What drugs are in development for Chronic graft-versus-host disease?

12 March 2025
Overview of Chronic Graft-Versus-Host Disease

Chronic graft-versus-host disease (cGVHD) is a major, long-term complication of allogeneic hematopoietic stem cell transplantation (HSCT) that substantially affects patient survival and quality of life. It is an immune-mediated syndrome in which donor immune cells attack host tissues, producing a clinical picture with multi-organ involvement and significant morbidity. Over recent decades, research has deepened our understanding of its complex pathophysiology, and novel therapeutic targets continue to emerge. In this section we provide a framework for understanding cGVHD from its definition and symptomatic manifestations to its current treatment landscape.

Definition and SymptomsChronic GVHDHD is defined as an immunologically mediated disorder that typically develops several months after transplantation. It involves immune dysregulation whereby mature donor T cells and B cells recognize host antigens as foreign, provoking ongoing inflammatory responses and tissue injury. Clinically, it can affect nearly every organ system—including the skin, liver, eyes, oral cavity, gastrointestinal tract, joints, fascia, and lungs—and present with heterogeneous clinical manifestations ranging from lichenoid changes in mucosal areas, sclerodermatous skin lesions, to progressive organ fibrosis. The disease often manifests with features such as erythema, dryness, skin tightening, and in severe cases, irreversible fibrosis that causes functional impairment. Patients may also experience systemic symptoms such as fatigue, pain, and a reduction in overall physical function. Because the spectrum of symptoms is so wide and often overlaps with those seen in autoimmune diseases, detailed clinical evaluation along with biomarker and histopathologic studies are essential for accurate diagnosis.

Current Treatment Landscape

Currently, the standard first-line treatment for cGVHD is systemic corticosteroid therapy, which, despite its widespread use, is often associated with significant adverse effects, including an increased risk of infections, metabolic complications, and long-term organ toxicity. For patients who are corticosteroid-refractory or dependent, several agents have emerged over recent years. Among these, ibrutinib (a Bruton's tyrosine kinase inhibitor) has received regulatory approval for both adults and pediatric patients in select jurisdictions. In addition, ruxolitinib, a Janus kinase (JAK) 1/2 inhibitor, has demonstrated efficacy in modulating the inflammatory cytokine cascade, thereby mitigating disease progression. More recently, belumosudil—a selective inhibitor of Rho-associated coiled-coil containing protein kinase 2 (ROCK2)—has been approved for treating patients with refractory cGVHD, representing a new step toward mechanism-based therapy. Although these drugs represent significant advances, a substantial proportion of patients do not experience sustained benefit, and considerable unmet needs remain. Therefore, research continues not only to optimize the use of currently approved therapies but also to investigate novel drug candidates with more favorable efficacy and tolerability profiles.

Drugs in Development for cGVHD

A vibrant and diverse pipeline of drug candidates is in development for the treatment of cGVHD. These candidate drugs are being designed on the basis of our increasing understanding of the immunopathogenesis of the disease. Their mechanisms of action target distinct pathways—for example, blocks of signaling pathways that drive cytokine production, modulation of immune cell subsets (such as T regulatory cells), and inhibition of pro-fibrotic processes. In this section we describe the novel drug candidates under development, discuss their mechanisms of action, and review the clinical trial phases in which they currently reside.

Novel Drug Candidates

A number of innovative drug candidates are currently emerging in the pipeline for cGVHD. Among the most noteworthy are:

1. Axatilimab
Axatilimab is a monoclonal antibody targeting the colony stimulating factor-1 receptor (CSF-1R). By blocking CSF-1R, axatilimab modulates the activity of monocytes and macrophages, cell types implicated not only in driving inflammation but also in promoting fibrosis—a key feature in cGVHD pathogenesis. Clinical trials (such as the ongoing global pivotal Phase 2 AGAVE-201 trial) are currently evaluating axatilimab’s safety and efficacy profile, with updated results expected in the near future.

2. ROCK2 Inhibitors (Rezurock™ and Analogues)
ROCK2 inhibitors represent one of the most promising new classes of drugs for cGVHD. These compounds, such as the one represented by Rezurock™, inhibit ROCK2—a kinase critical for T-cell differentiation and fibrotic processes. By suppressing ROCK2, these drugs may simultaneously attenuate inflammation and reduce pathological fibrosis, thereby addressing two central components of cGVHD. Several compounds with similar chemical structures are undergoing early-phase clinical trials.

3. Janus Kinase (JAK) Inhibitors
Although ruxolitinib is already approved in this setting, several next-generation JAK inhibitors are in development to improve selectivity and reduce side effects. These molecules target JAK-STAT signaling which plays a fundamental role in propagating inflammatory responses in cGVHD. Novel candidates at different clinical stages are being tested for their ability to offer more potent immunomodulation while minimizing the risk of cytopenias and infections.

4. Bruton's Tyrosine Kinase (BTK) Inhibitors
Ibrutinib has already paved the way for BTK inhibitors in cGVHD. However, second-generation BTK inhibitors are now under investigation, with efforts focused on improving efficacy and tolerability, particularly in patients who are refractory to ibrutinib. These drugs focus on targeting B-cell receptor signaling and modulating T-cell responses simultaneously.

5. Integrin α4β7 Targeting Agents
Novel agents that target integrin α4β7 are also in development. These drugs are designed to disrupt the trafficking of lymphocytes to the gastrointestinal tract and other tissues impacted by GVHD. By modulating cell adhesion and migration, they could reduce localized inflammatory damage and tissue injury.

6. CD80/CD86 Inhibitors
Some approaches are exploring the use of costimulatory blockade by targeting molecules such as CD80 and CD86. These drugs, often in the form of fusion proteins or monoclonal antibodies, reduce T-cell activation and mitigate the initiation of GVHD responses. Such agents may offer a more refined approach to immune modulation that spares beneficial graft-versus-leukemia (GVL) effects.

7. Other Emerging Targets and Combinations
In addition to the aforementioned classes, several other novel candidates are also under investigation. These include agents targeting cytokines like interleukin-6 (IL-6) receptor inhibitors, modulators of the NF-κB pathway, and cellular therapies that promote regulatory T-cell expansion. Some drug candidates aim to modulate the microenvironment by targeting fibrosis-associated pathways (for example, transforming growth factor-beta (TGF-β)). There is also interest in developing combined regimens that utilize agents with complementary mechanisms—for instance, combining a JAK inhibitor with a ROCK2 inhibitor—to achieve synergistic effects and enhance response rates.

Mechanisms of Action

The drugs in development for cGVHD are built upon a detailed understanding of the pathogenesis of the disease. Their mechanisms of action can be broadly classified into the following categories:

1. Immunomodulation and Cytokine Signal Blockade:
Many cGVHD drugs aim to blunt the overactive immune responses that cause tissue damage. For example, JAK inhibitors interrupt the JAK-STAT pathway, thereby reducing the production and action of key cytokines such as interferon-gamma and interleukins that drive inflammation. Similarly, BTK inhibitors reduce B-cell mediated activation and also modulate T-cell responses, ultimately decreasing the chronic inflammatory milieu.

2. Inhibition of Pro-Fibrotic Processes:
Fibrosis is a hallmark of advanced cGVHD. ROCK2 inhibitors, such as Rezurock™, work by interfering with intracellular signaling that leads to the differentiation of fibroblasts and the deposition of extracellular matrix components. By inhibiting ROCK2, these drugs help prevent the progression of tissue scarring and stiffness, thereby preserving organ function.

3. Modulation of Macrophage and Monocyte Function:
Axatilimab, which targets CSF-1R, exemplifies strategies aimed at reprogramming the behavior of monocytes and macrophages. These cells are key players in both the inflammatory and fibrotic aspects of cGVHD. Blocking CSF-1R may reduce the secretion of pro-inflammatory cytokines and limit the fibrotic cascade, resulting in improved tissue repair and reduced organ dysfunction.

4. Blockade of Costimulatory Signals:
Agents targeting costimulatory molecules such as CD80/CD86 function by preventing the required second signal for full T-cell activation. This blockade can dampen the alloreactive T-cell responses that underpin GVHD, while maintaining overall immune competence.

5. Disruption of Cellular Trafficking and Adhesion:
By targeting integrin α4β7, certain novel agents aim to reduce the migration of immune cells into vulnerable tissues such as the gut. This mechanism helps to limit localized inflammatory responses and tissue damage, ultimately contributing to a reduction in clinical GVHD manifestations.

Clinical Trial Phases

Drug candidates under development for cGVHD are at various stages in clinical testing:

1. Early-Phase Clinical Trials (Phase 1/2):
Many novel agents, including axatilimab and second-generation ROCK2 inhibitors, are currently being evaluated in early-phase studies. These trials primarily focus on determining safety, pharmacokinetics, and preliminary efficacy. For instance, the AGAVE-201 trial for axatilimab is in Phase 2 and is designed to assess its clinical efficacy and safety profile in patients with refractory cGVHD. Similarly, some of the newer JAK and BTK inhibitors are in Phase 1 or Phase 1/2 trials where dose escalation and safety endpoints are being defined.

2. Late-Stage Clinical Trials (Phase 3):
While some drugs have already received regulatory approval in select patient populations (e.g., ibrutinib and ruxolitinib), much of the drug pipeline remains in Phase 3 where efficacy is compared against best available therapies or placebo. The transition to Phase 3 trials is occurring as earlier studies have validated promising activity in smaller cohorts. These late-stage trials are critical not only for the assessment of clinical benefit across a broader patient population but also for confirming long-term outcomes, such as progression-free survival and overall survival.

3. Combination and Adaptive Clinical Trials:
In recognition of the multifactorial nature of cGVHD, several studies are now incorporating combination regimens into their design. These adaptive trials allow for the evaluation of multiple agents simultaneously to determine the most effective combination therapy. For example, trials combining JAK inhibitors with ROCK2 inhibitors or integrating costimulatory blockers with antifibrotic agents are on the horizon. In these studies, biomarkers are also being evaluated to stratify patients and personalize therapies, thereby increasing the chance of clinical success.

Challenges in Drug Development for cGVHD

Despite the ongoing progress, the development of novel drugs for cGVHD faces several challenges. These challenges span scientific, clinical, regulatory and market-related barriers.

Scientific and Clinical Challenges

1. Heterogeneity of Disease Manifestation:
One of the foremost challenges in cGVHD drug development is the heterogeneous nature of the disease. cGVHD can affect multiple organs simultaneously, and the pathophysiological mechanisms can vary significantly between patients—even among those with similar clinical grades. This heterogeneity complicates the design of clinical trials and the interpretation of endpoints since a drug’s efficacy may vary depending on the specific organ involvement and dominant pathogenic pathways.

2. Complex Pathogenesis:
The multifaceted immune dysregulation in cGVHD involves various cellular components—T cells, B cells, macrophages, and fibroblasts—as well as multiple cytokine and chemokine networks. The interplay between these factors not only drives disease progression but also affects treatment responsiveness. Drugs targeting one pathway may be insufficient to control the disease if compensatory mechanisms exist. For instance, inhibition of inflammatory cytokines may not fully address underlying fibrosis, and vice versa. This complexity necessitates the development of drugs with either broad-spectrum effects or the use of combination therapies, both of which present challenges in terms of safety and tolerability.

3. Biomarker Identification and Endpoint Assessment:
The lack of standardized biomarkers for diagnosis, prognosis, and therapeutic response is a significant hurdle. Although many potential biomarkers have been identified (e.g., autoantibodies, cytokine profiles, extracellular vesicles, microRNAs), their validation in multicenter clinical trials remains incomplete. This gap in biomarker-driven stratification makes it difficult to identify patient subsets that are most likely to benefit from targeted therapies, and complicates the assessment of clinical trial endpoints.

4. Safety and Tolerability Issues:
Many novel agents, especially those that modulate the immune system, carry the risk of increased infections and other immunosuppression-related toxicities. Balancing efficacy with an acceptable safety profile is a critical challenge, particularly in a patient population that is already vulnerable due to previous transplant-related complications and concurrent therapies. For instance, while JAK inhibitors are highly effective at reducing inflammation, they can also predispose patients to cytopenias and opportunistic infections.

Regulatory and Market Challenges

1. Regulatory Hurdles:
Given the complexity of cGVHD and the variability in patient responses, regulatory agencies require extensive data demonstrating both efficacy and long-term safety before granting approvals. This necessitates large, multicenter Phase 3 trials with robust endpoints—trials that are time-consuming and expensive to conduct. There is also the challenge of designing studies that can integrate adaptive endpoints or biomarker-based selection criteria without jeopardizing the trial’s integrity.

2. Market Competition and Cost Considerations:
The field of cGVHD is becoming increasingly competitive with several drugs already approved in various jurisdictions. New entrants must therefore demonstrate either superior efficacy or a significantly improved safety profile compared to existing therapies. Additionally, the high cost associated with the development and production of biologics and targeted therapies can be a barrier to market entry. Companies must consider strategies to optimize pricing and reimbursement to ensure that the new therapies are accessible to the broad patient population that needs them.

3. Challenges in Combining Therapies:
As the trend shifts toward combination therapies to address the multifactorial nature of cGVHD, regulatory pathways become more complex. Determining the optimal dosing, timing, and sequence of combination regimens requires carefully designed clinical trials. Moreover, demonstrating that the combined regimen offers a significant advantage over monotherapy is challenging, yet essential for regulatory approval and subsequent market adoption.

Future Directions and Conclusions

The future of drug development for cGVHD is promising, with several innovative approaches and research areas offering hope for improved patient outcomes. This final section discusses promising research areas, potential impacts on patient outcomes, and provides a detailed conclusion based on the diverse perspectives discussed herein.

Promising Research Areas

1. Personalized Medicine and Biomarker Integration:
One of the most exciting future directions is the integration of novel biomarkers into both clinical trial design and routine patient management. Advances in genomics, proteomics, and metabolomics have the potential to identify biomarkers that accurately predict treatment responsiveness and disease trajectory. These biomarkers can be used for patient stratification such that therapies are personalized: a tailored approach that matches the patient’s unique immune profile with the most effective drug or drug combination. This approach not only increases the likelihood of achieving therapeutic success but also minimizes unnecessary exposure to toxic agents.

2. Combination Therapies and Synergistic Approaches:
As emphasized earlier, cGVHD is driven by multiple pathogenic mechanisms. Future research is likely to focus on combination regimens that target more than one pathway simultaneously. For instance, combining a JAK inhibitor to blunt inflammatory cytokine signaling with a ROCK2 inhibitor to reduce fibrosis could provide synergistic benefits. Such combination strategies may also include pairing a costimulatory blockade agent with a macrophage-modulating drug, thereby covering both the cellular activation and the fibrotic arms of the disease. Ongoing adaptive trials are beginning to explore such combinations.

3. Novel Targets and Pathways:
The continuous elucidation of the molecular and cellular mechanisms involved in cGVHD is leading to the identification of new therapeutic targets. For example, novel agents aimed at modulating the CSF-1R pathway (such as axatilimab) or those that target integrin-mediated trafficking (such as integrin α4β7 inhibitors) are promising. In addition, emerging data on the roles of epigenetic regulators and metabolic pathways offer new angles for intervention. Targeting these pathways may lead to the development of drugs that offer more durable responses with fewer side effects.

4. Advanced Clinical Trial Designs:
The evolution of clinical trial methodology—including adaptive and umbrella trial designs—will be critical for accelerating drug development. These trial designs allow for the simultaneous evaluation of multiple drug candidates and flexible modifications based on interim analyses. They also facilitate the incorporation of biomarker-driven endpoints which can more accurately reflect patient responses. As trials become more efficient, the overall time to bring new drugs to market is expected to decrease, benefiting patients sooner.

Potential Impact on Patient Outcomes

The development of new drug candidates and therapeutic strategies for cGVHD holds enormous promise for transforming patient care. Improved therapies are expected to offer the following benefits:

1. Increased Efficacy and Response Rates:
By targeting critical pathways with more specificity, newly developed agents may produce higher overall response rates and durable remissions in patients with refractory or steroid-dependent cGVHD. This, in turn, can lead to prolonged survival and a reduction in transplant-related morbidity.

2. Enhanced Quality of Life:
Drugs with improved safety profiles and reduced toxicity can significantly enhance the quality of life for patients. Reducing corticosteroid-associated side effects, for example, could alleviate issues such as osteoporosis, hyperglycemia, and increased infection risk. Moreover, therapies that limit fibrosis can preserve organ function and reduce chronic pain and disability.

3. Personalized Treatment Approaches:
The integration of biomarker-based personalized medicine will ensure that the right drug is given to the right patient at the right time. This strategy is likely to improve response rates and reduce unnecessary exposure to ineffective therapies. In the long term, personalized treatment regimens could revolutionize the management of cGVHD by offering tailored interventions that align with each patient’s unique disease biology.

4. Reduced Healthcare Burden:
Effective management of cGVHD with novel agents not only improves patient outcomes but also has the potential to reduce healthcare costs by decreasing hospitalization rates, limiting the need for prolonged immunosuppressive therapy, and reducing the incidence of treatment-related complications.

Detailed and Explicit Conclusion

In conclusion, the current landscape of drug development for chronic graft-versus-host disease is characterized by a diverse array of novel candidates designed to target the complex and multifaceted mechanisms underlying the disease. Our understanding of cGVHD has evolved significantly over the past decade—from recognizing the heterogeneous clinical presentation and identifying key pathogenic pathways to the development of targeted therapies that modulate immune responses, reduce fibrosis, and improve patient outcomes.

Key novel drug candidates include axatilimab, a CSF-1R targeting monoclonal antibody; ROCK2 inhibitors, such as Rezurock™, which aim to attenuate both inflammatory and fibrotic processes; and emerging second-generation JAK and BTK inhibitors designed to refine the immunomodulation achieved with their first-generation counterparts. Additionally, agents that target integrin α4β7 and costimulatory molecules (such as CD80/CD86) offer promising avenues by disrupting cellular trafficking and T-cell activation, respectively. Many of these drugs are currently in early-phase or late-phase clinical trials, with adaptive trial designs further expediting the evaluation of combination therapies that tackle multiple aspects of cGVHD pathogenesis concurrently.

However, significant challenges remain. The inherent heterogeneity of cGVHD, the complexity of its immunopathogenesis, and the lack of universally accepted biomarkers for diagnosis and response assessment create obstacles to both drug development and clinical trial design. Regulatory hurdles and market competition further complicate the pathway to approval. Nevertheless, advances in the integration of biomarker-driven personalized medicine and novel clinical trial methodologies are poised to overcome these challenges.

From a general perspective, the future of cGVHD treatment appears brighter as research continues to yield promising candidates with the potential to transform patient outcomes. Specifically, the transition towards personalized, multi-targeted therapeutic regimens is expected to not only improve overall response rates and survival but also enhance quality of life by reducing treatment-related toxicities. On a specific level, drugs such as axatilimab and ROCK2 inhibitors are at the forefront of this therapeutic revolution, with ongoing studies poised to solidify their roles in the clinical management of cGVHD. Finally, from a global perspective, continued cross-disciplinary collaboration and adaptive clinical trial designs will accelerate progress and help ensure that patients benefit from the latest advances in the field.

In summary, the development of drugs for chronic graft-versus-host disease is approaching a transformative era. With novel agents targeting key pathways such as CSF-1R, ROCK2, JAK-STAT, BTK, integrins, and costimulatory molecules, the potential exists to significantly alter the treatment paradigm of cGVHD. Future drug development will likely move toward rational combinatory approaches tailored to the individual patient’s disease biology, ultimately reducing morbidity and enhancing survival and quality of life. The integration of robust biomarker panels and innovative clinical trial designs will be crucial in this endeavor, ensuring that these promising therapies translate into clinical practice efficiently and safely.

Each drug candidate in development is not only a testament to our evolving understanding of cGVHD but also a beacon of hope for patients who struggle with this debilitating disease. The continued integration of preclinical insights, clinical data, and personalized medicine strategies promises a future in which cGVHD is managed more effectively with fewer toxicities, leading to sustained remissions and improved long-term outcomes for transplant recipients.

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.