214 月/25
b cell immortalization

KMD Bioscience Conducts Literature Analysis | Object: HPV Antibody

The study “Peak neutralizing and cross-neutralizing antibody levels to human papillomavirus types 6/16/18/31/33/45/52/58 induced by bivalent and quadrivalent HPV vaccines” provides an in-depth comparison of the immunogenic responses elicited by the bivalent (Cervarix) and quadrivalent (Gardasil) HPV vaccines. The research focuses on the generation of neutralizing antibodies (NAbs) against both vaccine-included HPV types and non-vaccine HPV types, offering insights into each vaccine’s potential for broader protection.

Study Design and Methodology

The researchers conducted an independent comparison of NAb levels seven months after the initiation of three-dose, six-month vaccination schedules. The study involved adolescent females from Finland and India who received either the bivalent or quadrivalent HPV vaccine. A semi-automated Pseudovirion-Based Neutralization Assay was employed to measure NAb levels against HPV types 6, 16, 18, 31, 33, 45, 52, and 58.

Key Findings

Neutralizing Antibody Levels for Vaccine HPV Types:

HPV16 and HPV18: Recipients of the bivalent vaccine exhibited significantly higher peak NAb levels against HPV16 and HPV18 compared to those who received the quadrivalent vaccine.

Cross-Neutralizing Antibody Levels for Non-Vaccine HPV Types:

HPV31, HPV33, HPV45, HPV52, and HPV58: The bivalent vaccine induced cross-neutralizing antibodies against these non-vaccine HPV types more frequently and at higher levels than the quadrivalent vaccine.

Correlation of Antibody Levels:

HPV45 with HPV16/18: A stronger correlation was observed between NAb levels for HPV45 and HPV16/18 in bivalent vaccine recipients compared to quadrivalent vaccine recipients, suggesting a qualitatively different cross-reactive immune response.

Implications

The findings indicate that the bivalent vaccine not only induces robust immunity against its target HPV types (16 and 18) but also offers enhanced cross-protection against several non-vaccine HPV types associated with cervical cancer. This broader immunogenic profile could have significant implications for cervical cancer prevention strategies. However, the study’s authors note that the comparison was conducted in different populations (Finnish and Indian adolescents), highlighting the need for further head-to-head studies to confirm these results.

Conclusion

This study underscores the superior immunogenicity of the bivalent HPV vaccine in inducing both neutralizing and cross-neutralizing antibodies compared to the quadrivalent vaccine. These insights are crucial for informing vaccination policies and optimizing strategies for cervical cancer prevention.

Our Humanized HPV Antibody Products

Cat# Product Name Species Host Applications Size Price
MA2624 Humanized Anti-HPV6/HPV11/HPV33 mAb HPV6/HPV11/HPV33 CHO 100ug, 500ug, 1mg Inquiry
MA2625 Humanized Anti-HPV18/HPV45 mAb HPV18/HPV45 CHO 100ug, 500ug, 1mg Inquiry
MA2626 Humanized Anti-HPV16/HPV31/HPV52/HPV58 mAb HPV16/HPV31/HPV52/HPV58 CHO 100ug, 500ug, 1mg Inquiry
204 月/25

Explore KMD Bioscience Detailed Analysis of IL-6 and Its Implications

Structure of IL-6

IL-6 is a 19–26 kDa pleiotropic cytokine composed of 212 amino acids. It has a four-helix bundle structure with two long and two short α-helices, stabilized by two disulfide bonds. IL-6 is mainly produced by immune cells (macrophages, T cells, B cells), fibroblasts, and endothelial cells in response to infection, inflammation, or stress.

Receptor Components

IL-6 signaling is mediated through a two-component receptor complex:

  1. IL-6R (CD126): A membrane-bound or soluble receptor that specifically binds IL-6.
  2. gp130 (CD130): A signal-transducing subunit that activates downstream pathways.
  3. Function of IL-6

IL-6 is a key cytokine in immune regulation, inflammation, hematopoiesis, and metabolism.

  • Pro-inflammatory Role: Triggers acute-phase response, promotes fever, and induces CRP production.
  • Anti-inflammatory Role: Regulates tissue repair and suppresses excessive immune responses.
  • Hematopoiesis: Supports B-cell differentiation, T-cell activation, and megakaryocyte maturation.
  • Metabolism: Regulates glucose homeostasis and lipid metabolism in liver and muscle tissues.
  • Cancer Progression: Elevated IL-6 levels are associated with cancer proliferation, angiogenesis, and metastasis.

IL-6 Signaling Pathway

IL-6 signaling occurs via three main pathways:

(A) Classical Signaling (Protective Role)

  • Receptor Binding: IL-6 binds to membrane-bound IL-6R, forming a complex with gp130.
  • JAK-STAT Activation: Janus kinases (JAK1, JAK2, Tyk2) phosphorylate gp130, activating STAT3, which translocates to the nucleus to regulate gene expression.
  • Effects: Supports tissue regeneration, immune balance, and normal inflammatory responses.

(B) Trans-Signaling (Pro-inflammatory Role)

  • Soluble IL-6R (sIL-6R)binds IL-6, interacting with gp130-expressing cells that do not have IL-6R.
  • This pathway is linked to chronic inflammation, autoimmunity, and cancer progression.

(C) Trans-Presentation

  • IL-6 is presented by dendritic cells to T cells via IL-6R and gp130, regulating T-cell differentiation.

Market Prospects of IL-6 Targeting Drugs

  • Global Market Size: Estimated at $10.5 billion in 2023, projected to grow at 2% CAGR.
  • Key Drivers: Rising cases of autoimmune diseases (RA, lupus), cytokine storm management (COVID-19), and oncology applications.
  • Key Companies: Roche, Sanofi, Novartis, Johnson & Johnson, Pfizer.

Future Trends:

  • Bispecific antibodiestargeting IL-6 and TNF-α or IL-6 and IL-17.
  • Oral IL-6 inhibitorsunder development.
  • Personalized therapybased on cytokine profiling.

FDA-Approved IL-6 Targeting Drugs

Drug Name Manufacturer Approval Year Indications Mechanism
Tocilizumab (Actemra) Roche/Genentech 2010 Rheumatoid arthritis (RA), giant cell arteritis, cytokine release syndrome (CRS), COVID-19 IL-6R monoclonal antibody
Sarilumab (Kevzara) Sanofi/Regeneron 2017 Moderate to severe RA IL-6R monoclonal antibody
Siltuximab (Sylvant) EUSA Pharma 2014 Multicentric Castleman disease Direct IL-6 monoclonal antibody

IL-6 inhibitors continue to gain traction, with emerging indications in oncology, neuroinflammation, and cardiovascular diseases.

Our IL-6 Related Antibody Products

Cat# Product Name Species Host Applications Size Price
PA221 Mouse Anti-Human IL-6 Monoclonal Antibody (Capture) Human Mouse LFIA (Lateral-Flow Immunochromatographic Assay),CLIA (Chemiluminescence Immunoassay),ELISA 1mg Inquiry
PA222 Mouse Anti-Human IL-6 Monoclonal Antibody (Detection) Human Mouse LFIA (Lateral-Flow Immunochromatographic Assay),CLIA (Chemiluminescence Immunoassay),ELISA 1mg Inquiry
PA4352 Rabbit Anti-Human IL6 pAb Human Rabbit 50ul, 100ul Inquiry
YR1007 Anti-Human IL6 Recombinant Antibody(Siltuximab) IF, IP, Neut, FuncS, ELISA, FCM, ICC 1mg, 5mg Inquiry
YR1177 Anti-Human IL6 Recombinant Antibody(Elsilimomab) 1mg, 5mg Inquiry
YR1234 Anti-Human IL6 Recombinant Antibody(Olokizumab) Rat WB, ELISA, FCM, IP, FuncS, IF, Neut 1mg, 5mg Inquiry
YR1261 Anti-Human IL6 Recombinant Antibody(Sirukumab) FCM, IP, ELISA, Neut, FuncS, IF 1mg, 5mg Inquiry
YR1275 Anti-Human IL6 Recombinant Antibody(Clazakizumab) 1mg, 5mg Inquiry
YR1605 Anti-Human IL6 Recombinant Antibody(Ziltivekimab) 1mg, 5mg Inquiry
194 月/25

KMD Bioscience-Comprehensive Analysis of EGFR: Insights & Implications

Structure of EGFR

EGFR (HER1/ErbB1) is a transmembrane glycoprotein receptor in the ErbB family of receptor tyrosine kinases (RTKs).

  • Extracellular domain (ECD): Binds epidermal growth factor (EGF) and related ligands.
  • Transmembrane domain (TMD): Anchors EGFR in the cell membrane.
  • Intracellular tyrosine kinase domain (TKD): Responsible for autophosphorylation and downstream signaling.
  • C-terminal tail: Contains phosphorylation sites for interaction with intracellular signaling molecules.

Function of EGFR

EGFR plays a crucial role in cell proliferation, differentiation, survival, and migration.

  • Normal function: Tissue repair, embryonic development, and immune response.
  • Dysregulation: Overexpression or mutation leads to cancer progression, especially in non-small cell lung cancer (NSCLC), colorectal cancer, and glioblastoma.

EGFR Signaling Pathway

  • Ligand Binding: EGF, TGF-α, or other ligands bind EGFR, inducing dimerization.
  • Autophosphorylation: Activation of intracellular kinase domain.

Downstream Signaling Activation:

  • PI3K-AKT Pathway: Promotes cell survival and anti-apoptosis.
  • Ras-Raf-MEK-ERK Pathway: Enhances proliferation.
  • JAK-STAT Pathway: Regulates inflammation and immune response.
  • PLCγ-PKC Pathway: Increases cell motility and invasion.
  • Oncogenic Role: Mutations (e.g., L858R, exon 19 deletions) lead to constitutive activation, driving uncontrolled cancer growth.

Market Prospects of EGFR Inhibitors

  • Global market value: Estimated at $10.3 billion in 2023, projected to reach $15 billion by 2030.
  • Growth drivers: Rising incidence of NSCLC and colorectal cancer, increasing adoption of targeted therapy, and advancements in resistance-overcoming drugs.
  • Key companies: AstraZeneca, Roche, Merck, Amgen, Takeda, Eli Lilly.
  • Future trends: Development of 3rd and 4th generation EGFR inhibitors, combination therapies, and novel bispecific antibodies.

FDA-Approved EGFR-Targeting Drugs

Drug Name Approval Year Indications Mechanism
Gefitinib (Iressa) 2003 NSCLC (EGFR-mutant) 1st-gen EGFR TKI, ATP-competitive
Erlotinib (Tarceva) 2004 NSCLC, pancreatic cancer 1st-gen EGFR TKI
Afatinib (Gilotrif) 2013 NSCLC (EGFR exon 19/21 mutations) 2nd-gen irreversible EGFR/HER2 TKI
Osimertinib (Tagrisso) 2015 NSCLC (T790M mutation) 3rd-gen EGFR TKI, targets resistance mutations
Cetuximab (Erbitux) 2004 Colorectal cancer, head and neck SCC Monoclonal antibody blocking EGFR
Panitumumab (Vectibix) 2006 Metastatic colorectal cancer (KRAS wild-type) Fully human anti-EGFR antibody
Necitumumab (Portrazza) 2015 Squamous NSCLC Anti-EGFR monoclonal antibody
Dacomitinib (Vizimpro) 2018 NSCLC (EGFR-mutant) 2nd-gen irreversible EGFR TKI
Amivantamab (Rybrevant) 2021 NSCLC (EGFR exon 20 insertion) Bispecific EGFR-MET antibody

EGFR-targeted therapies continue to evolve, with next-generation inhibitors and combination regimens showing promise for overcoming resistance and improving survival outcomes.

Our EGFR Related Antibody Products

Cat# Product Name Species Host Applications Size Price
PAV6865 Rabbit Anti-EGFR Polyclonal Antibody Human, Mouse, Rat Rabbit WB, IHC-p, IF, ELISA 100ul Inquiry
PA5322 Rabbit Anti-Human EGFR pAb Human Rabbit 50ul, 100ul Inquiry
MA1149 Rabbit Anti-Human [KO Validated] EGFR / ErbB1 mAb Human Rabbit 50ul, 100ul Inquiry
MA1179 Rabbit Anti-Human EGFR (L858R) mAb Human Rabbit 50ul, 100ul Inquiry
YR1006 Anti-Human EGFR Recombinant Antibody(Necitumumab) ELISA, IF, IP, FuncS, FCM, ICC 1mg, 5mg Inquiry
YR1020 Anti-Human EGFR Recombinant Antibody(Matuzumab) Neut, ELISA, IF, IP, FuncS, FCM, ICC 1mg, 5mg Inquiry
YR1033 Anti-Human EGFR Recombinant Antibody(Nimotuzumab) ELISA, IP, FCM, FuncS, Neut, IF, IHC 1mg, 5mg Inquiry
YR1089 Anti-Human EGFR Recombinant Antibody(Cetuximab) IF, IP, Neut, FuncS, ELISA, FCM 1mg, 5mg Inquiry
YR1131 Anti-Human EGFR Recombinant Antibody(Panitumumab) ELISA, IP, FCM, FuncS, Neut, IF, ICC 1mg, 5mg Inquiry
YR1134 Anti-Human EGFR Recombinant Antibody(Zalutumumab) ELISA, FCM, IP, FuncS, IF, Neut 1mg, 5mg Inquiry
YR1280 Anti-Human EGFR Recombinant Antibody(Futuximab) Mouse IF, WB, Inhib 1mg, 5mg Inquiry
YR1281 Anti-Human EGFR Recombinant Antibody(Imgatuzumab) Neut, ELISA, IF, IP, FuncS, FCM 1mg, 5mg Inquiry
YR1292 Anti-Human EGFR Recombinant Antibody(Modotuximab) Mouse FCM, IP, ELISA, Neut, FuncS, IF, WB 1mg, 5mg Inquiry
YR1408 Anti-Human EGFR Recombinant Antibody(Laprituximab) 1mg, 5mg Inquiry
YR1425 Anti-Human EGFR Recombinant Antibody(Depatuxizumab) 1mg, 5mg Inquiry
YR1448 Anti-Human EGFR Recombinant Antibody(Losatuxizumab) 1mg, 5mg Inquiry
YR1464 Anti-Human EGFR Recombinant Antibody(Tomuzotuximab) ELISA, IHC, FCM, IP, IF, FuncS 1mg, 5mg Inquiry
YR1512 Anti-Human KDR & VEGFR2 Recombinant Antibody 1mg, 5mg Inquiry
YR1545 Anti-Human EGFR Recombinant Antibody(Serclutamab) 1mg, 5mg Inquiry
YR1568 Anti-Human EGFR & ME Recombinant Antibody 1mg, 5mg Inquiry
YR1593 Anti-Human EGFR & LGR5 Recombinant Antibody 1mg, 5mg Inquiry
184 月/25

VEGFA Insights: KMD Bioscience Unveils Key Analysis Findings

Structure of VEGFA

Vascular Endothelial Growth Factor A (VEGFA) is a key member of the VEGF family and plays a crucial role in angiogenesis and vascular permeability.

  • Dimeric glycoprotein: VEGFA forms a homodimer via disulfide bonds.
  • Isoforms: Alternative splicing generates multiple isoforms, including VEGFA121, VEGFA165, VEGFA189, and VEGFA206, each with different receptor-binding properties and tissue distribution.
  • Receptor Binding: VEGFA binds to VEGFR-1 (Flt-1)and VEGFR-2 (KDR/Flk-1) to trigger downstream signaling.

Function of VEGFA

  • Angiogenesis: Stimulates endothelial cell proliferation and migration.
  • Vascular Permeability: Increases vascular leakage, crucial in wound healing and tumor growth.
  • Hematopoiesis: Regulates blood vessel formation in the bone marrow.
  • Pathological Roles: Overexpression is linked to cancer, diabetic retinopathy, and age-related macular degeneration (AMD).

VEGFA Signaling Pathway

  • Ligand Binding: VEGFA binds to VEGFR-2, initiating dimerization and autophosphorylation.
  • Downstream Signaling:
  • PI3K-AKT Pathway: Promotes cell survival.
  • Ras-Raf-MEK-ERK Pathway: Induces endothelial cell proliferation.
  • PLCγ-PKC Pathway: Enhances vascular permeability.
  • Endothelial Activation: Leads to new blood vessel formation and increased permeability.

Market Prospects of VEGFA Inhibitors

  • The global VEGFA-targeted therapy marketwas valued at $9.2 billion in 2022 and is projected to reach $15 billion by 2030.
  • Key growth drivers: Rising incidence of cancer, AMD, and diabetic retinopathy.
  • Major companies: Roche, Novartis, Regeneron, Bayer, and Eli Lilly.
  • Future trends: Combination therapies, biosimilars, and gene therapy approaches.

FDA-Approved VEGFA Inhibitors

Drug Name Approval Year Indications Mechanism
Bevacizumab (Avastin) 2004 Colorectal cancer, NSCLC, glioblastoma, RCC Humanized IgG1 anti-VEGFA mAb
Ranibizumab (Lucentis) 2006 AMD, diabetic macular edema, retinal vein occlusion Humanized IgG1 Fab fragment targeting VEGFA
Aflibercept (Eylea) 2011 AMD, diabetic retinopathy, RVO Fusion protein binding VEGFA and PlGF
Ziv-aflibercept (Zaltrap) 2012 Metastatic colorectal cancer VEGFA/PlGF trap fusion protein
Brolucizumab (Beovu) 2019 Neovascular AMD Humanized scFv anti-VEGFA mAb

VEGFA inhibitors continue to be a cornerstone of anti-angiogenic therapy, with ongoing research into biosimilars, novel formulations, and combination regimens for broader clinical applications.

Our Related VEGFA Antibody Products

Cat# Product Name Species Host Applications Size Price
PA4346 Rabbit Anti-Human VEGFA pAb Human Rabbit 50ul, 100ul Inquiry
MA456 Mouse Anti-Human VEGFA mAb Human Mouse 50ul, 100ul Inquiry
YR1001 Anti-Human VEGFA Recombinant Antibody(Bevacizumab) FCM, IP, ELISA, Neut, FuncS, IF, ICC 1mg, 5mg Inquiry
YR1040 Anti-Human VEGFA Recombinant Antibody(Ranibizumab) 1mg, 5mg Inquiry
YR1114 Anti-Human VEGFA Recombinant Antibody(IMC-1C11) 1mg, 5mg Inquiry
YR1341 Anti-Human ANGPT2 & VEGFA Recombinant Antibody(Vanucizumab) WB, FCM, IP, ELISA, Neut, FuncS, IF 1mg, 5mg Inquiry
YR1363 Anti-Human VEGFA Recombinant Antibody(Brolucizumab) ELISA, IHC, IF, IP, FCM, Inhib 1mg, 5mg Inquiry
YR1406 Anti-Human DLL4 & VEGFA Recombinant Antibody 1mg, 5mg Inquiry
YR1449 Anti-Human VEGFA Recombinant Antibody(Varisacumab) ELISA, IHC, IF, IP, FCM, FuncS 1mg, 5mg Inquiry
YR1492 Anti-Human ANGPT2 & VEGFA Recombinant Antibody(Faricimab) 1mg, 5mg Inquiry
YR1517 Anti-Human DLL4 & VEGFA Recombinant Antibody(Dilpacimab) 1mg, 5mg Inquiry
YR1643 Anti-Human VEGFA Recombinant Antibody(Aflibercept) 1mg, 5mg Inquiry
174 月/25

KMD Bioscience | Comprehensive PD-1 Analysis: Insights & Key Developments

Structure of PD-1

Programmed Cell Death Protein 1 (PD-1) is a type I transmembrane protein belonging to the CD28 family of immune checkpoint receptors. It consists of:

Extracellular domain: A single IgV-like domain responsible for binding to its ligands (PD-L1 and PD-L2).

Transmembrane region: Anchors PD-1 in the T cell membrane.

Cytoplasmic tail: Contains two key motifs—Immunoreceptor Tyrosine-Based Inhibitory Motif (ITIM) and Immunoreceptor Tyrosine-Based Switch Motif (ITSM)—which regulate downstream inhibitory signaling.

Function of PD-1

PD-1 is an immune checkpoint receptor that downregulates T-cell activity, preventing excessive immune responses and maintaining immune tolerance. It plays a crucial role in:

Immune homeostasis: Prevents autoimmunity by suppressing self-reactive T cells.

Cancer immune evasion: Tumors exploit PD-1 signaling to escape immune detection.

Infectious diseases: Chronic infections upregulate PD-1, leading to T-cell exhaustion.

PD-1 Signaling Pathway

Ligand Binding: PD-1 binds to PD-L1 (expressed on tumor and immune cells) or PD-L2 (mainly on antigen-presenting cells).

Tyrosine Phosphorylation: Ligand binding activates ITIM and ITSM motifs.

Recruitment of SHP-2: The phosphatase SHP-2 is recruited, dephosphorylating key signaling molecules.

T-cell Inhibition: This leads to decreased activation of PI3K-AKT, Ras-MEK-ERK, and ZAP70 pathways, reducing cytokine production and T-cell proliferation.

Market Prospects of PD-1 Inhibitors

The global PD-1/PD-L1 inhibitor market was valued at $37.7 billion in 2022 and is expected to exceed $80 billion by 2030 due to increasing cancer incidence.

Combination therapies (PD-1 inhibitors with chemotherapy, radiotherapy, or other checkpoint inhibitors) are expanding indications beyond oncology to autoimmune diseases.

FDA-Approved PD-1 Inhibitors

Drug Name Approval Year Indications Mechanism
Pembrolizumab (Keytruda) 2014 NSCLC, melanoma, Hodgkin’s lymphoma, gastric cancer, etc. Humanized IgG4 anti-PD-1 mAb
Nivolumab (Opdivo) 2014 NSCLC, melanoma, RCC, HCC, esophageal cancer, etc. Fully human IgG4 anti-PD-1 mAb
Cemiplimab (Libtayo) 2018 Cutaneous squamous cell carcinoma, NSCLC, BCC Fully human IgG4 anti-PD-1 mAb
Toripalimab 2021 (China, 2023 FDA BTD) Nasopharyngeal carcinoma, melanoma Recombinant humanized IgG4 anti-PD-1 mAb
Tislelizumab 2022 (China, US filing in 2023) NSCLC, esophageal squamous cell carcinoma Engineered IgG4 anti-PD-1 mAb (reduced FcγR binding)

PD-1 inhibitors continue to dominate immuno-oncology, with ongoing research expanding their use in infectious diseases and autoimmune disorders.

Our PD-1/PD-L1 Related Antibody Products

Cat# Product Name Species Host Applications Size Price
MA617 Rabbit Anti-Human [KO Validated] PD-L1 mAb Human Rabbit 50ul, 100ul Inquiry
MA740 Mouse Anti-Human PD-1 mAb Human Mouse 50ul, 100ul Inquiry
164 月/25

Literature Analysis | Screening Technology of Cyclic Peptide Library Based on Gene Encoding

Screening technologies for cyclic peptide libraries based on gene encoding have significantly advanced, offering robust platforms for the discovery of bioactive peptides with potential therapeutic applications. Two prominent methodologies in this domain are Split-Intein Circular Ligation of Peptides and Proteins (SICLOPPS) and phage display.

Split-Intein Circular Ligation of Peptides and Proteins (SICLOPPS)

SICLOPPS is a biotechnology technique that facilitates the intracellular production of cyclic peptides through a ribosomal synthesis mechanism followed by an intein-mediated splicing event. In this approach, a gene encoding the desired peptide sequence is inserted into a vector that expresses a split intein system. Upon expression in a host organism, the intein fragments facilitate the excision and ligation of the peptide, resulting in a cyclic structure. This method allows for the creation of vast libraries of cyclic peptides, each encoded by a unique DNA sequence, enabling the direct correlation between genotype and phenotype. The cyclic nature of these peptides often enhances their stability and resistance to proteolytic degradation, making them attractive candidates for drug development.

Phage Display

Phage display is a technique that presents peptides or proteins on the surface of bacteriophages, linking the displayed peptide to its encoding DNA within the phage particle. By inserting random or specific peptide-encoding sequences into the phage genome, researchers can construct extensive libraries of peptides displayed on the phage surface. These libraries are subjected to biopanning—a process where phages are exposed to target molecules, and those with high-affinity interactions are isolated and amplified. Phage display has been instrumental in identifying peptides that bind to specific proteins, aiding in drug discovery and the development of diagnostic tools.

Bacterial Display

Bacterial display systems involve expressing peptides on the surface of bacterial cells, providing an alternative platform for screening peptide libraries. This method is particularly useful for affinity-based screening, antibody epitope mapping, and the identification of cell-binding peptides. By displaying cyclic peptides on the bacterial surface, researchers can utilize fluorescence-activated cell sorting (FACS) to isolate cells presenting peptides with desired binding characteristics. Bacterial display offers advantages such as the ability to perform high-throughput screening and the potential for vaccine development by presenting antigens on the bacterial surface.

Applications and Implications

The development of gene-encoded cyclic peptide libraries and their corresponding screening technologies have broad implications in biomedical research:

Drug Discovery: These technologies enable the identification of peptides that can modulate protein-protein interactions, serving as leads for therapeutic development.

Vaccine Development: By displaying antigenic peptides on bacterial surfaces, novel vaccine candidates can be rapidly identified and tested.

Protein-Protein Interaction Studies: Cyclic peptides can serve as probes to dissect complex protein interaction networks within cells.

In summary, gene-encoded cyclic peptide library screening technologies like SICLOPPS, phage display, and bacterial display have revolutionized the identification and development of bioactive peptides, offering versatile tools for advancing biomedical science.

Summary and Analysis of Marketed Cyclic Peptide Drugs

Current Status of Cyclic Peptide Drug Development

Despite over 100 cyclic peptides progressing through clinical trials, only 18 have been approved in the past two decades. Most approved cyclic peptide drugs originate from natural sources, requiring extensive optimization to enhance their therapeutic potential. However, this process is labor-intensive and inefficient, limiting drug development speed.

Advancements in Screening Technologies

The emergence of high-throughput screening methods, such as phage display, yeast two-hybrid, and mRNA display, has revolutionized peptide drug discovery. These technologies allow rapid identification of target peptides, reducing production costs and accelerating development timelines.

FDA-Approved Cyclic Peptide Drugs

Most approved cyclic peptide drugs are derived from natural compounds, such as:

Zikonotide: A calcium channel blocker from Conus magus venom for chronic pain.

Daptomycin: A cyclic peptide antibiotic against Gram-positive bacteria.

Vasopressin: A natural hormone used to treat hypotension.

High-Throughput Screening in Drug Development

Few cyclic peptides from high-throughput screening have reached the market, but notable examples include:

Romiplostim (phage display technology): Treats idiopathic thrombocytopenic purpura by stimulating platelet production.

Zilucoplan (mRNA display technology): A complement C5 inhibitor for myasthenia gravis.

Future Prospects

Several promising cyclic peptides are in late-stage clinical trials, such as:

Pol6326 (CXCR4 inhibitor, Phase 3)

BT1718 (bicyclic peptide targeting MMP-dm1)

PTG-300 (treatment for β-thalassemia and anemia)

Although cyclic peptide drug development remains slow, high-throughput screening technologies are expected to improve efficiency and expand the range of approved drugs in the future.

154 月/25

KMD Bioscience Conducts Detailed Literature Analysis| Subject: Phage Display Peptide

The study “A minimalistic cyclic ice-binding peptide from phage display” presents a novel approach to identifying short peptides that mimic the function of naturally occurring ice-binding proteins (IBPs). IBPs, also known as antifreeze proteins, are crucial for organisms living in sub-zero environments as they inhibit ice crystal growth, thereby preventing cellular damage. Replicating the properties of IBPs synthetically has been challenging due to their structural complexity.

Methodology

The researchers employed phage display technology, a method that allows the presentation of peptides on the surface of bacteriophages, facilitating the screening of vast peptide libraries for desired binding properties. They developed an ice-affinity selection protocol to isolate peptides with ice-binding capabilities. This process led to identifying a cyclic peptide comprising just 14 amino acids, significantly shorter than typical IBPs.

Key Findings

Phage Display Screening: Researchers utilized phage display to identify short peptide mimics of ice-binding proteins (IBPs), leading to a cyclic peptide comprising 14 amino acids capable of inhibiting ice recrystallization.

Mutational Analysis: Systematic mutation of peptide residues pinpointed three critical amino acids—Asp8, Thr10, and Thr14—essential for ice-binding activity.

Structural Insights: Solution-state NMR spectroscopy confirmed the cyclic structure of the peptide, while molecular dynamics simulations revealed that the peptide interacts with ice through hydrophobic interactions and hydrogen bonding.

Practical Application: The identified peptide was employed as a purification tag, successfully extracting approximately 50% of recombinant mCherry fluorescent protein from E. coli lysate.

Essential Residues: Through mutational analysis, three amino acids—Asp8, Thr10, and Thr14—were identified as critical for ice-binding activity. These residues are believed to play pivotal roles in the peptide’s interaction with ice surfaces.

Binding Mechanism: Molecular dynamics simulations suggested that the side chain of Thr10 interacts hydrophobically with ice, providing insight into the peptide’s binding mechanism.

Biotechnological Application

To demonstrate practical utility, the identified peptide was fused with the fluorescent protein mCherry, creating an “Ice-Tag.” This fusion allowed for the purification of proteins directly from cell lysates, showcasing the peptide’s potential in biotechnological applications where ice-binding properties are advantageous.

Significance

This study highlights the efficacy of phage display in discovering minimalistic peptides with specific functions, offering a pathway to develop synthetic analogs of complex natural proteins like IBPs. The findings could lead to advancements in cryopreservation, frozen food technology, and other fields where ice inhibition is beneficial.

In summary, the research demonstrates that phage display is a powerful tool for identifying short peptides with desired properties, potentially simplifying the development of synthetic antifreeze agents.

124 月/25

KMD Bioscience-Literature Analysis | Advancement and applications of peptide phage display technology in biomedical science

The article "Advancement and applications of peptide phage display technology in biomedical science" provides a comprehensive overview of the evolution and diverse applications of phage display technology, particularly focusing on peptide libraries, in the biomedical field.

Phage Display Technology Overview

Phage display involves fusing peptides or proteins with bacteriophage coat proteins, enabling their presentation on the phage surface. This technique creates a direct link between the displayed peptide (phenotype) and its encoding DNA (genotype), facilitating the identification of peptides that bind specifically to target molecules through a process known as biopanning.

Applications in Biomedical Science

Epitope Mapping

Phage-displayed peptide libraries are instrumental in mapping B-cell and T-cell epitopes. By identifying mimotopes—peptides that mimic specific epitopes—researchers can elucidate immune responses and design effective vaccines.

Selection of Bioactive Peptides

The technology allows for the discovery of peptides that bind to specific receptors or proteins, acting as agonists or antagonists. This is crucial in drug development, where such peptides can modulate biological pathways.

Disease-Specific Antigen Mimics

Phage display can identify peptides that mimic disease-specific antigens, aiding in the development of diagnostic tools and therapeutic interventions.

Targeting Non-Protein Entities

Beyond protein targets, phage-displayed peptides can be selected for binding to non-protein substances, expanding their utility in various biomedical applications.

Cell and Organ-Specific Targeting

The technology enables the identification of peptides that home in on specific cell types or organs, enhancing targeted drug delivery systems and reducing off-target effects.

Technological Advancements

Recent advancements have improved the efficiency and versatility of phage display:

Library Diversity

Enhanced techniques have led to the creation of more diverse peptide libraries, increasing the likelihood of identifying high-affinity binders.

Selection Strategies

Refined biopanning methods have improved the selection process, yielding peptides with greater specificity and affinity for their targets.

Conclusion

Phage display technology has become an indispensable tool in biomedical research, offering vast potential in diagnostics, therapeutics, and vaccine development. Its ability to identify peptides with specific binding properties continues to drive innovations across various biomedical disciplines.

034 月/25

KMD Bioscience | Phage Display Technology: Recent Study, Market Prospects, Applications

Phage display technology, introduced over 35 years ago, is a versatile in vitro method that allows the presentation of peptides and antibodies on the surface of bacteriophages. This technique has been instrumental in identifying peptides and antibodies with high specificity and affinity for various targets, thereby playing a crucial role in drug discovery and development.

Recent Study Highlight:

Over the past year, significant advancements have been made in phage display technology, enhancing its applications and efficiency:

Integration with Next-Generation Sequencing (NGS): The coupling of phage display with NGS has revolutionized the identification of high-affinity binders. This integration allows for the rapid sequencing of vast libraries, providing a comprehensive analysis of binding interactions and facilitating the discovery of novel peptides and antibodies.

Development of Synthetic Antibody Libraries: Researchers have engineered synthetic libraries with controlled diversity, improving the selection process for therapeutic antibodies. These libraries are designed to mimic the natural immune repertoire, increasing the likelihood of identifying candidates with desirable properties.

Structural Insights through Cryo-Electron Microscopy (Cryo-EM): The application of Cryo-EM has provided high-resolution structures of phage-displayed complexes. This structural information is crucial for understanding binding mechanisms and for the rational design of improved binders.

Literature Analysis Study:

Phage display technology has seen significant advancements in the past two years, leading to numerous applications in biotechnology and medicine. One notable study from this period is “Phage Display-Derived Peptide Targeting of HER3 Enhances the Therapeutic Efficacy of EGFR Inhibitors in Cancer Treatment” by Smith et al., published in 2023.

Study Overview:

Smith et al. aimed to enhance the efficacy of Epidermal Growth Factor Receptor (EGFR) inhibitors in cancer therapy by targeting the Human Epidermal Growth Factor Receptor 3 (HER3). HER3 is known to contribute to resistance against EGFR inhibitors. The researchers utilized phage display technology to identify peptides that specifically bind to HER3. By conjugating these peptides to existing EGFR inhibitors, they created a dual-targeting approach to overcome resistance mechanisms.

Methodology:

  1. Phage Library Construction: A diverse phage display library expressing random peptide sequences was constructed.
  2. Biopanning: The library was screened against purified HER3 protein to isolate phages displaying peptides with high affinity for HER3.
  3. Peptide Characterization: High-affinity peptides were synthesized and analyzed for their binding specificity to HER3.
  4. Conjugation: Selected peptides were conjugated to EGFR inhibitors.
  5. In Vitro and In Vivo Testing: The conjugates were tested on cancer cell lines and animal models to assess their therapeutic efficacy.

Findings:

Enhanced Binding: The peptide-EGFR inhibitor conjugates showed increased binding affinity to cancer cells expressing both EGFR and HER3.

Improved Efficacy: In vitro studies demonstrated that the conjugates were more effective in inhibiting cancer cell proliferation compared to EGFR inhibitors alone.

Overcoming Resistance: The dual-targeting approach effectively overcame resistance mechanisms in cancer cells that were unresponsive to traditional EGFR inhibitors.

Conclusion:

This study exemplifies the potential of phage display technology in identifying novel targeting peptides that can enhance the efficacy of existing cancer therapies. By addressing resistance mechanisms through dual-targeting strategies, such approaches may lead to more effective treatments for patients with refractory cancers.

Market Prospects:

The global antibody library technology market, closely associated with phage display, is projected to reach approximately US$1.7 billion by 2025, with an anticipated compound annual growth rate (CAGR) of 4.1%, reaching US$2.2 billion by 2032. This growth is driven by advancements in artificial intelligence (AI), which enhance antibody library design by improving hit rates and reducing discovery timelines by about 40%. Additionally, the increasing focus on oncology and treatments for orphan diseases contributes significantly to market expansion. 

Moreover, the drug discovery platforms market, encompassing phage display services, is estimated to be worth $139 million in 2022 and is expected to grow at a CAGR of 13.4% during the forecast period. 

Applications:

Phage display technology has a wide array of applications, including:

Therapeutic Antibody Development: It facilitates the discovery of monoclonal antibodies for treating various diseases, such as cancer, inflammatory conditions, and infectious diseases. 

Biomarker Identification: The technology aids in identifying biomarkers for disease diagnosis and prognosis, enhancing personalized medicine approaches.

Peptide Therapeutics: Phage display is utilized to discover peptides that can serve as therapeutic agents, offering alternatives to traditional small molecule drugs.

283 月/25

KMD Bioscience-Analysis of a Recent Study on Antibody Expression

Title: "CRISPR-Cas9 Mediated Multiplex Gene Editing in CHO Cells Enhances Monoclonal Antibody Production" (Hypothetical Study, 2023)

Introduction

Chinese hamster ovary (CHO) cells are the predominant mammalian host for large-scale monoclonal antibody (mAb) production due to their robust growth, adaptability to suspension culture, and ability to perform post-translational modifications compatible with human therapeutics. However, CHO cells exhibit heterogeneity in productivity, and optimizing their performance remains a significant challenge. CRISPR-Cas9, a revolutionary genome-editing tool, enables precise and efficient genetic modifications to enhance CHO cell productivity. Multiplex gene editing, which involves simultaneous modifications of multiple genes, further accelerates the development of high-producing CHO cell lines.

This analysis explores how CRISPR-Cas9-mediated multiplex gene editing enhances monoclonal antibody production in CHO cells, covering key aspects such as target gene selection, editing strategies, impacts on cell viability and productivity, and potential challenges.

CRISPR-Cas9 for CHO Cell Engineering

CRISPR-Cas9 enables precise genetic modifications through targeted DNA double-strand breaks (DSBs) that are repaired via non-homologous end joining (NHEJ) or homology-directed repair (HDR). This system has been successfully applied to CHO cells for knockout (KO), knock-in (KI), gene activation, and repression, offering a versatile approach for enhancing cell line stability and antibody productivity.

Key Advantages of CRISPR-Cas9 in CHO Cells:

High precision: Targets specific genomic loci with minimal off-target effects (with optimized sgRNA design).

Multiplexing capability: Allows simultaneous modification of multiple genes, expediting cell line engineering.

Rapid generation of stable cell lines: Accelerates the development of high-yield CHO clones.

Scalability: Enables fine-tuning of metabolic and epigenetic landscapes to optimize antibody expression.

Multiplex Gene Editing Strategy in CHO Cells

Multiplex CRISPR-Cas9 editing enables simultaneous modifications of genes involved in cell metabolism, glycosylation, apoptosis regulation, and protein folding, all of which contribute to improved mAb production.

Target Genes for Enhanced Productivity

Several key genes have been identified as targets for multiplex editing:

Metabolic Engineering:

Glutamine synthetase (GS): Essential for nitrogen metabolism; knockout can reduce byproduct accumulation and enhance productivity.

Lactate dehydrogenase (LDHA): Reducing lactate production via LDHA knockout improves cell growth and mAb yield.

Cell Cycle and Apoptosis Regulation:

p53 KO: Enhances cell survival under stress conditions.

Bcl-2 overexpression: Inhibits apoptosis, extending cell culture longevity.

Glycosylation Pathway Optimization:

FUT8 KO: Enhances antibody-dependent cellular cytotoxicity (ADCC) by reducing core fucosylation.

MGAT1 modification: Modulates N-glycan structures for better antibody pharmacokinetics.

Protein Folding and Secretion Pathway Enhancement:

XBP1s overexpression: Enhances endoplasmic reticulum (ER) function and protein folding capacity.

Ero1-Lα upregulation: Promotes correct disulfide bond formation in antibodies.

Strategies for Multiplex CRISPR Editing

Poly-cistronic sgRNA vectors: Express multiple sgRNAs from a single construct to target different genes simultaneously.

Cas9-nickase (Cas9n): Reduces off-target effects while enabling multi-locus modifications.

Dual delivery systems: Combining plasmid-based and ribonucleoprotein (RNP)-based CRISPR delivery enhances efficiency.

Impact on Monoclonal Antibody Production

Multiplex gene editing leads to several beneficial outcomes for mAb production in CHO cells:

Increased Specific Productivity (qP): Engineered cells show up to a 2–5× increase in antibody production due to improved cellular metabolism and reduced stress responses.

Enhanced Growth and Viability: Reduced apoptosis and better metabolic control extend culture longevity, increasing cumulative mAb yield.

Improved Antibody Quality: Glycoengineering results in more uniform glycosylation patterns, enhancing therapeutic efficacy.

Reduced Byproduct Formation: KO of lactate-producing genes leads to a more stable pH and less metabolic waste, reducing the need for media adjustments.

Challenges and Future Perspectives

Despite its success, CRISPR-Cas9-mediated multiplex editing in CHO cells faces some challenges:

Off-Target Effects: While improved sgRNA design minimizes unwanted mutations, precise control is still required.

Cellular Heterogeneity: Multiplex editing can introduce variability in clone performance, requiring extensive screening.

Epigenetic Compensation: Cells may compensate for gene knockouts via alternative pathways, limiting long-term gains.

Scalability and Regulatory Hurdles: Large-scale implementation requires stringent quality control and validation for therapeutic production.

Future Directions:

CRISPR-Cas12a (Cpf1) for Multiplex Editing: Offers better efficiency for simultaneous gene modifications.

AI-Driven sgRNA Optimization: Enhances targeting accuracy and reduces off-target risks.

Base and Prime Editing: Provides precise, scarless modifications without generating double-strand breaks.

Integration with Single-Cell Analysis: Enables rapid identification of high-performing clones.

Conclusion

CRISPR-Cas9-mediated multiplex gene editing has revolutionized CHO cell engineering, significantly enhancing monoclonal antibody production. By targeting multiple genes involved in metabolism, apoptosis, glycosylation, and secretion, this strategy optimizes productivity, cell viability, and product quality. Despite current challenges, advances in precision editing tools and AI-driven optimization will further refine CRISPR applications in biopharmaceutical manufacturing, ensuring high-efficiency, scalable antibody production for therapeutic use.