Macromolecule Drugs Mass Spectrometry Characterization Service
- Structural characterization of lead candidates (e.g., modification sites, glycoform distribution)
- Supporting target validation and molecular optimization for accelerated clinical translation
- Monitoring structural stability during purification and formulation steps
- Assessing batch-to-batch consistency and scale-up process robustness
- Evaluating the impact of temperature, humidity, and excipients on protein conformation and physicochemical properties
- Supporting formulation development and shelf-life determination
- Multi-dimensional structural comparability assessments (e.g., primary sequence, glycosylation profile, higher-order conformation)
- Demonstrating equivalence with the originator biologic, streamlining regulatory approvals
Macromolecule drugs refer to biotherapeutic molecules with a molecular weight exceeding 1 kDa, including monoclonal antibodies (mAbs), bispecific antibodies, recombinant proteins, peptides, and antibody-drug conjugates (ADCs). Their key advantage lies in their high specificity and ability to precisely regulate disease-related biological pathways, leading to breakthrough treatments in oncology, autoimmune diseases, rare disorders, and infectious diseases. For example, PD-1/PD-L1 monoclonal antibodies have revolutionized cancer therapy by activating T-cell anti-tumor responses, while long-acting recombinant coagulation factors have significantly improved the quality of life for hemophilia patients.
However, macromolecule drugs are far more complex than small-molecule drugs—their therapeutic functionality depends on higher-order structural stability (e.g., 3D conformation, disulfide bond arrangement), post-translational modification (PTM) precision (e.g., glycosylation, oxidation, deamidation), and product homogeneity (e.g., aggregate control). Comprehensive analytical characterization is essential to ensure drug safety, efficacy, and manufacturability. While traditional analytical methods (e.g., HPLC, capillary electrophoresis, and spectroscopic techniques) provide insight into some physicochemical properties, they often lack sensitivity for detecting low-abundance structural variants.
Qu, M. et al. Mass Spectrom Rev. 2017.
Figure 1. General Workflow for LC/MS Quantification of Biotherapeutics
Mass spectrometry (MS), with its high resolution, accuracy, and throughput, has become a standard tool for structural and functional characterization of macromolecule drugs. It enables deep insights into:
1. Molecular Weight & Purity Analysis-Identifying potential aggregates and fragment impurities
2. Amino Acid Sequence Verification-Ensuring sequence integrity throughout the production process
3. Post-Translational Modifications (PTMs) Profiling-Mapping glycosylation sites, glycoform ratios, oxidation, and deamidation
4. Higher-Order Structural Analysis-Utilizing hydrogen-deuterium exchange (H/DX-MS) and ion mobility spectrometry (IMS) to assess conformational stability
5. Pharmacokinetics (PK) Studies-Accurately quantifying drug concentration and metabolic products in biological matrices
Comprehensive mass spectrometry-based characterization is fundamental for process development, stability evaluation, comparability assessment.
MtoZ Biolabs offers end-to-end Macromolecule Drugs Mass Spectrometry Characterization Service for pharmaceutical, biotech, and academic research organizations, covering quality characterization from early-stage R&D to commercial production. With state-of-the-art high-resolution mass spectrometry platforms (Orbitrap Fusion Lumos, FT-ICR MS) and expertise in method development, we are committed to:
Analysis Project |
Applications |
Intact Molecular Weight Analysis |
Verification of protein purity, detection of aggregates (≥2% sensitivity) |
Peptide Mapping |
Sequence confirmation, misassembly detection, and untranslated region identification |
Glycosylation Analysis and Glycoform Quantification |
Characterization of Fc-region G0F/G1F/G2F ratios, optimization of ADCC/CDC effects |
Oxidation/Deamidation Site Identification |
Assessment of drug degradation risks under long-term storage or light exposure conditions |
Antibody-Drug Conjugate (ADC) Characterization |
DAR value determination, linker stability analysis, and conjugation site identification |
Higher-Order Structure Analysis (H/DX-MS) |
Evaluation of the impact of lyophilization processes or excipients on protein conformational stability |
Analysis Workflow
1. Consultation & Custom Experimental Design
Our technical team tailors the study based on the drug type (antibody/recombinant protein/ADC) and the required analytical dimensions (primary structure/PTMs/higher-order conformation).
2. Sample Preparation
Charge reduction, desalting, and subunit separation are applied to enhance MS signal quality, while optimized ionization sources, fragmentation techniques, and resolution settings are selected based on drug type.
3. High-Resolution Mass Spectrometry Analysis
Integrating Top-Down (intact protein analysis), Middle-Down (subunit analysis), and Bottom-Up (peptide mapping) strategies for comprehensive characterization.
4. Quality Control & Data Analysis
Internal standards and triplicate validation (n ≥ 3) ensure data reliability, generating a detailed report with raw data, modification maps, and structural annotations.
Why Choose MtoZ Biolabs?
1. Industry-Leading Analytical Capabilities
Leveraging high-resolution mass spectrometry, we precisely characterize macromolecule drugs’ structural heterogeneity, detecting even trace modifications or low-abundance variants with exceptional accuracy.
2. Full Lifecycle Support
From lead candidate selection, process development, stability studies, to commercial QC, we provide a one-stop solution that aligns with Quality by Design (QbD) principles.
3. Agile & Flexible Services
We offer rapid turnaround from consultation to data delivery, with customizable analytical strategies to meet urgent project timelines.
4. Transparent Pricing
We follow a One-Time-Charge model, ensuring clear, upfront pricing with no additional or hidden costs.
Applications
1. Biologic Drug Discovery & Development
2. Manufacturing Process Optimization & Quality Control
3. Stability & Storage Studies
4. Biosimilar Comparability Studies
Case Study
1. Rapid characterization of secreted recombinant proteins by native mass spectrometry
The quality assessment of recombinant proteins is essential for optimizing their design and production. However, traditional methods rely on labor-intensive purification processes, which are time-consuming and costly. This study introduces a native mass spectrometry (Native MS) approach that enables direct analysis of overexpressed proteins from culture media, eliminating the need for conventional purification steps. This method allows for the rapid characterization of solubility, molecular weight, folding state, assembly state, higher-order structure, post-translational modifications (PTMs), and biomolecular interactions. The study demonstrates its applicability to yeast, insect, and human cell expression systems and highlights its potential for high-throughput analysis, protein engineering, and mutant screening. Compared to traditional approaches, this method significantly reduces the time gap between protein production and characterization, facilitating improved protein quality and yield optimization. Macromolecule Drugs Mass Spectrometry Characterization Service utilizes high-resolution mass spectrometry, this service provides rapid and precise recombinant protein characterization, enabling the direct analysis of molecular weight, structure, post-translational modifications, and protein complex assembly. It supports researchers in optimizing protein expression systems, enhancing protein quality, and improving production efficiency.
Ben-Nissan, G. et al. Commun Biol. 2018.
Figure 2. Rapid Characterization of Recombinant Antibodies
2. Characterization of Classical Vaccines by Charge Detection Mass Spectrometry
Vaccines present antigens through different platforms (such as protein subunits or whole viruses) to induce immune responses, but due to the vast size differences of antigens, their analysis methods often rely on the specific platform. This study uses Charge Detection Mass Spectrometry (CDMS) to characterize three classic multivalent vaccines—the inactivated poliovirus vaccine (IPOL), rotavirus vaccine (RotaTeq), and nine-valent HPV vaccine (Gardasil-9). CDMS simultaneously measures the mass-to-charge ratio (m/z) and charge of individual ions, providing an accurate analysis of the mass distribution of large, heterogeneous samples. The results show that this method can directly assess the antigen purity in vaccine samples, revealing empty virus particles in whole virus vaccines. Additionally, CDMS can evaluate the formaldehyde crosslinking degree in IPOL, detect slightly lower mass in RotaTeq than theoretical values, indicating missing modifications in some pentamers. Gardasil-9 exhibited a broader mass distribution after buffer exchange, and a clearer spectrum peak was obtained after removing the aluminum adjuvant through exchange reactions. Based on high-resolution mass spectrometry, macromolecule drugs mass spectrometry characterization service offers precise characterization of vaccines and macromolecule drugs, allowing for the direct measurement of antigen purity, virus-like particle integrity, crosslink modifications, and high molecular weight distribution, providing critical data support for vaccine research and quality control.
Miller, LM. et al. Anal Chem. 2021.
Figure 3. CDMS Analysis of IPOL Vaccine
The development of macromolecule drugs is a pursuit of structural precision—every glycan branch, disulfide bond, and conformational shift can impact efficacy and safety. MtoZ Biolabs' Macromolecule Drugs Mass Spectrometry Characterization Service leverage high-resolution technologies to unveil molecular details, full-cycle analytical support to empower decision-making, and regulatory-grade data to accelerate drug approvals. Whether you're at the lead candidate selection, process development, or commercial QC stage, we provide scientific rigor and cutting-edge expertise to bring your molecular blueprint into therapeutic reality.
Contact MtoZ Biolabs today to unlock a new dimension in mass spectrometry-driven biologics characterization!
MtoZ Biolabs, an integrated chromatography and mass spectrometry (MS) services provider.
Related Services
How to order?