Recombinant Human Granulocyte-macrophage colony-stimulating factor protein (CSF2) (Active)

Shipped with Ice Packs
In Stock

Description

Definition and Nomenclature

Recombinant Human GM-CSF (CSF2) is a synthetic version of the naturally occurring cytokine produced in immune cells (e.g., T cells, macrophages, fibroblasts). It is classified as a monomeric glycoprotein with a molecular weight of ~15–22 kDa, depending on glycosylation status . The protein comprises 127–144 amino acids, with two glycosylation sites critical for in vivo activity .

Key Features:

ParameterDescription
Gene NameCSF2, GMCSF
AliasesColony-Stimulating Factor 2, Molgramostin, Sargramostim
ReceptorHeterodimeric complex (α-subunit + βc-subunit shared with IL-3/IL-5 receptors)
Species SpecificityHuman GM-CSF is inactive on murine cells and vice versa .

Structure and Production

The recombinant protein is produced via heterologous expression systems, including E. coli or HEK 293 cells, with purity exceeding 95% in some formulations . Non-glycosylated variants (e.g., E. coli-derived) retain in vitro activity but exhibit reduced in vivo efficacy compared to glycosylated forms .

Production Systems and Characteristics:

Host SystemGlycosylationPurityEndotoxin LevelBiological Activity
E. coliNo≥97%<0.1 ng/µg (1 EU/µg)ED₅₀ ≤0.1 ng/mL (TF-1 cells)
HEK 293Yes≥95%≤0.005 EU/µgED₅₀ ≤0.1 ng/mL (TF-1 cells)
Mammalian CellsYes≥85%Not specifiedED₅₀ ≤0.1 ng/mL (TF-1 cells)

Biological Activity and Mechanisms

Recombinant GM-CSF stimulates hematopoietic progenitor cells and modulates immune responses through STAT5 and STAT3 signaling pathways . Key activities include:

  • Hematopoiesis: Promotes proliferation and differentiation of granulocytes (neutrophils, eosinophils), monocytes, and macrophages .

  • Immune Activation: Enhances phagocytosis, reactive oxygen species (ROS) production, and cytokine secretion (e.g., IL-6, CCL17) .

  • Inflammatory Pathways: Drives pro-inflammatory macrophage polarization and antigen presentation .

Functional Assays:

Cell TypeEffectReference
Neutrophils↑ Survival, ↑ Phagocytosis, ↑ Superoxide Production
Eosinophils↑ Survival (enhanced by 9 hours), ↑ Cytotoxicity
Monocytes/Macrophages↑ CCL17 secretion, ↑ MHC-II expression, ↑ Antimicrobial activity

Clinical and Research Applications

Recombinant GM-CSF is used in immunotherapy, cancer research, and inflammatory disease modeling:

  • Immunotherapy: Enhances dendritic cell maturation for cancer vaccines .

  • Autoimmune/Inflammatory Diseases: Targeted by monoclonal antibodies (e.g., otilimab) to reduce GM-CSF-driven inflammation in rheumatoid arthritis or COVID-19 .

  • Cancer Research: Stimulates tumor-associated fibroblasts and osteogenic sarcoma cell lines, raising concerns about therapeutic off-target effects .

Ongoing Clinical Trials:

Target DiseaseApproachStatus
Rheumatoid ArthritisNeutralizing GM-CSF with monoclonal antibodiesPhase II/III
COVID-19Blocking GM-CSF to mitigate cytokine stormsPreclinical

Research Challenges and Future Directions

  • Species Specificity: Human GM-CSF is inactive in murine models, complicating preclinical studies .

  • Glycosylation Impact: Non-glycosylated variants lack in vivo efficacy, necessitating mammalian expression systems for clinical-grade products .

  • Dual-Edged Role: While enhancing antimicrobial responses, GM-CSF exacerbates chronic inflammation (e.g., colitis, asthma) .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered phosphate-buffered saline (PBS), pH 7.4.
Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Avoid repeated freeze-thaw cycles. Store reconstituted aliquots at 4°C for up to one week.
Reconstitution
Prior to opening, briefly centrifuge the vial to consolidate the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting to -20°C/-80°C. Our standard protocol utilizes 50% glycerol; this may serve as a guideline for customers.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and the inherent stability of the protein. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations maintain stability for 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
Colony stimulating factor 2 (granulocyte-macrophage); Colony Stimulating Factor 2; Colony stimulating factor; Colony-stimulating factor; CSF 2; CSF; CSF2; CSF2_HUMAN; GM-CSF; GMCSF; Granulocyte Macrophage Colony Stimulating Factor; Granulocyte-macrophage colony-stimulating factor; MGC131935; MGC138897; MGI1GM; Molgramostin; Pluripoietin-a; Sargramostim
Datasheet & Coa
Please contact us to get it.
Expression Region
18-144aa
Mol. Weight
14.5 kDa
Protein Length
Full Length of Mature Protein
Purity
>98% as determined by SDS-PAGE.
Research Area
Immunology
Source
E.coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a cytokine that stimulates the growth and differentiation of hematopoietic precursor cells from various lineages, including granulocytes, macrophages, eosinophils, and erythrocytes.

Gene References Into Functions

Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) Research Highlights:

  • Mycoplasma pneumoniae Infection: Elevated GM-CSF levels are associated with prolonged fever in children with M. pneumoniae infection, suggesting a role in neutrophil-mediated inflammation. (PMID: 30007865)
  • Rheumatoid Arthritis: Synovial fluid CD4 T cells show enrichment in GM-CSF production, implicating GM-CSF in disease pathogenesis. (PMID: 29142230)
  • Crohn's Disease: GM-CSF bioavailability is crucial for intestinal homeostasis. Reduced bioavailability, combined with genetic factors and/or smoking, contributes to aggressive ileal Crohn's disease. (PMID: 29434451)
  • Viral Decoy Receptor: ORF117 (GIF) acts as a competitive decoy receptor for GM-CSF and interleukin-2 (IL-2). (PMID: 27819269)
  • Tuberculosis (TB): Expansion of GM-CSF-producing T cells through vaccination may offer a prophylactic or therapeutic strategy for TB. (PMID: 29233902)
  • Cancer Immunotherapy: In some patients receiving GM-CSF, circulating myeloid-derived suppressor cells (MDSCs) and anti-GM-CSF-neutralizing antibodies were modulated. Antibody development correlated with improved relapse-free and overall survival. (PMID: 28536308)
  • Pregnancy: Cortisol inhibits CSF2 expression via DNA methylation, impacting trophoblast cell invasion. (PMID: 28846166)
  • HIV Infection: GM-CSF levels in gingival crevicular fluid (GCF) and serum may serve as biomarkers for HIV infection severity and response to antiretroviral therapy (ART). (PMID: 28043030)
  • Cancer Immunotherapy (Review): This review summarizes the anticancer immune response mediated by GM-CSF. (PMID: 27364892)
  • Renal Cell Carcinoma: Intratumoral GM-CSF expression may serve as an independent prognostic biomarker for recurrence in clear-cell renal cell carcinoma (ccRCC). (PMID: 27016418)
  • Breast Cancer: High GM-CSF expression is associated with breast cancer. (PMID: 28754674)
  • Gastric Cancer: Tumor-derived GM-CSF activates neutrophils, inducing PD-L1 expression and suppressing T-cell immunity in gastric cancer. (PMID: 28274999)
  • Fibroblast Differentiation: IL-3/GM-CSF influences the myofibroblastic differentiation of human adipose-derived stromal cells (hASCs) and human dermal fibroblasts (HDFs). (PMID: 28377320)
  • Breast Cancer Metastasis: Obesity enhances breast cancer metastasis via IL-5 and GM-CSF-mediated neutrophil infiltration in the lungs. (PMID: 28737771)
  • Recombinant GM-CSF Production: Pichia pastoris is an effective host for recombinant human GM-CSF (rhGM-CSF) production. (PMID: 28376863)
  • Skin Inflammation: Reduced filaggrin or loricrin expression increases IL-1α, IL-8, IL-18, and GM-CSF production in keratinocytes. (PMID: 26381575)
  • CARD9 Deficiency: Impaired RASGRF1/ERK-mediated GM-CSF response is a characteristic of CARD9 deficiency. (PMID: 26521038)
  • Honokiol and Rheumatoid Arthritis: Honokiol inhibits TNF-α-induced IL-1β, GM-CSF, and IL-8 production in rheumatoid arthritis patient peripheral blood mononuclear cells (PBMCs). (PMID: 26339358)
  • NF-κB Signaling and CSF2 Expression: Canonical NF-κB signaling in fibroblasts, but not tumor cells, is responsible for induced and constitutive CSF2 expression. (PMID: 25919140)
  • GM-CSF and IL-13 Production: GM-CSF primes IL-13 production by macrophages via protease-activated receptor-2 (PAR-2). (PMID: 25633855)
  • IL2RA Polymorphism and GM-CSF Production: An IL2RA polymorphism influences GM-CSF production in human T helper (Th) cells. (PMID: 25278028)
  • NFKBIZ and Bronchial Epithelial Cells: NFKBIZ gene knockdown in bronchial epithelial cells suppresses IL-1β-induced IL-6 and GM-CSF release. (PMID: 25629767)
  • CFH Polymorphism and Eye Inflammation: The CFH Y402H polymorphism is associated with elevated vitreal GM-CSF and choroidal macrophages. (PMID: 25814824)
  • Influenza Infection and Alveolar Epithelial Cells: Influenza infection stimulates IL-8 and GM-CSF secretion by alveolar epithelial cells. (PMID: 26033355)
  • PFAPA Syndrome: Febrile episodes in PFAPA syndrome (periodic fever, aphthous stomatitis, pharyngitis, and cervical adenopathy syndrome) are characterized by GM-CSF and IL-8 activation with Th1 skewing. (PMID: 24670131)
  • Chronic Lymphocytic Leukemia (CLL): STAT3 activation induces GM-CSF receptor α (GM-CSFRα) expression, protecting CLL cells from apoptosis. Targeting STAT3 or GM-CSFRα may be beneficial in CLL treatment. (PMID: 24836891)
  • Cancer and Cytokine-Mediated Immune Suppression (Review): G-CSF- or GM-CSF-secreting cancers are rapidly advancing due to cytokine-mediated immune suppression and angiogenesis. (PMID: 24692240)
  • Epithelial-Mesenchymal Transition (EMT): GM-CSF induces EMT in human HERS/ERM cells. (PMID: 24258001)
  • Atopic Dermatitis: GM-CSF polymorphisms may be involved in atopic dermatitis pathogenesis. (PMID: 24117406)
  • Juvenile Idiopathic Arthritis: GM-CSF expression is associated with systemic inflammation and Th17-related cytokines in juvenile idiopathic arthritis. (PMID: 24692225)
  • Macrophage Transformation: M-CSF induces macrophage transformation by upregulating c-Jun, synergistically with NF-κB. (PMID: 24100343)
  • Rheumatoid Arthritis and Rituximab Therapy: Basal levels of IgM rheumatoid factor and certain cytokines, including GM-CSF, are useful in predicting rituximab therapy effectiveness in rheumatoid arthritis. (PMID: 25080789)
  • Sepsis: Cytokines, including GM-CSF, predict clinical outcomes in sepsis. (PMID: 24244449)
  • G-CSF and Gene Expression: G-CSF induces long-term changes in gene and microRNA expression profiles in CD34+ cells. (PMID: 24056818)
  • Breast Cancer and Macrophages: Mesenchymal-like breast cancer cells activate macrophages to a tumor-associated macrophage-like phenotype via GM-CSF. (PMID: 24823638)
  • Essential Thrombocythemia (ET): ET patients show significantly higher levels of IL-4, IL-8, GM-CSF, interferon-γ, MCP-1, PDGF-BB, and VEGF compared to polycythemia vera (PV) patients. (PMID: 24463275)
  • Colorectal Cancer: Differentially expressed IL-17, IL-22, IL-23 levels are associated with K-ras in colorectal cancer, with an association between mutant K-ras and GM-CSF/IFN-γ. (PMID: 24040001)
  • Dengue Virus Infection and Type 2 Diabetes: Type 2 diabetes patients show higher IL-4, IL-10, and GM-CSF levels after dengue virus infection. (PMID: 24078930)
  • BMPR2 Loss and Aortic Aneurysms: BMPR2 loss leads to prolonged p38 MAPK phosphorylation, increased GADD34-PP1 phosphatase activity, and derepressed GM-CSF mRNA translation, contributing to aortic aneurysms. (PMID: 24446489)
  • Human Papillomavirus (HPV) and Pregnancy: Reduced GM-CSF levels in cervico-vaginal fluid of HPV-positive women may contribute to reduced live birth rates. (PMID: 23920355)
  • SECTM1 and Leukemic Cells: SECTM1 secreted from bone marrow stromal cells may interact with CD7 to influence GM-CSF expression in leukemic cells. (PMID: 24211252)
  • Autoimmune Pulmonary Alveolar Proteinosis (PAP): GM-CSF ELISA is used for routine clinical diagnosis of autoimmune PAP. (PMID: 24275678)
  • Acne Pathogenesis: The -677C/A polymorphism in the GM-CSF gene promoter does not appear to be involved in acne pathogenesis. (PMID: 23069312)
  • CBL Mutants and GM-CSF Signaling: CBL linker and RING finger mutants enhance GM-CSF signaling via elevated kinase expression, potentially inhibitable by small molecule inhibitors. (PMID: 23696637)
  • NFATC2-Calcineurin Interaction and Megakaryocyte Proliferation: Impaired NFATC2-calcineurin interaction promotes megakaryocyte proliferation through upregulation of CSF2 transcription. (PMID: 22911897)
  • MT1-MMP and CSF-2/CSF-3 Transcription: Overexpression of a nonphosphorylatable MT1-MMP mutant abrogates CSF-2 and CSF-3 transcriptional increases. (PMID: 23548906)
  • SMAD3 Deficiency and Aortic Aneurysms: GM-CSF contributes to aortic aneurysms arising from SMAD3 deficiency. (PMID: 23585475)
  • Pediatric Crohn's Disease and GM-CSF Autoantibodies: In pediatric Crohn's disease, neutrophil GM-CSF-dependent STAT5 activation and bacterial killing are reduced as GM-CSF autoantibodies increase. (PMID: 23600834)
  • Alzheimer's Disease: GM-CSF is downregulated in the brain of Alzheimer's disease patients. (PMID: 22430742)
  • IL-27 and GM-CSF Production: IL-27 suppresses GM-CSF production under non-polarizing and Th1, but not Th17, conditions. (PMID: 22837488)
Database Links

HGNC: 2434

OMIM: 138960

KEGG: hsa:1437

STRING: 9606.ENSP00000296871

UniGene: Hs.1349

Protein Families
GM-CSF family
Subcellular Location
Secreted.

Q&A

What are the primary biological functions of GM-CSF in the human immune system?

GM-CSF functions as a powerful stimulator of mature human eosinophils and neutrophils, exhibiting multiple immunomodulatory effects. It enhances the cytotoxic activity of neutrophils and eosinophils against antibody-coated targets and stimulates phagocytosis of serum-opsonized yeast by both cell types in a dose-dependent manner. Additionally, GM-CSF promotes neutrophil-mediated iodination in the presence of zymosan and enhances N-formylmethionylleucylphenylalanine (FMLP)-stimulated degranulation of Cytochalasin B pretreated neutrophils and FMLP-stimulated superoxide production. The protein induces morphological changes and significantly enhances the survival of both neutrophils and eosinophils (by 6 and 9 hours, respectively) .

Beyond these functions, GM-CSF plays a vital role in various immune system processes, including responses to inflammation and infection, as well as in hematopoiesis. It's important to note that despite its name suggesting specificity to granulocyte and macrophage development, GM-CSF demonstrates diverse biological effects extending beyond these cell lineages .

How does the receptor-binding mechanism of GM-CSF influence its biological activity?

The biological effects of GM-CSF are mediated through binding to specific receptors expressed on target cell surfaces. The GM-CSF receptor is expressed on multiple cell types, including granulocyte, erythrocyte, megakaryocyte, and macrophage progenitor cells, as well as mature neutrophils, monocytes, macrophages, dendritic cells, plasma cells, certain T lymphocytes, vascular endothelial cells, uterine cells, and myeloid leukemia cells .

Molecular studies have revealed that the GM-CSF receptor consists of two distinct subunits: α and common β (β). The α subunit provides specificity for GM-CSF binding, while the β subunit is shared with receptors for IL-3 and IL-5, explaining some of the overlapping functions of these cytokines. When GM-CSF binds to its receptor, it triggers intracellular signaling cascades that ultimately lead to the activation of various cellular functions, including proliferation, differentiation, and enhanced functional activity of mature effector cells .

What are the optimal methods for evaluating GM-CSF activity in research settings?

The TF-1 human erythroleukemia cell line provides an excellent system for detecting GM-CSF activity. This cell line of immature erythroid origin completely depends on interleukin-3 (IL-3) or GM-CSF for long-term growth, making it ideal for bioactivity assays. The standard protocol involves incubating TF-1 cells with various concentrations of recombinant human GM-CSF and observing cell proliferation using an inverted microscope over several days .

Another validated method utilizes the U937 human monoblastic leukemia cell line, where GM-CSF induces differentiation. Researchers typically incubate U937 cells with GM-CSF (10ng/mL is a standard concentration) for 5 days and observe morphological changes indicative of differentiation .

For quantitative assessment, researchers can employ:

  • Cell proliferation assays (MTT, XTT, or WST-1)

  • Flow cytometry to measure surface marker expression changes

  • Colony-forming assays using primary bone marrow cells

  • Western blot analysis to detect downstream signaling activation

These approaches provide complementary data on GM-CSF bioactivity from different perspectives, allowing for robust validation of experimental findings.

How should recombinant GM-CSF be reconstituted and stored to maintain optimal activity?

For optimal reconstitution, use 20mM Tris, 150mM NaCl (pH 8.0) to achieve a concentration of 0.1-1.0 mg/mL. It's critical to avoid vortexing during reconstitution as this can lead to protein denaturation and loss of activity. For short-term storage (up to one month), the reconstituted protein can be kept at 2-8°C .

For long-term storage, aliquot the reconstituted protein and store at -80°C for up to 12 months. Repeated freeze/thaw cycles should be strictly avoided as they significantly reduce protein activity. Thermal stability testing indicates that GM-CSF maintains stability when incubated at 37°C for 48 hours, with a loss rate of less than 5% .

Some preparations include stabilizers like trehalose and Proclin300, which enhance shelf-life. When preparing working dilutions, always use freshly reconstituted protein or thawed aliquots within the same day for consistent experimental results.

How can GM-CSF be utilized in dendritic cell-based immunotherapies and what are the protocol considerations?

GM-CSF serves as the principal mediator of proliferation, maturation, and migration of dendritic cells (DCs), which are crucial antigen-presenting cells that play a major role in inducing primary and secondary T-cell immune responses . For developing DC-based immunotherapies, researchers can follow these methodological guidelines:

  • DC Generation Protocol: Isolate CD14+ monocytes from peripheral blood using magnetic bead separation. Culture these cells with GM-CSF (50-100 ng/mL) and IL-4 (20-50 ng/mL) for 5-7 days to generate immature DCs. Add maturation factors (TNF-α, IL-1β, IL-6, and PGE2) for an additional 48 hours to obtain mature DCs.

  • Quality Control Parameters:

    • Flow cytometry should confirm high expression of CD80, CD86, HLA-DR, and CD83

    • Test for appropriate cytokine secretion profiles (IL-12p70, IL-10)

    • Assess T cell stimulatory capacity using mixed lymphocyte reactions

  • Antigen Loading:

    • For tumor therapies, pulse DCs with tumor lysates, specific tumor antigens, or transfect with tumor RNA

    • For infectious disease applications, load with pathogen-specific antigens

GM-CSF enhances DC functions through multiple mechanisms: increasing class II MHC expression, augmenting expression of costimulatory molecules (B7), enhancing adhesion molecule expression (ICAM), and promoting production of cytokines like IL-1, TNF, and IL-6 that facilitate expansion and differentiation of B and T lymphocytes .

What are the established protocols for using GM-CSF in peripheral blood progenitor cell mobilization?

Research has demonstrated that combining GM-CSF with G-CSF provides superior mobilization outcomes compared to either cytokine alone. The following protocol has demonstrated efficacy in clinical research settings:

  • Combination Regimen:

    • G-CSF: 10 μg/kg/day

    • GM-CSF: 5 μg/kg/day

    • Administration: Subcutaneous injection for 4-5 consecutive days

  • Collection Timing: Begin leukapheresis on day 5 of cytokine administration, when peripheral blood CD34+ cell counts typically peak.

  • Cell Characterization:

    • Flow cytometric analysis for CD34+CD38- and CD34+CD38-HLA-DR+ subpopulations

    • Colony-forming assays for CFU-GM and BFU-E potential

The combination mobilization approach yields significantly higher numbers of CD34+CD38-HLA-DR+ cells (1.41 × 10^6) compared to G-CSF alone (0.36 × 10^6) or GM-CSF alone (0.12 × 10^6). Additionally, the plating efficiency of colony-forming unit–granulocyte-macrophage (CFU-GM) and burst-forming unit-erythroid (BFU-E) is higher in cells stimulated by GM-CSF than in those stimulated by G-CSF .

Importantly, this combination approach may result in different immune cell compositions in the leukapheresis product, with potentially lower CD3+ T cell counts compared to G-CSF mobilization alone (160 versus 328 × 10^6/kg), which could impact graft-versus-host disease incidence in allogeneic transplantation settings .

How does GM-CSF enhance antitumor immunity and what are the optimal protocols for cancer immunotherapy applications?

GM-CSF enhances antitumor immunity through multiple mechanisms, making it valuable for cancer immunotherapy research. The protein enhances the cytotoxic activity of peripheral blood monocytes and lymphocytes, markedly increases antibody-dependent cellular cytotoxicity, and enhances monocyte cytotoxicity against malignant cell lines . Furthermore, GM-CSF augments IL-2–mediated lymphokine-activated killer (LAK) cell function and increases secretion of matrix metalloelastase in tumor-infiltrating macrophages, leading to angiostatin production that inhibits angiogenesis and suppresses metastatic growth .

Protocol for GM-CSF-based cancer immunotherapy research:

  • Tumor Vaccine Approach:

    • Engineer tumor cells to secrete GM-CSF (optimal concentration: 35-50 ng/10^6 cells/24h)

    • Irradiate cells (5000 cGy) to prevent proliferation while maintaining cytokine secretion

    • Administer subcutaneously, typically 1-5×10^6 cells per dose

    • Monitor immune responses via:

      • T cell proliferation assays

      • Cytokine production profiles (IFN-γ, TNF-α)

      • Antibody responses to tumor antigens

      • Tumor-infiltrating lymphocyte analysis

  • GM-CSF as an Adjuvant:

    • Combine GM-CSF (80-100 μg/day for 3-5 days) with:

      • Immune checkpoint inhibitors (anti-PD-1, anti-CTLA-4)

      • Tumor antigen vaccines

      • Adoptive cell therapies

In comparative studies, tumor cells engineered to secrete GM-CSF demonstrated superior stimulation of specific antitumor immunity compared to other cytokine-secreting tumor cells. This approach significantly enhances dendritic cell recruitment to the vaccination site and improves tumor antigen presentation .

What considerations are important when using GM-CSF in acute myeloid leukemia (AML) research?

Using GM-CSF in AML research requires careful consideration of its dual effects: potential stimulation of leukemia cells versus enhancement of anti-leukemic immune responses. Research methodologies should address these considerations:

  • Leukemic Cell Response Assessment:

    • Prior to therapeutic applications, evaluate whether patient AML blasts respond to GM-CSF with proliferation using colony formation assays

    • Determine if GM-CSF upregulates surface molecules (CD54/ICAM-1, CD58/LFA-3) on AML cells that might enhance their recognition by immune effectors

  • Post-Transplant Immunotherapy Protocol:

    • Administration timing: Typically 2-4 weeks after autologous bone marrow transplantation

    • Dosage: 5 μg/kg/day for 14-21 days

    • Monitoring parameters:

      • Activated killer cell function assays (baseline, during treatment, post-withdrawal)

      • Risk of relapse assessment

      • Correlation between activated killer cell activity and clinical outcomes

Research data demonstrates that GM-CSF treatment post-autologous bone marrow transplantation significantly enhances activated killer cell function (from 1.8% before transplant to 35% during treatment), with sustained effect after withdrawal (20%). This immunological enhancement correlates with reduced relapse risk (37.4% in GM-CSF-treated patients vs. 49.5% in controls) .

The mechanism appears to involve GM-CSF-induced upregulation of adhesion molecules on leukemia cells, particularly ICAM-1 (CD54) on leukemic CD34+ cells, making them more susceptible to immune recognition and killing. Experimental data shows that pre-exposure of AML cells to GM-CSF before incubation with immune effector cells significantly reduces their subsequent clonogenic activity .

How can researchers address variability in cellular responses to GM-CSF across different experimental systems?

Cellular responses to GM-CSF can vary considerably between different experimental systems due to multiple factors. To address this variability, researchers should implement the following methodological approaches:

  • Receptor Expression Characterization:

    • Quantify GM-CSF receptor (α and β chains) expression levels on target cells via flow cytometry or qRT-PCR

    • Correlate receptor density with functional responses

    • Consider that receptor polymorphisms may affect signaling efficiency

  • Standardization Procedures:

    • Use international standards for GM-CSF activity calibration

    • Establish dose-response curves for each cell system (typical effective range: 0.1-50 ng/mL)

    • Include positive control cell lines with well-characterized responses (TF-1 cells)

    • Normalize data to internal standards across experiments

  • Microenvironment Considerations:

    • Control serum factors that may contain GM-CSF inhibitors or synergistic factors

    • Standardize cell density, as overcrowding can alter responses

    • Document passage number for cell lines, as receptor expression may change with extended culture

  • Data Analysis Approaches:

    • Use EC50 values rather than single-point measurements

    • Implement area-under-curve analysis for time-dependent responses

    • Apply hierarchical statistical models to account for batch effects

When significant variability is observed despite these controls, investigate potential biological mechanisms, such as receptor downregulation after initial exposure, the presence of soluble receptors acting as decoys, or heterogeneity in downstream signaling pathways.

What are the common technical challenges in GM-CSF activity assays and how can they be overcome?

GM-CSF activity assays present several technical challenges that can affect experimental reproducibility and data interpretation. Here are the most common issues and recommended solutions:

  • Protein Stability Issues:

    • Challenge: Activity loss during reconstitution or storage

    • Solution: Reconstitute in buffer containing stabilizers (e.g., 0.1% human serum albumin), make single-use aliquots, and store at -80°C. Avoid multiple freeze-thaw cycles.

  • Endotoxin Contamination:

    • Challenge: Bacterial endotoxins can activate cells independently of GM-CSF

    • Solution: Use endotoxin-tested GM-CSF preparations (<0.1 EU/μg), include polymyxin B controls, and verify results with endotoxin inhibitors like LAL reagent.

  • Cell Responsiveness Variation:

    • Challenge: Diminished cell line responsiveness over passages

    • Solution: Maintain low-passage cell banks, regularly test receptor expression, and establish standard response curves for each new batch of cells.

  • Assay Endpoint Selection:

    • Challenge: Different endpoints yield varying sensitivity

    • Solution:

      • For proliferation: Compare MTT, tritiated thymidine incorporation, and direct cell counting

      • For functional assays: Combine multiple readouts (e.g., cytokine production, surface marker expression)

      • Use time-course experiments to identify optimal measurement windows

  • Data Interpretation Complexities:

    • Challenge: Distinguishing direct GM-CSF effects from secondary responses

    • Solution: Include appropriate blocking antibodies against GM-CSF receptor, use pathway-specific inhibitors, and perform parallel experiments with receptor-negative cell lines as controls.

How is GM-CSF being investigated in combination with immune checkpoint inhibitors?

GM-CSF's ability to enhance antigen presentation and immune cell activation makes it a promising candidate for combination with immune checkpoint inhibitors (ICIs). Recent research has focused on several methodological approaches:

  • Sequential Administration Protocols:

    • GM-CSF administration (3-5 days, 125-250 μg/m²/day) preceding ICI therapy (anti-PD-1, anti-CTLA-4)

    • Rationale: GM-CSF primes the immune system by increasing dendritic cell functionality and T cell activation before releasing checkpoint inhibition

  • GM-CSF-Secreting Cellular Vaccines with ICIs:

    • Irradiated autologous tumor cells engineered to secrete GM-CSF combined with systemic checkpoint inhibition

    • Assessment metrics:

      • Changes in tumor-infiltrating lymphocyte composition

      • Expansion of tumor-specific T cell clones

      • Broadening of T cell receptor repertoire diversity

      • Conversion of "cold" to "hot" tumor microenvironments

  • Biomarker Development:

    • Monitoring myeloid/lymphoid ratios in peripheral blood

    • Tracking changes in circulating monocyte subsets (classical, intermediate, non-classical)

    • Correlating these parameters with clinical response

This combinatorial approach leverages GM-CSF's ability to enhance T-cell immune responses by augmenting antigen presentation, increasing expression of costimulatory molecules (B7), and enhancing production of cytokines that promote expansion and differentiation of lymphocytes . The expectation is that GM-CSF-mediated immune priming will improve response rates to checkpoint inhibition, particularly in patients with low baseline tumor immunogenicity.

What new methodologies are being developed to enhance the therapeutic efficacy of GM-CSF in research settings?

Several innovative approaches are being developed to enhance GM-CSF's therapeutic potential:

  • Protein Engineering Strategies:

    • Site-directed mutagenesis to create GM-CSF variants with:

      • Enhanced receptor binding affinity

      • Extended half-life

      • Reduced immunogenicity

      • Cell type-specific targeting

    • Fusion proteins combining GM-CSF with:

      • Tumor-targeting antibody fragments

      • Additional cytokines (IL-2, IFN-γ) for synergistic effects

      • Immune checkpoint blocking domains

  • Advanced Delivery Systems:

    • Nanoparticle encapsulation for sustained release

    • Biomaterial scaffolds for localized delivery at tumor sites

    • mRNA delivery approaches for in situ GM-CSF production

  • Genetic Modification Techniques:

    • CRISPR/Cas9-mediated enhancement of GM-CSF receptor expression on effector cells

    • Chimeric antigen receptor (CAR) designs incorporating GM-CSF signaling domains

    • Regulatable GM-CSF expression systems using tetracycline-responsive promoters

  • Combination Strategy Development:

    • Systematic evaluation of GM-CSF with:

      • Radiation therapy (optimal timing: 24-48 hours post-radiation)

      • Chemotherapy agents that induce immunogenic cell death

      • Pattern recognition receptor agonists (TLR ligands)

    • Methodological assessment of sequence, dosing, and scheduling effects

These approaches are being evaluated using advanced preclinical models, including humanized mouse systems and three-dimensional organoid cultures, to better predict translational outcomes in human patients.

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.