TNF a Mutant Human

Tumor Necrosis Factor-Alpha Mutant Human Recombinant
Shipped with Ice Packs
In Stock

Description

Key Mutations and Their Rationale

TNF-α mutants are developed by altering residues critical for receptor binding or trimer stability. Common modifications include:

Mutant NameModificationsPurposeSource
rmhTNF (NAX®)Deletion of N-terminal 7 residues; substitutions: P8R, S9K, D10R, L157FEnhanced cytotoxicity, reduced toxicity
M3SL29S, S52I, Y56F, N-terminal 7-residue deletionReduced systemic toxicity
R1antTNFA84S, V85T, S86T, Y87H, Q88N, T89QSelective TNFR1 antagonism
XPro1595A145R, Y87HDominant-negative inhibition of TNF

These mutations disrupt receptor-binding interfaces (e.g., TNFR1 vs. TNFR2) or stabilize trimer conformations to alter signaling outcomes .

Receptor Selectivity and Signaling

  • Wild-type TNF-α: Binds both TNFR1 (ubiquitous) and TNFR2 (immune cells), activating NF-κB, MAPK, and apoptotic pathways .

  • Mutants:

    • rmhTNF: 50-fold higher cytotoxicity on L929 cells compared to wild-type, with reduced inflammatory toxicity in vivo .

    • R1antTNF: Binds TNFR1 with fast kinetics, promoting apoptosis over NF-κB activation .

    • XPro1595: Forms inactive heterotrimers with wild-type TNF, blocking receptor activation .

Anti-Tumor Efficacy

  • rmhTNF + Chemotherapy: In phase II trials for non-small cell lung cancer (NSCLC), response rates improved from 17.78% (chemotherapy alone) to 48.89% .

  • M3S: Demonstrates superior tumor suppression in murine models despite 10-fold lower systemic toxicity .

Toxicity Reduction

  • LD50: rmhTNF exhibits ≥50-fold higher LD50 than wild-type TNF in mice .

  • R1antTNF: PEGylation extends half-life and reduces immunogenicity in multiple sclerosis models .

Recombinant Expression

  • Host: E. coli (e.g., rmhTNF, M3S) .

  • Purity: >95% via ammonium sulfate precipitation and chromatography .

  • Bioactivity: Measured via L929 cell cytotoxicity assays (ED50 < 0.05 ng/mL for rmhTNF) .

Stability Enhancements

  • Thermal Stability: M3S shows increased resistance to denaturation .

  • Protease Resistance: Rigid loop structures in M3S reduce trypsin cleavage .

Challenges and Future Directions

  • Selectivity vs. Efficacy: Balancing TNFR1/TNFR2 binding remains challenging .

  • Delivery Systems: PEGylation and single-chain formats (e.g., scR1antTNF) improve pharmacokinetics .

  • Clinical Translation: Long-term safety and immunogenicity of mutants require further study .

Product Specs

Introduction
TNF-alpha (TNF-a) is a potent anti-tumor agent, but its clinical use is hampered by significant pro-inflammatory side effects. These adverse effects include fever, hypotension, liver damage, blood clots, and bleeding. To address these limitations, researchers have focused on developing modified TNF-a molecules (mutants) with reduced toxicity while preserving anti-tumor activity. Studies in mice have shown that human TNF-a, unlike murine TNF-a, binds to only one of the two murine TNF receptors (TNF-R55). This selective binding profile correlates with potent anti-tumor effects and reduced toxicity. Based on this finding, numerous TNF-a mutants that selectively bind to TNF-R55 have been created and demonstrated promising results. These mutants display cytotoxic activity against tumor cells in laboratory settings and exhibit a reduced toxicity profile in living organisms. This recombinant human TNF-a variant has specific changes in its amino acid sequence, including a deletion of amino acids 1-7 and substitutions at positions 8, 9, 10, and 157. These modifications are reported to enhance its activity and minimize inflammatory side effects in vivo.
Description
Recombinant Human TNF-alpha Variant is a non-glycosylated protein produced in E. coli. It consists of a single polypeptide chain with 151 amino acids, resulting in a molecular weight of 16598 Daltons. The purification process involves standard chromatographic techniques to ensure its quality and purity.
Physical Appearance
Sterile White lyophilized powder.
Formulation
The protein was freeze-dried after extensive dialysis in a 0.5x PBS solution at a pH of 7.
Solubility
To reconstitute the lyophilized TNF-alpha Variant, it is recommended to dissolve it in sterile 18 megaohm-centimeter (MΩ·cm) H2O at a concentration of at least 100 micrograms per milliliter (µg/ml). Once reconstituted, it can be further diluted in other aqueous solutions as needed.
Stability
Lyophilized TNF-alpha Variant is stable at room temperature for up to 3 weeks; however, it is recommended to store it desiccated at a temperature below -18 degrees Celsius (-0.4 degrees Fahrenheit) for optimal long-term storage. After reconstitution, the TNF-alpha Variant should be stored at 4 degrees Celsius (39.2 degrees Fahrenheit) for up to 7 days. For extended storage, freezing below -18 degrees Celsius (-0.4 degrees Fahrenheit) is advised. To further enhance stability during long-term storage, consider adding a carrier protein like HSA or BSA at a concentration of 0.1%. Avoid repeated freeze-thaw cycles to maintain protein integrity and activity.
Purity
The purity of this product is greater than 95%, as determined by two independent analytical methods: Reverse-Phase High-Performance Liquid Chromatography (RP-HPLC) and SDS-PAGE analysis.
Biological Activity
The biological activity of this TNF-alpha Variant was assessed by measuring its cytotoxic effect on murine L929 cells in the presence of Actinomycin D. The ED50, which represents the concentration required to achieve 50% cell lysis, was determined to be less than 0.05 nanograms per milliliter (ng/ml). This corresponds to a specific activity of 20,000,000 units per milligram (units/mg).
Synonyms
TNF-alpha, Tumor necrosis factor ligand superfamily member 2, TNF-a, Cachectin, DIF, TNFA, TNFSF2.
Source
Escherichia Coli.
Amino Acid Sequence
MRKRKPVAHV VANPQAEGQL QWLNRRANAL LANGVELRDN
QLVVPSEGLY LIYSQVLFKG QGCPSTHVLL THTISRIAVS YQTKVNLLSA IKSPCQRETP EGAEAKPWYE PIYLGGVFQL EKGDRLSAEI NRPDYLDFAE SGQVYFGIIAF.

Q&A

What is TNF-alpha and how was it originally identified?

TNF-alpha is a multifunctional cytokine first observed in the 1960s for its ability to induce tumor regression in mice treated with Serratia marcescens polysaccharide. The actual protein was not structurally identified until 1984, when researchers isolated two cytotoxic factors: a 17 kDa protein from macrophages (named TNF, later TNF-α) and a 20 kDa protein from lymphocytes (named lymphotoxin, later TNF-β) . These proteins exhibited approximately 50% amino acid sequence homology and bound to the same cell surface receptor despite being immunologically distinct . Their identification led to cDNA cloning and the discovery that TNF-alpha exhibits cytotoxic, cytostatic, and immunomodulatory effects on malignant tumors .

What signaling pathways does TNF-alpha activate in target cells?

TNF-alpha induces at least five different types of signaling pathways upon binding to its receptors . These include:

  • NF-κB activation - leading to inflammatory gene expression

  • Apoptosis pathways - mediated through death domain interactions

  • Extracellular signal-regulated kinase (ERK) activation

  • p38 mitogen-activated protein kinase (p38MAPK) signaling

  • c-Jun N-terminal kinase (JNK) activation

When TNF-alpha binds to TNFR1, it recruits TNFR-associated death domain (TRADD) through its death domain, which serves as a platform for subsequent signaling events . The specific outcome depends on cell type, receptor expression levels, and the presence of various adaptor proteins.

Why has clinical application of wild-type TNF-alpha been limited?

Despite its remarkable ability to selectively kill or inhibit malignant cell lines, especially in combination with interferon, clinical trials of TNF-alpha in cancer patients have been disappointing . The primary limitation is its high systemic toxicity, creating a narrow therapeutic window . It has been estimated that an effective TNF dose would be 5-25 times the maximum tolerated dose in humans . This toxicity profile has severely hampered TNF-alpha's utilization as an anti-cancer agent despite its promising anti-tumor effects .

What is the TNF superfamily and how extensive is it?

Based on sequence homology to TNF-alpha, a total of 19 members of the TNF superfamily have been identified, along with 29 interacting receptors and several molecules that interact with the cytoplasmic domains of these receptors . The TNF superfamily members play critical roles in:

  • Inflammation

  • Apoptosis

  • Proliferation

  • Invasion

  • Angiogenesis

  • Metastasis

  • Morphogenesis

Their involvement spans immunologic, cardiovascular, neurologic, pulmonary, and metabolic diseases . The therapeutic targeting of TNF superfamily members has created a market exceeding US $20 billion, particularly for conditions like rheumatoid arthritis, psoriasis, and Crohn's disease .

What diseases are associated with mutations in TNF-alpha and its receptors?

Various mutations in genes for TNF, its family members, and its receptors have been identified in humans . Specifically for TNF-alpha and its receptors:

GeneDisease Association
TNF-αCerebral infarction
TNF-β (LT-α)Cerebral infarction
TNFR1TNFR1-associated periodic syndrome (TRAPS), TRAPS associated with SLE, Crohn's disease
TNFR2Crohn's disease

Two biallelic polymorphisms in TNF-α (TNF-α–308) and TNF-β (TNF-β +252) genes have been associated with altered TNF production and susceptibility to inflammatory diseases . These genetic variations provide important insights into the physiological and pathological roles of TNF family members.

What specific amino acid modifications have proven most effective in enhancing TNF's anti-tumor activity?

Research has identified several key amino acid modifications that significantly enhance TNF's therapeutic properties. One well-documented mutant (recombinant mutated human TNF; rmhTNF) incorporated the following modifications :

  • Deletion of the first seven N-terminal amino acids

  • Substitution of amino acids at positions 8, 9, and 10 (Pro, Ser, and Asp) with Arg, Lys, and Arg

  • Substitution of C-terminal Leu157 with Phe

This engineered variant exhibited approximately 50-fold increased cytotoxicity against the mouse fibroblast cell line L929 compared to wild-type rhTNF in standard cytotoxicity tests . Importantly, it also demonstrated at least 50 times higher LD50 in mice, suggesting a substantially improved therapeutic window . This combination of heightened efficacy and reduced toxicity makes such mutants promising candidates for clinical development.

What expression systems and purification protocols yield optimal results for recombinant mutated TNF proteins?

For the production of recombinant mutated human TNF (rmhTNF), researchers have successfully employed prokaryotic expression systems using E. coli DH5alpha with the PLPR promoter . The expression results in partially soluble protein that can be purified using a multi-step approach:

  • Initial extraction from the supernatant of cell lysate

  • Ammonium sulfate precipitation for initial capture

  • Two sequential chromatographic steps for purification

This methodology yielded purified rmhTNF with >95% purity as determined by SDS-PAGE with silver staining and high-pressure size exclusion chromatography (SEC-HPLC) . The production efficiency was approximately 1.22 mg/g wet cell paste .

The choice of expression system significantly impacts yield, folding, and post-translational modifications. While bacterial systems offer high yield and simplicity, mammalian expression systems may provide advantages for more complex TNF variants requiring specific folding conditions.

How do in vivo and in vitro assays for TNF mutants differ methodologically?

The evaluation of TNF mutants requires complementary in vitro and in vivo approaches, each with distinct methodological considerations:

In vitro assays:

  • Standard cytotoxicity testing on cell lines (particularly L929 mouse fibroblasts)

  • Direct measurement of cell death, typically with sensitizing agents like actinomycin D

  • Controlled dose-response studies to determine EC50 values

  • Isolated assessment of specific mechanisms without systemic influences

In vivo assays:

  • Tumor cell transplanted mice models

  • Nude mice xenograft studies

  • Assessment of tumor growth inhibition or regression

  • Evaluation of systemic toxicity parameters (weight loss, cytokine levels)

  • Determination of maximum tolerated dose and LD50

Research has demonstrated that rmhTNF exhibits enhanced cytotoxicity compared to wild-type rhTNF in both systems, though the magnitude of effect may differ between in vitro and in vivo contexts . This highlights the importance of integrated assessment approaches when evaluating novel TNF mutants.

What mechanisms underlie the increased cytotoxicity of TNF mutants?

The dramatically enhanced cytotoxicity observed with engineered TNF mutants like rmhTNF (approximately 50-fold increase compared to wild-type) likely results from several molecular mechanisms:

  • Altered receptor binding dynamics:

    • Substitution of amino acids 8-10 with positively charged residues (Arg, Lys, Arg) may enhance receptor interactions

    • Deletion of N-terminal amino acids could reduce steric hindrance

  • Receptor selectivity changes:

    • Some mutants show selective activity on the p55 receptor (TNFR1)

    • Modified binding may preferentially activate cytotoxic pathways over pro-inflammatory ones

  • Altered protein stability:

    • C-terminal modifications (Leu157 to Phe) may enhance protein stability

    • Resistance to proteolytic degradation could increase effective concentration

  • Modified downstream signaling:

    • Enhanced activation of pro-apoptotic pathways

    • Potentially reduced activation of pro-survival NF-κB signaling

Understanding these mechanisms is crucial for the rational design of next-generation TNF variants with further optimized therapeutic properties.

How can researchers distinguish between receptor-specific effects when evaluating novel TNF mutants?

Distinguishing receptor-specific effects requires specialized experimental approaches:

  • Receptor expression manipulation:

    • Utilize cell lines with differential expression of TNFR1 (p55) versus TNFR2 (p75)

    • Employ receptor knockout/knockdown models

    • Use receptor-specific neutralizing antibodies

  • Pathway-specific readouts:

    • Monitor TNFR1-specific signaling (death domain interactions, TRADD recruitment)

    • Assess TNFR2-specific pathways (TRAF signaling)

    • Analyze differential gene expression profiles

  • Binding kinetics assessment:

    • Competitive binding assays with labeled wild-type TNF

    • Surface plasmon resonance to determine binding constants

    • Cross-linking studies to identify binding partners

This is particularly relevant given research demonstrating that TNF mutants with selective activity on the p55 receptor have been developed , which may provide enhanced therapeutic potential through more targeted biological activities with potentially reduced side effects.

What are the key considerations in designing TNF mutation studies?

When designing studies to create and evaluate TNF mutants, researchers should consider:

  • Structural targeting approach:

    • Focus on regions known to interact with receptors

    • Consider conserved versus variable regions across species

    • Target domains involved in oligomerization or stability

  • Mutation strategy:

    • Single point mutations versus multiple substitutions

    • Conservative versus non-conservative substitutions

    • Domain deletions or insertions

    • Fusion proteins with targeting moieties

  • Screening methodology:

    • High-throughput versus rational design approaches

    • Selection of appropriate bioassays for activity assessment

    • Comparative analysis with wild-type controls

  • Translation to therapeutic potential:

    • Balance between enhancing cytotoxicity and reducing systemic toxicity

    • Consider immunogenicity of heavily modified variants

    • Assess impact on pharmacokinetic properties

The successful development of rmhTNF demonstrates the value of a targeted approach focusing on specific N-terminal and C-terminal modifications to achieve a 50-fold increase in cytotoxicity with substantially reduced systemic toxicity .

How should dose-response data for TNF mutants be analyzed to determine therapeutic windows?

Rigorous analysis of dose-response data is essential for accurately determining the therapeutic window of TNF mutants:

  • Potency assessment:

    • Calculate EC50 values using non-linear regression

    • Determine relative potency compared to wild-type TNF

    • Assess maximum efficacy (Emax) and Hill coefficients

  • Toxicity evaluation:

    • Determine LD50 in appropriate animal models

    • Calculate therapeutic index (LD50/ED50 ratio)

    • Assess multiple toxicity parameters (weight loss, cytokine profiles)

  • Statistical considerations:

    • Use confidence intervals for potency ratios

    • Apply appropriate statistical tests for comparing mutants

    • Consider variability across experimental systems

For rmhTNF, the approximately 50-fold increase in cytotoxicity in the L929 assay combined with at least 50 times higher LD50 in mice compared to wild-type rhTNF suggests a dramatically improved therapeutic window that can be quantified through these analytical approaches.

What in vivo models are most appropriate for evaluating TNF mutant efficacy?

Selection of appropriate in vivo models is critical for translational evaluation of TNF mutants:

  • Xenograft models:

    • Human tumor cell lines in immunodeficient mice

    • Patient-derived xenografts for heterogeneity representation

    • Assessment of direct cytotoxic effects

  • Syngeneic models:

    • Mouse tumors in immunocompetent mice

    • Evaluation of immune-mediated effects

    • More representative of complex tumor-host interactions

  • Specialized models:

    • Orthotopic implantation for appropriate microenvironment

    • Metastatic models for systemic disease

    • Combination therapy models (with interferon, chemotherapy)

Research has demonstrated that both tumor cell transplanted mice and nude mice models effectively show the enhanced cytotoxicity of rmhTNF compared to wild-type rhTNF , validating these approaches for initial evaluation of engineered TNF variants.

How can researchers effectively measure the specific activity of TNF mutants?

Standardized assays for specific activity assessment are essential for comparing TNF variants:

  • Cell-based bioassays:

    • L929 mouse fibroblast cytotoxicity assay (the gold standard)

    • Actinomycin D sensitization for enhanced detection

    • Multiple endpoint options (MTT, neutral red, crystal violet)

    • Results expressed as units/mg (1 unit = concentration causing 50% cytotoxicity)

  • Biochemical characterization:

    • Receptor binding assays

    • Oligomerization state assessment

    • Thermal and chemical stability testing

  • Standardization approaches:

    • Direct comparison to international reference standards

    • Inclusion of wild-type TNF as internal control

    • Parallel line bioassay analysis

The rmhTNF mutant demonstrated approximately 50-fold increased specific activity compared to wild-type rhTNF in the standard L929 cytotoxicity assay , highlighting the importance of standardized bioassays for accurate potency comparison.

What are common pitfalls in interpreting TNF mutant activity across different experimental systems?

Researchers should be aware of several critical interpretation challenges:

  • Species-specific differences:

    • Human TNF is approximately 50-fold less toxic to mice than murine TNF

    • Receptor binding affinities vary across species

    • Immune response variations between species

  • Cell line variability:

    • Different receptor expression levels

    • Varied downstream signaling pathway activation

    • Cell-specific survival mechanisms

  • Experimental condition impacts:

    • Serum concentration effects

    • Presence/absence of sensitizing agents

    • Incubation time dependencies

  • In vitro to in vivo translation:

    • Pharmacokinetic factors absent in vitro

    • Tumor microenvironment complexity

    • Immune system interactions

Understanding these pitfalls is crucial when interpreting the remarkable 50-fold increase in cytotoxicity observed with rmhTNF and ensuring these findings translate appropriately to therapeutic applications.

What novel TNF engineering approaches show promise for enhanced therapeutic applications?

Several innovative approaches for TNF engineering warrant further investigation:

  • Receptor-selective variants:

    • Development of mutants with selective activity on either p55 or p75 receptors

    • Creation of receptor antagonists for inflammatory disease applications

    • Dual-receptor targeting with modified kinetics

  • Tumor-targeted delivery:

    • Fusion proteins with tumor-targeting domains

    • Antibody-TNF conjugates for localized delivery

    • Nanoparticle formulations for passive targeting

  • Controlled activation systems:

    • Protease-activated TNF mutants for tumor-specific activation

    • pH-sensitive variants for microenvironment-specific activity

    • Light-activated TNF for spatiotemporal control

  • Combination therapy optimization:

    • Synergistic combinations with checkpoint inhibitors

    • TNF mutants designed for enhanced interferon cooperation

    • Rational combinations with conventional chemotherapy

The dramatic improvements in therapeutic window achieved with rmhTNF (50-fold increased cytotoxicity with at least 50 times higher LD50) suggest that engineered TNF variants have significant untapped potential for cancer therapy.

How might genetics influence individual response to TNF-based therapies?

Genetic factors substantially impact response to TNF-targeted therapies:

  • TNF pathway polymorphisms:

    • TNF-α–308 and TNF-β+252 polymorphisms affect TNF production and disease susceptibility

    • These variants have been linked to cerebral infarction risk

    • Similar polymorphisms may influence therapeutic response

  • Receptor genetics:

    • TNFR1 mutations associated with TNFR1-associated periodic syndrome and Crohn's disease

    • TNFR2 variants linked to Crohn's disease

    • Receptor polymorphisms may alter binding of engineered TNF variants

  • Downstream signaling pathway variation:

    • Adaptors like TRAF3 and TRAF6 show disease-associated mutations

    • Genetic variation in NF-κB pathway components

    • Apoptosis pathway polymorphisms

This genetic landscape suggests that personalized approaches to TNF-based therapies may be necessary for optimal outcomes, potentially including genetic screening to predict response to specific TNF mutants.

What lessons from TNF research might apply to other cytokine engineering efforts?

The successful engineering of TNF mutants offers valuable insights for other cytokine optimization efforts:

  • Structure-function relationship principles:

    • Terminal modifications can dramatically alter activity

    • Charge modifications impact receptor interactions

    • Subtle changes can create receptor selectivity

  • Therapeutic window optimization strategies:

    • Balancing potency enhancement with toxicity reduction

    • Receptor selectivity as a mechanism for reducing side effects

    • Importance of comparative in vivo toxicity assessment

  • Expression and manufacturing considerations:

    • Bacterial expression systems can effectively produce non-glycosylated cytokines

    • Multi-step purification protocols yield high purity

    • Quality control via multiple orthogonal methods

  • Translational research approaches:

    • Bridging in vitro potency to in vivo efficacy

    • Importance of appropriate animal models

    • Recognition of species-specific differences

The remarkable 50-fold improvement in cytotoxicity achieved with rmhTNF demonstrates the tremendous potential of protein engineering for enhancing therapeutic properties of naturally occurring cytokines.

Product Science Overview

Structure and Function

The human TNF-α gene is located on chromosome 6p21.3 and spans approximately 3 kilobases. It encodes a 233-amino acid-long type II transmembrane protein that forms stable homotrimers. These trimers are the biologically active form of TNF-α. The primary role of TNF-α is to regulate immune cells, induce apoptosis, inhibit tumorigenesis, and retard the proliferation, angiogenesis, and metastasis of cancer cells .

Recombinant Mutant TNF-α

Recombinant mutant human TNF-α (rmhTNF) is a modified form of the natural TNF-α protein. This modification is achieved through genetic engineering techniques, such as polymerase chain reaction (PCR), to alter the TNF-α gene. The resulting product is a non-glycosylated single chain consisting of 151 amino acids with a molecular weight of approximately 16,598 Da .

One notable mutant is the R31D mutant, which has been engineered to preferentially bind to TNF receptor R1 with greater affinity compared to receptor R2. This selective binding is achieved by replacing the arginine residue at position 31 with aspartic acid. The R31D mutant has shown to have a higher affinity for receptor R1, which is associated with pro-inflammatory and apoptotic signaling, while having reduced binding to receptor R2, which is involved in anti-inflammatory and cell survival signaling .

Clinical Applications

Recombinant mutant TNF-α has been investigated for its potential therapeutic applications, particularly in cancer treatment. For example, thoracic perfusion of rmhTNF combined with cisplatin has been shown to improve the objective response rate and quality of life in patients with malignant pleural effusion (MPE) caused by lung cancer . This combination therapy has demonstrated efficacy in controlling MPE and alleviating symptoms such as dyspnea and chest pain.

Additionally, TNF-α mutants that selectively bind to specific TNF receptors have been designed to retain anti-tumor activity while reducing systemic toxicity. For instance, human TNF-α mutants that bind to the murine TNF-R55 receptor but not to the mouse TNF-R75 receptor have shown retained anti-tumor activity and reduced systemic toxicity in mice .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.