Alfa Cytology offers a comprehensive in vivo animal model development service for Metastatic Colorectal Cancer (mCRC), providing researchers and pharmaceutical partners with a robust platform to evaluate novel therapeutics, study tumor biology, and investigate mechanisms of metastasis and drug resistance. Our extensive portfolio features a wide range of clinically relevant mouse models, including patient-derived xenografts, syngeneic grafts, and genetically engineered systems, tailored to accelerate translational research and preclinical drug development.
Metastatic Colorectal Cancer remains a significant clinical challenge due to its aggressive nature and complex molecular landscape. Accurate animal models are essential for elucidating disease mechanisms and assessing therapeutic efficacy prior to clinical trials. Alfa Cytology utilizes Mus musculus (mouse) as the primary species, leveraging various strains such as Balb/c, C57BL/6, NOD, NSG, and athymic nude mice. These strains offer distinct immunological and genetic backgrounds, enabling the study of human tumor biology, immune interactions, and metastasis in a controlled environment. Our models incorporate key genetic mutations and resistance profiles observed in human mCRC, ensuring high translational relevance and predictive value for drug discovery and biomarker validation.
PDX models are established by implanting tumor tissues or cells directly derived from metastatic colorectal cancer patients into immunodeficient mice, such as NOD, NSG, or nude strains. This methodology preserves the heterogeneity, genetic mutations (e.g., BRAF, PIK3CA, KDR/VEGFR2), and drug resistance characteristics of the original tumor. Key advantages include high clinical relevance, faithful recapitulation of human tumor biology, and the ability to model therapeutic resistance. PDX models are primarily used for efficacy testing of novel agents, biomarker discovery, personalized medicine approaches, and studying mechanisms of metastasis and resistance.
Syngeneic models involve transplanting murine colon adenocarcinoma cell lines, such as CT26-FL3 or MC38, into immunocompetent mice of the same genetic background (e.g., Balb/c or C57BL/6). Allograft models use murine cells in genetically matched hosts, enabling the study of tumor-immune interactions. Orthotopic implantation (e.g., cecum or liver) enhances physiological relevance by mimicking the metastatic cascade. These models are advantageous for immunotherapy research, evaluation of tumor microenvironment, and preclinical screening of immunomodulatory agents.
These models utilize human or murine colon cancer cell lines that are genetically modified to express specific mutations (e.g., BRAF, PIK3CA), overexpress oncogenes (e.g., NRG1, FGF13, IGF2), or exhibit drug resistance (e.g., cetuximab- or vemurafenib-resistant). Cells are often transfected with reporter genes (e.g., luciferase, RFP) for non-invasive imaging and metastasis tracking. The main advantages are controlled genetic backgrounds, reproducibility, and suitability for mechanistic studies and high-throughput drug screening. Applications include evaluating targeted therapies, resistance mechanisms, and metastatic dissemination.
Alfa Cytology delivers a full-spectrum solution for in vivo mCRC model development and therapeutic testing. Our services encompass model selection and customization, tumor implantation (subcutaneous, orthotopic, or metastatic), drug administration, and comprehensive monitoring. Key efficacy endpoints include tumor growth inhibition, metastatic burden (quantified by bioluminescent imaging or histopathology), survival analysis, and assessment of drug resistance. Analytical capabilities span molecular profiling (genotyping, gene expression, protein analysis), pharmacokinetics/pharmacodynamics (PK/PD), and immune cell phenotyping. Stringent quality control measures are implemented at every stage, including authentication of cell lines, validation of genetic mutations, and standardized protocols to ensure reproducibility and data integrity.
Partnering with Alfa Cytology provides access to scientifically validated, highly translational animal models and expert-driven preclinical services tailored to your research objectives. Our commitment to quality, innovation, and collaboration ensures that your studies generate meaningful, actionable data to advance Metastatic Colorectal Cancer research and therapeutic development. Contact us today to discuss your project needs and accelerate your path to discovery.
| Species | Strain | Characteristic (Details) |
|---|---|---|
| Mus musculus (mouse) | Balb/c | Nude; Patient-derived xenograft (Colorectal carcinoma cells (metastatic), human (BRAF (V600E)-mutated) (encorafenib/cetuximab-resistant)) |
| Mus musculus (mouse) | Balb/c | Nude; Patient-derived xenograft (Colorectal carcinoma cells (metastatic), human (BRAF (V600E)-mutated)) |
| Mus musculus (mouse) | Balb/c | Orthotopic syngeneic graft (CT26-FL3 murine colon adenocarcinoma cells (liver-metastatic) transfected with RFP/luciferase) |
| Mus musculus (mouse) | C57BL/6 | Orthotopic syngeneic graft (MC38 murine colon adenocarcinoma cells transfected with luciferase) |
| Mus musculus (mouse) | C57BL/6J | Allograft (CT26-FL3 murine colon adenocarcinoma cells (liver-metastatic) (microsatellite stable)) |
| Mus musculus (mouse) | Foxn1 nu/nu | Athymic nude; Patient-derived orthotopic xenograft (Colorectal carcinoma cells (metastatic), human) |
| Mus musculus (mouse) | NOD | Patient-derived xenograft (Colon cancer cells, human (PIK3CA (H1047L) mutated)); Severe combined immune deficiency (SCID) |
| Mus musculus (mouse) | NOD | Patient-derived xenograft (Colon cancer cells, human (PIK3CA (H1047R) mutated) (cetuximab-resistant)); Severe combined immune deficiency (SCID) |
| Mus musculus (mouse) | NOD | Patient-derived xenograft (Colon cancer cells, human (PIK3CA (H1047R) mutated)); Severe combined immune deficiency (SCID) |
| Mus musculus (mouse) | NOD | Patient-derived xenograft (Colon cancer cells, human (PIK3CA (V344G) mutated) (cetuximab-resistant)); Severe combined immune deficiency (SCID) |
| Mus musculus (mouse) | NOD | Patient-derived xenograft (Colorectal carcinoma cells (metastatic), human (NRG1-overexpressing)); Severe combined immune deficiency (SCID) |
| Mus musculus (mouse) | NOD | Patient-derived xenograft (Colorectal carcinoma cells (metastatic), human); Severe combined immune deficiency (SCID) |
| Mus musculus (mouse) | NSG | Xenograft (HT29 human colon adenocarcinoma cells transfected with luciferase) |
| Mus musculus (mouse) | nu/nu | Nude; Patient-derived xenograft (Colorectal carcinoma cells (metastatic) (KDR/VEGFR2-mutated), human) |
| Mus musculus (mouse) | Immunosuppressed; Patient-derived xenograft (Colorectal carcinoma cells (metastatic), human) | |
| Mus musculus (mouse) | Knockin (HGF); Patient-derived xenograft (Colorectal carcinoma cells (metastatic), human); Severe combined immune deficiency (SCID) | |
| Mus musculus (mouse) | Nude; Orthotopic xenograft (RKO human colon carcinoma cells) | |
| Mus musculus (mouse) | Nude; Patient-derived xenograft (Colorectal carcinoma cells (metastatic), human (FGF13-overexpressing)) | |
| Mus musculus (mouse) | Nude; Patient-derived xenograft (Colorectal carcinoma cells (metastatic), human (IGF2-overexpressing)) | |
| Mus musculus (mouse) | Patient-derived xenograft (Colorectal carcinoma cells (metastatic), human (b-Raf mutated)) | |
| Mus musculus (mouse) | Xenograft (Colorectal carcinoma cells (metastatic)) | |
| Mus musculus (mouse) | Xenograft (Colorectal carcinoma cells (metastatic), human) | |
| Mus musculus (mouse) | Xenograft (RKO human colon carcinoma cells (b-Raf mutated/vemurafenib-resistant)) |
Make Order
Experimental Scheme
Implementation
Conclusion