MedPath

DS-1055 Advanced Drug Monograph

Published:May 16, 2025

Generic Name

DS-1055

DS-1055a: A Novel GARP-Targeting Immunotherapy for Solid Tumors

1. Executive Summary

DS-1055a is an investigational afucosylated human monoclonal antibody representing a novel approach in immuno-oncology. It is designed to selectively target Glycoprotein A Repetitions Predominant (GARP), a protein expressed on the surface of activated regulatory T cells (Tregs) within the tumor microenvironment. The primary mechanism of action of DS-1055a involves the depletion of these immunosuppressive GARP+ Tregs, thereby aiming to reverse tumor-induced immune tolerance and enhance the host's anti-tumor immune response. This therapeutic strategy is particularly relevant given the critical role Tregs play in inhibiting effective cancer immunosurveillance.

Developed through a collaboration between Daiichi Sankyo Co., Ltd. and BioInvent International AB, DS-1055a is currently undergoing Phase 1 clinical evaluation (NCT04419532) in patients with advanced or metastatic solid tumors. Preclinical studies have demonstrated its ability to deplete GARP+ Tregs and activate effector T cells, leading to significant anti-tumor activity in humanized mouse models. The afucosylation of DS-1055a is a key design feature intended to enhance its antibody-dependent cellular cytotoxicity (ADCC) capabilities, crucial for its Treg-depleting function. The development of DS-1055a signifies a focused effort to modulate the tumor microenvironment by specifically addressing Treg-mediated immunosuppression, offering a potentially complementary or alternative strategy to existing immunotherapies.

2. Introduction to DS-1055a

DS-1055a is emerging as a distinct investigational agent in the field of cancer immunotherapy, characterized by its unique molecular design and targeted approach.

2.1. Chemical and Therapeutic Classification

DS-1055a is an afucosylated, human monoclonal antibody.[1] The designation "monoclonal" indicates its origin from a single B-cell clone and its specificity for a single epitope on its target antigen. Its "human" nature is intended to minimize immunogenicity when administered to patients. A critical structural modification is its "afucosylation," a deliberate glycoengineering process where fucose sugar residues are omitted from the Fc region of the antibody. This modification is known to significantly enhance the antibody's binding affinity to FcγRIIIa receptors on immune effector cells, thereby potentiating ADCC, a key mechanism for cell depletion.[3]

Therapeutically, DS-1055a is classified as an immunomodulating and antineoplastic agent.[1] Its immunomodulatory role stems from its ability to alter the immune cell composition and activity within the tumor microenvironment, specifically by targeting and depleting immunosuppressive regulatory T cells. The antineoplastic effect is the anticipated outcome of this immune modulation, leading to an enhanced immune attack against tumor cells.

2.2. Origin and Developers

DS-1055a was discovered by Daiichi Sankyo Co., Ltd..[2] The discovery process was notably supported by a collaboration with BioInvent International AB, which provided access to its proprietary n-CoDeR® antibody library.[2] This collaboration underscores a common paradigm in pharmaceutical research where specialized discovery platforms contribute to the pipeline of larger development organizations. The terms of this collaboration include potential milestone payments and royalties from Daiichi Sankyo to BioInvent, reflecting the value of BioInvent's contribution to the program.[5] The global clinical development of DS-1055a is being led by Daiichi Sankyo Inc..[4] The intellectual property landscape and financial arrangements between these entities signify a structured, long-term strategic partnership built on the perceived early value and potential of DS-1055a.

Table 1: DS-1055a - Drug Profile Summary

FeatureDescription
Generic Name/Code NamesDS-1055a, DS-1055, anti-GARP monoclonal antibody DS-1055a, anti-glycoprotein A repetitions predominant monoclonal antibody DS-1055a
Drug TypeAfucosylated Human Monoclonal Antibody
Target AntigenGlycoprotein A Repetitions Predominant (GARP); Leucine-rich repeat-containing protein 32 (LRRC32)
Primary Mechanism of ActionSelective binding to GARP on activated Tregs, leading to their depletion via enhanced Antibody-Dependent Cellular Cytotoxicity (ADCC). This results in the reversal of Treg-mediated immunosuppression and enhancement of anti-tumor immune response.
Originators/DevelopersDaiichi Sankyo Co., Ltd.; BioInvent International AB
Highest Development PhasePhase 1
Primary Indication (Clinical)Relapsed or refractory, locally advanced or metastatic solid tumors
Clinical Trial IdentifierNCT04419532

Sources: [1]

The deliberate engineering of DS-1055a as an afucosylated antibody is a strong indication of a focused mechanistic strategy. This modification is pivotal for an antibody designed to deplete target cells, suggesting that direct and efficient cell killing of GARP+ Tregs via enhanced ADCC is a primary intended therapeutic mechanism.[3]

3. Mechanism of Action: Targeting the GARP-TGF-β Axis on Regulatory T cells

DS-1055a employs a targeted mechanism aimed at disrupting a key immunosuppressive pathway within the tumor microenvironment, focusing on the GARP protein expressed on activated regulatory T cells.

3.1. The Role of GARP and Tregs in Tumor Immune Evasion

Regulatory T cells (Tregs), typically identified by the expression of the transcription factor FOXP3, are a specialized subpopulation of CD4+ T lymphocytes that play a critical role in maintaining immune homeostasis and preventing autoimmunity.[8] However, their presence and activity within the tumor microenvironment (TME) are often detrimental to anti-tumor immunity. Tregs accumulate in tumors and actively suppress the function of effector immune cells, such as cytotoxic T lymphocytes and NK cells, thereby facilitating tumor immune evasion and promoting cancer progression.[3]

Glycoprotein A Repetitions Predominant (GARP), also known as Leucine-rich repeat-containing protein 32 (LRRC32), is a type I transmembrane protein that has emerged as a key surface marker and functional molecule on these immunosuppressive Tregs.[1] GARP expression is notably high on activated Tregs found within the TME across various human cancer types.[1] Importantly, studies have shown that GARP is selectively induced on peripheral Treg cells upon polyclonal stimulation, but not significantly on conventional effector T cells, suggesting a degree of specificity that could be exploited therapeutically.[2]

A critical function of GARP is its role as a cell surface docking receptor for latent Transforming Growth Factor-beta (TGF-β), particularly TGF-β1. GARP is essential for the presentation and subsequent activation of TGF-β on the surface of these activated Tregs.[1] TGF-β is a pleiotropic cytokine with potent immunosuppressive properties in the TME, contributing to the inhibition of effector T cell proliferation and function, and promoting an immune-tolerant environment conducive to tumor growth.[1] Thus, GARP-expressing Tregs represent a significant source of active TGF-β within tumors. The specific expression of GARP on activated Tregs, especially within the TME, makes it an attractive therapeutic target to selectively neutralize these immunosuppressive cells and their detrimental functions.

3.2. DS-1055a: Selective GARP Binding and Treg Depletion

DS-1055a is a human monoclonal antibody specifically designed and engineered to target and bind to GARP (LRRC32).[1] The core therapeutic strategy of DS-1055a revolves around the depletion of these GARP-positive activated Tregs from the TME.[1] By eliminating these cells, DS-1055a aims to reverse the profound immunosuppression they mediate, thereby creating a more favorable environment for the host's immune system to recognize and attack tumor cells.[1]

3.3. Impact of Afucosylation on Antibody-Dependent Cellular Cytotoxicity (ADCC)

A key molecular feature of DS-1055a is its afucosylated Fc region.[1] Afucosylation is a glycoengineering technique that involves the removal of fucose sugar moieties from the N-glycans attached to the Fc domain of the antibody. This modification has been well-documented to significantly enhance the binding affinity of the IgG1 Fc region to the activating Fc gamma receptor IIIa (FcγRIIIa, or CD16A), which is predominantly expressed on immune effector cells like Natural Killer (NK) cells and macrophages.[3]

The increased affinity for FcγRIIIa translates into a more potent Antibody-Dependent Cellular Cytotoxicity (ADCC) response. ADCC is a crucial immune mechanism whereby antibody-coated target cells are recognized and lysed by effector immune cells. In the context of DS-1055a, the enhanced ADCC mediated by its afucosylated structure is believed to be the primary mechanism responsible for the efficient and effective depletion of GARP+ Treg cells.[3] This targeted cell killing is central to the therapeutic efficacy of DS-1055a.

3.4. Modulation of the Tumor Microenvironment and Augmentation of Anti-Tumor Immunity

The depletion of GARP+ Tregs by DS-1055a is anticipated to induce a significant shift in the TME, transforming it from an immunosuppressive milieu to one that is immuno-permissive and conducive to effective anti-tumor responses.[1] By removing a major source of local immunosuppression (both the Tregs themselves and the TGF-β they present), DS-1055a is expected to lead to the activation and enhanced functionality of anti-tumor effector T cells and other immune cells.[2] The ultimate goal of DS-1055a therapy is to augment or restore the host's intrinsic anti-tumor immunity, enabling a more robust and sustained attack against malignant cells.[2]

The selective expression of GARP on activated Tregs, particularly within the TME, is a critical attribute that may provide DS-1055a with a favorable therapeutic index.[1] By preferentially targeting these highly immunosuppressive cells within the tumor, DS-1055a might minimize the disruption of systemic immune homeostasis and reduce the risk of widespread autoimmune-related adverse events that can be associated with less selective immunotherapies or broader Treg depletion strategies. Furthermore, the dual impact of depleting GARP+ Tregs—thereby removing the cells and reducing a key source of active TGF-β in the TME—suggests a multi-faceted approach to overcoming Treg-mediated immunosuppression.[1] This could prove more effective than strategies targeting only Treg presence or TGF-β signaling in isolation. This mechanism also positions DS-1055a as a candidate for overcoming resistance to existing checkpoint inhibitors, potentially by re-sensitizing tumors or by synergizing with such agents in combination therapies.[9]

4. Preclinical Profile of DS-1055a

The progression of DS-1055a into clinical trials was underpinned by a body of preclinical research validating its mechanism of action and demonstrating its anti-tumor potential.

4.1. In Vitro Activity

In vitro studies have been instrumental in characterizing the interaction of DS-1055a with its target and elucidating its effects on immune cell populations. It was confirmed that GARP is specifically expressed by highly suppressive Treg cells isolated from the TME of various human cancers. A key finding was that GARP expression is selectively induced in peripheral Treg cells upon polyclonal stimulation, a phenomenon not observed in effector T cells.[2] This selective upregulation reinforces GARP's utility as a target for specifically modulating activated Treg function.

Functionally, DS-1055a demonstrated its capacity to efficiently deplete GARP+ Treg cells in vitro. This depletion was directly correlated with a subsequent activation of effector T cells.[2] The afucosylation of DS-1055a is highlighted as a critical factor in achieving this efficient depletion, presumably through the enhancement of ADCC.[3] These in vitro results provided direct evidence of DS-1055a's ability to execute its intended biological functions in a controlled laboratory environment.

4.2. In Vivo Efficacy in Animal Models

The anti-tumor activity of DS-1055a was evaluated in vivo using humanized mouse models, which are designed to more closely mimic human immune responses to antibody-based therapies. In a model utilizing human colorectal cancer cells (HT-29) implanted into mice engrafted with human immune cells, administration of DS-1055a led to a discernible decrease in the population of FoxP3+CD4+ T cells (a standard marker for Tregs) within the TME.[2]

Concurrently with this reduction in intratumoral Tregs, DS-1055a treatment resulted in remarkable anti-tumor activity in this humanized mouse model.[2] This in vivo efficacy, observed in a system containing human immune components, provided strong preclinical proof-of-concept for the therapeutic potential of DS-1055a and its ability to modulate the TME by reducing Treg populations. The successful demonstration of efficacy in such a humanized model is particularly noteworthy as it offers a more relevant translational bridge to potential human clinical outcomes compared to studies in purely syngeneic mouse systems.

4.3. Summary of Preclinical Anti-Tumor Immunity Augmentation

Collectively, the preclinical data package for DS-1055a supports its proposed mechanism of augmenting anti-tumor immunity. By selectively targeting GARP on activated Tregs and mediating their efficient depletion (a process significantly enhanced by its afucosylated structure facilitating potent ADCC), DS-1055a effectively diminishes a key immunosuppressive component of the TME. This, in turn, unleashes or enhances the activity of effector immune cells against the tumor. These findings positioned DS-1055a as a novel and promising Treg-cell-targeted agent for cancer immunotherapy, warranting its advancement into clinical evaluation.[2] The preclinical data clearly established a causal link between GARP+ Treg depletion and the subsequent activation of effector T cells, indicating a functional reconfiguration of the TME towards an immune-permissive state.

Table 3: Summary of Key Preclinical Findings for DS-1055a

FeatureFindingImplication
Target Specificity (In Vitro)GARP specifically expressed on highly suppressive Tregs in human TME; selectively induced on peripheral Tregs upon stimulation.Validates GARP as a selective target on activated/suppressive Tregs, minimizing off-target effects on effector T cells.
Functional Activity (In Vitro)DS-1055a (afucosylated) efficiently depleted GARP+ Tregs, leading to activation of effector T cells.Demonstrates direct biological activity and intended immunomodulatory effect.
TME Modulation (In Vivo)In humanized HT-29 colorectal cancer model, DS-1055a decreased intratumoral FoxP3+CD4+ T cells.Confirms Treg depletion within the tumor context in a humanized system.
Anti-Tumor Efficacy (In Vivo)DS-1055a exhibited remarkable anti-tumor activity in the humanized HT-29 model.Provides preclinical proof-of-concept for therapeutic efficacy.
Overall MechanismDepletion of GARP+ Tregs via enhanced ADCC (due to afucosylation) leads to augmented anti-tumor immunity by effector T cells.Establishes a clear mechanistic rationale for clinical development.

Sources: [1]

5. Clinical Development Program: Phase 1 Study (NCT04419532)

The promising preclinical profile of DS-1055a led to its advancement into human clinical trials, with NCT04419532 being the first-in-human study.

5.1. Study Rationale and Design

The primary rationale for initiating the Phase 1 trial (DS1055-A-J101; NCT04419532) stems from the robust preclinical data indicating that DS-1055a can effectively target and deplete GARP-expressing activated Tregs, thereby restoring or enhancing anti-tumor immune responses.[9] This study aims to translate these preclinical findings into the clinical setting, evaluating DS-1055a as a novel immune intervention, particularly for patients with advanced solid tumors that are relapsed or refractory to standard therapies, including potentially those who have not responded to or have progressed after treatment with existing immune checkpoint inhibitors.[9]

The NCT04419532 study is designed as a first-in-human, global, multi-center, open-label, non-randomized, dose-escalation clinical trial.[5] This design is typical for early-phase oncology studies of novel therapeutic agents. The "first-in-human" designation signifies the initial administration of DS-1055a to human subjects. Its "global, multi-center" nature aims to facilitate patient recruitment and gather data from diverse populations. The "open-label" characteristic means that both the investigators and the participants are aware of the treatment being administered, which is common for initial safety-focused dose-escalation studies. The "non-randomized" aspect applies to the dose-escalation phase, where cohorts of patients sequentially receive increasing doses of DS-1055a. The core "dose-escalation" component is crucial for identifying a safe and tolerable dose range for subsequent investigation.

5.2. Primary and Secondary Objectives

The primary objectives of the NCT04419532 trial are:

  • To assess the safety and tolerability profile of DS-1055a in adult patients with relapsed or refractory locally advanced or metastatic solid tumors.[7]
  • To determine the Maximum Tolerated Dose (MTD) and/or identify the Recommended Dose for further study (RD) of DS-1055a.[7]

The secondary objectives include:

  • Evaluation of the pharmacokinetic (PK) properties of DS-1055a, including parameters such as maximum plasma concentration (Cmax), time to Cmax (Tmax), area under the plasma concentration-time curve (AUClast and AUCtau), and trough concentration (Ctrough).[7]
  • Assessment of the immunogenicity of DS-1055a, specifically the incidence of anti-drug antibodies (ADAs) and other relevant antibodies.[7]
  • Obtaining preliminary evidence of anti-tumor activity, measured by endpoints such as Objective Response Rate (ORR), Disease Control Rate (DCR), Duration of Response (DoR), Time To Response (TTR), Progression-Free Survival (PFS), and Overall Survival (OS).[7]
  • Assessment of various biomarker endpoints to understand the biological effects of DS-1055a.[9]

5.3. Key Eligibility Criteria

Inclusion Criteria:

Participants must be adults (generally ≥18 years, with specific age requirements like ≥20 years in Japan) with a histopathologically confirmed diagnosis of locally advanced or metastatic solid tumors. Specified tumor types include head and neck cancer, gastric cancer, esophageal cancer, non-small cell lung cancer (NSCLC), and melanoma, though patients with other solid tumor types may be considered eligible after consultation with the sponsor. Enrolled patients must have disease that is relapsed or refractory to standard curative therapies. Other key inclusion criteria include an Eastern Cooperative Oncology Group Performance Status (ECOG PS) of 0 or 1, the presence of measurable disease as defined by RECIST v1.1, and adequate organ function.7

Exclusion Criteria:

Key exclusion criteria include the presence of a concurrently active second malignancy (with certain exceptions such as adequately treated non-melanoma skin cancers or carcinoma in situ). Patients with a history of, or current, (non-infectious) interstitial lung disease (ILD) requiring steroid treatment, or suspected ILD, are excluded. Other exclusions involve a history of severe pulmonary compromise or the requirement for supplemental oxygen within six months prior to enrollment, active hepatitis B or C virus infection, and prior immunotherapy that resulted in a Grade 3 or higher immune-related adverse event (irAE) or any unresolved irAE of Grade 2 or higher.7 The stringent exclusion related to prior irAEs is particularly noteworthy, reflecting caution about potential cumulative immune toxicities with a new immunomodulatory agent.

5.4. Dosing and Administration

DS-1055a is administered via intravenous (IV) infusion.[7] The treatment is given on Day 1 of each 21-day cycle. The protocol includes a "priming dose period" involving low-dose administration(s) of DS-1055a before the commencement of the standard cyclical dosing. The initial infusion duration is set at 180 minutes, which may be reduced to 120 minutes for subsequent infusions if the first infusion is well-tolerated and does not result in infusion-related reactions.[7] This careful approach to infusion is a safety measure to manage potential acute reactions to the biologic.

5.5. Safety and Tolerability Profile (Anticipated Assessments)

The primary safety evaluation in the trial involves the monitoring and grading of Treatment-Emergent Adverse Events (TEAEs) according to the National Cancer Institute-Common Terminology Criteria for Adverse Events (NCI-CTCAE) Version 5.0.[7] A Dose-Limiting Toxicity (DLT) is specifically defined as any TEAE that occurs within the initial 21-day DLT evaluation period, is assessed as Grade 3 or higher, and is considered by the investigator to be related to DS-1055a. Toxicities clearly attributable to disease progression or other non-drug factors are not classified as DLTs.[7] The identification of DLTs is crucial for determining the MTD.

5.6. Current Status and Enrollment

  • Trial Identifiers: The trial is registered under NCT04419532 and also known by the sponsor ID DS1055-A-J101.[7]
  • Status: As of April 2025, the trial status is listed as "Active, not recruiting".[7] Earlier reports had indicated the trial was "Recruiting".[11] This change typically suggests that patient enrollment for the planned cohorts (likely dose-escalation and potentially initial expansion) has been completed, and enrolled patients are undergoing treatment or are in the follow-up phase.
  • Enrollment: The target enrollment for the study is approximately 40 participants.[7]
  • Locations: The study is being conducted at multiple sites globally, including in the United States, Japan, and Canada.[2]
  • Key Dates: The study start date was October 8, 2020.[7] Daiichi Sankyo announced the dosing of the first patient on October 22, 2020.[5] The estimated primary completion date and study completion date is January 30, 2026.[7] This extended timeline suggests a thorough long-term follow-up for safety, immunogenicity, and durability of any observed responses.

Table 2: Overview of Clinical Trial NCT04419532

ParameterDetails
Trial IdentifierNCT04419532 (Sponsor ID: DS1055-A-J101)
Official TitleA Phase 1, First In Human Study of DS-1055a in Subjects With Relapsed or Refractory Locally Advanced or Metastatic Solid Tumors
PhasePhase 1
Status (as of April 2025)Active, not recruiting
SponsorDaiichi Sankyo Co., Ltd.
Indication(s)Relapsed or refractory, locally advanced or metastatic solid tumors (including head and neck, gastric, esophageal, NSCLC, melanoma, and other solid tumors upon discussion with sponsor)
Primary ObjectivesAssess safety and tolerability of DS-1055a; Determine Maximum Tolerated Dose (MTD) and/or Recommended Dose for further study.
Key Secondary ObjectivesPharmacokinetics (Cmax, Tmax, AUClast, AUCtau, Ctrough); Immunogenicity (Anti-Drug Antibodies); Preliminary Efficacy (ORR, DCR, DoR, TTR, PFS, OS); Biomarker assessments.
Target EnrollmentApproximately 40 participants
Key LocationsUnited States, Japan, Canada
Study Start DateOctober 8, 2020
Estimated Study Completion DateJanuary 30, 2026

Sources: [2]

The strategic inclusion of diverse tumor types in this Phase 1 trial, such as head and neck cancer, gastric cancer, esophageal cancer, NSCLC, and melanoma, while also permitting enrollment of patients with other solid tumors, points towards an approach aimed at identifying early signals of activity across a spectrum of malignancies.[7] This broad initial screening can help prioritize indications for subsequent Phase 2 development based on observed responses and underlying biological rationale related to GARP expression or TME characteristics.

6. Therapeutic Potential and Future Outlook

DS-1055a, with its novel mechanism targeting GARP on activated Tregs, holds considerable therapeutic potential, particularly in addressing unmet medical needs in oncology.

6.1. Potential in Solid Tumors and Unmet Needs

The expression of GARP on activated Tregs within the TME of a wide array of solid tumors provides a broad rationale for the potential applicability of DS-1055a.[2] A significant unmet need exists for effective treatments for patients with advanced, metastatic, or recurrent/refractory solid tumors who have exhausted standard therapeutic options or have developed resistance to current treatments, including established immunotherapies like checkpoint inhibitors.[9] DS-1055a is being specifically evaluated in this challenging patient population [9], and its unique mechanism offers hope for a new line of defense. If DS-1055a demonstrates efficacy in patients who are resistant or refractory to current checkpoint inhibitors, it could fill a critical therapeutic gap and offer a novel strategy to overcome such resistance.

6.2. Considerations for Combination Therapies

The immunomodulatory action of DS-1055a, specifically the depletion of immunosuppressive Tregs, makes it an inherently attractive candidate for combination therapies. By alleviating Treg-mediated suppression within the TME, DS-1055a could create a more favorable environment for other anti-cancer agents to exert their effects. This is particularly relevant for combination with other immunotherapies, such as PD-1/PD-L1 checkpoint inhibitors. These inhibitors work by reinvigorating exhausted effector T cells; however, their activity can be significantly dampened by the presence of Tregs. Depleting these Tregs with DS-1055a could synergistically enhance the anti-tumor activity of checkpoint inhibitors, potentially leading to deeper and more durable responses, or even inducing responses in patients previously unresponsive to checkpoint monotherapy. This concept is supported by the preclinical findings where Treg depletion led to effector T cell activation [2] and the clinical trial rationale which includes evaluating DS-1055a in patients potentially resistant to checkpoint inhibitors.[9] Furthermore, combinations with conventional treatments like chemotherapy or targeted therapies might also be beneficial, as Treg infiltration can limit the efficacy of these modalities as well.

6.3. Future Research Directions

Following the completion and analysis of the ongoing Phase 1 trial, several key research directions will be critical for the further development of DS-1055a:

  • Biomarker Development: A crucial step will be the identification and validation of predictive biomarkers. This could include quantifying GARP expression levels on Tregs within the tumor or periphery, assessing the baseline density and phenotype of intratumoral Tregs, or characterizing broader tumor immune profiles. Such biomarkers, some of which are being explored in the current Phase 1 study [9], would be invaluable for selecting patients most likely to respond to DS-1055a, thereby optimizing its therapeutic application.
  • Tumor-Specific Efficacy: Based on signals of activity observed in the Phase 1 trial, focused Phase 2 studies in specific tumor types will be necessary to rigorously evaluate efficacy.
  • Combination Regimens: Designing and conducting well-controlled clinical trials to assess DS-1055a in combination with other anti-cancer agents, particularly checkpoint inhibitors, will be a high priority.
  • Mechanisms of Response and Resistance: Investigating the molecular and cellular mechanisms underlying response and potential resistance to DS-1055a will provide insights for optimizing treatment strategies and developing approaches to overcome resistance.

The clinical success of DS-1055a would significantly validate GARP as a druggable immuno-oncology target, potentially accelerating the development of other GARP-targeting agents and modalities currently in earlier stages of research.[10] Moreover, Daiichi Sankyo's application of afucosylation technology for DS-1055a [1] may indicate a broader strategic emphasis on advanced antibody engineering to enhance effector functions within their oncology portfolio, potentially providing a competitive edge in the development of next-generation antibody-based therapeutics.

7. Conclusion

DS-1055a is an afucosylated human monoclonal antibody representing a novel investigational approach in cancer immunotherapy. Its mechanism of action, centered on the selective targeting and depletion of GARP-expressing activated regulatory T cells within the tumor microenvironment, addresses a critical pathway of immune evasion utilized by cancer cells. Preclinical studies have provided a strong rationale for its development, demonstrating efficient Treg depletion, subsequent activation of effector T cells, and significant anti-tumor activity in relevant humanized mouse models.

The ongoing global Phase 1 clinical trial (NCT04419532) is a crucial step in translating these preclinical findings into the human setting. This study aims to establish the safety, tolerability, and optimal dosing of DS-1055a, while also gathering preliminary data on its pharmacokinetic profile, immunogenicity, and anti-tumor activity in patients with advanced solid tumors who have limited treatment options. The deliberate afucosylation of DS-1055a to enhance ADCC underscores a sophisticated antibody engineering strategy aimed at maximizing its Treg-depleting potential.

The therapeutic potential of DS-1055a is significant, particularly in its promise to overcome resistance to existing immunotherapies and to serve as an effective component in future combination treatment regimens. The journey of DS-1055a from discovery platforms to early-stage clinical evaluation highlights the complex but vital process of innovative drug development in oncology. The outcomes of the current and subsequent clinical trials will be pivotal in defining the ultimate clinical utility and role of DS-1055a in the evolving landscape of cancer treatment. The focus on depleting "highly suppressive" GARP+ Tregs suggests a refined strategy aimed at achieving meaningful immune reconstitution within the TME, potentially leading to durable clinical benefits for patients.

Works cited

  1. Definition of anti-GARP monoclonal antibody DS-1055a - NCI Drug Dictionary, accessed May 16, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/anti-garp-monoclonal-antibody-ds-1055a
  2. DS-1055 - Drug Targets, Indications, Patents - PatSnap Synapse, accessed May 16, 2025, https://synapse.patsnap.com/drug/01b00b23f957405ca4b37792562810ba
  3. Novel anti-GARP antibody DS-1055a augments antitumor immunity ..., accessed May 16, 2025, https://www.researchgate.net/publication/352164304_Novel_anti-GARP_antibody_DS-1055a_augments_antitumor_immunity_by_depleting_highly_suppressive_GARP_regulatory_T_cells
  4. DS 1055a - AdisInsight - Springer, accessed May 16, 2025, https://adisinsight.springer.com/drugs/800058954
  5. BioInvent receives €2 million milestone from Daiichi Sankyo - BioSpace, accessed May 16, 2025, https://www.biospace.com/bioinvent-receives-2-million-milestone-from-daiichi-sankyo
  6. BioInvent receives €2 million milestone from Daiichi Sankyo, accessed May 16, 2025, https://www.bioinvent.se/en/press/bioinvent-receives-eu2-million-milestone-daiichi-sankyo-1855679
  7. A Study Evaluating DS-1055a in Participants With Relapsed... - Clinical Trial Discovery, accessed May 16, 2025, https://clinicaltrial.be/en/details/55365?per_page=20&only_recruiting=0&only_eligible=0&only_active=0
  8. Therapeutic Targeting of Regulatory T Cells in Cancer - PMC - PubMed Central, accessed May 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9588644/
  9. Daiichi Sankyo Initiates Phase 1 Trial with Immuno-Oncology Therapy DS-1055 Targeting GARP on Activated Regulatory T Cells - Press Releases - Media, accessed May 16, 2025, https://www.daiichisankyo.com/media/press_release/detail/index_4081.html
  10. AbbVie sees a world according to GARP | ApexOnco - Oncology Pipeline, accessed May 16, 2025, https://www.oncologypipeline.com/apexonco/abbvie-sees-world-according-garp
  11. Clinical Trial: NCT04419532 - Solid Tumors - My Cancer Genome, accessed May 16, 2025, https://www.mycancergenome.org/content/clinical_trials/NCT04419532/
  12. A Study Evaluating DS-1055a in Participants With Relapsed or Refractory Locally Advanced or Metastatic Solid Tumors - NCI, accessed May 16, 2025, https://www.cancer.gov/research/participate/clinical-trials-search/v?id=NCI-2021-08880
  13. A Study Evaluating DS-1055a in Participants With Relapsed or Refractory Locally Advanced or Metastatic Solid Tumors - ClinicalTrials.Veeva, accessed May 16, 2025, https://ctv.veeva.com/study/a-study-evaluating-ds-1055a-in-participants-with-relapsed-or-refractory-locally-advanced-or-metastat

Published at: May 16, 2025

This report is continuously updated as new research emerges.

© Copyright 2025. All Rights Reserved by MedPath