What PDGFRα inhibitors are in clinical trials currently?

11 March 2025
Introduction to PDGFRα
PDGFRα, one of the two members of the platelet‐derived growth factor receptor family, plays a critical role in many physiological processes, including cellular growth, proliferation, differentiation, and the regulation of tissue development. It is a receptor tyrosine kinase (RTK) whose signaling cascade is often aberrantly activated in various malignancies. This activation can occur either through overexpression, gene amplification, or mutation and leads to the dysregulation of cell survival and proliferation pathways. As such, PDGFRα represents a high‐value target in oncology and several non‐oncologic settings where fibroproliferative and inflammatory processes contribute to disease pathology.

Role of PDGFRα in Disease
PDGFRα is found expressed in mesenchymal cells, and its ligand binding triggers receptor dimerization and activation of downstream signaling cascades, including the PI3K/AKT/mTOR and RAS/MAPK pathways. In cancers such as gastrointestinal stromal tumors (GISTs), gliomas, chordomas, and various sarcomas, mutations in PDGFRA can drive oncogenesis. PDGFRA-mutated tumors have distinct biological behaviors compared to those driven by other oncogenes and often display a differential response to conventional cytotoxic chemotherapy. In addition, PDGFRα’s involvement in aberrant angiogenesis and stromal interactions underscores its significance in tumor progression and metastasis formation.

Importance of Targeting PDGFRα
The rationale for targeting PDGFRα lies in its central role as a mediator of both oncogenic signaling and the tumor microenvironment. Inhibiting this receptor can disrupt autocrine and paracrine loops that foster tumor growth, angiogenesis, and metastasis. Moreover, because PDGFRα mutation or overexpression can serve as a molecular marker in select patient populations (for example, in PDGFRA-mutated GISTs), targeted inhibition offers the promise of personalized therapies with improved outcomes and reduced systemic toxicities compared to traditional chemotherapy. These inhibitors not only have the potential to induce direct antitumor effects but may also enhance the efficacy of combination regimens by modulating the tumor stroma and overcoming drug resistance mechanisms.

Overview of PDGFRα Inhibitors
A wide array of inhibitors have been developed to target PDGFRα, ranging from small-molecule tyrosine kinase inhibitors (TKIs) to monoclonal antibodies and, more recently, bispecific molecules engineered to enhance selectivity and efficacy. These agents are designed to interfere with PDGFRα signaling at different points, either by preventing ligand binding, blocking ATP binding to the kinase domain, or disrupting receptor dimerization and subsequent activation.

Mechanism of Action
PDGFRα inhibitors primarily function by competitively binding to the kinase domain of the receptor, thereby blocking the ATP binding site that is essential for autophosphorylation and activation of downstream signaling cascades. This interruption in signaling translates into reduced cell proliferation, induction of apoptosis, and diminution of angiogenic responses. In some instances, inhibitors may bind to an inactive “DFG-out” conformation of the kinase, thereby conferring a level of selectivity distinct from inhibitors that target the active conformation. Other approaches include the use of monoclonal antibodies or bispecific antibodies that prevent ligand-receptor interactions by binding directly to the extracellular domains of PDGFRα. Such strategies may additionally harness immune-mediated mechanisms to eliminate tumor cells or modulate the tumor microenvironment.

Types of PDGFRα Inhibitors
There are multiple classes of PDGFRα inhibitors currently under investigation, including:

• Small-Molecule Tyrosine Kinase Inhibitors (TKIs):
 – Many early studies focused on multi-targeted agents such as imatinib, sunitinib, and dasatinib. Though these drugs target multiple kinases including PDGFRα, their broad activity sometimes leads to off-target effects and limited clinical efficacy in certain cancers. Despite these limitations, imatinib has served as a benchmark for targeted therapy in PDGFR-driven pathologies and is often used as a comparator in clinical trials.

• Selective PDGFRα Inhibitors:
 – Recent drug discovery efforts have specifically focused on agents that predominantly target PDGFRα with minimal activity against other tyrosine kinases. According to recent comprehensive reviews, the main effective agents against PDGFRA-mutated tumors in gastrointestinal stromal tumors (GISTs) include avapritinib, ripretinib, and crenolanib. These agents have been designed to address the specific structural and conformational characteristics of PDGFRα and offer enhanced selectivity with improved clinical benefit in molecularly defined patient subsets.

• Monoclonal Antibodies and Bispecific Constructs:
 – Innovative approaches such as the development of PDGFRα-specific antibodies are also progressing. Patents detail compositions and methods for specifically inhibiting PDGFRα, including potential applications in demyelinating diseases and metastatic bone cancers. Although these antibodies are at earlier stages of development compared to small molecules, they represent a complementary modality that could synergize with other targeted therapies or be used in combination with immune checkpoint inhibitors.

Current Clinical Trials
Clinical trials investigating PDGFRα inhibitors are spearheading the advancement of precision oncology—particularly in tumor types that harbor PDGFRA alterations, such as certain gastrointestinal stromal tumors (GISTs), subsets of gliomas, and other rarer sarcomas. These trials aim to establish the safety, tolerability, pharmacodynamics, pharmacokinetics, and ultimately the efficacy of PDGFRα-targeted agents in defined patient populations.

Active Clinical Trials
Based on a synthesis of recent synapse-sourced literature and patent filings:

• Avapritinib:
 Avapritinib has emerged as a leading small-molecule inhibitor specifically designed to target PDGFRA mutations. In patients with GIST harboring the D842V mutation—a well-known alteration conferring resistance to other TKIs—avapritinib has demonstrated promising antitumor activity. Clinical trials investigating avapritinib have progressed through early-phase studies and now include expanded Phase II/III trials that focus on molecularly selected patients. These studies aim to validate its efficacy through improved objective response rates and progression-free survival compared to prior standard-of-care regimens.

• Ripretinib:
 Originally developed as a broad-spectrum TKI with activity that covers both wild-type and mutant forms of PDGFRA, ripretinib is currently investigated in multiple clinical trials. Although its primary indication has been in the context of advanced GISTs after prior treatments have failed, its effectiveness against PDGFRA-driven tumors makes it a candidate for focused trials to assess its selective inhibition potential. Ongoing clinical trials often use a basket design wherein patients with distinct PDGFRA mutations across different tumor types are enrolled.

• Crenolanib:
 Crenolanib is another selective TKI that has been reported to exhibit potent inhibitory activity against PDGFRA, particularly in cases where resistance mutations compromise the effectiveness of first-generation inhibitors. It is currently under clinical investigation in solid tumor basket trials and in specific phases for PDGFRA-mutated GISTs. Additionally, crenolanib is being assessed in combination studies with other targeted therapies to determine if synergistic effects can improve clinical outcomes and overcome acquired resistance mechanisms.

• Early-Stage Monoclonal Antibody Candidates:
 In parallel to small-molecule inhibitors, novel antibody-based PDGFRα inhibitors are being developed, as evidenced by recent patent disclosures. These therapies are in earlier phases of clinical development, often starting with Phase I safety and dose-escalation studies to determine their potential as anticancer agents. Because these molecules may have a different toxicity and efficacy profile compared to TKIs, early-phase clinical trials will be instrumental in assessing both their biological activity and potential combination strategies with other agents—such as immune checkpoint inhibitors or other kinase inhibitors.

• Combination Trials:
 Several ongoing clinical trials are evaluating PDGFRα inhibitors in combination with other targeted therapies or chemotherapeutic regimens. For example, trials have been designed to combine a PDGFRα inhibitor with an immunotherapy agent or with a VEGFR inhibitor, aiming to counteract the redundant signaling pathways that may contribute to inherent or acquired resistance. These combination trials are especially relevant in complex tumors where multiple RTKs are active, and the inhibition of PDGFRα alone may not be sufficient for an optimal antitumor response.

Phases of Clinical Trials
The clinical development landscape of PDGFRα inhibitors spans multiple phases:

• Phase I Trials:
 Early-phase studies primarily focus on determining the maximum tolerated dose (MTD), dose-limiting toxicities (DLTs), pharmacokinetics, and preliminary pharmacodynamic effects. For example, Phase I studies of avapritinib and crenolanib have established initial dosing regimens and safety profiles in patients with advanced PDGFRA-mutated tumors. The evaluation in Phase I trials also helps set the stage for biomarker-driven patient selection through genomic profiling for PDGFRA alterations.

• Phase II Trials:
 Phase II trials are mainly designed to test the efficacy of these inhibitors in selected patient populations with confirmed PDGFRA mutations or overexpression. These studies typically measure endpoints such as objective response rate (ORR), progression-free survival (PFS), and duration of response (DoR). For instance, expanded Phase II studies involving PDGFRA-mutated GIST patients have shown encouraging responses with avapritinib, ripretinib, and crenolanib. At this stage, adaptive trial designs and basket trials are frequently used to capture data across multiple tumor types driven by similar PDGFRα aberrations.

• Phase III Trials:
 Some agents, after demonstrating sufficient efficacy and a manageable safety profile in Phase II, proceed to Phase III randomized trials where they are compared with standard-of-care treatments. Even though some drugs like avapritinib have already received breakthrough designations or regulatory approvals in select indications, further Phase III studies in expanded patient cohorts provide critical data on long-term efficacy, overall survival benefit, and quality of life outcomes. These trials also help refine patient selection criteria and dosing schedules, ensuring that the benefits of PDGFRα inhibition are maximized in clinical practice.

Challenges and Future Directions
Despite the promising activity of PDGFRα inhibitors in early-stage clinical trials, several challenges remain in harnessing their full therapeutic potential. Both preclinical and clinical research have highlighted multiple obstacles in terms of patient selection, resistance mechanisms, and optimal drug combinations. Addressing these challenges is critical for advancing PDGFRα inhibition into a definitive therapeutic strategy for patients with PDGFRA-driven diseases.

Current Challenges in PDGFRα Inhibition
One of the primary challenges lies in the heterogeneity of tumors that express PDGFRα. Although PDGFRA mutations and overexpression are clearly identifiable in certain cancers, such as a subset of GISTs, the intratumoral genomic and phenotypic diversity often leads to variable clinical responses. This variability can be exacerbated by:

• Resistance Mechanisms:
 Acquired resistance, either through secondary mutations in the kinase domain or compensatory activation of alternative signaling pathways, poses a significant hurdle to the long-term efficacy of PDGFRα inhibitors. For example, mutations that alter the conformational dynamics of the receptor may render inhibitors less effective unless they are specifically designed to target these changes.

• Patient Selection and Biomarker Development:
 The success of these agents hinges on the ability to accurately stratify patients based on the molecular characteristics of their tumors. Although genomic profiling has improved, the sensitivity and specificity of these tests in detecting PDGFRA alterations still require optimization. Moreover, the presence of mixed populations of tumor cells with variable PDGFRα expression complicates response assessments.

• Off-target Effects and Toxicities:
 Even with selective inhibitors, off-target inhibition of kinases with structural similarities to PDGFRα is a concern. Such effects may manifest as adverse events that limit drug dosing or result in unforeseen toxicities. Since the PDGF pathway is also involved in normal physiological processes such as wound healing and tissue maintenance, the balance between effective inhibition of pathological signaling and preservation of normal function is delicate.

• Drug Delivery Challenges:
 For antibody-based PDGFRα inhibitors, ensuring adequate tumor penetration while maintaining stability in circulation is a recurring challenge. The pharmacokinetic properties of these larger molecules differ substantially from small-molecule TKIs, and their clinical trial designs must account for these differences.

Future Prospects and Research Directions
Looking forward, several strategies promise to enhance the effectiveness of PDGFRα-targeted therapies:

• Development of Next-generation Inhibitors:
 A key research direction is the design of inhibitors that can overcome resistance mechanisms by binding to alternative conformations of PDGFRα. Novel agents that target allosteric sites or that function as covalent inhibitors are being explored. These next-generation inhibitors have the potential to supplant current drugs, especially in cases where resistance mutations render standard treatments ineffective.

• Improved Biomarker-based Patient Selection:
 There is a strong drive toward integrating comprehensive genomic and proteomic profiling into clinical trial designs. By utilizing liquid biopsies and advanced sequencing techniques, clinicians can monitor PDGFRA mutation status and expression levels in real time. This approach enables more agile patient stratification and could lead to adaptive trial designs—akin to basket or umbrella trials—that are better suited to capture the diverse patient populations who might benefit from PDGFRα inhibition.

• Combination Therapy Approaches:
 Given the compensatory signaling pathways that often emerge during targeted therapy, combinations of PDGFRα inhibitors with agents that target complementary pathways (such as VEGFR, EGFR, or mTOR inhibitors) are actively being investigated. Early data from combination trials indicate that simultaneous targeting of multiple pathways can not only delay the onset of resistance but may also lead to synergistic efficacy. The integration of immunotherapy into these combination regimens is of great interest as well, particularly for tumors with an immunosuppressive microenvironment.

• Early Stage and Adaptive Clinical Trial Designs:
 Innovative clinical trial methodologies that incorporate real-time biomarker analysis and adaptive dosing strategies are expected to accelerate the development of PDGFRα inhibitors. Such designs will allow for mid-study adjustments based on emerging efficacy and safety data, helping to optimize dosing regimens and patient selection criteria. Adaptive trials minimize exposure of patients to suboptimal treatments while maximizing the chance of clinical benefit.

• Exploration of Non-oncologic Applications:
 Beyond oncology, there is an emerging interest in targeting PDGFRα in non-malignant conditions, such as fibrotic diseases and demyelinating disorders. Patent applications have described methods of using PDGFRα inhibitors for the treatment of demyelinating diseases. Expanding the scope of clinical trials to include these indications may provide additional insights into the broad biological implications of PDGFRα inhibition, ultimately improving our understanding and management of these diverse conditions.

Conclusion
In summary, PDGFRα inhibitors represent a promising class of targeted therapies that address the underlying molecular mechanisms driving several aggressive and treatment-resistant cancers, particularly PDGFRA-mutated gastrointestinal stromal tumors and other solid tumors. The landscape of agents in clinical investigation includes advanced small-molecule TKIs such as avapritinib, ripretinib, and crenolanib, all of which have demonstrated encouraging activity through disruption of key signaling pathways. In addition, emerging antibody-based therapies and bispecific constructs offer a complementary modality with the potential for improved selectivity and immune engagement.

Current clinical trials are designed across multiple phases—from early dose-escalation studies in Phase I to adaptive basket trials in Phase II and randomized controlled studies in Phase III—that aim to rigorously evaluate efficacy and optimize patient selection strategies. Nevertheless, challenges such as intrinsic and acquired resistance, variability in tumor heterogeneity, off-target toxicities, and pharmacokinetic hurdles remain. Future directions focus on next-generation inhibitor development, robust biomarker integration, combination therapy strategies, and adaptive clinical trial methodologies to enhance therapeutic outcomes and ultimately drive the field toward personalized medicine.

The current body of evidence suggests that the most advanced PDGFRα inhibitors in clinical trials include avapritinib, ripretinib, and crenolanib. These agents are being actively tested in defined patient cohorts and hold the promise to improve clinical management where PDGFRα plays a pivotal role. Innovation in trial design and combination therapies will be essential to overcome existing challenges and to translate preclinical successes into durable clinical benefits. Overall, while further research is needed to refine these approaches and validate long-term efficacy, the future of PDGFRα-targeted treatment remains optimistic, potentially transforming the treatment paradigm for multiple PDGFRA-driven diseases.

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.