Are you in search of a reliable method for discovering high-quality antibodies against cancer? Look no further than the world’s first immune phage display library derived from human cancer patient donors. These unique libraries are a rich source of high-affinity tumor-specific antibodies, ready to be developed into the next generation of cancer immunotherapies. With 3 therapeutic antibodies on the market and over 30 in pre-clinical and clinical development, you can trust ProteoGenix to consistently deliver outstanding results. Interested in customizing your anti-cancer antibody? We specialize in enhancing each antibody’s developability and offer advanced customization options tailored to your specific needs. ProteoGenix is your comprehensive solution for all your custom antibodies for cancer treatment. Schedule a call with our antibody experts today!

Custom Immune Checkpoint Antibody Discovery Workflow

Antigen procurement or design and production
Your cancer-specific antigen can be acquired by:

  • Delivering the antigen to ProteoGenix
  • Allowing ProteoGenix to make the antigen for you (preferred)

Immune Library Screening and Biopanning

  • Screening of our pre-built immune library for antigen binders
  • 4-6 rounds of biopanning

ELISA Screening of Single Phage Binders
Further validate binders by ELISA screening until at least 3-10 different binders have been identified.

DNA Extraction & Antibody Sequencing

Antibody Phage Display Service from Human Cancer Patients

Antibody phage display is one of the most powerful monoclonal antibody discovery methods on the market. Obtaining a source of high-affinity cancer-specific antibodies used to require the time-consuming and expensive act of engineering a custom library from immunized animal hosts.

These downsides are now a thing of the past. ProteoGenix is proud to offer the first-ever pre-built immune human antibody phage display library isolated from cancer patient donors. Each immune library (scFv and Fab libraries) offers high-affinity cancer-specific antibodies normally reserved to custom libraries built from immunized (non-human) hosts. However, ProteoGenix’s pre-built human immune cancer antibody library offers the rapid delivery and reduced cost of naïve libraries combined with the abundant high-affinity cancer-specific antibodies found in immunized hosts. Truly the best of both worlds.

Library Name Species Clone Number
LiAb-SFCANCERTM Library Human Donors (48 patients with 7 different cancers):
  • Prostate Cancer
  • Colorectal Cancer
  • Renal Cancer
  • Lung Cancer
  • Skin Cancer
  • Melanoma
  • Acute Myeloid Leukemia
3.81x1010 (scFv) 3.72x1010(Fab)

ProteoGenix’s pre-built immune cancer libraries are an abundant source of novel cancer immunotherapies waiting to be discovered. With a huge diversity of unique antibodies (>3×1010 different clones) and an excellent in-frame frequency (greater than 93%), each library is a rich source of novel cancer immunotherapies.

High-performing antibodies that bind cancer antigens with minimal cross-reactivity are notoriously difficult to design. However, our immune human cancer libraries are especially useful for creating monoclonal antibodies that target highly conserved antigens found in human cancers. These include:

  • Cancer neoantigens like MHC-peptide complexes
  • Highly conserved proteins
  • Membrane proteins
  • Glycoproteins

Antibody Phage Display Deliverables and Project Timeline

Developing your custom anti-tumor monoclonal antibody using ProteoGenix’s naïve human cancer libraries means you receive intellectual property (IP)-free DNA sequences of three high-affinity binders to your target cancer antigen.

Each project takes approximately 8 weeks from antigen design and purification to delivery of your antibody DNA sequences. That is one of the fastest deliverable timelines on the market.

ProteoGenix also offers several other additional services for maximized convenience. This includes:

Combining our custom anti-cancer antibody development service with our additional antibody expression, developability, and customization services means you can outsource your toughest cancer immunotherapy projects to our antibody experts, freeing up your time, money, and resources while accelerating market entry.

High Antibody Clone Diversity with Minimal Construction Bias

Unlock the full potential of antibody phage display with our proprietary library. Unlike other methods that combine the variable regions of kappa and lambda light chains together, our library separates their processing to generate a higher diversity of unique antibody clones and reduce construction bias.

This breakthrough technique increases the chances of discovering high-affinity antibodies with no cross-reactivity against challenging antigens such as cancer neoantigens and highly conserved human antigens. This technique allows ProteoGenix to produce higher quality antibodies, increasing the potential your antibody makes it to the advanced clinical stages of development.

Why are Immune Human Cancer Libraries An Untapped Source of Future Immunotherapies?

The immune system of cancer patients is unable to recognize and eliminate cancer cells during the earliest stages of transformation. This represents an inability of the immune system to distinguish self from non-self-antigens resulting in cancer cell escape of immune detection.

Eventually, the adaptive immune response recognizes the non-self-antigens expressed by the cancer cell, but unfortunately, the opportunity to completely eliminate the cancer has passed allowing the transformed cells rapidly proliferate and become a tumor.

Although early cancer cells initially evade the immune system, their evasion is short-lived. The immune system eventually recognizes and mounts an adaptive immune response against the cancer. A immune antibody phage display library from cancer patients is therefore a rich source of novel anti-cancer antibodies. Never before has anyone been able to access these invaluable antibody repertoires, until now.

Affinity Maturation Produces Anti-Tumor Antibodies with High Affinity

An antibody phage display library sourced from a cancer patient offers a repertoire of antibodies that have undergone natural affinity maturation and selection to cancer antigens. This provides a source of antibodies with enhanced antigen binding affinity and therapeutic potential compared to a naïve library constructed from healthy donors.

By utilizing a human-derived library, the risk of immunogenicity and adverse reactions during therapeutic applications is minimized. This breakthrough offers a transformative platform for accelerating the discovery and development of novel, effective, and patient-specific antibody-based treatments in the field of cancer immunotherapy.

Immune Human Cancer Phage Display Library Antibody Characteristics

Antibodies developed from our immune human cancer phage display libraries hold immense value due to their inherent ability to target tumor-specific antigens. They offer the potential for more precise and potent therapeutic interventions, as they have likely undergone the affinity maturation processes driven by the complexities of the cancer microenvironment.

Thus, each library offers an untapped source of diverse and highly specific antibodies, unlocking new avenues for the development of innovative and effective antibodies against cancer.

Below is a table summarizing the antibody characteristics of naïve donors with cancer compared to that of healthy human donors and immunized hosts:

Naive Antibody Library (Human Donor) Immune Antibody Library (Human Donor) Immune Library (Non-Human Host)
Source Healthy Human Donors Cancer patients Immunized Animals (non-human)
Antibody Diversity Broad Cancer Antigen Skewed Immunized Antigen Skewed
Target Specificity Limited Antigen-specific Antibodies Increased Cancer-specific Antibodies Increased Antigen-specific Antibodies
Affinity Maturation Minimal Affinity Maturation Affinity Maturation to Cancer Antigens Affinity Maturation to Immunized Antigen
Therapeutic Applications Potential for Broader Applications Potential for Cancer-Specific Therapeutic Antibodies Primarily Focused on the Immunizing Antigen
Immunogenicity Risk Fully Human – Minimal Immunogenicity Risk Fully Human – Minimal Immunogenicity Risk Risk of an Immune System Reaction
Clinical Trials May Require Additional Optimization and Engineering Accelerated Leads for Cancer-Specific Therapies May require further characterization and optimization

What is Antibody Phage Display

Antibody phage display is a method used in the laboratory to investigate the interactions between antibodies and other molecules such as antigens. This method genetically manipulates the DNA of bacteriophages, viruses that infect bacteria, by fusing genes encoding antibodies with a phage coat protein.

This causes the phage to display the antibody on its exterior while containing the gene for the antibody inside the phage. As a result, this establishes a link between the genotype and phenotype. The displaying phages can then be screened against unique antigens to identify antibodies that interact with the antigen. This allows for the screening and amplification of large antibody libraries through a process similar to natural selection, known as in vitro selection.

How is a Phage Display Library Made?

An antibody phage display library is made by cloning antibody variable region genes into a phage display vector. This process begins by isolating mRNA from B-cells of an animal or human patient, and then reverse transcribing it into cDNA. Next, primers are designed which hybridize the conserved antibody cDNA sequences. The antibody cDNA is used as a template to amplify the variable regions of the heavy and light chains of each antibody using PCR.

The amplified variable regions are then cloned into a phage display vector that has been modified to express the antibody variable regions as a fusion protein with a coat protein on the surface of the phage. This allows the antibodies to be displayed on the surface of the phage, while the DNA encoding the antibody is contained within the phage particle.

The library can then be screened against a target antigen of interest to identify phages displaying antibodies that bind to the target. The bound phages can be eluted, and the DNA encoding the antibody can be sequenced and used to produce the desired antibody for further study or therapeutic purposes.

Immune Libraries and Naïve Libraries

A naive antibody phage display library is constructed using genes from an organism that has not been exposed to the antigen of interest. In contrast, an immune antibody phage display library is constructed using genes from an organism that has been immunized with the antigen of interest.

Naive libraries contain a diverse array of antibodies that have not undergone antigen-driven selection, which can lead to the identification of novel antibodies with unique binding properties. Immune libraries, on the other hand, contain antibodies that have been selected for their ability to bind the antigen of interest and can be useful for the rapid identification of high-affinity antibodies.

Overall, the main difference between naive and immune antibody phage display libraries is the source of the genes used to construct them. While naive libraries contain a diverse array of antibodies that have not been exposed to the antigen of interest, immune libraries contain antibodies that have been selected for their ability to bind to that antigen.

Single Chain Variable Fragment (scFv) Antibody Format

A single-chain variable fragment (scFv) is a fusion protein that consists of the variable regions of the heavy (VH) and light chains (VL) of immunoglobulins, connected by a short peptide linker that contains approximately 10 to 25 amino acids. It is not an actual fragment of an antibody. The linker peptide is often rich in glycine for flexibility and contains serine or threonine for solubility. The linker can connect the N-terminus of the variable heavy chain (VH) with the C-terminus of the variable light chain (VL) or vice versa.

Single Chain Variable Fragment (scFv) Format Advantages

Single-chain variable fragments (scFv) offer a range of advantages compared to the parental monoclonal antibody (mAb) format, as highlighted below:

  • Smaller size: scFv molecules have a compact structure, enabling better tissue penetration and improved access to target sites.
  • Easy production and purification: The smaller size of scFv simplifies production and purification processes, resulting in higher yields and cost savings.
  • Recombinant DNA technology: scFv can be produced using recombinant DNA technology, allowing for efficient and scalable production in various expression systems.
  • Versatile engineering options: scFv molecules can be engineered to have multimeric forms and/or multivalency, enhancing their binding affinity and specificity compared to the parental mAb.

 

These advantages make scFv an attractive choice for therapeutic and diagnostic applications, offering improved properties and potential for optimization in antibody-based treatments.

Fragment Antigen Binding (Fab) Antibody Format

The fragment antigen-binding (Fab) region is a segment of an antibody that links to antigens. It comprises one variable and one constant domain from both the heavy and light chains. The variable domain encompasses the paratope, which is part of the antibody that binds to the antigen and contains a group of complementarity-determining regions located at the N-terminus of the monomer. This arrangement enables each arm of the Y-shaped antibody to attach to an epitope on the antigen.

Fragment Antigen Binding (Fab) Format Advantages

The fragment antigen-binding (Fab) antibody format provides several advantages compared to full-length monoclonal antibodies (mAbs), as outlined below:

  • Smaller size: Fab fragments are significantly smaller than full-length mAbs, enabling improved tissue penetration and accessibility to target sites.
  • Reduced immunogenicity: The smaller size of Fab fragments reduces the likelihood of triggering an immune response, making them less immunogenic.
  • Cost-effective production: Fab fragments can be produced more efficiently and cost-effectively compared to full-length mAbs, leading to potential cost savings.
  • High specificity: Fab fragments exhibit high specificity for their target antigens, allowing for precise and targeted binding.
  • Versatile applications: Fab fragments can be engineered for various applications, including drug delivery, imaging, and targeting specific antigens in tumors or other tissues.
  • Diagnostic and therapeutic potential: The high specificity and tissue penetration of Fab fragments make them valuable in diagnostic assays and as potential therapeutics for diseases such as cancer and infectious diseases.
  • Broad applicability: The advantages of the Fab antibody format make it a promising candidate for diverse applications in cancer therapy, infectious disease diagnosis, and other areas of biomedical research.

 

The unique features of the Fab antibody format position it as an attractive option for various applications, offering improved performance, versatility, and cost-effectiveness compared to full-length mAbs.