Aptamer Screening Services for Multiple Targets At KMD Bioscience, we understand that the future of diagnostics, therapeutics, and targeted research lies in high-affinity, specific molecular recognition. While antibodies have long been the standard, aptamers—often termed "chemical antibodies"—offer a superior, versatile alternative. Our comprehensive Aptamer Screening Services are designed to discover and develop these powerful single-stranded DNA or RNA molecules against a diverse range of your targets. Why Choose Aptamers? Aptamers bind to their targets, from small molecules and proteins to whole cells and viruses, with exceptional specificity and affinity. They offer distinct advantages: High Specificity & Affinity: Selected through an iterative process to precisely recognize unique epitopes. Chemical Stability: Unlike proteins, aptamers are thermally stable and can be easily regenerated. Easy Modification: Simple chemical synthesis allows for easy labeling and conjugation without loss of activity. Low Immunogenicity: Ideal for in vivo therapeutic and diagnostic applications. Our Multi-Target Screening Platform: SELEX Evolved We employ and continuously optimize state-of-the-art SELEX (Systematic Evolution of Ligands by EXponential enrichment) technologies to cater to the unique nature of each target. Our platform is not one-size-fits-all; it is a flexible, sophisticated system capable of handling multiple target types: Protein Targets: For biomarker detection, assay development, and therapeutic blocking. We screen against purified recombinant proteins, complex protein mixtures,…
Aptamer Screening Services: Unlocking Precision with KMD Bioscience At KMD Bioscience, we are at the forefront of molecular innovation, providing cutting-edge aptamer screening services that empower researchers and industries to discover high-affinity, high-specificity nucleic acid ligands for their most challenging targets. What are Aptamers? Aptamers are single-stranded DNA or RNA oligonucleotides that bind to specific target molecules—from small ions and metabolites to proteins and whole cells—with antibody-like precision. Often termed "chemical antibodies," they offer unique advantages: smaller size, superior stability, minimal immunogenicity, and effortless chemical modification. Why Choose KMD Bioscience for Aptamer Development? Our state-of-the-art facility and expert team specialize in the Systematic Evolution of Ligands by Exponential Enrichment (SELEX) process, the gold standard for aptamer selection. We have refined this technology to deliver aptamers with exceptional performance for diagnostics, therapeutics, and targeted delivery applications. Our Comprehensive Service Portfolio Custom SELEX Screening: Target Flexibility: We work with diverse targets including purified proteins, peptides, small molecules, cells, and even complex structures. Advanced Library Design: Utilize naive or customized libraries for optimal starting diversity. Multiple SELEX Platforms: Choose from Magnetic Bead-based SELEX, Capillary Electrophoresis-SELEX (CE-SELEX) for superior stringency, or Cell-SELEX for live cell surface targets. High-Throughput Sequencing & Bioinformatics: Next-Generation Sequencing (NGS) to analyze selection rounds comprehensively. Advanced bioinformatics pipelines to identify enriched sequences, predict…
Aptamer Screening Services for Protein Targeting Precision Targeting, Unlocking New Dimensions in Protein Research In the fields of life science research and biopharmaceutical development, there is a growing demand for molecular tools with high affinity and specificity that target specific proteins. Leveraging our advanced SELEX (Systematic Evolution of Ligands by Exponential Enrichment) technology platform, KMD Bioscience provides professional and efficient aptamer screening services for protein targets, empowering your research to break through technical barriers. What are Aptamers? Aptamers are single-stranded DNA or RNA oligonucleotides obtained through in vitro screening techniques. They can bind to specific target molecules—including proteins, small molecules, and cells—with high affinity and specificity. Compared to antibodies, aptamers offer unique advantages such as small molecular weight, high stability, ease of chemical modification, no immunogenicity, and minimal batch-to-batch variation, earning them the title of "chemical antibodies." Our Technological Advantages 1. Advanced SELEX Technology Platform Multiple SELEX Variants: Including magnetic bead SELEX, capillary electrophoresis SELEX, cell-SELEX, and more, selecting the optimal screening strategy based on target characteristics. Next-Generation Sequencing (NGS) Support: Combined with high-throughput sequencing technology for in-depth analysis of screening libraries, significantly improving screening efficiency and success rates. Microfluidic Chip Technology: Enables ultra-low volume reaction systems, reducing sample consumption…
Excellent topic. Aptamer Screening refers to the process of identifying specific, high-affinity nucleic acid ligands (DNA or RNA aptamers) that bind to a target molecule of interest. It's often called SELEX (Systematic Evolution of Ligands by EXponential enrichment). Here’s a comprehensive breakdown of the screening process, its applications, and key considerations. 1. The Core Principle: SELEX SELEX is an iterative, in vitro combinatorial chemistry technique. The fundamental idea is to start with a vast, random library of nucleic acid sequences (up to 10^15 different molecules), expose them to the target, separate the binders from non-binders, amplify the binders, and repeat the cycle until a population of strong, specific binders is enriched. 2. General SELEX Workflow (Step-by-Step) A typical screening cycle involves: Step 1: Library Preparation A synthetic oligonucleotide library is created with a central random region (20-60 nucleotides) flanked by constant primer regions for PCR amplification. Library Diversity: Key to success. A 40-nucleotide random region represents ~10^24 possible sequences. Step 2: Incubation & Binding The library is incubated with the target molecule (protein, small molecule, cell, etc.). Conditions (buffer, temperature, ionic strength) are controlled to influence selection pressure. Step 3: Partitioning (The Most Critical Step) This step physically separates target-bound sequences from unbound ones. The…
In early drug discovery, hit identification is the disciplined search for molecules that measurably affect a biological target or disease-relevant system, while lead compound selection is the subsequent decision to elevate the best validated “hits” into lead compounds that are strong enough—scientifically and operationally—to justify an optimization campaign. This “hit-to-lead” logic sits between assay development/high-throughput screening and full lead optimization, and its quality strongly influences downstream success. 1) Core Definitions (so the team argues less) What is a “Hit”? A hit is an initial compound (or series) that shows reproducible activity in a primary screen and survives basic confirmation steps. Hits often begin with modest potency (commonly micromolar range) and uncertain mechanism until validated. What is a “Lead Compound”? A lead compound is a more mature chemical starting point: typically a hit-derived molecule (or series) with improved potency and enough evidence for selectivity, developability, and tractable chemistry to justify systematic optimization toward a clinical candidate. Lead optimization then focuses on balancing potency with ADMET (absorption, distribution, metabolism, excretion, toxicity) and related properties. 2) Why Hit Identification Is Harder Than “Finding Actives” Modern discovery can generate many actives quickly, but the bottleneck is identifying…
Epitope Mapping (also called antibody epitope mapping) is the set of experimental and computational approaches used to identify the precise antigen features an antibody recognizes and binds—down to specific amino acids, structural patches, or even interaction “hot spots.” In immunology terms, the epitope is the binding site on the antigen, while the antibody’s complementary binding surface is the paratope. Knowing exactly where binding occurs is foundational for understanding immune recognition, improving biologics, and designing better diagnostics and vaccines. Why Epitope Mapping Matters (Beyond “It Binds”) Antibodies can bind the same antigen in very different ways. Two antibodies may both “hit” the same protein yet differ dramatically in neutralization strength, cross-reactivity, or tolerance to mutations. Epitope mapping turns binding into actionable knowledge, helping teams: Differentiate antibodies that otherwise look similar by affinity alone (e.g., classifying binding regions and overlap patterns). Explain potency and mechanism of action, especially when blocking a receptor site or preventing conformational changes. Reduce off-target risk by detecting binding to conserved motifs shared across proteins. Guide design decisions for vaccines and diagnostics by focusing on minimal, protective, or assay-relevant epitopes. Two Big Epitope Types: Linear vs Conformational A key concept for practical…
Diagnostics increasingly relies on biomarkers—measurable molecular signals such as proteins, peptides, nucleic acids, metabolites, or enzymatic activities—that correlate with disease presence, stage, or treatment response. To read those signals reliably in real samples (blood, saliva, urine, tissue), modern assays need a recognition element that can find the target selectively, bind strongly enough, and produce a measurable output. Alongside antibodies and nucleic acids (aptamers), peptide probes have become a powerful option because they are chemically programmable, compatible with many detection platforms, and can be engineered for stability and surface attachment. This article explains how peptide probes are developed for biomarker detection, which design strategies are most common, and what technical pitfalls matter most in real diagnostic workflows. 1) What Is a “Peptide Probe” in Diagnostics? A peptide probe is a designed short amino-acid sequence that either: Binds a biomarker (affinity peptide / targeting peptide / peptide aptamer concept), or Responds to a biomarker-related activity (for example, a protease-cleavable peptide that changes signal after enzymatic cutting), or Acts as a capture element on a surface to pull a biomarker out of complex samples for readout. Compared with antibodies, peptides are usually easier to synthesize and modify (labels, linkers, anchors),…
Computational/AI-aided Peptide Screening (also called in silico peptide screening) is a modern discovery workflow that uses physics-based simulation, statistical learning, and deep learning to search large peptide sequence spaces for candidates likely to meet a target function—such as binding a protein pocket, disrupting an interface, penetrating cells, or achieving a desired bioactivity—while simultaneously filtering for “developability” (solubility, stability, toxicity, immunogenicity risk, and manufacturability). The core advantage is leverage: instead of testing millions of peptides experimentally, teams can prioritize a small, high-quality shortlist by combining virtual screening, ML prediction, and iterative optimization loops. 1) What “Peptide Screening” Means in the AI + Computational Era A peptide screening problem usually has one (or more) of these goals: Function-first screening: find sequences predicted to perform a biological function (e.g., antimicrobial, signaling, inhibitory, cell-penetrating). Target-first screening: find peptides predicted to bind a defined target (enzyme active site, receptor pocket, protein–protein interface). Property-first screening: find peptides with favorable developability characteristics, then verify function. Historically, wet-lab screening approaches (e.g., library panning) dominate discovery. Computational/AI-aided peptide screening complements these by (a) generating/curating large virtual libraries and (b) ranking them using scoring functions and predictive models before committing to experiments. 2) Data Foundations: Where “Learning” Comes…
SPOT Synthesis (often written as SPOT peptide synthesis or SPOT synthesis technique) is a positionally addressable, parallel solid-phase peptide synthesis method where many peptides are built simultaneously as discrete “spots” on a derivatized cellulose membrane. Instead of synthesizing one peptide at a time on resin beads, SPOT Synthesis dispenses activated amino acid solutions onto predefined membrane coordinates, enabling rapid generation of peptide libraries and arrays for downstream screening.  ⸻ What Makes SPOT Synthesis Unique? 1) Parallel synthesis on a planar cellulose support In SPOT Synthesis, the membrane acts as a flat solid support. Each printed droplet is absorbed into the porous cellulose and behaves like a tiny reaction “micro-compartment,” allowing hundreds to thousands of peptides to be synthesized in parallel on one sheet.  2) Addressable peptide libraries (arrays you can map by position) Every spot corresponds to a known sequence (or sequence mixture), which makes SPOT arrays especially useful when you need systematic coverage—such as scanning a protein sequence with overlapping peptides or exploring sequence–activity relationships.  3) Scale and throughput The method is widely described as supporting very high spot counts (from hundreds up to many thousands, depending on format and spot size). This density makes it…
1. Breakdown of Core Concepts XNA (Xeno Nucleic Acids): Refers to all nucleic acid analogs whose chemical structures differ from natural DNA and RNA. Common examples include: HNA (Hexitol Nucleic Acid), FANA (2'-Fluoro Arabino Nucleic Acid), LNA (Locked Nucleic Acid), CeNA (Cyclohexene Nucleic Acid), etc. Key Features of XNA: They typically exhibit greatly enhanced nuclease resistance (higher stability in biological fluids), higher thermal stability, and potentially a more diverse three-dimensional structural space, providing a foundation for discovering high-performance aptamers. Aptamer: A short, single-stranded DNA, RNA, or XNA oligonucleotide that can bind specifically and with high affinity to a target molecule (e.g., a protein, small molecule, cell). It can be considered a "chemical antibody." Functionally Enhanced: Here, it specifically refers to aptamers discovered using an XNA backbone, which inherently possess superior functional properties compared to natural nucleic acid aptamers, such as: Extremely high stability in vivo and in vitro (resistant to degradation). Stronger binding affinity and specificity. Broader tolerance to physicochemical conditions (e.g., pH, temperature range). Parallelized Library Screening: Refers to the use of high-throughput, automated experimental platforms (e.g., microfluidic chips, droplet microfluidics, next-generation sequencing-coupled techniques) to simultaneously screen an XNA random library containing an enormous number of sequences (typically 10^13 - 10^15). This dramatically accelerates the discovery process. 2. Overview of the Technical Workflow The entire discovery process is a…