Anti-Mouse DR5 (CD262) (Clone MD5-1) – Purified in vivo PLATINUM™ Functional Grade

Anti-Mouse DR5 (CD262) (Clone MD5-1) – Purified in vivo PLATINUM™ Functional Grade

Product No.: D231

- -
- -
Clone
MD5-1
Target
DR5
Formats AvailableView All
Product Type
Hybridoma Monoclonal Antibody
Alternate Names
TRAIL-R2, KILLER, TRICK2, TNFRSF10B, Ly98, CD262
Isotype
Armenian Hamster IgG κ
Applications
Agonist
,
FA
,
FC
,
IP
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Mouse
Host Species
Armenian Hamster
Recommended Dilution Buffer
Immunogen
Mouse DR5-Ig fusion protein
Product Concentration
≥ 5.0 mg/ml
Endotoxin Level
<0.5 EU/mg as determined by the LAL method
Purity
≥98% monomer by analytical SEC
>95% by SDS Page
Formulation
This monoclonal antibody is aseptically packaged and formulated in 0.01 M phosphate buffered saline (150 mM NaCl) PBS pH 7.2 - 7.4 with no carrier protein, potassium, calcium or preservatives added. Due to inherent biochemical properties of antibodies, certain products may be prone to precipitation over time. Precipitation may be removed by aseptic centrifugation and/or filtration.
State of Matter
Liquid
Product Preparation
Functional grade preclinical antibodies are manufactured in an animal free facility using in vitro cell culture techniques and are purified by a multi-step process including the use of protein A or G to assure extremely low levels of endotoxins, leachable protein A or aggregates.
Pathogen Testing
To protect mouse colonies from infection by pathogens and to assure that experimental preclinical data is not affected by such pathogens, all of Leinco’s Purified Functional PLATINUM<sup>TM</sup> antibodies are tested and guaranteed to be negative for all pathogens in the IDEXX IMPACT I Mouse Profile.
Storage and Handling
Functional grade preclinical antibodies may be stored sterile as received at 2-8°C for up to one month. For longer term storage, aseptically aliquot in working volumes without diluting and store at ≤ -70°C. Avoid Repeated Freeze Thaw Cycles.
Regulatory Status
Research Use Only
Country of Origin
USA
Shipping
2 – 8° C Wet Ice
Additional Applications Reported In Literature ?
Agonist,
FA,
FC,
IP,
WB
Each investigator should determine their own optimal working dilution for specific applications. See directions on lot specific datasheets, as information may periodically change.

Description

Description

Specificity
MD5-1 activity is directed against mouse DR5 (CD262).
Background
DR5 is a member of the TNF-related apoptosis-inducing ligand (TRAIL) receptor family and acts as an agonist receptor that transmits death signals, it is also a transcriptional target of the known oncogene p531,2,3. Additionally, DR5 negatively regulates innate immune responses in dendritic cells4. DR5 is expressed in solid tumors and hematological malignancies in both mouse and humans1, where it induces apoptosis via its functional cytoplasmic death domains2. DR5 initiates apoptosis signals when its ligand, TRAIL, or an agonist monoclonal antibody, e.g., MD5-1, triggers a functional trimer configuration of its transmembrane helices and cytosolic domains1. Apoptosis is induced in many types of transformed cells but not in normal cells5. Since TRAIL death receptors are elevated in a wide range of solid tumors, they are being investigated for the treatment of cancer1,2.

MD5-1 was generated by immunizing an Armenian hamster with mouse DR5-Ig fusion protein5. Resulting splenocytes were fused with P3U1 mouse myeloma cells and screened for reactivity to mouse DR5-transfected BHK cells by flow cytometry. MD5-1 reacts with all TRAIL-sensitive tumor cells, including 4T1 mammary carcinoma and R331 renal carcinoma. MD5-1 acts as a death-inducing agonist when cross-linked by streptavidin, anti-hamster Ig monoclonal antibody, or FcR. Cytotoxic activity is completely abrogated by pan-caspase inhibitor z-VAD-fmk.

MD5-1 has been tested as a cancer therapy in mouse models of human cancer5,6. MD5-1 inhibits TRAIL-sensitive tumor cell growth in vivo without toxicity and also primes tumor-specific T cells5. Additionally, MD5-1 in combination with anti-CTLA-4 retards tumor growth7.

Antigen Distribution
DR5 (also known as CD262, TRAIL-Receptor 2, TNFRSF10b) is a plasma membrane bound receptor that also localizes to the cytoplasm and nucleus.
Ligand/Receptor
TRAIL (CD253); cytoplasmic domain interacts with TRADD and RIP
NCBI Gene Bank ID
UniProt.org
Research Area
Apoptosis
.
Cell Biology
.
Cell Death
.
Immunology
.
Tumor Suppressors

Leinco Antibody Advisor

Powered by AI: AI is experimental and still learning how to provide the best assistance. It may occasionally generate incorrect or incomplete responses. Please do not rely solely on its recommendations when making purchasing decisions or designing experiments.

Clone MD5-1 is an agonistic antibody against mouse DR5 (CD262/TRAIL-R2) with several key in vivo applications in preclinical mouse models, primarily focused on cancer research and immunotherapy.

Cancer Therapy and Tumor Rejection

MD5-1 has been extensively tested as a cancer therapeutic agent in mouse models. The antibody induces TRAIL-mediated apoptosis in tumor cells when administered in vivo. Studies have demonstrated its efficacy against various tumor types, including mammary carcinoma (4T1) and renal carcinoma (R331) in syngeneic tumor models. When administered at doses of 200 μg on days 0, 4, and 8 following tumor inoculation, MD5-1 can promote complete tumor rejection in immunocompetent mice.

Induction of Tumor-Specific T Cell Immunity

Beyond direct cytotoxic effects, MD5-1 generates lasting antitumor immunity. Mice that reject tumors after MD5-1 treatment develop immunological memory, becoming resistant to rechallenge with the same tumor cells. This protective immunity is mediated by both CD8+ and CD4+ T cells and involves death ligands including FasL and TRAIL. The antibody effectively primes T cell responses, with the kinetics of this priming critical for establishing durable antitumor immunity.

Selective Depletion of Myeloid-Derived Suppressor Cells

A particularly important application is the selective depletion of myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment. A single dose of MD5-1 significantly reduces MDSC accumulation, decreasing granulocytic MDSCs by approximately 40% and monocytic MDSCs by approximately 76% in tumor tissues. This depletion also occurs systemically in the spleen, reducing gMDSCs by about 26% and mMDSCs by about 34%. Importantly, MD5-1 selectively targets MDSCs without affecting other myeloid populations such as macrophages and dendritic cells. The reduction in immunosuppressive MDSCs leads to increased CD3+ T cell infiltration in tumors, thereby enhancing antitumor immune responses.

Combination Therapies

MD5-1 has been investigated in combination with other therapeutic agents, including histone deacetylase inhibitors like panobinostat, in multiple myeloma models. These combination approaches aim to enhance the therapeutic efficacy by targeting multiple pathways simultaneously.

Commonly used antibodies or proteins that are co-administered or studied along with MD5-1 (an anti-mouse DR5 monoclonal antibody) include:

  • DNA vaccines encoding tumor antigens, particularly HPV-16 E7 antigen fused to calreticulin (CRT/E7), are frequently combined with MD5-1 in cancer immunotherapy studies. This combination boosts antigen-specific CD8+ T cell responses and enhances antitumor effects.
  • Isotype-matched control antibodies are routinely used alongside MD5-1 as negative controls in flow cytometry or in vivo experiments to validate the specificity of MD5-1.
  • Other DR5 antibodies such as AMG655 and KMTR2 have been engineered in dual specificity constructs with MD5-1 for comparative or synergistic studies targeting DR5-mediated apoptosis pathways.
  • Antibodies against apoptosis pathway proteins, such as caspase-8, are used in immunoprecipitation or western blotting to assess downstream signaling and apoptotic activity when cells are treated with MD5-1.
  • E7-specific cytotoxic T lymphocytes (CTLs) (not antibodies, but relevant immune effectors) are often examined for their activity in conjunction with MD5-1 treatment, as MD5-1 can sensitize tumor cells to CTL-mediated lysis.

Additional frequently used reagents in experiments involving MD5-1 include:

  • Antibodies against mouse DR5/CD262 for flow cytometric detection.
  • TRAIL (TNF-related apoptosis-inducing ligand), the endogenous ligand for DR5, as a positive control or comparative agent in apoptosis studies.

These combinations are common in experimental oncology, particularly when studying synergistic or additive effects on tumor rejection, apoptosis, or immune responses.

The key findings from scientific literature citing clone MD5-1—an agonistic anti-mouse DR5 (Death Receptor 5, CD262) antibody—are as follows:

  • Induction of DR5-mediated apoptosis and cytotoxicity: MD5-1 efficiently induces apoptosis in DR5-expressing murine tumor cells, but is typically effective when Fc crosslinking occurs, mimicking immune effector cell engagement. MD5-1 cytotoxicity is strictly caspase-dependent and closely parallels sensitivity to TRAIL-mediated apoptosis.

  • Antimetastatic and antitumor effects: In murine models, MD5-1 significantly inhibits spontaneous metastasis and prolongs survival, even when administered after large tumor burdens have already formed. The antimetastatic effect is independent of endogenous TRAIL or perforin pathways, instead relying on DR5 death signaling.

  • Induction of anti-tumor immunity: Treatment with MD5-1 eradicates susceptible tumors and induces a CD8+ T cell–mediated immune response. Remarkably, this immunization effect enables mice to subsequently reject not only the original tumor but also resistant variants, emphasizing its role in immunological memory.

  • Selective depletion of immunosuppressive cells: MD5-1 selectively induces apoptosis in polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs), sparing normal neutrophils (PMNs) and other myeloid cells. This reduces tumor-induced immune suppression and enhances CD8+ T cell-mediated antitumor activity in vivo. In tumor-bearing mice, administration of MD5-1 decreases MDSC numbers and delays tumor progression.

  • Mechanistic insights and limitations:

    • The apoptotic effect of MD5-1 is lost in cells with blocked caspase 8 signaling or in the absence of Fc crosslinking.
    • While the antitumor effects are robust, they depend on the susceptibility of target cells to DR5-mediated apoptosis—cells overexpressing FLIP (an inhibitor of caspase-8) or otherwise resistant to DR5 signaling are not affected by MD5-1.
  • Safety and targeting:

    • The antitumor and potential hepatotoxic effects of MD5-1 are context-dependent and require Fc receptor engagement.
    • MD5-1 specifically targets DR5-expressing cells, making it useful in dissecting the function of DR5 and studying DR5-targeted therapies in mouse models.

These conclusions are grounded in published mechanistic studies and preclinical mouse tumor models, which collectively establish MD5-1 as a valuable tool for exploring DR5 signaling, tumor immunotherapy, and selective depletion of immunosuppressive cells in vivo.

Dosing regimens of clone MD5-1, an anti-mouse DR5 monoclonal antibody, vary across mouse models primarily in dose amount, frequency, administration route, mouse strain, and combination with other agents.

Key dosing regimens in published studies include:

  • Wild-type BALB/c, SCID, and immune-deficient models (tumor growth inhibition):

    • Dose: 200 μg per mouse, intraperitoneally (i.p.)
    • Schedule: Days 0, 4, and 8 after tumor cell inoculation.
    • Variants: Used alone or in combination with immune cell depletion or rechallenge experiments.
  • C57BL/6 mice (TC-1 tumor model):

    • Dose: 250 μg per mouse, i.p. (as 100 μl of a 2.5 mg/ml solution)
    • Schedule: Single dose on day 8 after tumor inoculation.
  • 615 mice (MFC tumor model):

    • Dose: 50 μg per mouse, i.p.
    • Schedule: Once every 3 days, for a total of three doses.
  • C57BL/6 (dermal myofibroblast targeting):

    • Dose: 100 μg per mouse, i.p.
    • Schedule: Dosing frequency not always specified, but typically resembles the above repeat dosing strategies.
  • Transgenic and syngeneic tumor models (combination therapy, e.g., panobinostat+MD5-1):

    • Dose: 50 μg per mouse, i.p.
    • Schedule: Specific timing varies depending on the study design and combination partner.

Factors influencing regimen differences:

  • Mouse strain and genetic background: Some regimens use wild-type, immune-deficient (SCID, perforin knockout), or transgenic mice to test for on-target toxicity or immune dependency.
  • Tumor model: The choice of tumor cell line (e.g., 4T1, TC-1, MFC, Vk*MYC) and tumor location affect the dose and schedule.
  • Combination therapy: Lower doses are sometimes used in combination regimens due to additive toxicities (e.g., with histone deacetylase inhibitors like panobinostat or vorinostat).

General principles:

  • The most common dose range for MD5-1 is 50–250 μg per mouse, usually administered i.p.
  • Monotherapy regimens tend to use higher single or repeat doses; combination therapy regimens often reduce dose frequency or amount due to enhanced or synergistic toxicity.
  • Treatment schedules typically start at or shortly after tumor inoculation, with injections spaced several days apart (often 3–4 days).

Summary Table: MD5-1 Dosing Regimens Across Mouse Models

Mouse Model/StrainTumor/IndicationDose (μg)RouteFrequencyReference
BALB/c, SCID4T1, R331 tumors200i.p.Days 0, 4, 8
C57BL/6TC-1 lung carcinoma250i.p.Day 8 (single)
615MFC gastric carcinoma50i.p.q3d ×3
C57BL/6Dermal myofibroblasts study100i.p.Not always specified
C57BL/6, transgenic modelsVk*MYC myeloma (combo)50i.p.Per protocol

Regimen selection should be tailored to the mouse strain, tumor type, and experimental goals, with close attention to toxicity, especially in combination therapy.

References & Citations

1. Piechocki MP, Wu GS, Jones RF, et al. Int J Cancer. 131(11):2562-2572. 2012.
2. Mert U, Sanlioglu AD. Cell Mol Life Sci. 74(2):245-255. 2017.
3. Willms A, Schupp H, Poelker M, et al. Cell Death Dis. 12(8):757. 2021.
4. Iyori M, Zhang T, Pantel H, et al. J Immunol. 187(6):3087-3095. 2011.
5. Takeda K, Yamaguchi N, Akiba H, et al. J Exp Med. 199(4):437-448. 2004.
6. Haynes NM, Hawkins ED, Li M, et al. J Immunol. 185(1):532-541. 2010.
7. Condamine T, Kumar V, Ramachandran IR, et al. J Clin Invest. 124(6):2626-2639. 2014.
8. Dufour F, Rattier T, Shirley S, et al. Cell Death Differ. 24(3):500-510. 2017.
9. Mondal T, Shivange GN, Tihagam RG, et al. EMBO Mol Med. 13(3):e12716. 2021.
10. Park C, Choi EO, Hwangbo H, et al. Nutr Res Pract. 16(3):330-343. 2022.
Agonist
FA
Flow Cytometry
Immunoprecipitation Protocol
General Western Blot Protocol

Certificate of Analysis

Formats Available

- -
- -
Disclaimer AlertProducts are for research use only. Not for use in diagnostic or therapeutic procedures.