GET THE APP

Journal of Nanomedicine & Biotherapeutic Discovery

Journal of Nanomedicine & Biotherapeutic Discovery
Open Access

ISSN: 2155-983X

+44 1300 500008

Opinion Article - (2022)Volume 12, Issue 2

A Short Note on Antibody-Drug Conjugates

Vander Sluis*
 
*Correspondence: Vander Sluis, Department of nanomedicine, University of Amsterdam, Amsterdam, The Netherlands, Email:

Author info »

About the Study

Antibody-drug conjugates are a type of biopharmaceutical medicine that is used to treat cancer as a targeted treatment. ADCs, unlike chemotherapy, are designed to specifically target and destroy tumour cells while leaving healthy cells intact. Antibody-drug conjugates are complex molecules that combine an antibody with a physiologically active cytotoxic (anticancer) payload or medication. Bio conjugates and immuno conjugates are examples of antibody-drug conjugates. ADCs combine the cancer-killing capacity of cytotoxic medicines with the targeting capabilities of monoclonal antibodies. They can be programmed to distinguish the difference between healthy and malignant tissue.

The Components of an Antibody-Drug Conjugate There are three components to an antibody-drug conjugate: Antibody-a substance that binds to the ADC and can cause a therapeutic response. The therapeutic response is elicited by the payload. The payload is attached to the antibody via a linker, which should be stable in circulation and only release the payload at the targeted target. For attachment to the antibody, many techniques of conjugation have been devised and evaluated. The drug to antibody ratio, or DAR, reflects the degree of payload loading on the ADC. Mechanism of action of an anticancer medication is combined with an antibody that particularly targets a tumour antigen (e.g. a protein that, ideally, is only to be found in or on tumour cells). Antibodies bind to the antigens on the surface of malignant cells. The biochemical interaction between the antibody and the target protein (antigen) causes a signal to be sent to the tumour cell, which absorbs or internalizes the antibody along with the associated cytotoxin. The cytotoxin destroys the cancer after the ADC is absorbed. This targeting reduces adverse effects while providing a larger treatment window than conventional chemotherapeutic drugs.

An ADC must have a stable connection between the antibody and the cytotoxic (anti-cancer) chemical. A stable ADC linker ensures that less of the cytotoxic payload is lost before reaching a tumour cell, hence enhancing safety and minimizing doses. Chemical motifs such as disulfides, hydra zones or peptides (cleavable), or thioethers are used to create linkers (noncleavable). In preclinical and clinical testing, cleavable and noncleavable linkers were shown to be safe. Brentuximab vedotin has an enzyme-sensitive cleavable linker that transports the synthetic antineoplastic drug Mono Methyl Auristatin E (MMAE) to human-specific CD30-positive malignant cells. MMAE inhibits cell division by preventing tubulin polymerization. MMAE cannot be employed as a single-agent chemotherapeutic medication due to its severe toxicity. MMAE coupled to an anti- CD30 monoclonal antibody (cAC10, a cell membrane protein of the Tumour Necrosis Factor or TNF receptor) was, on the other hand, stable in extracellular fluid. It is cathepsin-cleavable and therapy-safe. Trastuzumab emtansine is a stable, non-cleavable linker-based combination of the microtubule-formation inhibitor mertansine (DM-1) and the antibody trastuzumab.

The availability of better and more stable linkers has altered the chemical bond's function. The kind of linker, cleavable or noncleavable, influences the cytotoxic drug's characteristics. A non-cleavable linker, for example, retains the medicine within the cell. As a result, the complete antibody, linker, and cytotoxic (anti-cancer) substance enter the specific cancer cell and are digested into amino acids. The active drug is thought to be the resultant combination of amino acid, linker, and cytotoxic agent. Cleavable linkers, on the other hand, are broken down by enzymes in the cancer cell. The cytotoxic payload can then escape from the targeted cell and assault surrounding cells in a process known as "bystander killing."

Another sort of cleavable linker that is currently being developed involves the addition of an additional molecule between the cytotoxin and the cleavage site. This enables researchers to develop ADCs with greater flexibility without altering the cleavage kinetics. Researchers are working on a new peptide cleavage technique based on Edman degradation, which is a way of sequencing amino acids in a peptide. TDCs and innovative conjugation procedures are also being developed to increase the stability and therapeutic index of immuno conjugates, antibodyconjugated nanoparticles, and antibody-oligonucleotide conjugates. As the science of antibody-drug conjugates has progressed, a more precise definition of ADC is currently Anything-Drug Conjugate. Multiple smaller antibody fragments, such as diabodies, Fab, SCFV, and bicyclic peptides, are now available as antibody targeting components.

Author Info

Vander Sluis*
 
Department of nanomedicine, University of Amsterdam, Amsterdam, The Netherlands
 

Citation: Sluis V (2022) A Short Note on Antibody-Drug Conjugates. J Nanomedine Biotherapeutic Discov. 12:154.

Received: 22-Mar-2022, Manuscript No. JNBD-22-16719; Editor assigned: 24-Mar-2022, Pre QC No. JNBD-22-16719 (PQ); Reviewed: 07-Apr-2022, QC No. JNBD-22-16719; Revised: 13-Apr-2022, Manuscript No. JNBD-22-16719(R); Published: 20-Apr-2022 , DOI: DOI:10.4172/2155-983X.1000153

Copyright: © 2022 Sluis V. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Top