• Home
  • PrimePDX™: Next-Gen PDX for Clinical Simulation
Alfa Cytology - Cancer Drug R&D Services

PrimePDX™: Next-Gen PDX for Clinical Simulation

Inquiry

Patient-derived xenografts (PDX) preserve tumor histology and heterogeneity in vivo, making them a critical bridge from in vitro studies to clinical translation. Building on this foundation, Alfa Cytology's PrimePDX™ combines a PBMC-humanized immune baseline with single-cell/small-aggregate initiation and early-passage use, plus optional human CAF microenvironment. The result is an integrated in vitro-to-in vivo workflow that accelerates confident decisions on candidate selection, combinations, and immune mechanisms.

Introduction to PrimePDX™ Platform

The PrimePDX™ establishes tumor PDX on a PBMC-humanized immune background, bridging in vitro clinical simulation ( PDC/ 3D) with in vivo studies to deliver immuno-oncology readouts that are closer to real-world settings. Methodologically, we use single-cell/small-aggregate initiation to improve starting uniformity and conduct efficacy in early passages to balance heterogeneity with statistical robustness; an optional human CAF xenograft increases stromal content and immune-suppressive features to emulate the "tumor fortress."

The platform is powered by a biobank of over 2,000 live tumor samples spanning major indication. It is integrated with a tumor database (pathology & multi-omics) and a patient database (clinical data & longitudinal follow-up), alongside GWAS/PheWAS, NGS, medical imaging, plasma/DNA/genotyped samples, and real-world data. This data backbone enables biomarker- and history-aware model selection, faster hypothesis testing, and a seamless in vitro-to-in vivo concordance workflow that strengthens decisions on candidate selection, dosing & scheduling, combinations, immune mechanisms, and patient stratification.

Advantages to PrimePDX™ Platform

Modeling

  • Single-cell/small-aggregate inoculation improves starting uniformity and facilitates paired in vitro/in vivo mechanism testing.
  • Efficacy in P2–P5 balances heterogeneity and clinical relevance with clearer group separation and stable statistics.
  • Optional human CAF co-implantation heightens stromal content and immune suppression to better model antibody/cell-therapy challenges.

From In-Vitro to In-Vivo

  • Use PDC/ 3D for high-throughput in-vitro stratification, then proceed to matched in vivo PDX; emphasize trend concordance and hit rate.
  • Supports small molecules, antibodies/ADCs, checkpoint inhibitors, cell therapies, cancer vaccines, and oncolytic viruses.

Dual humanization

  • Perform PDX efficacy and immune readouts on PBMC-humanized mice.
  • Autologous pairing—same-patient PBMC × same-patient tumor in one mouse, is technically feasible and can be evaluated and custom-run subject to sample availability and ethics.

Comparison of Patient-Derived Xenograft (PDX) Model

The table compares conventional PDX, PDO, and PrimePDX™ across methodology, in vitro throughput, clinical simulation, in vivo efficacy, immune integration, and end-to-end application. Conventional PDX remains dependable for general in-vivo efficacy; PDO excels at high-throughput in-vitro screening. PrimePDX™ adds a PBMC-humanized baseline, single-cell/small-aggregate initiation, and early-passage (P2–P5) use, with optional human CAF to enhance stromal realism—delivering stronger from in vitro to in-vivo concordance, immune-relevant readouts, and a more time- and cost-efficient integrated workflow for immuno-oncology and translational decision-making.

PrimePDX™Model Traditional PDX Model PDO
Key Technical Aspects Patient-derived tumors are engrafted in mice to generate PDX models, followed by tissue dissociation and cryopreservation to create primary cell banks. PDX Fragment Biobank. Generation of a Biobanked Patient-Derived Organoid (PDO) Library.
In Vitro High-Throughput Analysis Capability Primary Cell Bank & High-Throughput Assays.
Strong In Vivo Data Correlation.
Cost-Competitive vs. Conventional Cell Lines.
A key limitation is the absence of relevant in vitro models, creating a bottleneck for high-throughput screening. Suitable for high-throughput in vitro screening, albeit at a higher cost than primary cell-based assays.
Clinical Trial Simulation Capability Cost-effective, high-throughput clinical simulation studies, followed by verification in relevant small-scale animal models. Expenses and duration are multi-fold to ten-fold higher than in vitro high-throughput models. Suitable for in vitro clinical simulation studies, but lacks in vivo validation and is not sufficient for regulatory submission studies.
In Vivo Efficacy Testing Capability Superior Model Stability.
Enhanced Clinical Predictive Power.
Poor quality control and model reproducibility
Loss of Clinical Relevance.
No
In Vivo Model Efficacy Pre-Validation of Drug Efficacy in Candidate Models via In Vitro Screening.
>80% Concordance Between In Vivo and In Vitro Data.
Inefficient Selection Strategy: Reliance on target profiling alone ignores key resistance pathways.
Major Project Delays: High rate of unresponsive models necessitates repeated in vivo studies, slowing progress.
No
Degree of Humanized Immune System-Tumor Integration The vast majority of models can be established as dual-humanized and are suitable for drug efficacy testing. Poor compatibility with humanized immune system models. TME Modeling via Immune Co-Culture.
In Vitro, IND-Enabling, Clinical-Simulation Integrated Trilogy Universal Application: Suitable for both in vitro and in vivo studies.
Reutilization & Data Integration: A platform approach with a closed-loop data system.
Dramatic Cost Reduction: Cuts project costs to 1/2 - 1/3 of traditional PDX pathways.
Solely For In Vivo Research Solely For In Vitro Research.

Workflow for Generating a PDX Model in PBMC Humanized Mice

PBMC Humanization Modeling

  • Isolate PBMCs, condition recipients, engraft and reconstitute.
  • Proceed when human leukocyte reconstitution meets the project-defined threshold.

Tumor Processing

  • Gently dissociate patient tumor to single-cell or small aggregates.
  • Verify viability/phenotype and align with matched in vitro models ( PDC/ 3D).

PDX Engraftment

  • Inoculate via Subcutaneous / Orthotopic / IV systemic / Brain.
  • Optional human CAF co-implantation to boost stromal context.
  • Enable IVIS if applicable.

Early-passage Expansion & QC

  • Track P0→P2–P5.
  • Confirm growth stability, histology/molecular retention, and group separation suitable for efficacy statistics.

Case Study

Case 1 PrimePDX™ -PDC (Patient-Derived Tumor Cell) In Vitro Efficacy Platform

  • Model Introduction
    This study evaluates the concordance of PDC-based in-vitro stratification for PD-L1 high NSCLC with in vivo efficacy on PBMC-humanized PrimePDX™. The aim is to test whether PDC results predict tumor response to immune checkpoint inhibitors (ICI) in the PD-L1 high, EGFR/ALK wild-type NSCLC model.
  • Model Information
    • Model: PrimePDX™ – NSCLC in PBMC-humanized mice
    • Age:6–8 weeks
    • Weight:18–22 g
    • Passage:P2–P5 for efficacy

Model Characteristics

  • Simulated Clinical Efficacy Testing
  • Applicable for Small Molecule Drugs, Antibodies, Oncolytic Viruses, Immunotherapies, and Cancer Vaccines
  • Cost-effective (Comparable to Tumor Cell Line Assays)
  • Rapid Turnaround (<2 Weeks)
  • High-throughput Cohort Screening

The in vitro and in vivo efficacy of the same model demonstrate high consistency

Case 2: PrimePDX™ -Next-Generation PDX Models with Enhanced Stability

  • Model Introduction
    This case study tests the efficacy of an ADCC-enabled antibody in PBMC-NK PrimePDX™ for HER2-positive breast cancer, focusing on dose optimization and immune response. The model evaluates immune-mediated efficacy and tumor infiltration dynamics, with in vivo readouts aligned to the ADCC mechanism.
  • Model Information
    • Model: PrimePDX™ – HER2-Positive Breast Cancer in PBMC-humanized mice
    • Age:6–8 weeks
    • Weight:18–22 g
    • Passage:P2–P5 for efficacy

Model Characteristics

Stable tumor growth

  • Tumor growth stability similar to CDX models.
  • Allow usage of early passages.

Early passage for efficacy study

  • Normally using P2-P5 for efficacy study.
  • Early passages have a higher similarity to patient tumors.

Dual humanized models

  • Stable PDX growth and PBMC reconstitution
  • T, NK reconstitution.

Guaranteed High Success Rates

  • Pre-screening with in vitro PDC models identifies optimal PDX models, ensuring a high success rate in subsequent in vivo efficacy studies

Superior Stability

  • Demonstrates improved tumor growth stability and more significant intergroup differences for precise efficacy evaluation.

Preserved Clinical Relevance

  • Utilizes early-passage PDX modeling to better retain the original tumor heterogeneity and drug response profile.

Streamlined Workflow Integration

  • Enables efficient in vitro screening to select the most predictive models, boosting in vivo study success.

Why Choose Us

  • Complies with the Standards of the Human Genetic Resources Regulations
  • Comprehensive Patient Clinical Background
  • Enhanced Usability & Stability: >50% vs. Conventional PDX Models
  • Unique PBMC Compatibility: Enabling PBMC-PDX Dual-Humanized Models

FAQs

Do you support "same-patient PBMC × same-patient tumor in one mouse"?

It is research-feasible but limited by PBMC yield and patient condition. Our standard offering is PDX on PBMC-humanized mice; autologous designs require dedicated feasibility assessment.

How do you manage window and variability?

Via humanization subtype selection and SOP control with predefined dosing/observation windows, plus early-passage use and redundancy in readouts.

When is PrimePDX the best fit?

For immune-mechanism-centric programs—checkpoint, ADCC, NK modulation, bi/tri-specifics, cell therapies, vaccines—requiring integrated in vitro-to-in vivo evidence.

Leverage Alfa Cytology's PrimePDX™ Platform for reproducible, clinically relevant immuno-oncology readouts. From model provisioning and study design to dosing & monitoring, immune profiling, multi-omics/pathology, IVIS imaging, and statistical reporting, our expert team delivers standardized yet configurable workflows that align in-vitro findings with in-vivo evidence. Whether you're validating mechanisms, prioritizing candidates, optimizing combinations, or building biomarker packages, we help you move faster—and smarter. Contact us to explore collaboration and request a custom study proposal

For research use only.

Online Inquiry