What drugs are in development for hemophagocytic lymphohistiocytosis?

12 March 2025
Overview of Hemophagocytic Lymphohistiocytosis (HLH)

Definition and Pathophysiology
Hemophagocytic lymphohistiocytosis (HLH) is a rare, life‐threatening syndrome defined by an uncontrolled and hyperinflammatory immune activation. The condition is characterized by the aberrant activation of cytotoxic T lymphocytes, natural killer (NK) cells, and macrophages that leads to an overwhelming cytokine storm and multiorgan dysfunction. At its core, HLH involves an imbalance between immune activation and regulatory mechanisms, often triggered by infections, malignancies, or autoimmune processes, with some cases being familial (primary HLH) due to genetic defects in cytotoxic pathways and others being acquired (secondary HLH). The excessive release of pro-inflammatory cytokines such as interferon-γ (IFN-γ), interleukins (IL-1, IL-6, IL-18), tumor necrosis factor-α (TNF-α), and other mediators contributes to clinical features that include persistent high fever, pancytopenia, hepatosplenomegaly, and coagulopathy. Diagnosis is typically based on established criteria (HLH-2004) that include laboratory findings (elevated ferritin, hypertriglyceridemia, and hypofibrinogenemia) and clinical evidence such as organomegaly and hemophagocytosis on biopsy.

Current Treatment Approaches
Historically, the management of HLH has revolved around aggressive immunosuppressive and cytotoxic regimens. The HLH-94 and HLH-2004 protocols represent cornerstones in treatment—these regimens employ combinations of corticosteroids, etoposide, and, in some cases, cyclosporine A to dampen the hyperactive immune response. For certain patients, particularly those with familial HLH or refractory disease, allogeneic hematopoietic stem cell transplantation (HSCT) is considered the definitive curative option. More recently, clinicians have also repurposed targeted biologicals and immunomodulatory drugs to mitigate the adverse effects of an excessive cytokine storm. Among these, interferon-γ blockade (using agents like emapalumab), interleukin antagonists (such as anakinra targeting IL-1), and Janus kinase (JAK) inhibitors (e.g., ruxolitinib) have been introduced in off-label or experimental settings to better control inflammation, although not all are yet formally approved for HLH. These newer approaches have begun to shift treatment paradigms toward more targeted immunotherapy, prompting the exploration of drugs in development specifically for HLH.

Drug Development Landscape for HLH

Overview of Drugs in Development
Given the complex and rapidly evolving nature of HLH, a substantial research effort is underway to identify and develop agents that can more specifically target the underlying hyperinflammatory processes while reducing treatment‐related toxicity. The strategic focus of drug development in HLH is twofold: (1) to modulate cytokine signaling pathways that drive immune dysregulation, and (2) to provide potent immunomodulatory effects that can restore balance in the immune system before irreversible organ damage occurs.

One of the leading strategies has been the development of cytokine-targeting biologicals. Emapalumab, an anti–IFN-γ monoclonal antibody, represents a milestone in HLH therapy and is the first agent approved specifically for refractory, recurrent, or progressive primary HLH. Building on this success, newer agents and next-generation cytokine-targeted therapies are in various stages of development. For example, Electra Therapeutics is advancing ELA026, a monoclonal antibody that selectively depletes pathological immune cell populations, particularly in secondary HLH (sHLH) triggered by malignancies, infections, or autoimmune processes. ELA026 is currently undergoing early-phase clinical studies as part of a broader effort to tackle HLH through precise immune modulation.

In parallel, drug development efforts are exploring the repurposing and refinement of known immunomodulatory agents for HLH. Janus kinase (JAK) inhibitors such as ruxolitinib, which are approved for other hyperinflammatory and myeloproliferative conditions, have shown promising activity in controlling cytokine release in HLH. Their ability to inhibit multiple downstream cytokine signals makes them attractive candidates for HLH intervention, particularly when standard chemoimmunotherapy is not tolerated or is unsuccessful. Additionally, interleukin (IL)-1 inhibitors (for example, anakinra) and potentially IL-6 inhibitors are being evaluated for their efficacy in dampening the inflammatory cascade that underpins HLH.

Combination therapies are also on the agenda. Researchers are investigating synergistic effects when combining targeted biologicals with conventional therapies, such as adding cytokine-blocking antibodies to reduced-intensity chemotherapy or incorporating JAK inhibitors with corticosteroids for improved early response while maintaining a favorable safety profile. This multi-pronged approach is aimed at addressing both the massive inflammatory burden and the rapid, life-threatening deterioration that characterizes HLH.

Clinical Trial Phases and Key Players
A significant portion of current drug development involves early-phase clinical trials aimed at assessing safety, tolerability, and preliminary efficacy. ELA026, for instance, is in initial clinical testing, with early data expected to inform later phase studies. In addition to ELA026, repurposed agents like ruxolitinib are being tested in controlled clinical settings to define their role in HLH therapy. Several multicenter, prospective trials are underway, including studies designed to evaluate salvage treatments in adult HLH.

Key pharmaceutical and biotechnology companies are emerging as leaders in this space. Electra Therapeutics is notable for its targeted approach with ELA026, while other companies previously focused on cytokine or JAK inhibitors for autoimmune diseases and hematologic malignancies (e.g., Novartis, AbbVie, and Incyte Corp.) are now extending their research portfolios to include HLH indications. Moreover, academic institutions and collaborative research networks are playing a pivotal role in designing and implementing innovative clinical trials that specifically address the HLH patient population.

These studies not only assess pharmacokinetics and safety profiles but also aim to define biomarkers that can predict treatment response and guide personalized therapy. With HLH being a condition associated with high unmet need and mortality, regulatory agencies are showing increasing interest in supporting accelerated approval pathways based on promising early-phase data. The overall trend indicates a move away from blanket immunosuppressive regimens to more nuanced, mechanism-based therapies that offer improved efficacy with a reduced risk profile.

Mechanisms of Action

Targeted Therapies
The mechanistic rationale behind many novel agents for HLH centers on precise interruption of the cytokine networks that fuel the hyperinflammatory state. In primary HLH, defects in cytolytic pathways lead to unchecked T-cell and macrophage activation; therefore, one strategy is to directly inhibit the key cytokines that mediate these effects. Emapalumab’s mechanism—binding and neutralizing IFN-γ—epitomizes this approach. IFN-γ is a central mediator that activates macrophages and propagates further cytokine production, and its neutralization has been shown to improve survival in primary HLH cases.

Similarly, next-generation drugs like ELA026 are designed to deplete or modulate selected immune cell subsets that are excessively activated in HLH. By targeting specific surface markers or pathways unique to these pathological cells, such agents aim to restore a semblance of immune homeostasis without broadly suppressing the entire immune system. This targeted depletion strategy is particularly appealing in secondary HLH, where the underlying trigger may require simultaneous treatment.

Another component of targeted therapy in HLH is the use of agents inhibiting intracellular signaling mediators. Janus kinase inhibitors, such as ruxolitinib, work by blocking the JAK-STAT pathway, a critical conduit for a number of cytokines that drive the inflammatory process, including IFN-γ, IL-6, and others. By impeding this pathway, JAK inhibitors can potentially reduce the cytokine storm and mitigate downstream tissue damage. Their pleiotropic inhibition offers a broader dampening effect on hypercytokinemia compared to single cytokine blockade, making them suitable for cases that may involve multiple overlapping cytokine pathways.

Immunomodulatory Agents
Beyond direct cytokine blockade, immunomodulatory agents are being explored for their ability to fine-tune the immune response. Corticosteroids remain the backbone of HLH treatment; however, their non-specific immunosuppressive actions can lead to significant side effects. In contrast, agents such as anakinra—an IL-1 receptor antagonist—offer a more targeted approach by specifically inhibiting the pro-inflammatory actions of IL-1, which is known to be highly elevated in HLH. Anecdotal reports and small case series have demonstrated that anakinra can be effective in reducing inflammation and improving clinical outcomes in certain HLH cases, especially when used as part of combination regimens.

Other intermediate approaches include the use of IL-6 inhibitors (like tocilizumab) and therapies targeting other interleukins or costimulatory molecules involved in T-cell activation. These agents, while not yet specifically approved for HLH, are undergoing investigation either as monotherapy or in combination with other immunosuppressants to determine their potential role in controlling the inflammatory cascade in HLH patients.

A further promising area is the exploration of combination strategies that incorporate both targeted cytokine blockade and broader immunomodulatory agents. For example, combining low-dose chemotherapy (or cytotoxic agents) with targeted anti-cytokine therapy may allow for rapid control of the inflammatory process while minimizing the risk of relapse. This approach reflects a broader trend in immunotherapy, where multi-agent regimens are increasingly recognized as necessary to achieve sustained disease remission in complex inflammatory disorders.

Challenges and Future Directions

Current Challenges in Drug Development
Despite significant progress, several challenges persist in the development of drugs for HLH. One of the primary hurdles is the heterogeneity of the patient population. HLH can arise in a variety of contexts—ranging from genetic predisposition to triggers such as infections, malignancies, or autoimmune diseases—which means that a “one-size-fits-all” therapeutic approach is rarely appropriate. This heterogeneity complicates clinical trial design, patient enrollment, and the identification of robust biomarkers for treatment response. As a result, many clinical trials must contend with small sample sizes and diverse underlying pathologies, making it difficult to draw definitive conclusions.

Safety is another major concern. Given that HLH treatments involve potent immune suppression, there is always a risk of opportunistic infections and other complications. The balance between sufficiently dampening the hyperinflammatory response and avoiding excessive immunosuppression is delicate. As new agents—especially those targeting specific cytokines or signaling pathways—enter clinical trials, their safety profiles need to be rigorously evaluated and compared with existing regimens. Adverse events such as secondary infections, cytopenias, and organ toxicity remain significant barriers to broader adoption.

Regulatory challenges also exist. Because HLH is a rare disease, obtaining large-scale, statistically powered clinical trial data is inherently challenging. Many promising agents are currently only supported by early-phase data or small case series, which can make regulatory approval pathways less straightforward. There is an ongoing need for adaptive trial designs and international collaborations to pool data and accelerate the development process.

Lastly, the rapid evolution of diagnostic criteria and the emergence of novel biomarkers further complicate drug development. Accurate and early diagnosis is essential for effective treatment; however, the diagnostic landscape for HLH is continually evolving. This can result in variability in patient selection across trials and affect the overall assessment of therapeutic efficacy.

Future Prospects and Research Directions
Looking ahead, the road to more effective therapies for HLH is promising but will require a multi-pronged approach that spans basic research, translational studies, and clinical development. Future research is likely to focus on the following key areas:

1. Biomarker Development and Personalized Medicine:
The identification and validation of reliable biomarkers are crucial for early diagnosis, patient stratification, and monitoring treatment response. Future studies are expected to refine molecular and immunologic markers (e.g., cytokine profiles, NK cell activity, and genetic mutations) that can help tailor therapies to individual patients.

2. Advanced Combination Therapies:
Combination regimens that integrate targeted biologicals (such as anti–IFN-γ antibodies or JAK inhibitors) with conventional treatments are a major focus of ongoing research. These combinations aim to synergistically suppress the cytokine cascade while reducing toxicity. Adaptive clinical trial designs that allow for real-time intervention adjustments based on early biomarker responses are likely to play a central role in future therapeutic strategies.

3. Next-Generation Cytokine-Targeting Agents:
Building on the success of emapalumab, next-generation monoclonal antibodies and fusion proteins that target other key cytokines (such as IL-1, IL-6, and IL-18) or their receptors are under active investigation. These agents may offer a broader therapeutic window and could be used either alone or in combination with other immunomodulatory agents.

4. Cellular Therapies and Gene Editing:
Although still in early stages, cellular therapies—including engineered T cells or NK cells—offer intriguing possibilities for restoring immune regulation in HLH, particularly in familial cases. Concurrently, advances in gene therapy and gene editing technologies (such as CRISPR-Cas9) may ultimately provide curative approaches for primary HLH by correcting underlying genetic defects.

5. International Collaboration and Regulatory Innovation:
Due to the rarity of HLH, international multicenter collaborations are essential to conduct sufficiently powered clinical trials. Regulatory agencies are increasingly open to adaptive trial designs and accelerated approval pathways for rare diseases. Future regulatory strategies may be informed by real-world evidence and biomarker-driven outcomes, thereby shortening the time from discovery to clinical use.

6. Safety and Tolerability Improvements:
Addressing the safety concerns that have historically limited treatment outcomes remains a priority. Future research is likely to focus on identifying the optimal dosing regimens, mitigating off-target effects, and developing risk management strategies to prevent adverse events such as severe infections. Tailoring treatment intensity based on individual patient risk profiles will be key to maximizing therapeutic efficacy while minimizing harm.

Detailed Conclusion

In conclusion, the drug development landscape for hemophagocytic lymphohistiocytosis (HLH) is witnessing an exciting evolution from non-specific, high-toxicity regimens toward precision immunotherapy. Current research efforts are focusing on a wide array of drugs, including next-generation cytokine-targeted agents, repurposed immunomodulatory therapies, and innovative combination strategies designed to restore immune balance while minimizing adverse effects. Agents like ELA026—an emerging monoclonal antibody from Electra Therapeutics—as well as the continued exploration of JAK inhibitors (such as ruxolitinib) and interleukin antagonists (such as anakinra) represent major advances in our evolving understanding of HLH pathophysiology.

From a clinical trial perspective, early-phase studies are paving the way for more extensive multicenter trials, despite challenges posed by disease heterogeneity, safety concerns, and regulatory hurdles. The integration of biomarker development and adaptive clinical designs is expected to refine patient selection and enhance treatment personalization. Furthermore, advances in cellular therapies and gene editing offer a glimpse into a future where definitive cures for primary HLH may become a reality.

The hurdles in drug development for HLH—ranging from small patient populations and diagnostic variability to balancing efficacy with safety—necessitate international cooperation and innovative trial methodologies. However, the progress to date, bolstered by promising early-phase data and strategic repurposing of existing agents, suggests that the next few years may well see significant improvements in patient outcomes.

Overall, the future of HLH treatment lies in a generalized shift—from broad-based immunosuppression to targeted immunomodulation—enabling clinicians to achieve rapid control of hyperinflammation while preserving necessary immune function. As research continues to unravel the molecular underpinnings of HLH and lead to the development of more specific therapies, patients are likely to benefit from more effective, less toxic treatment regimens that are tailored to both the underlying cause and the individual patient’s profile. This integrated, multi-angle strategy—spanning targeted therapies, combination regimens, and personalized medicine—points toward a brighter future for the management of this challenging, life-threatening syndrome.

In summary, drugs currently in development for HLH include next-generation cytokine-targeted therapies (such as ELA026 and other agents that modulate IFN-γ and related cytokines), repurposed immunomodulatory agents (including JAK inhibitors like ruxolitinib and IL-1 inhibitors such as anakinra), and innovative combination therapies that seek to optimize both efficacy and safety. Future research directions aim to overcome existing challenges through enhanced biomarker-driven approaches, adaptive clinical trial designs, and potentially curative cellular or gene therapies. These developments are critical for improving outcomes in HLH, a disease where early, targeted intervention can make the difference between life and death.

Curious to see how Eureka LS fits into your workflow? 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.