Small Molecule
C30H34N8
1037624-75-1
Bemcentinib is an investigational small molecule drug classified as a first-in-class, selective inhibitor of the AXL receptor tyrosine kinase.[1] The compound has been developed under several code names and synonyms that reflect its history. It was originally discovered by Rigel Pharmaceuticals as R428 and is frequently referred to by this designation in preclinical literature.[3] Following its licensing by BerGenBio ASA for clinical development, it became widely known by the code BGB324 (or BGB-324) and was assigned the generic name Bemcentinib.[3]
The compound is cataloged across major chemical and pharmacological databases under a consistent set of identifiers, ensuring its unambiguous tracking in scientific literature and regulatory filings.
Bemcentinib is a complex synthetic organic compound belonging to the aralkylamine class, characterized by an alkyl group substituted with an aromatic hydrocarbyl group.[1] Its structure incorporates several heterocyclic moieties, including pyridazine, triazole, and N-alkylpyrrolidine components, which are crucial for its interaction with the target kinase.[1]
The computed physicochemical properties of Bemcentinib provide insight into its drug-like characteristics, suggesting properties consistent with an orally administered agent. These parameters are summarized in Table 1.
Table 1: Key Chemical and Physical Properties of Bemcentinib
| Property | Value | Source(s) |
|---|---|---|
| Molecular Formula | $C_{30}H_{34}N_{8}$ | 1 |
| Average Molecular Weight | 506.658 g·mol⁻¹ | 1 |
| Monoisotopic Mass | 506.29064312 Da | 1 |
| XLogP / logP | 5.05 – 6.2 | 1 |
| pKa (Strongest Acidic) | 11.76 | 1 |
| pKa (Strongest Basic) | 10.55 | 1 |
| Hydrogen Bond Acceptor Count | 7 | 1 |
| Hydrogen Bond Donor Count | 2 | 1 |
| Rotatable Bond Count | 4 | 1 |
| Topological Polar Surface Area (TPSA) | 97.26 – 97.8 Ų | 1 |
| Water Solubility (predicted) | 0.0081 mg/mL | 1 |
Bemcentinib's primary molecular target is the AXL receptor tyrosine kinase, a key member of the TAM family of receptors, which also includes TYRO3 and MER.[2] These receptors are transmembrane proteins that play a fundamental role in transducing signals from the extracellular environment into the cell's cytoplasm, thereby regulating a host of critical physiological processes, including cell survival, proliferation, migration, and differentiation.[1]
The activation of AXL is initiated by the binding of its primary ligand, Growth Arrest-Specific 6 (GAS6).[1] This binding event induces receptor homodimerization and subsequent autophosphorylation of tyrosine residues within the intracellular domain, creating docking sites for various downstream signaling molecules.[1] Key adaptors and enzymes recruited to the activated AXL receptor include the p85 regulatory subunit of phosphatidylinositol 3-kinase (PI3K), GRB2, and phospholipase C-gamma (PLCG1).[1] The recruitment of PI3K is a pivotal step, as it leads to the activation of the pro-survival serine/threonine kinase AKT, a central node in pathways that promote cell survival and proliferation.[1]
In the context of cancer, the GAS6/AXL signaling axis is frequently dysregulated. Overexpression of AXL is a well-documented negative prognostic factor across a wide array of human malignancies, including non-small cell lung cancer (NSCLC), breast cancer, ovarian cancer, melanoma, and glioblastoma.[6] This overexpression is functionally linked to the hallmarks of cancer; AXL signaling drives tumor cell proliferation, enhances survival by inhibiting apoptosis, promotes invasion and metastasis, and is a key mediator of the epithelial-mesenchymal transition (EMT)—a cellular program that endows cancer cells with migratory and invasive properties.[2] Furthermore, AXL activation has emerged as a critical mechanism of acquired resistance to both conventional chemotherapy and modern targeted therapies, making it an attractive therapeutic target for overcoming treatment failure.[3]
Bemcentinib functions as an orally available and highly selective ATP-competitive inhibitor of AXL.[2] Its mechanism of action involves targeting and binding to the intracellular catalytic kinase domain of the AXL protein.[2] By occupying the ATP-binding pocket, Bemcentinib prevents the autophosphorylation of AXL, thereby blocking the initiation of all downstream signal transduction pathways mediated by the receptor.[2]
The compound demonstrates a high degree of selectivity for its primary target. Biochemical and cell-based assays have established an IC50 (half-maximal inhibitory concentration) for AXL of approximately 14 nM.[12] Importantly, Bemcentinib exhibits significant selectivity over other kinases, including those within its own family. It is reported to be over 100-fold more selective for AXL than for the Abl kinase and maintains 50-fold and over 100-fold selectivity against the other TAM family members, Mer and Tyro3, respectively.[12] This selectivity profile is critical for minimizing off-target toxicities and focusing the therapeutic effect on the intended pathway.
The functional consequences of this inhibition are profound. By blocking AXL signaling, Bemcentinib has been shown to inhibit AKT phosphorylation, reduce cancer cell invasion, suppress angiogenesis, and decrease the production of pro-inflammatory cytokines.[9] A key outcome of AXL inhibition is the reversal of resistance to other anticancer agents, effectively re-sensitizing tumor cells to treatments like chemotherapy.[2]
Beyond its direct role in tumor cell signaling, the AXL kinase has been identified as a crucial suppressive immune checkpoint, particularly within the innate immune system.[14] AXL signaling within the tumor microenvironment (TME) contributes significantly to immune evasion.[3] It achieves this by downregulating the activity of antigen-presenting cells like dendritic cells, modulating the process of efferocytosis (the clearance of apoptotic cells), and fostering an immunosuppressive cytokine profile that favors the expansion of myeloid-derived suppressor cells (M-MDSCs).[14] This creates a TME that is hostile to an effective anti-tumor immune response.
This immunomodulatory function provides a compelling rationale for combining AXL inhibition with immuno-oncology (I-O) agents, such as PD-1/PD-L1 checkpoint inhibitors. The central hypothesis is that by inhibiting AXL, Bemcentinib can dismantle the immunosuppressive shield around the tumor, thereby rendering it more susceptible to immune-mediated attack elicited by checkpoint blockade. Preclinical models have provided strong support for this concept, demonstrating that AXL inhibition with Bemcentinib reverses multiple immune suppressive mechanisms. This leads to a more inflamed, "hot" TME, characterized by increased infiltration of cytotoxic T-lymphocytes (CTLs), Natural Killer (NK) cells, and NKT cells, and a concurrent decrease in M-MDSCs.[14] Preliminary clinical data from a trial in acute myeloid leukemia (AML) patients treated with Bemcentinib provided the first human evidence of this immune modulation, showing a diversification of the T-cell and B-cell receptor repertoires in peripheral blood following treatment.[14]
While AXL inhibition is the primary and most well-characterized mechanism of Bemcentinib, a growing body of evidence suggests that the drug may exert significant anti-cancer effects through pathways that are independent of its action on AXL. This discovery adds a critical layer of complexity to the drug's biological profile and has important implications for its clinical application and biomarker development.
Research has revealed that Bemcentinib can directly impair the endo-lysosomal and autophagy systems within cancer cells. Autophagy is a cellular recycling process that tumor cells often co-opt to survive metabolic stress and resist therapy. By impairing autophagic flux, Bemcentinib can induce a form of cellular stress that leads to immunogenic cell death, a type of apoptosis that is particularly effective at stimulating an anti-tumor immune response. This effect was observed in NSCLC cells resistant to EGFR inhibitors and was shown to be AXL-independent.
Furthermore, in models of pediatric rhabdomyosarcoma (RMS), pharmacologic blockade with Bemcentinib induced rapid cell death and acted as a potent chemosensitizer, augmenting the efficacy of vincristine and cyclophosphamide.[8] This activity was also determined to be independent of AXL expression or signaling in the RMS cells.[8] The existence of these AXL-independent mechanisms suggests that Bemcentinib's clinical activity may not be solely predicted by the level of AXL expression in a tumor. It raises the possibility that its efficacy in some contexts could be driven by these alternative effects on cellular homeostasis and stress pathways. This could help explain some of the variable clinical results observed across different tumor types and provides new avenues for exploring its therapeutic potential, particularly in combination with agents that also modulate autophagy or induce cellular stress.
The clinical development of Bemcentinib was underpinned by a robust body of preclinical evidence demonstrating its potent anti-cancer activity. In vitro studies consistently showed that the compound effectively inhibits cancer cell migration and invasion across various cell lines.[7]
This activity translated effectively into in vivo models. In multiple mouse models of metastatic breast cancer, oral administration of Bemcentinib (as R428) significantly blocked tumor dissemination, reduced the burden of metastases, and prolonged overall survival.[9] The drug also demonstrated potent anti-angiogenic effects, suppressing both tumor angiogenesis in xenograft models and vascular endothelial growth factor (VEGF)-induced neovascularization in a corneal micropocket assay.[9] The therapeutic potential was further validated in models of other aggressive cancers, including highly invasive esophageal adenocarcinoma and esophageal squamous cell carcinoma (ESCC), where Bemcentinib treatment reduced key cellular and molecular functions associated with malignancy.[9]
A central theme of the preclinical research was the potent synergy observed when Bemcentinib was combined with other anti-cancer agents. This provided the foundational rationale for the combination-focused clinical development strategy.
The biological role of AXL extends beyond oncology, and preclinical research has identified Bemcentinib as a potential agent for other diseases. AXL has been identified as a host cell entry factor for several viruses. Consequently, Bemcentinib was shown to inhibit the entry of Zika virus into human glial cells and to block the entry of SARS-CoV-2 into host cells.[1] This latter finding provided the rationale for its investigation as a potential treatment for COVID-19.[3] Additionally, the drug has been explored preclinically for a wide range of other indications, including influenza, respiratory syncytial virus (RSV) infections, Ebola virus infections, idiopathic pulmonary fibrosis, and non-alcoholic steatohepatitis (NASH), though these have not progressed to late-stage clinical development.[20]
Bemcentinib (as R428) was originally discovered and developed preclinically by Rigel Pharmaceuticals, Inc..[4] The clinical development program has been led by BerGenBio ASA, a clinical-stage biopharmaceutical company based in Bergen, Norway, which licensed the compound from Rigel.[3] BerGenBio has advanced the drug, under the identifier BGB324, through a broad Phase II clinical program.[3]
The clinical development of Bemcentinib has been extensive, exploring its potential as both a monotherapy and a combination agent across a diverse range of diseases. The primary focus has been on oncology, targeting both solid tumors and hematological malignancies where AXL overexpression is implicated in poor prognosis and treatment resistance. Key indications investigated include Non-Small Cell Lung Cancer (NSCLC), Acute Myeloid Leukemia (AML), Myelodysplastic Syndrome (MDS), Metastatic Melanoma, Triple-Negative Breast Cancer (TNBC), and Pancreatic Cancer.[1]
Beyond oncology, Bemcentinib was rapidly advanced into clinical trials for COVID-19, based on preclinical data showing its ability to inhibit SARS-CoV-2 viral entry. It was the first candidate selected for the UK Government's ACCORD (Accelerating COVID-19 Research & Development) platform for Phase II trials in hospitalized patients.[3] The major clinical trials that have defined the development trajectory of Bemcentinib are summarized in Table 2.
Table 2: Summary of Major Clinical Trials for Bemcentinib
| NCT Identifier | Trial ID | Phase | Indication(s) | Combination Agent(s) | Status | Source(s) |
|---|---|---|---|---|---|---|
| NCT03184571 | BGBC008 | II | Advanced NSCLC (2L+) | Pembrolizumab | Completed | 15 |
| NCT02922777 | SPOC 2015-01 | I | Advanced NSCLC (2L+) | Docetaxel | Completed | 15 |
| NCT05469178 | BGBC016 | I/II | Advanced NSCLC (1L, STK11m) | Pembrolizumab + Chemotherapy | Terminated | 5 |
| NCT02488408 | BGBC003 | I/II | R/R AML & High-Risk MDS | Monotherapy / Low-Dose Cytarabine | Completed | 14 |
| NCT02872259 | BGBIL006 | I/II | Metastatic Melanoma | Pembrolizumab or Dabrafenib/Trametinib | Completed | 11 |
| NCT06516887 | CTMS 24-0020 | Ib/II | Advanced Lung Adenocarcinoma (2L+) | Pacritinib (JAK2 inhibitor) | Active, Not Recruiting | 24 |
| NCT06469138 | 8479217 | I | Healthy Volunteers (ADME Study) | ¹⁴C-labeled Bemcentinib | Completed | 26 |
The clinical story of Bemcentinib is one of notable successes in specific, hard-to-treat patient populations, contrasted with clear failures in other settings. This divergence highlights the critical importance of tumor biology, treatment line, and biomarker selection in defining the drug's therapeutic window.
NSCLC has been the most extensively studied indication for Bemcentinib, with multiple trials exploring its role in combination with both immunotherapy and chemotherapy.
This Phase II, open-label, single-arm study was a cornerstone of the NSCLC program, evaluating Bemcentinib plus the PD-1 inhibitor pembrolizumab in patients with advanced adenocarcinoma of the lung who had progressed on prior platinum-based chemotherapy and/or immune checkpoint inhibitor (ICI) therapy.[10] The combination was found to be well-tolerated and demonstrated promising clinical efficacy compared to historical controls.[17] The first stage of the trial met its primary efficacy endpoint, triggering expansion into a second stage.[29]
The most compelling findings emerged from biomarker analyses. While the overall response rate (ORR) across 29 evaluable patients was 24% (7 partial responses), the efficacy was significantly enriched in patients whose tumors were AXL-positive.[28] In this subgroup, the ORR was 40%, and the median progression-free survival (mPFS) was 5.9 months, compared to 4.0 months in the overall population.[28] Furthermore, an exploratory sub-analysis revealed a survival benefit in patients harboring mutations associated with a poor prognosis and resistance to ICI therapy, such as STK11, KEAP1, and KRAS.[17] This provided strong clinical validation for the preclinical hypothesis that AXL inhibition could reverse immune suppression and restore sensitivity to checkpoint blockade in an immunologically "cold" TME.[17]
This Phase I dose-escalation trial assessed the safety and preliminary efficacy of Bemcentinib combined with the chemotherapeutic agent docetaxel in previously treated, advanced NSCLC patients.[15] The combination demonstrated clear anti-tumor activity. Among 17 patients evaluable for radiographic response, 6 (35%) achieved a partial response (PR) and 8 (47%) had stable disease (SD), resulting in a high disease control rate of 82%.[15] The maximum tolerated dose (MTD) was established as docetaxel at 60 mg/m² administered with prophylactic G-CSF support, combined with Bemcentinib at a 400 mg loading dose followed by 200 mg daily.[15] This study confirmed the preclinical synergy between Bemcentinib and taxane chemotherapy and established a safe and effective dose for further investigation.
The promising signal observed in the STK11-mutated subgroup of the BGBC008 trial prompted BerGenBio to initiate the BGBC016 trial, a high-stakes study designed to move Bemcentinib into the first-line (1L) setting for this specific, biomarker-defined population.[5] However, this strategic move proved unsuccessful. Despite demonstrating acceptable safety in the Phase Ib portion and observing one durable complete response, a preliminary analysis of the first 10 efficacy-evaluable patients in the Phase IIa expansion cohort showed no additional responses.[5]
This lack of efficacy was a significant setback. Citing the weak results and the consequent inability to secure the necessary funding to complete the trial, BerGenBio announced the discontinuation of the study in February 2025 and initiated a process to explore strategic alternatives for the company.[5] The failure of this trial suggests a critical distinction in Bemcentinib's role: while it appears effective as a "resistance-breaker" in heavily pre-treated, second-line or later (2L+) settings where AXL-mediated resistance is an established driver, it may lack efficacy as a primary anti-tumor agent in the first-line setting, even in a supposedly favorable biomarker-selected population.
The clinical trial NCT02488408 investigated Bemcentinib in older patients with relapsed/refractory (R/R) AML or high-risk MDS who were ineligible for intensive chemotherapy.[14] The study evaluated the drug both as a monotherapy and in combination with low-dose cytarabine (LDAC).[30]
The combination with LDAC yielded promising results. In newly diagnosed (ND) AML patients, the ORR was reported to be between 40% and 50%, with a median overall survival (mOS) of 16.1 months.[31] In the more difficult-to-treat R/R AML cohort, the ORR was 20% with an mOS of 7.8 months.[31] A key finding was the observation of durable responses, particularly in very elderly patients (over 75 years of age) and those with adverse cytogenetic features.[32] The strength of these data was sufficient to earn Bemcentinib a Fast Track Designation from the U.S. FDA for the treatment of R/R AML, validating its potential in this area of high unmet medical need.[33]
In stark contrast to the positive signals in NSCLC and AML, the trial in metastatic melanoma was a definitive failure from an efficacy perspective. This Phase Ib/II study evaluated the addition of Bemcentinib to standard-of-care (SOC) therapies—either pembrolizumab or the targeted combination of dabrafenib plus trametinib.[11]
Despite being well-tolerated, the addition of Bemcentinib to SOC did not lead to any improvement in ORR, PFS, or OS when compared to SOC alone.[11] Critically, preplanned biomarker analyses also failed to identify any subgroup of patients who derived an increased benefit from the Bemcentinib-containing combinations.[11] This outcome provides a crucial boundary for the drug's activity. Melanoma is often an immunologically "hot" tumor where ICIs are highly effective. The failure of Bemcentinib to enhance this activity suggests that AXL signaling may not be a dominant mechanism of immune resistance in this specific disease context, unlike in certain subsets of NSCLC. This result underscores that the therapeutic utility of AXL inhibition is highly dependent on the specific tumor biology and the prevailing mechanisms of immune evasion in a given cancer type.
Across its extensive clinical development program, Bemcentinib has demonstrated a generally manageable and predictable safety profile, both as a monotherapy and in combination with other anti-cancer agents. The nature and severity of adverse events (AEs) are often influenced by the partner drug in combination regimens.
The most frequently reported treatment-related adverse events (TRAEs) are consistent across studies and are typically low-grade and manageable.
The profile of serious adverse events (SAEs) and dose-limiting toxicities (DLTs) is highly dependent on the combination regimen.
A consolidated summary of the most significant adverse events across key combination trials is presented in Table 3.
Table 3: Consolidated Summary of Key Treatment-Related Adverse Events
| Adverse Event | Combination with Pembrolizumab (NSCLC) | Combination with Docetaxel (NSCLC) | Combination with LDAC (AML) |
|---|---|---|---|
| All Grades (%) | ≥G3 (%) | All Grades (%) | |
| Diarrhea | 24 - 29 | N/A | 57 |
| Fatigue / Asthenia | 30 | 8 | 57 |
| Nausea | N/A | N/A | 52 |
| Increased ALT/AST | 38 | 14 | N/A |
| Neutropenia | N/A | N/A | 86 |
| Febrile Neutropenia | N/A | N/A | 38 |
| Anemia | N/A | 2 | N/A |
| Thrombocytopenia | N/A | N/A | N/A |
| QTc Prolongation | N/A | N/A | N/A |
| Data compiled from sources.15 N/A indicates data not specifically reported in the provided sources for that category. |
Bemcentinib is described as an orally bioavailable and orally active agent, a key property that facilitates its use in outpatient settings.[3] Pharmacokinetic (PK) data has been collected as part of its clinical development. In the Phase I trial combining Bemcentinib with docetaxel, a key finding was that the PK profile of Bemcentinib was highly similar when administered alone versus when given in combination with docetaxel.[15] This suggests a low potential for clinically significant pharmacokinetic drug-drug interactions between the two agents, simplifying co-administration. However, detailed public data on specific PK parameters such as absorption rate, volume of distribution, and clearance from most clinical trial publications remains limited.
To address the gaps in the understanding of Bemcentinib's absorption, distribution, metabolism, and excretion (ADME) profile, BerGenBio sponsored a dedicated Phase I human mass balance study.[26] This type of study is a critical component of a drug development program and is typically required for regulatory submissions like a New Drug Application (NDA).
The study (NCT06469138) was an open-label trial in healthy male subjects designed to investigate the disposition of a single 200 mg oral dose of Bemcentinib labeled with carbon-14 ($^{14}$C).[26] The primary objectives were to determine the mass balance recovery by quantifying the total amount of radioactivity excreted in urine and feces, to characterize the pharmacokinetic profiles of total radioactivity and unchanged Bemcentinib in the bloodstream, and to identify the major metabolic pathways by profiling the breakdown products (metabolites) in plasma and excreta.[26] The initiation and completion of this study is a significant developmental milestone, indicating that the necessary data to fully characterize the drug's human ADME profile has been generated, which would be essential for any future regulatory filings.
Bemcentinib has received multiple Fast Track Designations from the U.S. FDA, a program designed to facilitate the development and expedite the review of drugs that treat serious conditions and fill an unmet medical need.[37] These designations are a strong signal of regulatory recognition of the drug's potential in specific, hard-to-treat patient populations.
These designations make Bemcentinib eligible for more frequent meetings with the FDA, rolling review of its NDA, and potential accelerated approval, significantly streamlining its path to market in these specific indications.[37]
Information regarding Bemcentinib's status with other major regulatory agencies is more limited. BerGenBio has publicly stated its intention to seek regulatory advice from the European Medicines Agency (EMA) regarding the optimal development path for Bemcentinib in AML.[37] However, a review of publicly available EMA documentation does not indicate any active marketing authorization application or formal opinion for Bemcentinib at this time.[39] Similarly, there is no evidence to suggest that the drug has been submitted for review or is listed on the Australian Register of Therapeutic Goods (ARTG) by the Therapeutic Goods Administration (TGA) of Australia.[42]
The corporate strategy of BerGenBio has been profoundly impacted by recent clinical trial outcomes. The company had built a strong development narrative around biomarker-driven therapy, leveraging the promising data in 2L+ NSCLC and the multiple FDA Fast Track Designations to pursue a high-risk, high-reward strategy in the larger first-line NSCLC market with the BGBC016 trial.
The failure and subsequent discontinuation of this pivotal trial in February 2025 represented a major strategic setback.[5] The lack of efficacy in the 1L STK11m population undermined the company's primary value proposition and, critically, its ability to secure near-term funding to continue its operations as planned. As a direct consequence of this clinical failure, the Board of Directors announced that it had initiated a process to explore strategic alternatives, which could include a sale of the company, a merger, or other strategic transactions.[5] This places the future of Bemcentinib at a critical inflection point, where its continued development is likely dependent on the outcome of this corporate strategic review.
The extensive clinical development program for Bemcentinib has painted a complex and nuanced picture of its therapeutic potential. The collective evidence suggests that Bemcentinib is not a single entity but rather functions differently depending on the biological context of the disease.
On one hand, the data provides strong support for Bemcentinib as an effective "resistance-breaker" in later lines of therapy. Its successes in ICI-refractory, AXL-positive NSCLC, in combination with chemotherapy in pre-treated NSCLC, and in elderly/relapsed AML all point to a mechanism that is most effective in tumors that have evolved complex resistance and survival pathways. In these settings, AXL signaling often becomes a critical dependency, creating a vulnerability that Bemcentinib can exploit to re-sensitize tumors to therapy.
On the other hand, the program has clearly defined the drug's limitations. The failures in first-line STK11m NSCLC and in metastatic melanoma demonstrate that Bemcentinib is not an effective broad-spectrum or front-line agent. In these contexts, where AXL signaling may not be the primary driver of tumor progression or immune evasion, its inhibition provides little to no clinical benefit. Furthermore, the emerging preclinical data on AXL-independent mechanisms, such as the disruption of autophagy, adds a layer of complexity that must be reconciled with the clinical findings to fully understand its activity and optimize its use.
Based on the available evidence, the most viable path forward for Bemcentinib lies in a focused development strategy targeting well-defined, niche patient populations with high unmet medical needs. The most promising indications are:
However, several critical questions remain. The true clinical contribution of the AXL-independent mechanisms is unknown and requires further investigation. Most pressingly, the future of the entire program hinges on the outcome of BerGenBio's strategic review. Without a new partner or acquirer to provide the necessary funding, the final stages of development and potential commercialization of this promising agent may not be realized.
In conclusion, Bemcentinib is unlikely to become the broad "cornerstone of cancer therapy" that was once envisioned. However, it remains a scientifically compelling and clinically valuable investigational drug. It has a well-defined mechanism of action, a manageable safety profile, and has demonstrated clear and meaningful clinical activity in specific, biomarker-defined patient populations where treatment options are limited. Its journey serves as an important case study in the development of targeted therapies, illustrating the critical importance of patient selection, the challenges of translating preclinical synergy into clinical success, and the unforgiving financial realities of biopharmaceutical innovation. The ultimate success of Bemcentinib will depend on a disciplined, biomarker-driven clinical strategy and, most immediately, on a corporate resolution that allows its development to continue.
Published at: October 27, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.