VEGF/TGF-β axis. It could address the substantial unmet medical need of primary and acquired resistance to current ICIs. If successful, it could become a foundational therapy across multiple solid tumors, both as a monotherapy and as a combination partner.
  • Threats: The high efficacy and safety bar set by competitors, particularly ivonescimab, presents a major threat. The entire field of TGF-β targeting may be viewed with skepticism by clinicians and investors following previous failures. The rapid pace of innovation in oncology means that other therapeutic modalities could emerge and supersede this approach.

7.2 Critical Unanswered Questions

As DR30206 progresses through clinical trials, several critical questions must be answered to determine its ultimate value:

  1. What is the clinical safety and tolerability profile? This is the most significant near-term hurdle. The key question is whether the potential toxicities associated with each target—immune-related adverse events (PD-L1), hypertension, bleeding, and proteinuria (VEGF), and potential cardiac or other off-target effects (TGF-β)—will be manageable when combined in a single agent.
  2. Does the preclinical synergy translate to clinical efficacy? The core value proposition of DR30206 rests on its preclinical superiority over combination therapies. The clinical trials must demonstrate a clear and meaningful improvement in efficacy outcomes (e.g., ORR, duration of response, progression-free survival) over the established PD-(L)1/VEGF bispecific standard.
  3. Is there a predictive biomarker strategy? The current trials are enrolling relatively broad patient populations. The identification of biomarkers—whether related to PD-L1 expression, TME vascularity, or TGF-β pathway activation—that can predict which patients are most likely to respond would significantly de-risk late-stage development and optimize the drug's clinical application.

7.3 Future Outlook and Projected Trajectory

The near-term trajectory of the DR30206 program will be dictated by the initial data readouts from the dose-escalation portions of the NCT06132828 and NCT07056777 trials. A favorable safety profile will be the first and most critical catalyst.

Should strong efficacy signals emerge, particularly in the monotherapy setting, the company will likely move rapidly to fully enroll the planned dose-expansion cohorts in specific tumor types and engage with regulatory authorities to define an accelerated path to approval. If, however, monotherapy activity proves to be modest but the combination with chemotherapy shows significant promise, the strategic focus will likely shift toward positioning DR30206 as a chemo-enhancing agent in first- or second-line settings for high-prevalence indications like gastrointestinal cancers.

In conclusion, DR30206 stands as a scientifically ambitious and highly innovative next-generation immuno-oncology agent. Its trispecific mechanism is at the cutting edge of antibody engineering and is rationally designed to address the fundamental challenge of TME-mediated immunotherapy resistance. While the preclinical evidence is compelling, the program carries substantial clinical risk, primarily centered on the potential for cumulative toxicity. The initial Phase 1 safety and efficacy data will therefore represent a critical inflection point, determining whether this advanced therapeutic concept can be translated into a clinical reality. Success would position DR30206 as a potentially transformative therapy for patients with solid tumors, but the path forward is challenging and contingent on demonstrating a clear and superior benefit-risk profile in a fiercely competitive therapeutic landscape.

Works cited

  1. Pipeline - Zhejiang Doer Biologics Corporation, accessed October 31, 2025, https://www.doerbio.com/english.php/Product/product/nid/3
  2. Definition of anti-PD-L1/VEGF/TGF-beta trispecific antibody DR30206 - NCI Drug Dictionary, accessed October 31, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/anti-pd-l1-vegf-tgf-beta-trispecific-antibody-dr30206
  3. An innovative antibody fusion protein targeting PD-L1, VEGF and TGF-β with enhanced antitumor efficacies - PubMed, accessed October 31, 2025, https://pubmed.ncbi.nlm.nih.gov/38377856/
  4. Hangzhou Zhongmeihuadong Pharmaceutical Co., Ltd. - Drug pipelines, Patents, Clinical trials - Patsnap Synapse, accessed October 31, 2025, https://synapse.patsnap.com/organization/4151879e2be683699c95135e4af229c4
  5. Study Details | NCT07056777 | Evaluation of the Preliminary Efficacy and Safety of DR30206 in Combination With Standard Therapy in Patients With Gastrointestinal Cancer | ClinicalTrials.gov, accessed October 31, 2025, https://clinicaltrials.gov/study/NCT07056777
  6. Study Details | NCT06132828 | Evaluate the Safety, Tolerability, Pharmacokinetics of DR30206 in Patients With Advanced or Metastatic Solid Tumors | ClinicalTrials.gov, accessed October 31, 2025, https://www.clinicaltrials.gov/study/NCT06132828
  7. PD-1 x VEGF and PD-L1 x VEGF Bispecific Antibodies Business, Pipeline and Competitor Analysis Report 2025 - GlobeNewswire, accessed October 31, 2025, https://www.globenewswire.com/news-release/2025/03/03/3035532/0/en/PD-1-x-VEGF-and-PD-L1-x-VEGF-Bispecific-Antibodies-Business-Pipeline-and-Competitor-Analysis-Report-2025-Comprehensive-Overview-of-R-D-and-Partnering-Activities-of-Pharma-and-Techn.html
  8. PD-1 x VEGF and PD-L1 x VEGF bispecific antibodies: a business, pipeline and competitor analysis from an industry perspective - La Merie Publishing, accessed October 31, 2025, https://lamerie.com/report/pd-1-x-vegf-and-pd-l1-x-vegf-bispecific-antibodies-a-business-pipeline-and-competitor-analysis-from-an-industry-perspective/
  9. Bispecific antibody targeting TGF-β and PD-L1 for synergistic cancer immunotherapy, accessed October 31, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2023.1196970/full
  10. Therapeutic targeting of VEGF and/or TGF-β to enhance anti-PD-(L)1 therapy: The evidence from clinical trials - PubMed, accessed October 31, 2025, https://pubmed.ncbi.nlm.nih.gov/35957885/
  11. Dual inhibition of TGF‐β and PD‐L1: a novel approach to cancer treatment - PMC, accessed October 31, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9168966/
  12. Recent Advances in the Development of TGF-β Signaling Inhibitors for Anticancer Therapy - Journal of Cancer Prevention, accessed October 31, 2025, https://www.jcpjournal.org/journal/download_pdf.php?doi=10.15430/JCP.2020.25.4.213
  13. Therapeutic targeting of VEGF and/or TGF-β to enhance anti-PD-(L)1 therapy: The evidence from clinical trials - PMC - NIH, accessed October 31, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9360509/
  14. TROP2 gets more crowded | ApexOnco - Clinical Trials news and analysis, accessed October 31, 2025, https://www.oncologypipeline.com/apexonco/trop2-gets-more-crowded
  15. Zhejiang Doer Biologics Corporation, accessed October 31, 2025, https://doerbio.com/english.php
  16. DR30206 / Huadong Medicine - LARVOL DELTA, accessed October 31, 2025, https://delta.larvol.com/Products/?ProductId=f277bbe5-708a-469f-83f5-2e558d285221
  17. DR-30206 - Drug Targets, Indications, Patents - Patsnap Synapse, accessed October 31, 2025, https://synapse.patsnap.com/drug/1e41377947f24bb294b5b7231b27337b
  18. Zhejiang Doer Biologics Corporation, accessed October 31, 2025, https://doerbio.com/english.php/News/detail/nid/2/id/105/type/1
  19. Evaluate the Safety, Tolerability, Pharmacokinetics of DR30206 in Patients With Advanced or Metastatic Solid Tumors | TrialScreen, accessed October 31, 2025, https://app.trialscreen.org/trials/phase-1-evaluate-safety-tolerability-pharmacokinetics-dr30206-patients-trial-nct06132828
  20. Evaluate the Safety, Tolerability, Pharmacokinetics of DR30206 in Patients With Advanced or Metastatic Solid Tumors - CareAcross, accessed October 31, 2025, https://www.careacross.com/clinical-trials/trial/NCT06132828
  21. Evaluate the Safety, Tolerability, Pharmacokinetics of DR30206 in Patients With Advanced or Metastatic Solid Tumors (NCT06132828) - Ancora.ai, accessed October 31, 2025, https://www.ancora.ai/it/details/NCT06132828
  22. Study Details | NCT07056777 | Evaluation of the Preliminary Efficacy and Safety of DR30206 in Combination With Standard Therapy in Patients With Gastrointestinal Cancer | ClinicalTrials.gov, accessed October 31, 2025, https://www.clinicaltrials.gov/study/NCT07056777?term=AREA%5BBasicSearch%5D(AREA%5BLastUpdatePostDate%5D%20RANGE%5B2020-06-05,MAX%5D%20AND%20SEARCH%5BLocation%5D(AREA%5BLocationCountry%5DChina))&rank=10
  23. Evaluation of the Preliminary Efficacy and Safety of DR30206 in Combination With Standard Therapy in Patients With Gastrointestinal Cancer - ClinConnect, accessed October 31, 2025, https://clinconnect.io/trials/NCT07056777
  24. Anti-PD-1/TGF-βRII bispecific antibody fusion protein LBL-015 in patients with advanced malignant tumors: A phase I, first-in-human, open-label, multicenter, dose-escalation study. - ASCO Publications, accessed October 31, 2025, https://ascopubs.org/doi/10.1200/JCO.2024.42.16_suppl.2592

Published at: October 31, 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.