Anti-mouse PD-1 Antibody (CD279, RMP1-14.1) | PA007162.r2a
$150.00 – $700.00
Recombinant anti-mouse PD 1/ CD279 antibody(RMP1-14.1), which share the same variable region sequences with the rat anti-mouse PD-1 antibody (clone: RMP1-14), are produced from mammalian cells. Anti-mouse PD-1 / CD279 antibody (RMP1-14.1, rat IgG2a kappa) ‘s affintiy to the mouse PD-1 protein is <2 nM. The recombinant rat IgG2a isotype control available.
- Details & Specifications
- References
- More Offers
Catalog No. | PA007162.r2a |
---|---|
Product Name | Anti-mouse PD-1 Antibody (CD279, RMP1-14.1) | PA007162.r2a |
Supplier Name | Syd Labs, Inc. |
Brand Name | Syd Labs |
Synonyms | Mouse Anti-Mouse PD 1 Monoclonal Antibodies, Murinized Anti-Mouse PD 1 Monoclonal Antibodies |
Summary | The Anti-mouse PD-1 Antibody (RMP1-14.1, Rat IgG2a Kappa ) was produced in mammalian cells. |
Clone | RMP1-14.1, the same variable region and constant region sequences as the rat anti-mouse PD-1 monoclonal antibody (clone number: RMP1-14) |
Isotype | Rat IgG2a, kappa |
Applications | immunohistochemistry (IHC), Flow Cytometry (FC), and various in vitro and in vivo functional assays. |
Immunogen | The original rat hybridoma (clone name: RMP1-14) was generated by immunizing Sprague Dawley rats with mouse PD-1-transfected BHK cells and using a P3U1 myeloma as the fusion partner. |
Form Of Antibody | 0.2 μM filtered solution of 1x PBS. |
Endotoxin | Less than 1 EU/mg of protein as determined by LAL method. |
Purity | >95% by SDS-PAGE under reducing conditions. |
Shipping | The In Vivo Grade Recombinant Anti-mouse PD-1 Antibody (RMP1-14.1) are shipped with ice pack. Upon receipt, store it immediately at the temperature recommended below. |
Stability & Storage | Use a manual defrost freezer and avoid repeated freeze-thaw cycles. 1 month from date of receipt, 2 to 8°C as supplied. 3 months from date of receipt, -20°C to -70°C as supplied. |
Note | Recombinant mouse anti-mouse PD 1 / CD279 monoclonal antibodies, whose variable region sequences are murined from the rat anti-mouse PD-1 monoclonal antibody (clone number: RMP1-14), are produced from mammalian cells. The recombinant rat and chimeric mouse versions of the RMP1-14 antibody are also available. |
Order Offline | Phone: 1-617-401-8149 Fax: 1-617-606-5022 Email: message@sydlabs.com Or leave a message with a formal purchase order (PO) Or credit card. |
Description
PA007162.r2a: Recombinant Anti-mouse PD-1 Antibody(Clone: CD279, RMP1-14.1), Rat IgG2a Kappa, In Vivo Grade
The anti mouse PD-1 antibody is widely recognized as a pivotal reagent in immunological studies, with the RMP1-14 antibody being a standout clone for targeting mouse PD-1 (CD279). This anti mouse pd 1 antibody is designed to interrupt the PD-1/PD-L1 pathway, a critical mechanism in immune checkpoint regulation, making it essential for research in cancer immunotherapy. Scientists often employ the anti mouse cd279 antibody in mouse models, such as those for lung cancer, to investigate how blocking PD-1 enhances T-cell activation and combats tumor growth. The RMP1-14 antibody also supports studies on immune tolerance in chronic infections and autoimmune conditions, offering insights into therapeutic development. Its precise binding to mouse PD-1 ensures reliable results, cementing its role as a cornerstone in advancing our understanding of immune responses.
Syd Labs provides anti-mouse PD1 monoclonal antibodies. The in vivo grade recombinant anti-mouse PD-1 monoclonal antibody (MAb Rat IgG2a Kappa) was produced in mammalian cells. Syd Labs presents a comprehensive array of anti-mouse PD 1 monoclonal antibody (mab) subtypes. These subtypes are crafted to meet different research needs and include Rat IgG2a Kappa, Rabbit IgG, and several Mouse IgG variants. Each of these antibody subtypes has distinct biochemical and functional characteristics, making them well – suited for various applications in immunology research.
References about Anti-mouse PD-1 (CD279, RMP1-14.1) MAb,please click: anti-mouse PD-1 monoclonal antibody (clone RMP1-14) referenced literature.
References for Anti-mouse PD-1 Antibody (RMP1-14):
1、Myeloid Antigen-Presenting Cell Niches Sustain Antitumor T Cells and License PD-1 Blockade via CD28 Costimulation
Jaikumar Duraiswamy,et al.Cancer Cell. 2022.PMCID: PMC8861565
“The mechanisms regulating exhaustion of tumor-infiltrating lymphocytes (TIL) and responsiveness to PD-1 blockade remain partly unknown. In human ovarian cancer we show that tumor-specific CD8+ TIL accumulate in tumor islets, where they engage antigen and upregulate anti-mouse PD-1 antibody, which restrains their functions. Intraepithelial PD-1+CD8+ TIL can be however polyfunctional. cd279 antibody PD-1+ TIL indeed exhibit a continuum of exhaustion states, with variable levels of CD28 costimulation, which is provided by antigen-presenting cells (APC) in intraepithelial tumor myeloid niches. CD28 costimulation is associated with improved effector fitness of exhausted CD8+ TIL and is required for their activation upon PD-1 blockade, which also requires tumor myeloid APCs. Exhausted TIL lacking proper CD28 costimulation in situ fail to respond to PD-1 blockade, and their response may be rescued by local CTLA-4 blockade and tumor APC stimulation via CD40L.”
2、Immune tolerance against infused FVIII in hemophilia A is mediated by PD-L1+ Tregs
Janine Becker-Gotot,et al.J Clin Invest. 2022.PMCID: PMC9663153
“A major complication of hemophilia A therapy is the development of alloantibodies (inhibitors) that neutralize intravenously administered coagulation factor VIII (FVIII). Immune tolerance induction therapy (ITI) by repetitive FVIII injection can eradicate inhibitors, and thereby reduce morbidity and treatment costs. However, ITI success is difficult to predict and the underlying immunological mechanisms are unknown. Here, we demonstrated that immune tolerance against FVIII under nonhemophilic conditions was maintained by programmed death (PD cd279 antibody) ligand 1–expressing (PD-L1–expressing) regulatory T cells (Tregs) that ligated mouse PD-1 antibody on FVIII-specific B cells, causing them to undergo apoptosis. FVIII-deficient mice injected with FVIII lacked such Tregs and developed inhibitors. Using an ITI mouse model, we found that repetitive FVIII injection induced FVIII-specific PD-L1+ Tregs and reengaged removal of inhibitor-forming B cells. We also demonstrated the existence of FVIII-specific Tregs in humans and showed that such Tregs upregulated PD-L1 in patients with hemophilia after successful ITI. Simultaneously, FVIII-specific B cells upregulated PD-1 rmp1 14 antibody and became killable by Tregs. In summary, we showed that PD-1–mediated B cell tolerance against FVIII operated in healthy individuals and in patients with hemophilia A without inhibitors, and that ITI reengaged this mechanism. These findings may impact monitoring of ITI success and treatment of patients with hemophilia A.”
3、Immune receptor inhibition through enforced phosphatase recruitment
Ricardo A Fernandes,et al.Nature. 2021.PMCID: PMC7875542
“Antibodies that antagonize extracellular receptor–ligand interactions are used as therapeutic agents for many diseases to inhibit signalling by cell-surface receptors1. However, this approach does not directly prevent intracellular signalling, such as through tonic or sustained signalling after ligand engagement. Here we present an alternative approach for attenuating cell-surface receptor signalling, termed receptor inhibition by phosphatase recruitment (RIPR). This approach compels cis-ligation of cell-surface receptors containing ITAM, ITIM or ITSM tyrosine phosphorylation motifs to the promiscuous cell-surface phosphatase CD452,3, which results in the direct intracellular dephosphorylation of tyrosine residues on the receptor target. As an example, we found that tonic signalling by the programmed cell death-1 receptor (PD-1) results in residual suppression of T cell activation, but is not inhibited by ligand-antagonist antibodies. We engineered a PD-1 molecule, which we denote RIPR-PD1, that induces cross-linking of PD-1 rmp1 14 antibody to CD45 and inhibits both tonic and ligand-activated signalling. RIPR-PD1 demonstrated enhanced inhibition of checkpoint blockade compared with ligand blocking by anti-PD1 antibodies, and increased therapeutic efficacy over anti-PD1 in mouse tumour models. We also show that the RIPR strategy extends to other immune-receptor targets that contain activating or inhibitory ITIM, ITSM or ITAM motifs; for example, inhibition of the macrophage SIRPα ‘don’t eat me’ signal with a SIRPα–CD45 RIPR molecule potentiates antibody-dependent cellular phagocytosis beyond that of SIRPα blockade alone. RIPR represents a general strategy for direct attenuation of signalling by kinase-activated cell-surface receptors.”
4、Distinct antibody clones detect PD-1 checkpoint expression and block PD-L1 interactions on live murine melanoma cells
Christina Martins,et al.Sci Rep. 2022.PMCID: PMC9304406
“Monoclonal antibodies (abs) targeting the programmed cell death 1 (PD-1) immune checkpoint pathway have revolutionized tumor therapy. Because T-cell-directed PD-1 blockade boosts tumor immunity, anti-PD-1 abs have been developed for examining T-cell-PD-1 functions. More recently, PD-1 expression has also been reported directly on cancer cells of various etiology, including in melanoma. Nevertheless, there is a paucity of studies validating anti-PD-1 ab clone utility in specific assay types for characterizing tumor cell-intrinsic PD-1. Here, we demonstrate reactivity of several anti-murine PD-1 ab clones and recombinant PD-L1 with live B16-F10 melanoma cells and YUMM lines using multiple independent methodologies, positive and negative PD-1-specific controls, including PD-1-overexpressing and PD-1 knockout cells. Flow cytometric analyses with two separate anti-PD-1 ab clones, 29F.1A12 and RMP1-30, revealed PD-1 surface protein expression on live murine melanoma cells, which was corroborated by marked enrichment in PD-1 gene (Pdcd1) expression. Immunoblotting, immunoprecipitation, and mass spectrometric sequencing confirmed PD-1 protein expression by B16-F10 cells. Recombinant PD-L1 also recognized melanoma cell-expressed PD-1, the blockade of which by 29F.1A12 fully abrogated PD-1:PD-L1 binding. Together, our data provides multiple lines of evidence establishing PD-1 expression by live murine melanoma cells and validates ab clones and assay systems for tumor cell-directed PD-1 pathway investigations.”
5、Targeting HIF-1α abrogates PD-L1–mediated immune evasion in tumor microenvironment but promotes tolerance in normal tissues
Christopher M Bailey,et al.J Clin Invest. 2022.PMCID: PMC9057613
“A combination of anti–CTLA-4 plus anti–PD-1/PD-L1 is the most effective cancer immunotherapy but causes high incidence of immune-related adverse events (irAEs). Here we report that targeting of HIF-1α suppressed PD-L1 expression on tumor cells and tumor-infiltrating myeloid cells, but unexpectedly induced PD-L1 in normal tissues by an IFN-γ–dependent mechanism. Targeting the HIF-1α/PD-L1 axis in tumor cells reactivated tumor-infiltrating lymphocytes and caused tumor rejection. The HIF-1α inhibitor echinomycin potentiated the cancer immunotherapeutic effects of anti–CTLA-4 therapy, with efficacy comparable to that of anti–CTLA-4 plus anti–PD-1 antibodies. However, while anti–PD-1 exacerbated irAEs triggered by ipilimumab, echinomycin protected mice against irAEs by increasing PD-L1 levels in normal tissues. Our data suggest that targeting HIF-1α fortifies the immune tolerance function of the PD-1/PD-L1 checkpoint in normal tissues but abrogates its immune evasion function in the tumor microenvironment to achieve safer and more effective immunotherapy.”
6、Diphtheria toxin‐derived, anti‐PD‐1 immunotoxin, a potent and practical tool to selectively deplete PD‐1+ cells
Tianxiao Zhang,et al.Protein Sci. 2023.PMCID: PMC10443333
“Programmed death‐1 (PD‐1), an immune checkpoint receptor, is expressed on activated lymphocytes, macrophages, and some types of tumor cells. While PD‐1+ cells have been implicated in outcomes of cancer immunity, autoimmunity, and chronic infections, the exact roles of these cells in various physiological and pathological processes remain elusive. Molecules that target and deplete PD‐1+ cells would be instrumental in defining the roles unambiguously. Previously, an immunotoxin has been generated for the depletion of PD‐1+ cells though its usage is impeded by its low production yield. Thus, a more practical molecular tool is desired to deplete PD‐1+ cells and cd279 antibody to examine functions of these cells. We designed and generated a novel anti‐PD1 diphtheria immunotoxin, termed PD‐1 rmp1 14 antibody DIT, targeting PD‐1+ cells. PD‐1 DIT is comprised of two single chain variable fragments (scFv) derived from an anti‐PD‐1 antibody, coupled with the catalytic and translocation domains of the diphtheria toxin. PD‐1 DIT was produced using a yeast expression system that has been engineered to efficiently produce protein toxins. The yield of PD‐1 DIT reached 1–2 mg/L culture, which is 10 times higher than the previously reported immunotoxin. Flow cytometry and confocal microscopy analyses confirmed that PD‐1 DIT specifically binds to and enters PD‐1+ cells. The binding avidities between PD‐1 DIT and two PD‐1+ cell lines are approximately 25 nM. Moreover, PD‐1 DIT demonstrated potent cytotoxicity toward PD‐1+ cells, with a half maximal effective concentration (EC50) value of 1 nM. In vivo experiments further showed that PD‐1 DIT effectively depleted PD‐1+ cells and enabled mice inoculated with PD‐1+ tumor cells to survive throughout the study. Our findings using PD‐1 DIT revealed the critical role of pancreatic PD‐1+ T cells in the development of type‐1 diabetes (T1D). Additionally, we observed that PD‐1 DIT treatment ameliorated relapsing–remitting experimental autoimmune encephalomyelitis (RR‐EAE), a mouse model of relapsing–remitting multiple sclerosis (RR‐MS). Lastly, we did not observe significant hepatotoxicity in mice treated with PD‐1 DIT, which had been reported for other immunotoxins derived from the diphtheria toxin. With its remarkable selective and potent cytotoxicity toward PD‐1+ cells, coupled with its high production yield, PD‐1 DIT emerges as a powerful biotechnological tool for elucidating the physiological roles of PD‐1+ cells. Furthermore, the potential of PD‐1 DIT to be developed into a novel therapeutic agent becomes evident.”
7、Decoupling FcRn and tumor contributions to elevated immune checkpoint inhibitor clearance in cancer cachexia
Trang T Vu,et al.Pharmacol Res. 2024.PMCID: PMC10798214
“High baseline clearance of immune checkpoint inhibitors (ICIs), independent of dose or systemic exposure, is associated with cachexia and poor outcomes in cancer patients. Mechanisms linking ICI clearance, cachexia and ICI therapy failure are unknown. Here, we evaluate in four murine models and across multiple antibodies whether altered baseline catabolic clearance of administered antibody requires a tumor and/or cachexia and whether medical reversal of cachexia phenotype can alleviate altered clearance. Key findings include mild cachexia phenotype and lack of elevated pembrolizumab clearance in the MC38 tumor-bearing model. We also observed severe cachexia and decreased, instead of increased, baseline pembrolizumab clearance in the tumor-free cisplatin-induced cachexia model. Liver Fcgrt expression correlated with altered baseline catabolic clearance, though elevated clearance was still observed with antibodies having no (human IgA) or reduced (human H310Q IgG1) FcRn binding. We conclude cachexia phenotype coincides with altered antibody clearance, though tumor presence is neither sufficient nor necessary for altered clearance in immunocompetent mice. Magnitude and direction of clearance alteration correlated with hepatic Fcgrt, suggesting changes in FcRn expression and/or recycling function may be partially responsible, though factors beyond FcRn also contribute to altered clearance in cachexia.”
8、Lymph node and tumor-associated PD-L1+ macrophages antagonize dendritic cell vaccines by suppressing CD8+ T cells
Jenny Sprooten,et al.Cell Rep Med. 2024.PMCID: PMC10829875
“Current immunotherapies provide limited benefits against T cell-depleted tumors, calling for therapeutic innovation. Using multi-omics integration of cancer patient data, we predict a type I interferon (IFN) responseHIGH state of dendritic cell (DC) vaccines, with efficacious clinical impact. However, preclinical DC vaccines recapitulating this state by combining immunogenic cancer cell death with induction of type I IFN responses fail to regress mouse tumors lacking T cell infiltrates. Here, in lymph nodes (LNs), instead of activating CD4+/CD8+ T cells, cd279 antibody, DCs stimulate immunosuppressive programmed death-ligand 1-positive (PD-L1+) LN-associated macrophages (LAMs). Moreover, DC vaccines also stimulate PD-L1+ tumor-associated macrophages (TAMs). This creates two anatomically distinct niches of PD-L1+ macrophages that suppress CD8+ T cells. Accordingly, a combination of PD-L1 blockade with DC vaccines achieves significant tumor regression by depleting PD-L1+ macrophages, suppressing myeloid inflammation, and de-inhibiting effector/stem-like memory T cells. Importantly, clinical DC vaccines also potentiate T cell-suppressive PD-L1+ TAMs in glioblastoma patients. We propose that a multimodal immunotherapy and vaccination regimen is mandatory to overcome T cell-depleted tumors.”
9、Dual targeting of RANKL and PD‐1 with a bispecific antibody improves anti‐tumor immunity
William C Dougall,et al.Clin Transl Immunology. 2019.PMCID: PMC6763724
“Objectives
The addition of RANKL/RANK blockade to immune checkpoint inhibitors (ICIs) such as anti‐PD‐1/PD‐L1 and anti‐CTLA4 antibodies is associated with increased anti‐tumor immunity in mice. Recent retrospective clinical studies in patients with advanced melanoma and lung cancer suggest the addition of anti‐RANKL antibody to ICI increases the overall response rate relative to ICI treatment alone. Based on this rationale, we developed a novel bispecific antibody (BsAb) co‐targeting RANKL and PD‐1.
Methods
We characterized target binding and functional activity of the anti‐RANKL/PD‐1 BsAb in cell‐based assays. Anti‐tumor activity was confirmed in experimental lung metastasis models and in mice with established subcutaneously transplanted tumors.”
10、Type I MET inhibitors cooperate with PD-1 blockade to promote rejection of hepatocellular carcinoma
Ricardo DeAzevedo,et al.J Immunother Cancer. 2024.PMCID: PMC11529525
“Blockade of the immune checkpoints programmed death-1 (PD-1) and cytotoxic lymphocyte antigen 4 has improved outcomes for patients with hepatocellular carcinoma (HCC), yet most still fail to achieve objective clinical benefit. MET plays key roles in both HCC tumorigenesis and immunosuppressive conditioning; however, inhibition of MET causes upregulation of PD-ligand 1 (PD-L1) suggesting the use of these inhibitors in the context of PD-1 blockade. We sought to investigate across the Hepa1-6, HCA-1 and diethylnitrosamine (DEN) models of HCC whether the combination of more specific type I versus more pleiotropic type II MET inhibitors would confer superior outcomes in combination with PD-1 blockade. While MET inhibition demonstrated cooperativity with αPD-1 across all three models, the type I MET inhibitor capmatinib showed optimal activity in combination and statistically significantly outperformed the combination with the type II inhibitor cabozantinib in the αPD-1 refractory DEN model. In both HCA-1 and DEN HCC, the capmatinib and αPD-1 combination enhanced CD8 T cell frequency and activation state while limiting intratumoral myeloid immune suppression. In vitro studies of antigen-specific T cell activation reveal significantly less inhibition of effector cytokine production and proliferation by capmatinib versus by type II or type III MET inhibitors. These findings suggest significant potential for clinical HCC combination studies of type I MET inhibitors and PD-1 blockade where prior trials using type II inhibitors have yielded limited benefit.”
11、Synergistic anticancer activity of a novel oral chemotherapeutic agent containing trifluridine and tipiracil in combination with anti-PD-1 blockade in microsatellite stable-type murine colorectal cancer cells
Norihiko Suzuki,et al.Am J Cancer Res. 2017.PMCID: PMC5665850
“Trifluridine/tipiracil (FTD/TPI) is a combination of FTD, an antineoplastic thymidine-based nucleoside analog, and TPI, which acts to enhance the bioavailability of FTD in vivo. It is used to treat patients with unresectable advanced or recurrent colorectal cancer that is refractory to standard therapies. We investigated the anticancer activity of FTD/TPI combined with anti-mouse programed cell death 1 (PD-1, cd279 antibody) monoclonal antibody (mAb) against CMT-93 cells, which are microsatellite stable (MSS)-type murine colorectal cancer cells. Tumor growth inhibition (TGI) after treatment with anti-mouse PD-1 mAb monotherapy (0.1 mg, i.p., days 1, 5, 9) and FTD/TPI monotherapy (150 mg/kg/day, p.o., days 1-14) were 86.7% and 52.7%, respectively, and that of the combination was 98.4%. The TGI of the combination therapy was significantly greater than that of each monotherapy (P<0.05). The combination therapy caused complete tumor regression in four out of five mice without body-weight reduction, but neither of the monotherapies resulted in complete tumor regression. Low dose FTD/TPI (75 and 100 mg/kg) combined with anti-mouse PD-1 mAb also showed significant antitumor activity against CMT-93 tumors. Flow cytometric analysis revealed that a higher CD8+ T cell ratio among total lymphocytes and a lower regulatory T cells (Tregs) ratio in CD4+ T cells in the combination group compared with that in the control group. These results suggested that the combination therapy induced a cytotoxic response from infiltrated cytotoxic CD8+ T cells and reduced immunosuppressive activity as indicated by decreased Tregs. In this study, the combination therapy was found to have synergistically greater antitumor activity against CMT-93 cells. These preclinical findings indicated that FTD/TPI and anti-mouse PD-1 mAb combination therapy may be a promising treatment option, even for MSS-type colorectal cancer.”
12、Antibody-mediated depletion of programmed death 1-positive (PD-1+) cells
Yujia Zhai,et al.J Control Release. 2023.PMCID: PMC10699550
“PD-1 immune checkpoint has been intensively investigated in pathogenesis and treatments for cancer and autoimmune diseases. Cells that express PD-1 (PD-1+ cells) draw ever-increasing attention in cancer and autoimmune disease research although the role of PD-1+ cells in the progression and treatments of these diseases remains largely ambiguous. One definite approach to elucidate their roles is to deplete these cells in disease settings and examine how the depletion impacts disease progression and treatments. To execute the depletion, we designed and generated the first depleting antibody (D-αPD-1) that specifically ablates PD-1+ cells. D-αPD-1 has the same variable domains as an anti-mouse PD-1 blocking antibody (RMP1–14). The constant domains of D-αPD-1 were derived from mouse IgG2a heavy and κ-light chain, respectively. D-αPD-1 was verified to bind with mouse PD-1 rmp1 14 antibody as well as mouse FcγRIV, an immuno-activating Fc receptor. The cell depletion effect of D-αPD-1 was confirmed in vivo using a PD-1+ cell transferring model. Since transferred PD-1+ cells, EL4 cells, are tumorigenic and EL4 tumors are lethal to host mice, the depleting effect of D-αPD-1 was also manifested by an absolute survival among the antibody-treated mice while groups receiving control treatments had median survival time of merely approximately 30 days. Furthermore, we found that D-αPD-1 leads to elimination of PD-1+ cells through antibody-dependent cell-mediate phagocytosis (ADCP) and complement-dependent cytotoxicity (CDC) mechanisms. These results, altogether, confirmed the specificity and effectiveness of D-αPD-1. The results also highlighted that D-αPD-1 is a robust tool to study PD-1+ cells in cancer and autoimmune diseases and a potential therapeutic for these diseases.”
13、Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo
Sarwish Rafiq,et al.Nat Biotechnol. 2019.PMCID: PMC6126939
“The efficacy of CAR-T cell therapy against poorly responding tumors has been enhanced by administering the cells in combination with immune checkpoint blockade inhibitors. Alternatively, the CAR construct has been engineered to co-express factors that boost CAR-T cell function in the tumor microenvironment. Here we modified CAR-T cells to secrete PD-1-blocking single-chain variable fragments (scFv). These scFv-secreting CAR-T cells work in both a paracrine and autocrine manner to improve the anti-tumor activity of CAR-T cells and bystander tumor-specific T cells in clinically relevant syngeneic and xenogeneic mouse models of PD-L1+ hematologic and solid tumors. Efficacy was similar or better to that achieved by combination therapy with CAR-T cells and a checkpoint inhibitor. This approach could improve safety as the secreted scFv remained localized to the tumor, protecting CAR-T cells from PD-1 inhibition, which could potentially avoid toxicities associated with systemic checkpoint inhibition.”
14、Targeting PD-L2/RGMb overcomes microbiome-related immunotherapy resistance
Joon Seok Park,et al.Nature. 2023.PMCID: PMC10219577
“The gut microbiota has been shown to be a critical regulator of anti-tumor immunity during immune checkpoint inhibitor therapies. Several bacteria that promote an anti-tumor response to immune checkpoint inhibitors have been identified in mice(1-6), and transplantation of fecal specimens from responders has been shown to improve the efficacy of anti-PD-1 therapy in melanoma patients(7, 8). However, the increased efficacy from fecal transplants is variable and how gut bacteria promote anti-tumor immunity remains unclear. Here, we show that the gut microbiome downregulates expression of PD-L2 and its binding partner Repulsive Guidance Molecule b (RGMb) to promote anti-tumor immunity and identify bacterial species that mediate this effect. PD-L1 and PD-L2 share PD-1 as a binding partner, but PD-L2 can also bind RGMb. We demonstrate that blockade of PD-L2/RGMb interactions can overcome microbiome-dependent resistance to PD-1(cd279 antibody) pathway inhibitors: antibody-mediated blockade of the PD-L2/RGMb pathway or conditional deletion of RGMb in T cells combined with anti-PD-1 or anti-PD-L1 promotes anti-tumor responses in multiple mouse tumor models that do not respond to anti-PD-1 rmp1 14 antibody or anti-PD-L1 alone (Germ Free mice, antibiotic treated mice, and even mice colonized with non-responder patient stool). These studies identify downregulation of the PD-L2/RGMB pathway as a specific mechanism by which the gut microbiota can promote responses to PD-1 checkpoint blockade and define a potentially effective immunological strategy for treating patients who do not respond to PD-1 cancer immunotherapy.”
15、Sources of inter-individual variability leading to significant changes in anti-PD-1 and anti-PD-L1 efficacy identified in mouse tumor models using a QSP framework
Jessica C Leete,et al.Front Pharmacol. 2022.PMCID: PMC9760747
“While anti-PD-1 and anti-PD-L1 [anti-PD-(L)1] monotherapies are effective treatments for many types of cancer, high variability in patient responses is observed in clinical trials. Understanding the sources of response variability can help prospectively identify potential responsive patient populations. Preclinical data may offer insights to this point and, in combination with modeling, may be predictive of sources of variability and their impact on efficacy. Herein, a quantitative systems pharmacology (QSP) model of anti-PD-(L)1 was developed to account for the known pharmacokinetic properties of anti-PD-(L)1 antibodies, their impact on CD8+ T cell activation and influx into the tumor microenvironment, and subsequent anti-tumor effects in CT26 tumor syngeneic mouse model. The QSP model was sufficient to describe the variability inherent in the anti-tumor responses post anti-PD-(L)1 treatments. Local sensitivity analysis identified tumor cell proliferation rate, PD-1 expression on CD8+ T cells, PD-L1 expression on tumor cells, and the binding affinity of PD-1 rmp1 14 antibody:PD-L1 as strong influencers of tumor growth. It also suggested that treatment-mediated tumor growth inhibition is sensitive to T cell properties including the CD8+ T cell proliferation half-life, CD8+ T cell half-life, cytotoxic T-lymphocyte (CTL)-mediated tumor cell killing rate, and maximum rate of CD8+ T cell influx into the tumor microenvironment. Each of these parameters alone could not predict anti-PD-(L)1 treatment response but they could shift an individual mouse’s treatment response when perturbed. The presented preclinical QSP modeling framework provides a path to incorporate potential sources of response variability in human translation modeling of anti-PD-(L)1.”
16、Angiogenic inhibitor pre‐administration improves the therapeutic effects of immunotherapy
Mineyoshi Sato,et al.Cancer Med. 2023.PMCID: PMC10166916
“In lung cancer, immune checkpoint inhibitors (ICIs) are often inadequate for tumor growth inhibition. Angiogenic inhibitors (AIs) are required to normalize tumor vasculature for improved immune cell infiltration. However, in clinical practice, ICIs and cytotoxic antineoplastic agents are simultaneously administered with an AI when tumor vessels are abnormal. Therefore, we examined the effects of pre‐administering an AI for lung cancer immunotherapy in a mouse lung cancer model. Using DC101, an anti‐vascular endothelial growth factor receptor 2 (VEGFR2) monoclonal antibody, a murine subcutaneous Lewis lung cancer (LLC) model was used to determine the timing of vascular normalization. Microvessel density (MVD), pericyte coverage, tissue hypoxia, and CD8‐positive cell infiltration were analyzed. The effects of an ICI and paclitaxel after DC101 pre‐administration were investigated. On Day 3, increased pericyte coverage and alleviated tumor hypoxia represented the highest vascular normalization. CD8+ T‐cell infiltration was also highest on Day 3. When combined with an ICI, DC101 pre‐administration significantly reduced PD‐L1 expression. When combined with an ICI and paclitaxel, only DC101 pre‐administration significantly inhibited tumor growth, but simultaneous administration did not. AI pre‐administration, and not simultaneous administration, may increase the therapeutic effects of ICIs due to improved immune cell infiltration.”
17、Prophylactic IL-23 blockade uncouples efficacy and toxicity in dual CTLA-4 and PD-1 immunotherapy
Mingyi Ju,et al.J Immunother Cancer. 2024.PMCID: PMC11293404
“Background
Immune-related adverse events (irAEs), characterized by targeted inflammation, occur in up to 60% of patients with melanoma treated with immune checkpoint inhibitors (ICIs). Evidence proved that the baseline peripheral blood profiles of patients at risk for severe irAEs development paralleled clinical autoimmunity. Interleukin (IL)-23 blockade with risankizumab is recommended for cases that are suffering from autoimmune disease, such as autoimmune colitis. However, currently, the role of IL-23 in irAEs onset and severity remains poorly understood.
Methods
The pro-inflammatory cytokines most associated with severe irAEs onset were identified by retrospective analysis based on GSE186143 data set. To investigate the efficacy of prophylactic IL-23 blockade administration to prevent irAEs, refer to a previous study, we constructed two irAEs murine models, including dextran sulfate sodium salt (DSS)-induced colitis murine model and a combined-ICIs-induced irAEs murine model. To further explore the applicability of our findings, murine models with graft-versus-host disease were established, in which Rag2−/−Il2rg−/− mice were transferred with human peripheral blood mononuclear cells and received combined cytotoxic T-lymphocyte associated antigen 4 (CTLA-4) and programmed cell death protein-1 (PD-1) treatment. Human melanoma cells were xenografted into these mice concomitantly.”
18、Macrophages Impair TLR9 Agonist Antitumor Activity through Interacting with the Anti-PD-1 Antibody Fc Domain
Simone Camelliti,et al.Cancers (Basel). 2021.PMCID: PMC8391891
“Background. A combination of TLR9 agonists and an anti-PD-1 antibody has been reported to be effective in immunocompetent mice but the role of innate immunity has not yet been completely elucidated. Therefore, we investigated the contribution of the innate immune system to this combinatorial immunotherapeutic regimens using an immunodeficient mouse model in which the effector functions of innate immunity can clearly emerge without any interference from T lymphocytes. Methods. Athymic mice xenografted with IGROV-1 human ovarian cells, reported to be sensitive to TLR9 agonist therapy, were treated with cytosine–guanine (CpG)-oligodeoxynucleotides (ODNs), an anti-PD-1 antibody or their combination. Results. We found that PD-1 blockade dampened CpG-ODN antitumor activity. In vitro studies indicated that the interaction between the anti-PD-1 antibody fragment crystallizable (Fc) domain and macrophage Fc receptors caused these immune cells to acquire an immunoregulatory phenotype, contributing to a decrease in the efficacy of CpG-ODNs. Accordingly, in vivo macrophage depletion abrogated the detrimental effect exerted by the anti-PD-1 antibody. Conclusion. Our data suggest that if TLR signaling is active in macrophages, coadministration of an anti-PD-1 antibody can reprogram these immune cells towards a polarization state able to negatively affect the immune response and eventually promote tumor growth.”
19、PD-L1 signaling selectively regulates T cell lymphatic transendothelial migration
Wenji Piao,et al.Nat Commun. 2022.PMCID: PMC9023578
“Programmed death-1 (PD-1) and its ligand PD-L1 are checkpoint molecules which regulate immune responses. Little is known about their functions in T cell migration and there are contradictory data about their roles in regulatory T cell (Treg) function. Here we show activated Tregs and CD4 effector T cells (Teffs) use PD-1 rmp1 14 antibody/PD-L1 and CD80/PD-L1, respectively, to regulate transendothelial migration across lymphatic endothelial cells (LECs). Antibody blockade of Treg PD-1, Teff CD80 (the alternative ligand for PD-L1), or LEC PD-L1 impairs Treg or Teff migration in vitro and in vivo. PD-1/PD-L1 signals through PI3K/Akt and ERK to regulate zipper junctional VE-cadherin, and through NFκB-p65 to up-regulate VCAM-1 expression on LECs. CD80/PD-L1 signaling up-regulates VCAM-1 through ERK and NFκB-p65. PD-1 and CD80 blockade reduces tumor egress of PD-1high fragile Tregs and Teffs into draining lymph nodes, respectively, and promotes tumor regression. These data provide roles for PD-L1 in cell migration and immune regulation.”
20、PD-L1/PD-1 checkpoint pathway regulates hippocampal neuronal excitability and learning and memory behavior
Junli Zhao,et al.Neuron. 2024.PMCID: PMC10529885
“Programmed death protein 1 (PD-1) and its ligand PD-L1 constitute an immune checkpoint pathway. We report that neuronal PD-1 signaling regulates learning/memory in health and disease. Mice lacking PD-1 (encoded by Pdcd1) exhibit enhanced long-term potentiation (LTP) and memory. Intraventricular administration of anti-mouse PD-1 monoclonal antibody (RMP1-14, cd279 antibody) potentiated learning and memory. Selective deletion of PD-1 rmp1 14 antibody in excitatory neurons (but not microglia) also enhanced LTP and memory. Traumatic brain injury (TBI) impairs learning and memory, which is rescued by Pdcd1 deletion or intraventricular PD-1 blockade. Conversely, re-expression of Pdcd1 in PD-1 deficient hippocampal neurons suppresses memory and LTP. Exogenous PD-L1 suppressed learning/memory in mice and the excitability of mouse and NHP hippocampal neurons through PD-1. Notably, neuronal activation suppressed PD-L1 secretion, and PD-L1/PD-1 signaling is distinctly regulated by learning and TBI. Thus, conditions that reduce PD-L1 levels or PD-1 signaling could promote memory in both physiological and pathological conditions.”
Related Recombinant IgG Reference Antibodies:
Recombinant Mouse IgG1 Isotype Control Antibody and Mutants, In vivo Grade
Recombinant Mouse IgG2a Isotype Control Antibody and Mutants, In vivo Grade
Recombinant Mouse IgG2c Isotype Control Antibody and Mutants, In vivo Grade
Recombinant Rat IgG2a Isotype Control Antibody, In vivo Grade
Syd Labs provides the following anti-mouse PD-L1 / PD-1 antibodies:
Recombinant anti-mouse PD1 antibodies (Clone 29F.1A12.1), In vivo grade
Recombinant anti-mouse PD-1 antibodies (Clone RMP1-14.1), In vivo grade
Recombinant anti-mouse PD-L1 antibodies (Clone 10F.9G2.1), In vivo grade
Recombinant anti-mouse PD-1 / PD-1 bispecific antibodies (Clone RMP1-14.1 / 29F.1A12.1), In vivo grade
Recombinant anti-mouse PD-1 / PD-1 bispecific antibodies (Clone 29F.1A12.1 / RMP1-14.1), In vivo grade
Recombinant anti-mouse PD-1 / PD-L1 bispecific antibodies (Clone RMP1-14.1 / 10F.9G2.1), In vivo grade
Recombinant anti-mouse PD-L1 / PD-1 bispecific antibodies (Clone 10F.9G2.1 / RMP1-14.1), In vivo grade
Recombinant anti-mouse PD-1 / PD-L1 bispecific antibodies (Clone 29F.1A12.1 / 10F.9G2.1), In vivo grade
Recombinant anti-mouse PD-L1 / PD-1 bispecific antibodies (Clone 10F.9G2.1 / 29F.1A12.1), In vivo grade
Questions and Answers about recombinant anti-mouse PD-1 (CD279, Clone RMP1-14) antibody:
Question: Which anti-mouse PD1 antibody works the best?
Answer: (by AI) There are several anti-mouse PD1 antibodies available in the market: Clones RMP1-14, 29F.1A12, and J43. All three of these antibodies are commonly used to block PD-1 signaling in vivo in murine tumor models and other mouse models. These three clones all have extensive multi-year publication records supporting them. The RMP1-14 antibody has been reported to block the binding of PD-1 to its ligands (B7-H1 and B7-DC) and to inhibit T cell proliferation and cytokine production costimulated by macrophages (but not by dendritic cells and B cells). (by Syd Labs) Syd Labs offers anti-mouse PD-1 monoclonal antibodies based on the sequences of clones RMP1-14 and 29F.1A12. Syd Labs provides in vivo grade recombinant antibodies including engineered antibodies for the clones RMP1-14 and 29F.1A12. Even though mouse and rat are close, rat antibodies may still induce immunogenecity in mice. Antibodies with murinized variable regions and mouse constant regions behave like humanized antibody drugs in animal models using mice. In addition, the mouse IgG2c antibody is produced in certain inbred strains such as C57BL/6, C57BL/10, SJL, and NOD, which does not express the mouse IgG2a antibody; the mouse IgG2a antibody is produced in other inbred strains such as BALB/c and Swiss Webster mice, which does not express the mouse IgG2c antibody. If one uses the C57BL/6 mouse strain for animal model research, it is better to use the IgG2c antibodies rather than the IgG2a antibodies. The format with the Fc silenced, Fc silent, or Fcs with silenced effector function, such as LALAPG mutation, is the most popular for anti-mouse PD1 antibodies (clones RMP1-14 and 29F.1A12) and anti-mouse PD-L1 antibodies (clone 10F.9G2).
Question: Do you produce any recombinant Fc-silenced RMP1-14 antibody?
Answer: Sure, we provide various recombinant Fc silent RMP1-14 antibodies, such as mIgG2c LALAPG, mIgG2a LALAPG, and mIgG1 D265A. We also provide custom recombinant antibody production service to produce other engineered versions of recombinant RMP1-14 antibodies. We have a promotion program running: We provide 1 mg PA007162.m2cLA (In Vivo Grade Recombinant Anti-mouse PD-1 Mouse IgG2c-LALAPG Kappa Monoclonal Antibody (Clone RMP1-14.1)) for free in exchange of results. Please contact us to know more about the free RMP1-14 antibody.
Question: What is the difference among PA007162.r2a, PA007162.m2cLA, and PA007162.mm2cLA?
Answer: PA007162.r2a is the recombinant anti-mouse PD-1 monoclonal antibody (rat IgG2a kappa, clone RMP1-14.1) produced in CHO cells or HEK293 cells if needed. It has the same variable region and constant region sequences as the rat anti-mouse PD-1 monoclonal antibody from the hybridoma clone of RMP1-14. Rat antibodies may cause high immuogenicity in mice; thus, at least recombinant antibodies with mouse antibody constant regions should be used to replace the rat antibody constant regions. PA007162.m2cLA is the recombinant anti-mouse PD-1 antibody (clone RMP1-14.1) whose constant regions are mouse IgG2c LALAPG kappa. We further murinize the antibody variable region sequences of PA007162.m2cLA to produce PA007162.mm2cLA.
anti-mouse PD-1 Antibody (CD279, RMP1-14.1) from: Anti-Mouse PD1 Monoclonal Antibodies: PA007162.r2a Syd Labs