Altmetrics
Downloads
414
Views
145
Comments
0
A peer-reviewed article of this preprint also exists.
This version is not peer-reviewed
Submitted:
04 September 2023
Posted:
06 September 2023
You are already at the latest version
Technologies | |
---|---|
Technology | Application |
Mass Spectrometry (MS) | Characterization and post-translational modification analysis[54]. |
Surface Plasmon Resonance (SPR) | Protein interaction studies[55] |
Capillary Electrophoresis (CE) | Biosimilar comparability studies.[56] |
Two-Dimensional Gel Electrophoresis | Separation and identification of proteins [57] |
Enzyme-Linked Immunosorbent Assay (ELISA) | Specific protein quantification and immunogenicity studies.[58] |
X-ray Crystallography and Nuclear Magnetic Resonance (NMR) Spectroscopy | Structural analysis and 3D modeling.[59] |
Protein Microarrays | High-throughput analysis of protein functions and interactions.[60] |
Circular Dichroism | In vivo and in vitro stability analysis -[61]. |
Immunoassays | Pharmacokinetic and pharmacodynamic studies.[62] |
Differential Scanning Calorimetry | Thermal stability analysis.[63] |
Stable Isotope Labeling by Amino acids in Cell culture | Quantitative proteomics for expression analysis.[64] |
Yeast Two-Hybrid System | Protein-protein interaction mapping.[65] |
Liquid Chromatography-Mass Spectrometry | Comprehensive protein characterization [66] |
Hydrogen-Deuterium Exchange Mass Spectrometry | Conformational dynamics and higher-order structure analysis.[67] |
Multi-Angle Light Scattering (MALS) | Molar mass and size distribution.[68] |
Size-Exclusion Chromatography | Protein aggregation and purity assessment.[69] |
Matrix-Assisted Laser Desorption/Ionization Time-of-Flight MS | Rapid identification and characterization of proteins [70] |
Isoelectric Focusing (IEF) | Protein separation based on isoelectric point -[71]. |
Reversed-Phase High-Performance Liquid Chromatography | Analysis of Protein purity and heterogeneity [72] |
Chemical Cross-Linking Coupled with MS | Studying spatial arrangement and interactions within protein complexes.[73] |
Fourier Transform Infrared Spectroscopy (FTIR) | Secondary structure analysis and stability monitoring.[74] |
Flow Cytometry | Cell line development and monitoring of protein expression.[75] |
Biacore (SPR-based technology) | Label-free interaction analysis.[76] |
Selected Reaction Monitoring MS | Targeted protein quantification in biosimilar development.[77] |
Fluorescence Spectroscopy | Folding and conformational analysis.[78] |
Hydrophobic Interaction Chromatography (HIC) | Analysis of hydrophobicity and aggregation.[79] |
Ion Exchange Chromatography | Charge heterogeneity analysis.[80] |
Native MS | Structural characterization and complex formation analysis.[81] |
Affinity Chromatography | Purification and target binding analysis. |
N-Terminal Sequencing | Analysis of protein sequence and modifications.[82] |
Dynamic Light Scattering (DLS) | Size and stability analysis.[83] |
Peptide Mapping and Fingerprinting | Identification and characterization of proteins.[84] |
Immunoprecipitation and Pull-Down Assays | Protein interaction studies.[85] |
Chromatography Coupled with Multi-Angle Light Scattering | Absolute molar mass, size, and conformation.[86] |
NMR Spectroscopy in Conjunction with Hydrogen Exchange | Conformational dynamics and structural analysis.[87] |
Examples of Use of Biomarkers | |
Antibody Drug Conjugates (ADCs) | Brentuximab vedotin, an ADC used for Hodgkin's lymphoma and systemic anaplastic large cell lymphoma, delivers the cytotoxic drug monomethyl auristatin E (MMAE) to CD30-expressing cells, and the measurement of MMAE can serve as a marker of target engagement[88]. |
Antigen-antibody complex | The formation of antigen-antibody complexes provides direct evidence of target engagement. For example, in the case of adalimumab, an anti-tumor necrosis factor (TNF)-α antibody, the serum levels of the adalimumab-TNFα complex can be measured as evidence of the drug binding to its target[89]. |
Antigenic Modulation | This refers to the downregulation or loss of antigen expression on the cell surface in response to antibody binding and can be used as a marker of monoclonal antibody (mAb) engagement. Rituximab, a monoclonal antibody against the CD20 antigen on B cells, causes antigenic modulation, decreasing CD20 expression and indicating rituximab engagement[90]. |
Binding of mAbs to Fc Receptors | The Fc region of mAbs can bind to Fc receptors on immune cells. This binding can modulate the activity of these cells, making Fc receptor occupancy a valuable PD marker. The occupancy of RIIIa on natural killer cells by rituximab can be used as a PD marker[91]. |
Cell Proliferation Markers | mAbs may also be designed to inhibit cell proliferation. Here, decreased cell proliferation markers, such as Ki-67, can indicate successful target engagement[92]. |
Circulating Tumor Antigen Levels | In cancer therapy, mAbs are often designed for binding to specific tumor antigens. Reduction in the levels of these circulating antigens following mAb therapy can serve as a marker of target engagement. For instance, CA-125 levels in patients with ovarian cancer have been treated with mAbs targeting the CA-125 antigen[93]. |
Complement System Alterations | mAbs can modulate the complement system. Eculizumab, a mAb that inhibits complement component C5, reduces hemolytic activity and can be used as a PD marker[94]. |
Cytokine Release Syndrome | mAbs, particularly those targeting immune cells, increase the release of specific cytokines. For instance, administration of the anti-CD28 mAb TGN1412 releases many cytokines, such as interleukin (IL)-2 and interferon (IFN)-γ, which could be monitored as PD markers[95]. Measuring cytokines, such as IL-2, IL-6, or TNF- α, can estimate target engagement. This is particularly relevant for immunomodulatory mAbs such as ipilimumab, which can increase circulating cytokine levels upon engagement with its target, cytotoxic T lymphocyte-associated (CTLA-4)[96]. |
Fluorescent Tag | Flow cytometry can be a valuable tool for assessing target engagement when the target of a mAb is expressed on cell surfaces. Labeling the mAb with a fluorescent tag confirms its binding to the target cells in a sample. This has been utilized in therapies, such as those using rituximab, wherein binding to CD20+ B cells can be confirmed using flow cytometry[97]. |
Gut Microbiota Alterations | Specific mAb therapies can alter gut microbiota, serving as functional response markers. Vedolizumab, a mAb against the α4β7 integrin used in treating inflammatory bowel disease, can restore gut microbial diversity, indicating a functional response to therapy[98]. |
Immune Response Markers | Some mAbs stimulate immune responses against specific antigens. Hence, increased antibodies against the target antigen in the patient's serum can serve as a target engagement marker. For instance, palivizumab, a mAb that prevents respiratory syncytial virus (RSV) infection in high-risk infants, engages its target through anti-RSV antibodies in the patient's serum[99]. Immune response can be measured as a functional response marker. For instance, ipilimumab, a mAb that targets the immune checkpoint protein CTLA-4, is the most widely used mAb. |
Omics Technology | Role | Rationale |
Proteomics[205] | Determine the protein expression profile, post-translational modifications (like glycosylation), and protein-protein interactions of the biosimilar compared to the reference product. | Minor differences in protein structure or modifications can impact the efficacy. |
Transcriptomics | Analyze the gene expression profile of cells producing the biosimilar, ensuring that the cellular machinery has the therapeutic protein in a manner consistent with the reference product. | Differences in gene expression may hint at differences in protein product production, folding, or modification. |
Metabolomics[206] | Examine the metabolic profile of the biosimilar-producing cells. | The metabolic state of a cell can influence the final product's quality and consistency. For instance, changes in nutrient levels can influence glycosylation patterns of proteins. |
Genomics | Ensures genetic stability of the cell line producing the biosimilar. | Over time, cell lines might undergo genetic drift, which can impact the product's quality, consistency, and efficacy. |
Microbiomics | Understanding the microbiome can be essential if the biological product has a microbial origin (like some recombinant proteins produced in bacteria). | Microbial contaminants or shifts in the microbial population can influence the final product's quality and safety. |
Phosphoproteomics | Analyze phosphorylation patterns on proteins, which can be critical for some biologics' function or stability. | Changes in phosphorylation can affect protein activity, stability, or interaction with other proteins. |
mAb (Brand) | Receptor |
Abciximab (ReoPro)[210] | GPIIb/IIIa |
Adalimumab (Humira)[211] | TNFα |
Alemtuzumab (Lemtrada)[212] | CD52 |
Atezolizumab (Tecentriq)[213] | PD-L1 |
Basiliximab (Simulect)[214] | CD25 |
Belimumab (Benlysta)[215] | BLyS |
Bevacizumab (Avastin)[216] | VEGF |
Cetuximab (Erbitux)[217] | EGFR |
Daclizumab (Zinbryta)[218] | CD25 |
Daratumumab (Darzalex)[219] | CD38 |
Denosumab (Prolia)[220] | RANKL |
Dupilumab (Dupixent)[221] | IL-4Rα |
Eculizumab (Soliris)[222] | C5 |
Infliximab (Remicade)[223] | TNFα |
Ipilimumab (Yervoy)[224] | CTLA-4 |
Nivolumab (Opdivo)[225] | PD-1 |
Obinutuzumab (Gazyva)[226] | CD20 |
Ofatumumab (Arzerra)[227] | CD20 |
Omalizumab (Xolair)[228] | IgE |
Palivizumab (Synagis)[229] | RSV F protein |
Pembrolizumab (Keytruda)[230] | PD-1 |
Rituximab (Rituxan)[231] | CD20 |
Sarilumab (Kevzara)[232] | IL-6R |
Secukinumab (Cosentyx)[233] | IL-17A |
Tocilizumab (Actemra)[234] | IL-6R |
Trastuzumab (Herceptin)[235] | HER2/neu |
Vedolizumab (Entyvio)[236] | α4β7 integrin |
Type | Count |
Fab | 1 |
Toxin | 1 |
Carrier Protein | 1 |
Single-Domain Antibody | 1 |
Fusion Proteins | 1 |
Bispecific Antibody | 3 |
Coagulation Factor | 4 |
Cytokine | 4 |
Peptide | 4 |
Growth Factor | 4 |
Enzyme | 9 |
Enzyme Inhibitor | 11 |
Hormone | 11 |
Monoclonal Antibody Conjugate | 13 |
Monoclonal Antibody | 96 |
Total | 164 |
Mode of Action | Biomarker Potential |
AMPK and mTORC1 Signaling | Monitoring these central energy sensors and regulators can be vital for drugs targeting cellular energy status or metabolic health.[257] |
Angiogenesis Indicators | If a protein drug affects blood vessel formation, angiogenic factors like VEGF can be used as biomarkers.[258] |
Apoptosis Markers | Evaluation of cell death can be instrumental for drugs designed to induce or inhibit apoptosis. Markers such as caspase activation or phosphatidylserine externalization can be employed.[259] |
Autophagy Markers | LC3-II and p62/SQSTM1, for drugs modulating autophagic activity.[260] |
Autophagy-lysosomal Pathway Markers | Monitoring markers like p62/SQSTM1 or LAMP1 can give insights into the autophagy-lysosomal activity upon drug treatment.[261] |
Blood Coagulation Factors | For protein drugs affecting hemostasis, measurement of specific clotting factors or clotting times might be used.[262] |
Bone Turnover Markers | For protein drugs acting on the skeletal system, bone resorption or formation markers can provide insight into their effect.[263] |
Calcium Signaling | Monitoring intracellular calcium flux and associated proteins can be important for drugs that modulate calcium homeostasis or signaling pathways.[264] |
Cell Cycle Regulators | Drugs aiming at modulating the cell cycle might alter levels or activities of cyclins, cyclin-dependent kinases, or associated inhibitors.[265] |
Cell Metabolism | Assessing the metabolic profile of cells or tissues after drug treatment, for instance, glucose uptake, lactate production, or ATP levels.[266] |
Cell Surface Markers | These markers can be evaluated for drugs targeting cell surface proteins or for those that induce phenotypic changes in cells.[267] |
Cellular Apoptosis or Proliferation | Some protein drugs may induce or inhibit apoptosis or cell proliferation, which can be quantified.[268] |
Cellular Signaling Pathways | Assessment of downstream or upstream signaling pathways that might be affected by the protein drug. MAPK, PI3K/AKT, or JAK/STAT pathways.[269] |
Changes in specific cell populations | Especially in immunology, a protein drug can lead to the proliferation or reduction of specific cell populations.[270] |
Circadian Rhythms | For protein drugs affecting cellular or physiological rhythms, markers related to circadian clock genes such as PER, CRY, or CLOCK might be relevant.[271] |
Complement Activation | For specific therapeutic proteins, activation or inhibition of the complement system can serve as a pharmacodynamic readout.[272] |
Cytokine Levels | Many protein drugs target specific cytokines or have effects on cytokine levels.[273] |
DNA Damage and Repair Markers | γH2AX and other proteins associated with DNA damage response can be relevant for drugs targeting genomic stability.[274] |
Drug Concentration | Although this is more of a pharmacokinetic parameter, the concentration of the drug in the bloodstream can sometimes serve as a surrogate for its pharmacodynamic effects, significantly when the concentration closely correlates with the drug's effect.[275] |
Endocannabinoid System Markers | Components include CB1 and CB2 receptors or endocannabinoids (anandamide, 2-AG) for drugs affecting this system.[276] |
Endocrine Biomarkers | For protein drugs affecting the endocrine system, hormones or hormone precursors might be potential pharmacodynamic indicators.[277] |
Endocytosis and Exocytosis Metrics | Protein drugs targeting cell trafficking mechanisms might alter the rates of endocytosis or exocytosis, which can be tracked using various cellular assays.[278] |
Endogenous Antioxidant Enzymes | Superoxide dismutase (SOD), catalase, and glutathione peroxidase levels can be tracked for oxidative stress modulation.[279] |
Endoplasmic Reticulum (ER) Stress Markers | GRP78/BiP, CHOP, XBP1, ATF6 for drugs influencing ER homeostasis or targeting diseases related to protein misfolding.[280] |
Endosome Trafficking | Protein drugs that interfere with endosomal pathways can be monitored for their effects using markers of early, late, and recycling endosomes.[281] |
Endothelial Activation Markers | For drugs impacting vascular inflammation or barrier function, such as E-selectin, ICAM-1, and VCAM-1.[282] |
Enzyme Activity | If the protein drug targets an enzyme, measuring the change in enzyme activity can be an effective biomarker.[283] |
Epigenetic Markers | Changes in DNA methylation, histone modification, or other epigenetic markers might indicate a response to certain protein drugs.[284] |
Exosome Release and Composition | Certain protein drugs can influence exosomes and their cargo (RNA, protein, lipids), especially those impacting intercellular communication.[285] |
Extracellular Matrix (ECM) Components | Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) are relevant for tissue remodeling or cancer invasion.[286] |
Fatty Acid Oxidation (FAO) Rates | Drugs targeting metabolic states might shift cells between carbohydrate and fatty acid metabolism.[287] |
Flow Cytometry | This is particularly relevant for drugs that target cells of the immune system. Flow cytometry can provide insights into cell numbers, phenotypes, and functions.[288] |
Functional Assays | Depending on the intended drug action, functional assays can be developed. For instance, if a protein drug aims to inhibit a specific cellular function, assays can be set up to measure that specific function.[289] |
Gene Expression Profiles | Transcriptomics can reveal the downstream effects of a protein drug on cellular gene expression.[290] |
Glycolytic versus Oxidative Metabolism | Assessing the switch between glycolytic and oxidative metabolism can be crucial for drugs targeting metabolic diseases or cancer.[291] |
Glycosylation Patterns | Alterations in the glycosylation patterns of cells or proteins can directly or indirectly affect some protein drugs.[292] |
Gut Microbiota Composition | Sequencing or metabolomic profiles of gut bacteria can be helpful for drugs impacting the gut environment.[293] |
Heat Shock Proteins (HSPs) | As molecular chaperones, changes in HSP levels can indicate cellular stress responses or protein homeostasis disruptions.[294] |
Heat Shock Proteins (HSPs) | These proteins respond to cellular stress and can be targets or indicators for several drugs, especially in protein misfolding diseases.[295] |
Histone Modifications | Epigenetic changes, like histone acetylation or methylation, can be markers for drugs targeting chromatin remodeling or gene expression.[296] |
Hormone Levels | Assessment of specific hormone levels, like insulin, glucagon, or thyroid hormones, can indicate drug impact on endocrine systems.[297] |
Hypoxia Indicators | For protein drugs affecting cellular responses to oxygen deprivation, markers like HIF-1α can be interesting.[298] |
Imaging Biomarkers | Techniques like MRI, PET, and CT can be used to measure the effects of protein drugs at the tissue or organ level.[299] |
Immune Response Markers | The immune system might mount an antibody response for protein drugs, especially those foreign to the human body. Monitoring anti-drug antibodies (ADAs) can be a biomarker for potential immunogenicity issues.[300] |
Inflammasome Activation | Monitoring inflammasome components can be helpful in drugs targeting inflammatory conditions or diseases like Alzheimer's.[301] |
Ion Channel Activity | For protein drugs targeting ion channels, the measurement of ion flux or electrical properties of cells could directly indicate drug action.[302] |
Iron Metabolism Markers | Ferritin, transferrin, and hepcidin for drugs modulating iron homeostasis.[303] |
Levels of circulating drug target | If the target of the protein drug circulates in the bloodstream (like a soluble receptor or ligand), measuring its levels can serve as a biomarker.[304] |
Ligand-Receptor Interactions | Investigating a protein drug's binding dynamics and affinity to its target receptor can provide insights into its effectiveness.[305] |
Lipidomic Profile | Analyzing the cellular lipid composition can be informative, especially for drugs impacting lipid metabolism or signaling.[306] |
Lipophagy Markers | Indicators of lipid droplet autophagy crucial for lipid metabolism-related conditions.[307] |
Lysosomal Enzymes | The levels and activity of the specific lysosomal enzymes can be essential biomarkers for enzyme replacement therapies in lysosomal storage disorders.[308] |
Markers of Fibrosis | In conditions like liver or lung fibrosis, protein drugs might target fibrogenesis, and thus, markers such as tissue collagen or specific matrix proteins can serve as indicators.[309] |
Metabolic Enzymes | Monitoring the levels or activities of critical metabolic enzymes, such as those involved in glycolysis or the TCA cycle, can provide insights into the metabolic state of cells upon drug treatment.[310] |
MicroRNAs (miRNAs) | Changes in the expression of specific miRNAs can serve as biomarkers since they play pivotal roles in gene regulation and might be influenced by protein drugs.[311] |
Mitochondrial Dynamics | Assessing mitochondrial morphology and dynamics can indicate cellular health and metabolism, especially for drugs targeting these organelles.[312] |
Mitophagy Indicators | Monitoring mitophagy, a process to degrade damaged mitochondria, can be helpful in drugs targeting cellular health.[313] |
mRNA Splicing Markers | Such as components of the spliceosome for drugs modulating RNA splicing or targeting splicing-related diseases.[314] |
mTOR Signaling | The mechanistic target of the rapamycin (mTOR) pathway, central to cell growth and metabolism, might be affected by certain protein drugs. Monitoring components like p70S6 kinase or 4E-BP1 can be informative.[315] |
Myelination Markers | Proteins like MBP or PLP can be tracked for drugs targeting neurodegenerative diseases or demyelinating conditions.[316] |
NAD+/NADH Ratio | A marker for cellular redox state and metabolism, especially relevant for aging |
Neural Activity Markers | c-Fos, Arc, or immediate early genes can be indicators of neural activity and synaptic plasticity.[317] |
Neurotransmitter Levels | If the drug has a neurological target, neurotransmitter levels in the central nervous system or peripheral tissues can be evaluated.[318] |
Neurotrophic Factors | In neurodegenerative diseases, protein drugs might aim to modulate the levels of neurotrophic factors like BDNF, NGF, or GDNF.[319] |
NO (Nitric Oxide) Production | Relevant for cardiovascular or immunomodulatory drugs, NO levels can indicate endothelial function and inflammatory states.[320] |
Nrf2-Keap1 Pathway | Tracking the Nrf2-Keap1 pathway components can be vital for drugs that modulate oxidative stress.[321] |
Nucleotide Metabolites | Monitoring the levels of specific nucleotide metabolites can indicate cellular activity or stress in response to certain protein drugs.[322] |
Oxidative Phosphorylation (OXPHOS) Metrics | For protein drugs targeting mitochondrial function, OXPHOS or mitochondrial health markers can be relevant.[323] |
Oxidative Stress Markers | Oxidative stress plays a role in numerous diseases, and markers like reactive oxygen species (ROS) or antioxidant levels can be used to assess drug effects.[324] |
Oxysterols | Such as 24(S)-hydroxycholesterol 27-hydroxycholesterol for drugs targeting cholesterol metabolism or diseases like Niemann-Pick type C.[325] |
Peroxisome Proliferators-Activated Receptors (PPARs) | As metabolic regulators, PPARs can be markers for drugs impacting lipid metabolism or inflammation.[326] |
Pharmacogenomic Biomarkers | Some patients might respond differently to protein drugs based on genetic variations. Exploring these can provide insights into efficacy and safety.[327] |
Phosphorylation status of proteins | The activation or deactivation of specific signaling pathways can be tracked by looking at the phosphorylation status of essential proteins.[328] |
Proteasome Activity | Assessing proteasomal activity can be insightful for protein drugs that modulate protein degradation.[329] |
Proteomic Analysis | To assess broader proteome for changes in protein levels or post-translational modifications upon drug treatment.[330] |
Reactive Oxygen Species (ROS) Levels | As an indicator of oxidative stress, ROS can be monitored for drugs that either induce or counteract cellular stress.[331] |
Receptor Occupancy | Measuring the degree to which a protein drug binds to its target receptor can be a direct biomarker of its activity.[332] |
Senescence-associated Secretory Phenotype (SASP) Factors | Monitoring factors associated with cellular senescence might be relevant for drugs targeting aging or oncogenesis.[333] |
Sirtuin Activity | Monitoring sirtuin proteins can be essential for drugs modulating cellular longevity or metabolic health.[334] |
Telomerase Activity | Telomerase activity or telomere length might be relevant biomarkers for drugs targeting cancer or aging processes.[335] |
Tight Junction Proteins | Markers like claudins, occludin, and ZO-1 are relevant for drugs targeting barrier integrity, such as in gut or blood-brain barrier conditions.[336] |
Tissue Repair and Regeneration Markers | For protein drugs that facilitate tissue healing, markers of tissue repair or stem cell activation might be relevant.[337] |
Tumor Microenvironment Components | Factors like TGF-beta, PD-L1, and various cytokines/chemokines for drugs targeting cancer immune evasion.[338] |
Unfolded Protein Response (UPR) in the ER | For protein drugs that might induce ER stress, tracking components of the UPR can be informative.[339] |
Wnt Signaling Pathway Components | Tracking this pathway can be crucial for drugs that modulate developmental processes, tissue regeneration, or certain cancers.[340] |
No | Protein Drug | Pharmacodynamic Marker |
1. | Abatacept (Orencia) | T cell proliferation & co-stimulation[341] |
2. | Adalimumab (Humira) | TNF-alpha levels & inflammatory cytokine reduction[342] |
3. | Aducanumab (Aduhelm) | Beta-amyloid plaques in brain[343] |
4. | Aflibercept (Eylea) | VEGF levels, Central retinal thickness[344] |
5. | Agalsidase Alfa (Replagal) | Lyso-Gb3 levels, Kidney function[345] |
6. | Agalsidase Beta (Fabrazyme) | Lyso-Gb3 levels, Kidney function[346] |
7. | Albiglutide (Tanzeum) | Blood glucose & GLP-1 levels[347] |
8. | Albutrepenonacog Alfa (Idelvion) | Factor IX activity levels[348] |
9. | Aldesleukin (Proleukin) | T cell count, IL-2 levels[349] |
10. | Alefacept (Amevive) | CD4 and CD8 memory T-cell count[350] |
11. | Alemtuzumab (Lemtrada) | CD52-expressing cell count[351] |
12. | Alglucerase (Ceredase) | Gaucher cell count, Chitotriosidase levels [352] |
13. | Alirocumab (Praluent) | LDL cholesterol levels[353] |
14. | Alpha-1-proteinase inhibitor (Prolastin, etc.) | Alpha-1 antitrypsin levels, Neutrophil elastase activity[354] |
15. | Alteplase (Activase) | Fibrinolytic activity, clot dissolution[355] |
16. | Amivantamab (Rybrevant) | EGFR and MET signaling inhibition[356] |
17. | Anakinra (Kineret) | IL-1β levels[357] |
18. | Ancestim (Stemgen) | CD34+ cell count in peripheral blood[358] |
19. | Andexanet Alfa (Andexxa) | Reversal of factor Xa inhibitors[359] |
20. | Anifrolumab (Saphnelo) | Type I interferon gene signature[360] |
21. | Anistreplase (Eminase) | Fibrinolytic activity[361] |
22. | Ansuvimab (Ebanga) | Reduction in viral load of Ebola virus[362] |
23. | Atezolizumab (Tecentriq) | PD-L1 expression on tumor & immune cells [363] |
24. | Avelumab (Bavencio) | PD-L1 expression & T cell activation [364] |
25. | Benralizumab (Fasenra) | Reduction in eosinophil counts[365] |
26. | Bermekimab (Xilonix) | IL-1α levels[366] |
27. | Bevacizumab (Avastin) | VEGF level & microvessel density[367] |
28. | Bezlotoxumab (Zinplava) | Reduction in C. difficile infection recurrence[368] |
29. | Bimekizumab (Bimzelx) | IL-17A and IL-17F levels[369] |
30. | Bivalirudin (Angiomax) | Thrombin activity[370] |
31. | Blinatumomab (Blincyto) | CD19+ B cell count[371] |
32. | Bone Morphogenetic Proteins (BMPs) | Bone density or new bone formation[372] |
33. | Botulinum Toxin Type A (Botox) | Neuromuscular transmission inhibition[373] |
34. | Botulinum Toxin Type B (Myobloc) | Neuromuscular transmission inhibition[374] |
35. | Brodalumab (Siliq) | IL-17 receptor A occupancy [375] |
36. | Brolucizumab (Beovu) | VEGF levels, Central retinal thickness[376] |
37. | Burosumab (Crysvita) | Serum phosphorus levels[377] |
38. | Calaspargase pegol (Asparlas) | Asparagine levels[378] |
39. | Canakinumab (Ilaris) | IL-1β levels & CRP[379] |
40. | Cetuximab (Erbitux) | EGFR expression & phosphorylation[380] |
41. | Chymopapain (Chymodiactin) | Disc volume reduction[381] |
42. | Coagulation factor IX (BeneFIX) | Factor IX clotting activity[382] |
43. | Coagulation Factor VIIa (NovoSeven) | Clotting activity[383] |
44. | Collagenase (Santyl) | Degrades necrotic tissue[384] |
45. | Conestat alfa (Ruconest) | Bradykinin levels[385] |
46. | Corticotropin (Acthar) | Adrenal gland stimulation[386] |
47. | Cosyntropin-ACTH(1-24) (Cortrosyn) | Adrenal gland stimulation[387] |
48. | Crizanlizumab (Adakveo) | P-selectin inhibition[388] |
49. | Darbepoetin alfa (Aranesp) | Hemoglobin or hematocrit level[389] |
50. | Denosumab (Prolia, Xgeva) | RANKL inhibition & bone turnover markers [390] |
51. | Denosumab (Prolia) | Bone mineral density & serum C-telopeptide[391] |
52. | Dupilumab (Dupixent) | IL-4 and IL-13 signaling pathways [392] |
53. | Durvalumab (Imfinzi) | PD-L1 expression in tumor cells[393] |
54. | Eculizumab (Soliris) | Complement component C5 activity [394] |
55. | Edrecolomab (Panorex) | EpCAM expression[395] |
56. | Efalizumab (Raptiva) | CD11a expression[396] |
57. | Efgartigimod alfa | IgG reduction[397] |
58. | Elapegademase (Revcovi) | ADA enzyme activity[398] |
59. | Elosulfase Alfa (Vimizim) | GAG reduction[399] |
60. | Elotuzumab (Empliciti) | SLAMF7 expression in myeloma cells[400] |
61. | Emapalumab (Gamifant) | IFNγ levels[401] |
62. | Emicizumab (Hemlibra) | Factor IXa and factor X bridging[402] |
63. | Enfortumab vedotin (Padcev) | Nectin-4 expression[403] |
64. | Erenumab (Aimovig) | CGRP receptor binding and inhibition [404] |
65. | Erythropoietin (EPO) | Hemoglobin or hematocrit level[405] |
66. | Eteplirsen (Exondys 51) | Dystrophin production in muscle tissue[406] |
67. | Evolocumab (Repatha) | LDL cholesterol levels [407] |
68. | Fibrinolysin (Elase) | Fibrin degradation[408] |
69. | Filgrastim (Neupogen) | Neutrophil count[409] |
70. | Follitropin (Follistim, Gonal-f) | Follicular development, Estradiol levels[410] |
71. | Fremanezumab (Ajovy) | CGRP levels[411] |
72. | Galcanezumab (Emgality) | CGRP levels[412] |
73. | Galcanezumab (Emgality) | CGRP binding[413] |
74. | Galsulfase (Naglazyme) | Urinary glycosaminoglycan levels[414] |
75. | Gemtuzumab ozogamicin (Mylotarg) | CD33 antigen expression[415] |
76. | Girentuximab | CAIX expression[416] |
77. | Glatiramer acetate (Copaxone) | Immune modulation; T-cell response[417] |
78. | Glucagon recombinant (GlucaGen) | Blood glucose elevation[418] |
79. | Glucarpidase (Voraxaze) | Methotrexate levels reduction[419] |
80. | Golimumab (Simponi) | TNFα inhibition[420] |
81. | Growth Hormone | IGF-1 (Insulin-like Growth Factor 1) level[421] |
82. | Guselkumab (Tremfya) | IL-23 levels & PASI score [422] |
83. | Human C1-esterase inhibitor (Berinert, Cinryze) | C1-INH levels and activity[423] |
84. | Ibalizumab (Trogarzo) | HIV-1 viral load & CD4+ T-cell count [424] |
85. | Imiglucerase (Cerezyme) | Glucocerebroside levels, macrophage activity[425] |
86. | Inebilizumab (Uplizna) | B-cell depletion[426] |
87. | Infliximab (Remicade) | TNF-alpha levels & CRP[427] |
88. | Inotuzumab ozogamicin (Besponsa) | CD22 expression[428] |
89. | Insulin Regular (Humulin R, etc) | Glucose levels[429] |
90. | Interferons | Expression of interferon-responsive genes[430] |
91. | Ipilimumab (Yervoy) | T-cell activation[431] |
92. | Isatuximab (Sarclisa) | CD38 expression[432] |
93. | Itolizumab (Alzumab) | CD6 expression[433] |
94. | Ixekizumab (Taltz) | PASI score, serum IL-17A levels[434] |
95. | Lanadelumab (Takhzyro) | Plasma kallikrein activity[435] |
96. | Lanadelumab (Takhzyro) | Plasma kallikrein inhibition[436] |
97. | Laronidase (Aldurazyme) | Reduction of glycosaminoglycans[437] |
98. | Lepirudin (Refludan) | Inhibition of thrombin[438] |
99. | Leuprolide (Lupron) | Reduction in testosterone or estradiol[439] |
100. | Liraglutide (Victoza) | Blood glucose & GLP-1 levels[440] |
101. | Lixisenatide (Adlyxin) | GLP-1 receptor activation |
102. | Loncastuximab tesirine (Zynlonta) | CD19 expression[441] |
103. | Lucinactant (Surfaxin) | Improved lung compliance[442] |
104. | Luspatercept-aamt (Reblozyl) | Erythroid maturation[443] |
105. | Lutropin alfa (Luveris) | LH receptor activation[444] |
106. | Margetuximab (Margenza) | HER2 expression[445] |
107. | Mecasermin (Increlex) | IGF-1 receptor activation[446] |
108. | Menotropins (Menopur) | FSH and LH receptor activation[447] |
109. | Mepolizumab (Nucala) | IL-5 neutralization[448] |
110. | Metreleptin (Myalept) | Leptin receptor activation[449] |
111. | Mirvetuximab Soravtansine | Folate receptor alpha targeting[450] |
112. | Mogamulizumab (Poteligeo) | CCR4 targeting[451] |
113. | Moxetumomab pasudotox (Lumoxiti) | CD22 expression[452] |
114. | Muromonab (Orthoclone OKT3) | CD3 expression[453] |
115. | Natalizumab (Tysabri) | α4-integrin saturation[454] |
116. | Naxitamab (Danyelza) | GD2 expression[455] |
117. | Necitumumab (Portrazza) | EGFR targeting[456] |
118. | Nesiritide (Natrecor) | Natriuretic peptide receptor A activation[457] |
119. | Netakimab (Netakimab) | IL-17A inhibition[458] |
120. | Nimotuzumab (Theraloc, h-R3) | EGFR targeting[459] |
121. | Nivolumab (Opdivo) | PD-1 receptor occupancy & T cell function [460] |
122. | Nofetumomab merpentan (Verluma) | Carcinoembryonic antigen (CEA) targeting[461] |
123. | Obiltoxaximab (Anthim) | Protective antigen (PA) binding of Bacillus anthracis [462] |
124. | Obinutuzumab (Gazyva) | CD20 targeting[463] |
125. | Ocrelizumab (Ocrevus) | CD20 targeting[464] |
126. | Ocriplasmin (Jetrea) | Vitreomacular adhesion dissolution[465] |
127. | Ofatumumab (Arzerra) | CD20 targeting[466] |
128. | Olaratumab (Lartruvo) | PDGFRα phosphorylation levels[467] |
129. | Olipudase alfa (Xenpozyme) | Acid sphingomyelinase replacement[468] |
130. | Omalizumab (Xolair) | Free serum IgE levels & FcεRI expression on basophils [469] |
131. | Oportuzumab monatox (Vicineum) | N-acetylgalactosamine-4-sulfatase targeting[470] |
132. | Oprelvekin (Neumega) | Thrombopoietin receptor activation[471] |
133. | Oxytocin (Pitocin) | Oxytocin receptor activation[472] |
134. | Palifermin (Kepivance) | Keratinocyte growth factor receptor activation[473] |
135. | Palivizumab (Synagis) | RSV neutralization in serum[474] |
136. | Pancrelipase amylase (Creon) | Pancreatic enzyme replacement[475] |
137. | Panitumumab (Vectibix) | EGFR receptor occupancy & phosphorylation [476] |
138. | Parathyroid/Preotact (Preos) | Parathyroid hormone (PTH) receptor activation[477] |
139. | Pegademase bovine (Adagen) | ADA enzyme replacement[478] |
140. | Pegaspargase (Oncaspar) | Asparagine depletion[479] |
141. | Pegcetacoplan (Empaveli) | Complement C3 inhibition[480] |
142. | Peginterferon alfa-2a (Pegasys) | Interferon alpha receptor activation[481] |
143. | Peginterferon alfa-2b (PegIntron) | Interferon alpha receptor activation[482] |
144. | Pegloticase (Krystexxa) | Uric acid metabolism[483] |
145. | Pegvisomant (Somavert) | Growth hormone receptor antagonist[484] |
146. | Pembrolizumab (Keytruda) | PD-1 receptor occupancy & PD-L1 expression[485] |
147. | Pertuzumab (Perjeta) | HER2 receptor dimerization inhibition [486] |
148. | Pertuzumab (Perjeta) | HER2/neu targeting[487] |
149. | Pramlintide (Symlin) | Amylin analogue[488] |
150. | Protein S human (PROS) | Protein S supplementation[489] |
151. | Ramucirumab (Cyramza) | VEGFR2 targeting[490] |
152. | Ranibizumab (Lucentis) | VEGF level & macular thickness[491] |
153. | Rasburicase (Elitek) | Uric acid conversion to allantoin[492] |
154. | Reteplase (Retavase) | Plasminogen activation[493] |
155. | Rilonacept (Arcalyst) | Interleukin-1 blockade[494] |
156. | Rituximab (Rituxan) | CD20+ B cell depletion [495] |
157. | Romiplostim (Nplate) | Thrombopoietin receptor stimulation[496] |
158. | Romosozumab (Evenity) | Sclerostin levels & bone mineral density[497] |
159. | Sacrosidase (Sucraid) | Sucrase replacement for sucrose digestion[498] |
160. | Sargramostim (Leukine) | GM-CSF receptor activation[499] |
161. | Sarilumab (Kevzara) | IL-6 receptor blockade & CRP[500] |
162. | Sebelipase alfa (Kanuma) | Lysosomal acid lipase replacement[501] |
163. | Secretin (SecreFlo) | Secretin receptor activation[502] |
164. | Secukinumab (Cosentyx) | PASI score, serum IL-17A levels[503] |
165. | Sermorelin (Geref) | GHRH receptor activation[504] |
166. | Siltuximab (Sylvant) | IL-6 targeting[505] |
167. | Somatotropin (Genotropin) | GH receptor activation[506] |
168. | Streptokinase (Streptase) | Plasminogen activation[507] |
169. | Tagraxofusp (Elzonris) | CD123-directed cytotoxin[508] |
170. | Taliglucerase alfa (Elelyso) | Glucocerebrosidase enzyme replacement[509] |
171. | Teduglutide (Gattex) | GLP-2 receptor activation[510] |
172. | Tenecteplase (TNKase) | Plasminogen activation[511] |
173. | Teriparatide (Forteo) | Bone formation stimulation[512] |
174. | Terlipressin (Varpress) | Vasoconstriction through V1 receptor activation[513] |
175. | Tesamorelin (Egrifta) | GH-releasing hormone receptor activation[514] |
176. | Thymalfasin (Zadaxin) | Immunomodulation, T-cell stimulation[515] |
177. | Thyrotropin Alfa (Thyrogen) | Thyroid-stimulating hormone receptor activation[516] |
178. | Tirzepatide (Mounjaro) | Dual GLP-1 and GIP receptor agonism[517] |
179. | Tisotumab vedotin (Tivdak) | ADC targeting tissue factor[518] |
180. | Tocilizumab (Actemra) | IL-6 receptor blockade[519] |
181. | Tositumomab (Bexxar) | CD20 targeting[520] |
182. | Trastuzumab (Herceptin) | HER2/neu receptor expression [521] |
183. | Urofollitropin (Bravelle) | Follicle-stimulating hormone stimulation[522] |
184. | Urokinase (Abbokinase) | Plasminogen activation[523] |
185. | Ustekinumab (Stelara) | Inhibition of the p40 subunit of interleukin-12 (IL-12) and interleukin-23 (IL-23).[524] |
186. | Vasopressin (Vasostrict) | V1 and V2 receptor activation[525] |
187. | Vedolizumab (Entyvio) | α4β7 integrin receptor occupancy[526] |
188. | Velaglucerase alfa (Vpriv) | Glucocerebrosidase enzyme replacement[527] |
Function | Usage | Example |
---|---|---|
Demonstrate Drug Activity | Early evidence of a drug's effect before overt clinical outcomes manifest. | In treating chronic myeloid leukemia (CML), the BCR-ABL tyrosine kinase inhibitor imatinib is used. The decline in BCR-ABL transcript levels in patients' blood is a functional PD marker of the drug's activity on its target.[34] |
Guide Dosing | Dose-response relationship, ensuring optimal drug dosing. | For cholesterol-lowering drugs like statins, the low-density lipoprotein cholesterol (LDL-C) levels in the blood serve as a PD marker to guide dosing and assess efficacy.[35] |
Select Patients | To identify patients likely to benefit from a specific treatment. | In some breast cancers, overexpression of the HER2 protein is observed. HER2 status serves as a functional PD marker to select patients who might benefit from trastuzumab, which targets HER2.[36] |
Monitor Resistance | Track the development of resistance to treatments. | In HIV treatment, the emergence of specific viral mutations can serve as PD markers indicating resistance to certain antiretroviral drugs.[37] |
Determine Drug Mechanism of Action | Confirmation of action through its intended mechanism. | In Alzheimer’s disease, the buildup of beta-amyloid plaques is considered a hallmark. Drugs designed to reduce beta-amyloid levels in the brain might use CSF (cerebrospinal fluid) levels of beta-amyloid as a PD marker to show the drug's effect.[38] |
Validate Target Engagement | Demonstrate that a drug is successfully engaging with and modulating its target. | For multiple sclerosis drugs like fingolimod, a PD marker such as the number of circulating lymphocytes can indicate the drug's effect on immune cell egress from lymph nodes.[39] |
Evaluate Drug-induced Toxicity | Monitor potential adverse effects of a drug. | In chemotherapy, monitoring the levels of liver enzymes like AST and ALT in the blood can serve as PD markers for drug-induced liver damage.[40] |
Optimize Therapeutic Window | Establish the range between the minimum effective dose and the onset of adverse effects. | For anticoagulant drugs like warfarin, the INR (International Normalized Ratio) serves as a PD marker to ensure the drug's effect is within a therapeutic range that minimizes the risk of bleeding and clot formation.[41] |
Predict Long-term Drug Effects | Early changes in PD markers can predict longer-term therapeutic or adverse effects. | In osteoporosis treatments, reducing bone resorption markers like CTX (C-terminal telopeptide) can predict longer-term benefits in bone mineral density and fracture risk.[42] |
Assess Immune Response | For immunotherapies, to gauge the body's immune response to the treatment. | In cancer immunotherapy, the presence and proliferation of tumor-infiltrating lymphocytes (TILs) in the tumor microenvironment can serve as a PD marker to indicate the activation and targeting of the immune system against tumor cells.[43] |
Indicate Drug Combination Efficacy | In combination therapies, to show the synergistic or additive effects of the combined drugs. | In treatments for tuberculosis, monitoring bacterial load in sputum samples can serve as a PD marker for the combined efficacy of multiple antimicrobial agents.[44] |
Track Reversal of Disease Progression | Indicate whether a drug is not just halting but reversing disease progression. | In fibrotic diseases like idiopathic pulmonary fibrosis, measuring levels of collagen-derived peptides in blood or bronchoalveolar lavage fluid can act as PD markers, indicating the repair or degradation of fibrotic tissue.[45] |
Evaluate Neural Activity and Plasticity | PD markers can track neural activity or connection changes in neurologic disorders and treatments. | For treatments aimed at Alzheimer's or other neurodegenerative conditions, the levels of synaptic proteins or neuronal activity markers in CSF can indicate neural activity and synaptic plasticity.[46] |
Monitoring Metabolic Responses | To help track changes in metabolic pathways. | In diabetes management, measuring C-peptide levels alongside insulin can give insights into endogenous insulin production and pancreatic function.[47] |
Monitoring Cellular Senescence and Aging | In treatments aiming to affect aging processes or cellular health, to track cellular senescence. | Measuring levels of senescence-associated beta-galactosidase or p16^INK4a expression can act as PD markers for cellular aging or the efficacy of anti-aging treatments.[48] |
Evaluating Epigenetic Changes | Track changes in DNA methylation, histone modification, or other epigenetic markers. | In oncology, when treating with drugs targeting DNA methyltransferases, the global or gene-specific changes in DNA methylation levels can serve as PD markers.[49] |
Assessing Drug-induced Autophagy | For therapies inducing autophagy as a mechanism, to monitor the process. | Monitoring LC3B lipidation, a critical step in autophagosome formation, can serve as a PD marker for autophagy activation.[50] |
Monitoring Immune Checkpoint Inhibition | Cancer immunotherapy targets immune checkpoints to gauge the effectiveness of checkpoint inhibition. | In patients receiving PD-1 or PD-L1 inhibitors, monitoring circulating tumor DNA (ctDNA) levels can serve as a PD marker to indicate response to therapy.[51] |
Test Method | Application |
Mass Spectrometry (MS) | Structural analysis of glycan [102] |
High-Performance Liquid Chromatography (HPLC) | Separation of glycan structures - [103] |
Capillary Electrophoresis (CE) | High-resolution separation of glycans - [104] |
NMR Spectroscopy | Detailed structural analysis of glycan[105] |
Matrix-assisted laser desorption/ionization (MALDI-MS) | Mass determination of glycan[106] |
Glycan Microarrays | High-throughput analysis of protein-glycan interactions[107] |
Lectin Affinity Chromatography | Separation of glycans using specific binding proteins[108] |
FTIR | Analysis of glycan structure[109] |
Enzyme-linked Lectin Assay | Quantitative glycan analysis[110] |
Exoglycosidase Sequencing | Structural characterization of glycans[111] |
Stable Isotope Labeling | Quantitative glycomics[112] |
SPR | Real-time glycan-protein interaction analysis[113] |
Gas Chromatography-Mass Spectrometry | Analysis of volatile derivatives of glycans[114] |
Tandem Mass Spectrometry | Sequential fragmentation for glycan structure elucidation[115] |
Hydrophilic Interaction Liquid Chromatography | Separation of polar glycans[116] |
Fluorescence Detection | Sensitivity enhancement in glycan analysis[117] |
ELISA | Detection of specific glycan-protein interactions[118] |
MALS | Characterizing size and composition[119] |
Immunohistochemistry | Localization of specific glycans in tissues[120] |
IEF | Separation of glycoproteins by isoelectric point[121] |
Reverse Phase Liquid Chromatography | Separation of glycopeptides and glycoproteins[122] |
Glycoproteomic Analysis | Integrative approach for comprehensive glycoprotein study[123] |
Electron Microscopy | Visualizing glycan structures and localization[124] |
Optical Glycan Biosensors | Real-time monitoring of glycan-protein interactions[125] |
X-ray Crystallography | 3D structure determination of glycoproteins[126] |
DLS | Size distribution analysis of glycoproteins[127] |
Glycan Sequencing using Edman Degradation | Sequential identification[128] |
Bottom-up MS | Mass spectrometric analysis for primary sequence analysis, evaluation of N/O-glycosylation sites, and quantification of methionine oxidation.[129] |
Bioanalytical and bioinformatics | Data integration at different structural levels to identify various glycoforms of recombinant Human Chorionic Gonadotropin (r-hCG), urinary hCG (u-hCG), and recombinant Follicle Stimulating Hormone (r-hFSH) revealed that these biopharmaceuticals differ considerably in their glycosylation patterns.[130] |
Resolution Discrepancies | Between high-resolution native and glycopeptide-centric mass spectrometric approaches for the glycosylation of erythropoietin variants.[131] |
Glycan microheterogeneity | To identify multiple glycosylation sites in the vascular endothelial growth factor IgG (VEGFR-IgG) fusion protein to understand the functional significance of each glycosylation pattern.[132] |
Glycoforms | Several glycoforms using hybrid high-performance liquid chromatography-MS approaches.[133], such as 24 glycoengineered erythropoietin variants with varying glycan branching and sialylation levels, which are crucial parameters for biotherapeutic efficacy. |
NMR | Identification of glucose-induced glycation in mAbs and other proteins using NMR spectroscopy. [134] |
Novel glycoforms | Identification of novel glycosylations in human serum-derived factor IX. [135] |
Mass spectral profiling | The N-linked, O-linked, ganglioside, and glycosaminoglycan compound classes and the tandem mass spectrometry of glycans have led to spectral glycoproteomics.[136] |
N-glycosylation profile | Analysis of trastuzumab biosimilar candidates using normal-phase liquid chromatography and matrix-assisted laser desorption/ionization-time of flight-MS.[137] |
Microheterogeneity | Composite glycosylation profiles and other microheterogeneities in intact mAbs via high-resolution native MS using a modified Orbitrap.[138] |
Targeted site-specific quantitation | N-and O-glycopeptides using 18O-labeling and product ion-based MS.[139] |
Hybrid MS | Approaches in glycoprotein analysis and their usage in scoring biosimilarity.[140] |
Gene Expression Profiling | Simultaneous measurement of the expression levels of several genes to create a global picture of cellular functions. |
Non-coding RNA Analysis | Identifying non-coding RNAs. |
Alternative Splicing | Events wherein exons of a gene are joined in multiple ways during protein synthesis, thus producing numerous proteins from a single gene. |
Disease Diagnosis and Treatment | Disease diagnosis, prognosis, and determining the treatment response. |
Optimization of Host Cells for Protein Expression | Optimize host cells to improve recombinant protein expression.[143] |
Identifying Suitable Expression Systems | Selection of optimal expression system.[144] |
Understanding protein function and interactions | How proteins function and interact within cellular networks.[145] |
Monitoring Quality Control in Biopharmaceutical Production | Manufacturing QC.[146] |
Improving Protein Solubility and Folding | Enhance the solubility and folding of recombinant.[147] |
Tailoring Protein Post-Translational Modifications (PTMs) | To produce proteins with the desired PTMs.[148] |
Enhanced Yield in Industrial Protein Production | To enhance the yield of recombinant proteins for industrial applications.[149] |
Studying Protein Stability and Degradation | Design recombinant proteins with enhanced stability.[150] |
Personalized Medicine and Therapeutics | Patient-specific proteins based on individual gene expression profiles.[151] |
Improving Expression Systems | Researchers can optimize expression systems for recombinant protein production by studying host cell genomes.[161] |
Designing Recombinant Proteins | Genomics can be used to identify and design recombinant proteins with the desired functions.[162] |
Enhancing Protein Stability | Understanding the genomic context can aid in designing recombinant proteins with enhanced stability and activity.[163] |
Personalized Medicine | Personalized genomics allows the development of recombinant proteins tailored to the genetic profiles of individual patients.[164] |
Metabolic Engineering | Genomic insights can guide the redesign of metabolic pathways to produce recombinant proteins in microbial systems efficiently.[165] |
Biomarker Discovery | Genomics aids in the identification of biomarkers that can be targeted with recombinant proteins for diagnostic or therapeutic purposes.[166] |
Understanding Protein Function | Comparative genomics can elucidate the functions of proteins by identifying conserved sequences and structures.[167] |
Enhanced Protein Folding | Genomic data can be used to understand and improve the folding of recombinant proteins.[168] |
Development of Novel Therapeutics | Genomics is used to identify potential targets of therapeutic recombinant proteins.[169] |
Cell Line Optimization | Cell line optimization results from (i) research applied to parental, non-recombinant cell lines; (ii) systems-level datasets generated with recombinant cell lines; (iii) datasets linking phenotypic traits to relevant biomarkers; (iv) data depositories and bioinformatics tools; and (v) in silico model development.[170] |
Optimization of Expression Systems | Understanding and manipulating epigenetic markers can enhance recombinant proteins' expression in host cells.[174] |
Production of Recombinant Proteins with Specific Modifications | Epigenomic control enables the production of recombinant proteins with functionally essential PTMs.[175] |
Development of recombinant proteins for epigenetically targeted therapies | Epigenomics guides the discovery and development of recombinant proteins that target specific epigenetic modifications involved in various diseases.[176] |
Studying the Epigenetic Control of Protein Function | Recombinant proteins can be used to study how epigenetic modifications regulate endogenous proteins, providing insights into their functions and control.[177] |
Recombinant Epigenetic Modifiers for Research | Producing recombinant proteins involved in epigenetic modifications, such as methyltransferases, may be helpful for research and drug development.[178] |
Modeling Diseases | Using epigenomic information for modifying host cells to produce recombinant proteins allows the creation of more accurate disease models, particularly for conditions in which epigenetic alterations play a crucial role.[179] |
Quality Control and Stability of Biopharmaceuticals | Epigenomic control may improve the quality and stability of recombinant proteins for biopharmaceutical applications.[180] |
The Optimization of Expression Systems for Protein Production | MS and NMR.[184] |
Improving Industrial Protein Production Yields | Liquid Chromatography-Mass Spectrometry.[185] |
Understanding Host-Pathogen Interactions for Recombinant Vaccine Development | Gas Chromatography-Mass Spectrometry.[186] |
Investigating Protein-Metabolite Interactions | NMR Spectroscopy.[187] |
Quality Control and Stability of Recombinant Proteins in Biopharmaceuticals | Capillary Electrophoresis-Mass Spectrometry.[188] |
Assessing the impact of protein therapeutics on metabolic pathways | Using ultra-performance liquid chromatography-mass spectrometry.[189] |
Designing More Effective Protein-Based Therapies Through Metabolite Profiling | Fourier Transform Mass Spectrometry.[190] |
Environmental Stress Response in Recombinant Protein Production | Liquid Chromatography-Quadrupole Time-of-Flight Mass Spectrometry.[191] |
Immune response against tumor cells in patients with melanoma | increases in absolute lymphocyte counts are observed in response to therapy and can serve as a marker of functional immune response[192]. |
Lab Values Alteration | Changes in specific laboratory values can serve as functional response markers for certain conditions. For example, in patients with rheumatoid arthritis treated with the anti-TNFα mAb, adalimumab, reductions in serum C-reactive protein levels and erythrocyte sedimentation rate, which are markers of inflammation, indicate a positive response to therapy[193]. |
Modulation of T-cell Response | Some mAbs, particularly immune checkpoint inhibitors such as pembrolizumab, function by modulating the T-cell response, which can be measured using T-cell activation markers, such as CD137, or by enumerating antigen-specific T-cells.[194] |
Alteration in Serum Immunoglobulin Levels | Some mAbs can cause changes in the serum levels of immunoglobulins, which can serve as PD markers. Rituximab, an anti-CD20 mAb, decreases serum immunoglobulin levels, which can be monitored clinically.[195] |
PK and PD Correlation | The correlation between PK properties, such as serum drug concentration and PD markers, is a significant component of mAb engagement. This elucidates the dose-response relationship for adjusting the dosing regimens. For instance, with infliximab, an anti-TNFα mAb used in treating autoimmune diseases, measuring the serum drug concentration and correlating this with the clinical response and anti-TNFα activity can indicate effective drug-target engagement[196]. |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 MDPI (Basel, Switzerland) unless otherwise stated