Small Molecule
PRTX-100, also known by the International Nonproprietary Name (INN) Bevifimod, is an investigational biologic therapeutic agent developed for the treatment of autoimmune diseases.[1] It is a highly-purified, native form of Staphylococcal Protein A (SpA), a 47 kDa protein derived from the A676 strain of the bacterium Staphylococcus aureus.[3] Contrary to its classification as a small molecule in some databases, PRTX-100 is a protein biologic intended for intravenous administration.[4]
The therapeutic rationale for PRTX-100 was based on a novel, dual-pathway immunomodulatory mechanism designed to restore immune system balance rather than induce broad immunosuppression.[5] The mechanism targeted both the adaptive and innate immune systems. First, by binding to the VH3-family of immunoglobulins, PRTX-100 was intended to selectively modulate the activity of B-cells responsible for producing pathogenic autoantibodies, a key driver in many autoimmune conditions.[2] Second, by forming immune complexes with IgG, it aimed to induce a suppressor phenotype in macrophages, thereby inhibiting the phagocytosis of antibody-coated cells, a mechanism directly relevant to the pathology of Immune Thrombocytopenia (ITP).[8]
The clinical development program, sponsored by Protalex, Inc., focused primarily on two indications: Rheumatoid Arthritis (RA) and ITP.[5] The program encompassed at least eight human clinical studies, including multiple Phase 1 and Phase 1/2 trials, which established a generally acceptable safety and tolerability profile at the low microgram-per-kilogram doses administered.[7] Despite this, the program was ultimately unsuccessful and has been discontinued.[12]
The failure of PRTX-100 can be attributed to a confluence of critical factors. Scientifically, the drug was hampered by high immunogenicity. As a foreign bacterial protein, it induced a significant anti-drug antibody (ADA) response in most patients. This ADA response led to rapid drug clearance upon repeated administration, severely compromising the pharmacokinetic exposure necessary to achieve a durable therapeutic effect.[4] Clinically, this pharmacological limitation manifested as weak and inconsistent efficacy signals across both the RA and ITP programs, which were insufficient to warrant progression to pivotal Phase 3 trials.[14] The final determinant in the program's demise was the corporate failure of its sponsor, Protalex, Inc., which ceased operations and had its securities registration revoked by the U.S. Securities and Exchange Commission in 2021 for failure to comply with periodic filing requirements.[16] This confluence of scientific, clinical, and corporate challenges led to the complete termination of the PRTX-100 development program.
The investigational drug is primarily identified as PRTX-100, with the assigned INN of Bevifimod.[1] It is associated with DrugBank Accession Number DB05947 and CAS Number 2223113-32-2.[1] A comprehensive list of its identifiers and synonyms is provided in Table 1.
PRTX-100 is a biologic therapeutic protein, not a small molecule.[5] It is a highly purified preparation of native Staphylococcal Protein A (SpA), a virulence factor protein produced by the bacterium Staphylococcus aureus.[3] The specific formulation used in clinical trials was prepared from S. aureus Strain A676 under Good Manufacturing Practices (GMP).[3]
The molecular weight of SpA has been reported with some variability, which can be reconciled by considering the context of the measurement. The theoretical molecular weight of the core single polypeptide chain is approximately 42 kDa.[2] However, the specific clinical formulation of PRTX-100 was characterized as a 47 kDa protein composed of 432 amino acids.[4] Furthermore, due to its structure, SpA is known to migrate anomalously during SDS-PAGE analysis, resulting in a higher apparent molecular weight of 55-56 kDa.[19] Structurally, SpA consists of a single polypeptide chain containing five highly homologous, tandem immunoglobulin-binding domains, commonly designated E, D, A, B, and C.[18]
PRTX-100 was formulated for parenteral administration and was delivered as a short intravenous (IV) infusion in clinical trials. The infusion duration varied depending on the specific study protocol and dose level, ranging from a rapid injection over 10-90 seconds to a more standard infusion over 30-60 minutes.[4]
Table 1: Drug Identification and Physicochemical Properties of PRTX-100
| Property | Detail | Source(s) |
|---|---|---|
| DrugBank ID | DB05947 | [5] |
| Primary Name | PRTX-100 | [5] |
| INN | Bevifimod | [1, 2, 22] |
| Key Synonyms | Staphylococcus aureus protein A, SpA PRTX-100, PRTX 100 | [2, 5, 23] |
| CAS Number | 2223113-32-2 | [1, 24] |
| Classification | Biologic Therapeutic Protein | [4, 5] |
| Source Organism | Staphylococcus aureus (Strain A676) | 3 |
| Molecular Weight | 47 kDa (Clinical Formulation) | 4 |
| Formulation Route | Intravenous (IV) Infusion | [3, 10] |
PRTX-100 was developed as a novel immunomodulatory compound intended to restore immune system homeostasis rather than cause broad immunosuppression.[5] Its proposed mechanism of action is multifaceted, engaging both the adaptive and innate branches of the immune system through its unique ability to bind human immunoglobulins at two distinct sites.
PRTX-100 exhibits high-affinity binding to the Fab framework region of immunoglobulins that utilize genes from the VH3 family to encode their heavy chain variable region.[4] This property also allows it to bind to the IgM B-cell receptor on the surface of all B-lymphocytes of the VH3 lineage, leading to its characterization as a "B-cell superantigen".[4] The therapeutic rationale for this targeting is based on the observation that in numerous autoimmune diseases, including ITP, the pathogenic autoantibodies responsible for the disease are predominantly derived from this specific VH3 gene family.[5] By selectively targeting this subset of B-cells, PRTX-100 aimed to prevent their activation, induce apoptosis, and thereby reduce the production of disease-causing autoantibodies without affecting the broader B-cell repertoire essential for normal immune function.[2]
In addition to its Fab binding, PRTX-100 binds with high affinity to the Fc region of immunoglobulin G (IgG), leading to the formation of SpA-IgG immune complexes.[4] These immune complexes subsequently interact with monocytes and macrophages, which express Fc receptors.[7] This interaction was shown to induce a "suppressor" phenotype in these phagocytic cells.[8] This mechanism is particularly relevant for the treatment of ITP, a disease characterized by the premature destruction of antibody-coated (opsonized) platelets by macrophages, primarily in the spleen.[10] In vitro preclinical studies provided direct evidence for this mechanism, demonstrating that pretreating human monocytes with PRTX-100 significantly inhibited the phagocytosis of opsonized platelets in a dose-dependent manner.[8]
This dual mechanism provides a synergistic therapeutic hypothesis for ITP. The modulation of B-cells addresses the root cause of the disease by targeting the "supply" of pathogenic autoantibodies. Concurrently, the suppression of macrophage activity addresses the primary effector pathology by reducing the "demand" for platelet destruction. This elegant, two-pronged approach positioned PRTX-100 as a potential disease-modifying therapy, rather than a purely symptomatic one.[5]
As a natural virulence factor of S. aureus, SpA has evolved to help the bacterium evade the host immune response.[3] One of its key immune evasion strategies is the potent inhibition of the classical complement pathway. This is achieved through a direct steric hindrance mechanism. The initiation of the classical pathway requires the binding of the C1q protein to arrays of IgG molecules on a target surface. This binding is facilitated by the self-association of target-bound IgG into hexamers via noncovalent Fc-Fc interactions.[3] SpA binds directly to the interface on the IgG Fc region that is required for this hexamerization.[3] By competitively occupying this site, SpA effectively blocks the formation of IgG hexamers and, consequently, prevents the binding of C1q and all downstream complement activation events.[3] While this is a mechanism of pathogenesis in the context of a bacterial infection, it contributes to the overall immunomodulatory profile of PRTX-100 in a therapeutic setting.
The therapeutic hypothesis for PRTX-100 was supported by positive results in relevant animal models of autoimmune disease.
The development of PRTX-100 represents a therapeutic paradox: the harnessing of a bacterial virulence factor for beneficial immunomodulation. The very properties that make SpA an effective immune evasion tool for S. aureus—its ability to bind IgG at both the Fc and Fab regions and to block complement—are the same properties that were exploited for its therapeutic effect. This approach, while mechanistically elegant, also carried an inherent and substantial risk. The human immune system is evolutionarily primed to recognize and mount a vigorous response against foreign bacterial proteins, making a high degree of immunogenicity and the formation of neutralizing anti-drug antibodies a highly probable clinical challenge.
The clinical development of PRTX-100 was conducted by Protalex, Inc. and involved at least eight human studies with more than 160 subjects, including healthy volunteers and patients with RA and ITP.[7] The program's primary goals were to establish the safety, tolerability, and pharmacokinetic profile of PRTX-100 and to identify a therapeutic dose for the target autoimmune indications.
The initial clinical focus for PRTX-100 was on RA, with a series of Phase 1 and 2 studies designed to assess its potential as an add-on therapy for patients with active disease.
ITP became the lead indication for PRTX-100, bolstered by strong preclinical data and the granting of Orphan Drug Designation by both the U.S. Food and Drug Administration (FDA) and European authorities.[10] The IND for ITP was accepted by the FDA in early 2015.[38] However, the clinical program in ITP was marked by a series of prematurely terminated trials.
The collective clinical data paints a consistent picture. Across all studies, PRTX-100 demonstrated an acceptable safety profile, suggesting it did not fail due to overt toxicity. However, the efficacy signals were weak and inconsistent. The RA program showed modest effects in small, and in one case biased, patient populations. More critically, the ITP program, despite its Orphan Drug status and strong preclinical rationale, failed to produce robust evidence of clinical benefit, leading to the termination of all its key trials. This pattern suggests a drug that was safe enough to continue testing but ultimately not effective enough to progress to later-stage development.
Table 2: Summary of Clinical Trials for PRTX-100
| NCT Identifier | Study Title/Code | Phase | Indication | Design | Patients (Enrolled) | Dose Range | Final Status |
|---|---|---|---|---|---|---|---|
| NCT01749787 | SPARTA | Ib | Rheumatoid Arthritis | Randomized, Placebo-Controlled, Dose-Escalation | 61 | 1.5 - 12 µg/kg; 240/420 µg fixed | Completed |
| NCT02330445 | SPARTA-II | I/II | Rheumatoid Arthritis | Open-Label, Continuation | 11 | Fixed Dose | Completed |
| NCT00571467 | Phase I Safety and Tolerability Study | I | Immune Thrombocytopenia | Open-Label, Dose-Escalation | 9 | 0.075 - 0.30 µg/kg | Terminated |
| NCT02401061 | PRTX-100-202 | I/II | Immune Thrombocytopenia | Open-Label, Dose-Escalation | N/A | 1 - 24 µg/kg | Terminated |
| NCT02566603 | PRTX-100-203 | Ib | Immune Thrombocytopenia | Open-Label, Dose-Escalation | 15 | 3 - 24 µg/kg | Terminated |
| NCT00517855 | Phase 1 Safety and PK Study | I | Healthy Volunteers | Randomized, Placebo-Controlled, Single Dose | 20 | 0.30 - 0.45 µg/kg | Completed |
Sources: [12]
The clinical pharmacology of PRTX-100 was evaluated in healthy volunteers and in patient populations during the Phase 1 programs.[4] Pharmacokinetic parameters were secondary endpoints in most of the clinical trials.[14]
Analysis of data from the Phase Ib RA study after the first intravenous dose provided key insights into the drug's disposition.[4]
Table 3: Pharmacokinetic Parameters of PRTX-100 in Patients with Rheumatoid Arthritis (First Dose, Day 0)
| PRTX-100 Dose (µg/kg) | Cmax (ng/mL) Arithmetic Mean (SD) | AUC(0–168) (h·ng/mL) Arithmetic Mean (SD) | Vz (mL/kg) Mean (SD) | Half-life (hours) Mean (SD) |
|---|---|---|---|---|
| 0.15 | 4.25 (1.11) | 40.8 (55.8) | 40.3 (9.43) | 2.5 (1.23) |
| 0.45 | 16.06 (2.80) | 268.2 (255.0) | 46.4 (20.3) | 3.5 (1.8) |
| 0.90 | 27.97 (10.72) | 210.9 (124.2) | 57.6 (43.0) | 8.4 (5.9) |
| 1.50 | 52.84 (11.15) | 1132.8 (N/A) | 57.8 (43.2) | 15.1 (20.5) |
Adapted from Bernton et al., 2014 [4]
The development of anti-drug antibodies was a major and ultimately decisive challenge for the PRTX-100 program. As a non-human protein derived from bacteria, a high rate of immunogenicity was a predictable risk.
This quantitative evidence reveals the central scientific reason for the drug's failure. The development of ADAs was not a minor issue but a fundamental pharmacological barrier. A 95-96% reduction in drug exposure and half-life by the fourth weekly dose indicates that the drug was being cleared from circulation almost immediately upon infusion in patients with a robust immune response. An immunomodulatory drug that requires sustained interaction with its cellular targets cannot exert a meaningful or durable clinical effect under such conditions. This ADA-mediated neutralization of the drug provides a clear mechanistic explanation for the weak and inconsistent efficacy signals observed throughout the clinical program.
Across the entire clinical program, which involved more than 160 subjects in eight studies, PRTX-100 was consistently described as having an acceptable safety profile and being generally well-tolerated at the intravenous doses tested, which went up to 24 µg/kg weekly in the ITP trials.[7] The majority of adverse events (AEs) considered related to PRTX-100 administration were mild to moderate in severity.[7]
The pattern of common AEs was consistent across the different patient populations, with many events suggestive of a transient, systemic inflammatory response.
This consistent reporting of inflammatory-type symptoms like arthralgia, muscle pain, and disease flares is significant. These are likely not off-target toxicities but rather on-target pharmacodynamic consequences of the drug's mechanism of action. As a B-cell superantigen and a former of immune complexes, PRTX-100 is designed to activate specific components of the immune system. The observed AEs suggest that this activation, even at very low doses, can lead to a counterproductive, transient pro-inflammatory state. This points to a potentially narrow therapeutic window, where the dose required for beneficial immunomodulation is very close to a dose that triggers undesirable inflammatory side effects, which could confound or even negate any therapeutic benefit.
No deaths were attributed to PRTX-100, and no treatment-related SAEs were reported in the larger RA studies or the later ITP trials.[7] Infusion reactions were observed in a minority of patients but were generally manageable with symptomatic therapy.[7] One notable safety event was reported from an early ITP study, where a patient developed a vasculitic reaction after two doses and was discontinued from the trial.[47] Preclinical data also point to a potential mechanism for toxicity, showing that SpA-IgG complexes can induce necrosis in human B-cells and monocytes, a phenomenon dependent on the drug's ability to bind immunoglobulins.[47]
Table 4: Summary of Common Treatment-Related Adverse Events Across Clinical Programs
| Adverse Event | Indication(s) Reported In | Reported Frequency/Severity |
|---|---|---|
| Nausea | RA, ITP | Common, Mild-to-Moderate |
| Headache | RA, ITP | Common, Mild-to-Moderate |
| Muscle Spasms / Pain | RA, ITP | Common, Mild-to-Moderate |
| Arthralgia / RA Flare | RA, ITP | Common, Mild-to-Moderate |
| Dizziness | RA | Common, Mild-to-Moderate |
| Flushing | RA | Common, Mild-to-Moderate |
| Fatigue | RA | Common, Mild-to-Moderate |
| Infusion Reactions | ITP | Common, Mild-to-Moderate |
| Abdominal Pain | ITP | Common, Mild-to-Moderate |
| Rash | ITP | Common, Mild-to-Moderate |
Sources: [4]
The PRTX-100 program achieved several important regulatory milestones, particularly for the ITP indication.
Despite these regulatory achievements, the entire clinical development program for PRTX-100 has been discontinued. Evidence for this is definitive and comes from multiple sources. All key clinical trials for ITP are officially listed on ClinicalTrials.gov with a status of "Terminated," indicating they were stopped early and would not restart.[12] Furthermore, multiple independent pharmaceutical development databases explicitly list the status of Bevifimod as "Discontinued".[12]
The ultimate factor sealing the fate of PRTX-100 was the collapse of its sponsoring company, Protalex, Inc. (OTCQB: PRTX).[10] It is essential to distinguish this entity from the currently active and unrelated biopharmaceutical company, Protalix BioTherapeutics, Inc. (NYSE: PLX).[52]
On June 25, 2021, the U.S. Securities and Exchange Commission (SEC) issued an opinion formally revoking the registration of Protalex, Inc.'s securities.[16] The basis for this action was the company's persistent failure to comply with its periodic filing obligations under the Securities Exchange Act of 1934, including required annual (Form 10-K) and quarterly (Form 10-Q) reports. The company was found to be in default after failing to respond to the SEC's OIP, indicating that it was no longer a functioning corporate entity.[16]
The timeline of these events suggests a cascade of failure. The clinical trials were terminated in the 2018-2019 period, likely due to the mounting evidence of high immunogenicity and weak efficacy, which would have made securing additional financing or partnerships exceptionally difficult. This financial and scientific pressure likely led to the company's operational collapse and its inability to meet its basic corporate filing requirements, which in turn triggered the SEC's enforcement action. The corporate demise was therefore not an isolated event but the final consequence of the preceding scientific and clinical failures of its lead asset.
The history of PRTX-100 (Bevifimod) is a compelling case study of an investigational therapeutic with a strong, elegant scientific rationale that ultimately failed to translate into a viable clinical product. The concept of harnessing a bacterial virulence factor for targeted immunomodulation was innovative. The dual mechanism of action, which aimed to simultaneously reduce the production of pathogenic autoantibodies and inhibit the effector cells responsible for tissue damage, was well-supported by preclinical data and represented a potentially disease-modifying approach for conditions like ITP and RA.
However, this promising concept was confronted by the harsh realities of human clinical pharmacology. While the drug's safety profile was acceptable at the low doses tested, it was not entirely benign, with on-target inflammatory side effects suggesting a narrow therapeutic window. More importantly, the clinical efficacy signals were consistently weak and failed to demonstrate a clear, robust benefit. The central scientific flaw was the drug's profound immunogenicity. The predictable and potent ADA response in most patients led to rapid drug clearance, preventing the sustained exposure required for an immunomodulatory agent to work effectively.
PRTX-100 did not fail for a single reason but rather from an interconnected cascade of scientific, clinical, and corporate shortcomings.
The story of PRTX-100 offers valuable lessons for the field of biopharmaceutical development. It underscores the immense challenge of translating therapies derived from foreign proteins, particularly those with inherent, potent immunological activity like bacterial superantigens. While such molecules offer novel mechanisms, their inherent immunogenicity may represent a fundamental barrier to success. Future attempts to develop therapeutics from similar sources would likely require advanced protein engineering techniques, such as de-immunization or PEGylation, to mitigate the host immune response.
Ultimately, PRTX-100 is a discontinued asset with no future clinical or commercial potential. Its development serves as a cautionary tale, illustrating how a promising preclinical concept can be defeated by the complex interplay of human pharmacology, clinical trial outcomes, and the critical importance of sustained corporate and financial viability.
Published at: October 31, 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.