How many FDA approved Immune stimulating antibody conjugate (ISAC) are there?

17 March 2025
Immune stimulating antibody conjugates (ISACs) represent a novel and innovative class of biopharmaceutical agents that are being engineered to not only target tumor cells with high specificity but also actively stimulate the immune system to recognize and attack these cancer cells. In contrast to conventional antibody–drug conjugates (ADCs), which deliver cytotoxic payloads with the primary aim of killing the tumor cell directly, ISACs incorporate immune stimulatory elements designed to recruit and activate components of the innate and adaptive immune systems. This dual mode of action holds the promise of establishing a long‐lasting antitumor immune response that may both eliminate cancer cells and imbue the immune system with memory against tumor-associated antigens.

ISACs are defined as antibody conjugates that are modified to include immune-stimulating molecules along with their targeting antibody. The targeting component is typically a monoclonal antibody (mAb) engineered to bind to a specific antigen on neoplastic cells. However, rather than being linked to a cytotoxic payload—as is the case with traditional ADCs—ISACs are linked to immune-stimulatory molecules such as Toll-like receptor (TLR) agonists. The conjugate is designed so that after binding and internalization into the tumor or tumor microenvironment, the immune stimulant is released to activate local antigen-presenting cells (APCs), such as dendritic cells and macrophages, triggering a cascade of immune activation events. This activation promotes the phagocytosis of tumor cells, primes T cell responses, and encourages epitope spreading—a process where the immune system learns to recognize multiple tumor-associated antigens.

The core idea behind ISACs is to convert immunologically “cold” tumors into “hot” ones that are more susceptible to immune attack. This is achieved by using the antibody as a guide to the tumor site while leveraging immune stimulation to overcome the immunosuppressive tumor microenvironment. In doing so, ISACs aim to generate durable anti-tumor immunity that not only targets the primary tumor cells but may also prevent recurrence by inducing long-term immunological memory.

The evolution of immuno-oncology has led to significant breakthroughs in recent years, with monoclonal antibodies and checkpoint inhibitors dramatically shifting treatment paradigms in oncology. Early antibody conjugation strategies focused on linking cytotoxins to antibodies (forming ADCs) to harness the specificity of antibodies while delivering potent chemotherapeutic agents directly to tumor cells. During this period, the concept of conjugating antibodies with immune-stimulatory agents was explored as a logical extension of ADC technology.

One notable milestone in this evolution was the development of ISACs by pharmaceutical innovators. For example, Bolt Biotherapeutics has been pioneering this approach with its Boltbody ISAC platform. Detailed descriptions of the Boltbody ISACs indicate that the engineered conjugates comprise a tumor-specific antibody, a non-cleavable linker, and a proprietary TLR7/8 agonist. Their mechanism involves targeting tumor cells and, upon binding, activating the local myeloid cells to create a feed-forward loop of immune activation and recruitment of adaptive immune cells. Although these early developments have generated significant scientific and clinical interest, they remain at the preclinical or early clinical phase, with no approved products on the market as of the latest reports.

The pathway to FDA approval for any biotherapeutic, and ISACs in particular, involves rigorous testing of safety, efficacy, and manufacturing consistency. The FDA approval process is designed to ensure that any new therapy provides a favorable benefit-to-risk ratio and meets strict quality and safety standards before it is made available for clinical use.

The FDA approval process for biotherapeutics, including innovative modalities like ISACs, generally follows several key stages:

1. Preclinical Studies: Extensive laboratory testing, including in vitro assays and animal model studies, is conducted to assess the fundamental pharmacological properties, toxicity profile, and mechanism of action of the candidate ISAC. These studies help in establishing a proof of concept and determining safe starting doses for human trials.

2. Investigational New Drug (IND) Application: Based on promising preclinical data, developers submit an IND application to the FDA. This submission includes detailed information on the manufacturing process, preclinical study outcomes, and a proposed clinical trial protocol. Only after the IND is cleared can clinical trials in humans begin.

3. Clinical Trials (Phase I-III):
- Phase I trials focus primarily on safety, tolerability, and pharmacokinetics in a small group of healthy volunteers or patients.
- Phase II trials provide preliminary data on efficacy while continuing to assess safety in a larger patient cohort.
- Phase III trials are pivotal studies designed to confirm efficacy and monitor adverse effects on a large scale. For novel modalities like ISACs, these phases are crucial in determining not only the therapeutic benefit but also the durability of immune responses generated.

4. New Biologic License Application (BLA): Upon successful completion of clinical trials, an applicant submits a New Biologic License Application to request formal FDA approval. The application presents all clinical data, manufacturing details, and other supporting documentation needed for a regulatory review.

5. FDA Review and Approval: The FDA reviews the BLA and, if the evidence demonstrates an acceptable benefit-to-risk profile along with robust manufacturing consistency and safety data, it approves the product for clinical use. This process is extensive and involves multiple expert evaluations and advisory committee meetings.

For ISACs to receive FDA approval, several specific evaluation criteria must be met:

- Safety and Tolerability: The ISAC must show an acceptable safety profile in both early-phase and pivotal clinical trials. Immune activation can sometimes lead to adverse events, such as cytokine release syndrome or off-target inflammatory effects, which need to be carefully managed and minimized.

- Efficacy: The immune stimulatory component of the ISAC must demonstrably induce a clinically meaningful antitumor immune response. This involves not only direct tumor regression but also indications that the therapy may promote ongoing immunologic memory or synergize with existing immunotherapies.

- Pharmacokinetics and Pharmacodynamics: The distribution, metabolism, and clearance of the ISAC, along with its immune activation metrics, are rigorously evaluated. Consistent and predictable behavior in the human body is a key requirement.

- Manufacturing Consistency and Quality: Batch consistency, purity, and stability of the ISAC are major concerns. The product must be manufactured under good manufacturing practices (GMP) and meet strict standards for potency and quality.

- Benefit-to-Risk Ratio: Ultimately, the FDA considers the overall clinical benefit relative to any risks posed by the therapy. ISACs must provide clear therapeutic advantages compared to available treatments while maintaining a manageable safety profile.

When addressing the specific question of “How many FDA approved Immune stimulating antibody conjugate (ISAC) are there?” it is crucial to distinguish between the conceptual promise of ISACs and the current regulatory status. Despite promising preclinical studies and early clinical evaluations exploring the potential of ISACs, as of the most up-to-date evidence available from the provided references, no ISAC has yet received FDA approval.

Both the scientific literature and the news articles provided within our references clearly articulate the innovative potential and the ongoing clinical development of ISACs. For example, reference details the biological rationale and early-stage research into immune-stimulating antibody conjugates without mentioning any products approved by the FDA. Similarly, reference discusses the Boltbody ISAC technology, including the lead candidate BDC-1001, but at the time of reporting, it remains under investigation in clinical trials rather than having achieved regulatory approval.

Thus, based on these structured sources from synapse, the answer is that there are currently zero (0) FDA approved immune stimulating antibody conjugates (ISACs).

Since none of the ISACs have been approved by the FDA at this time, there are no official indications or established clinical uses sanctioned by the regulatory agency. However, it is important to note that the clinical evaluation of ISACs is typically targeted at solid tumors that historically exhibit resistance to conventional chemotherapy and even some immune checkpoint inhibitors. Early clinical trials are investigating these agents in tumor types such as HER2-positive cancers and other malignancies where converting an immunologically “cold” tumor into a “hot” one could significantly improve response rates.

The design of ISACs implies potential use either as monotherapy or in synergistic combinations with other immunomodulatory treatments, such as immune checkpoint inhibitors, to potentially achieve enhanced antitumor activity. Nonetheless, these applications remain investigational and are not yet part of clinical practice due to the absence of any FDA approval for the ISAC class.

Although no ISACs are currently FDA approved, the preclinical and early clinical research on these conjugates has already made a significant impact on the oncology landscape. Their unique mechanism of action—which seeks to combine precise tumor targeting with potent immune system activation—is considered one of the most promising avenues in modern cancer immunotherapy. Early data suggest that ISACs might be capable of overcoming limitations seen with conventional therapies. For instance, by actively stimulating local immune responses, ISACs may help bypass the immunosuppressive mechanisms that often limit the efficacy of standard ADCs or checkpoint inhibitors. This could lead to more durable responses and potentially lower rates of tumor relapse.

The promise of ISACs also lies in their ability to harness both innate and adaptive immunity. Activated APCs can process tumor antigens and present them to T cells, thereby potentially generating a broad and long-lasting antitumor immune response. Additionally, the feed-forward loop established by the release of cytokines and chemokines might recruit further immune effector cells to the tumor microenvironment, amplifying the therapeutic effect. The clinical impact of such a mechanism could translate into improved survival outcomes for patients who otherwise have limited options, particularly in tumors that are resistant to current cytotoxic and targeted therapies.

Looking ahead, the future research for ISACs is likely to focus on several key areas:

1. Optimization of Target Selection: To maximize therapeutic efficacy, researchers are working on identifying new tumor-specific targets that are highly expressed on cancer cells but have limited expression in normal tissues. Such selective targeting will be critical in minimizing off-target immune activation and associated toxicities.

2. Refinement of Linker Chemistry and Payload Engineering: Future ISAC development will undoubtedly involve advancing linker technologies that allow for the controlled and efficient release of immune-stimulatory agents. Engineered payloads must be potent enough to activate immune responses without causing systemic inflammation. Innovations in this area may significantly enhance the therapeutic index of ISACs.

3. Combination Therapies: Given that the clinical landscape of cancer treatment is moving toward combination regimens, future trials may evaluate ISACs in tandem with other immunotherapies—such as checkpoint inhibitors—and even targeted therapies. Studies examining these combination approaches will be crucial in determining whether synergistic effects can be consistently achieved in a clinical setting.

4. Biomarker Development and Patient Stratification: As with all immunotherapies, identifying predictive biomarkers that can stratify patients based on their likely response to ISACs is a high priority. Such biomarkers would allow clinicians to select appropriate cohorts of patients, maximizing the benefit-to-risk ratio of the treatment. Genomic, proteomic, and even immune cell profiling strategies may be incorporated into future clinical studies.

5. Addressing Safety and Toxicity Profiles: One of the greatest challenges facing ISAC development is managing immune-related adverse events. Research initiatives are anticipated to explore refined dosing strategies, mitigate potential cytokine release syndromes, and develop supportive care protocols to handle any adverse reactions arising from excessive immune activation.

6. Regulatory Strategies and Accelerated Approvals: Given the pressing need for novel cancer therapies, innovative regulatory strategies such as accelerated approvals or breakthrough therapy designations may eventually be applied to promising ISAC candidates. Collaboration between developers and regulatory agencies will be critical to streamline the evaluation process while ensuring patient safety.

Overall, while ISACs have not yet crossed the finish line to FDA approval, the preclinical and early clinical data are highly encouraging. Their potential to redefine cancer treatment—particularly for patients with immunologically “cold” tumors—is driving substantial research momentum in this field.

In a general sense, immune stimulating antibody conjugates (ISACs) embody the next generation of immunotherapy by integrating antibody targeting with immune activation. Detailed mechanistic studies have elucidated how these agents can induce tumor-specific immune responses through the engagement of antigen-presenting cells and subsequent T-cell activation. Historically, the evolution from conventional ADCs to ISACs marks a significant milestone, reflecting ongoing progress in the field of immuno-oncology. However, when considering the rigorous FDA approval process—spanning preclinical investigations, clinical trials, and detailed evaluations of safety, efficacy, and manufacturability—it becomes evident that the ISAC concept, although promising, remains in the developmental stage.

Specifically, the examination of the available literature and news reports from reliable synapse sources shows that, as of now, there are no FDA approved ISACs—that is, the number of FDA approved immune stimulating antibody conjugates currently stands at zero. Thus, while ISACs continue to be a dynamic area of research with significant potential for future breakthroughs, no product in this category has yet met the stringent criteria for FDA approval for clinical use.

In summary, based on the current evidence and structured information from the synapse sources, the answer to the question "How many FDA approved Immune stimulating antibody conjugates (ISAC) are there?" is that there are none approved by the FDA at this time. Looking forward, continued research, improved design strategies, and innovative regulatory pathways may eventually lead to the approval of ISACs, potentially revolutionizing cancer therapies and improving clinical outcomes for patients with refractory malignancies.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.