Advancement in Cancer Detection: A Leap Forward with Novel Priming Agents in Liquid Biopsies

Significance 

Circulating tumor DNA (ctDNA) refers to fragments of DNA that are released from cancer cells into the bloodstream. These DNA fragments are shed into the blood either through the apoptosis (programmed cell death) or necrosis (cell death due to injury) of tumor cells. The fragments of ctDNA are usually short, often around 160-180 base pairs. This is in contrast to circulating free DNA (cfDNA) from healthy cells, which is typically longer. The presence of ctDNA in the blood offers a unique opportunity for non-invasive cancer diagnosis and monitoring, a method often referred to as a “liquid biopsy.”  The amount of ctDNA in the blood can vary widely among patients and depends on factors such as the type and stage of cancer, the tumor burden, and the effectiveness of treatments.

Since ctDNA carries the genetic signature of the tumor, analyzing ctDNA can help in the early detection of cancer, potentially even before clinical symptoms appear. Moreover, changes in the levels of ctDNA can indicate how well a patient is responding to treatment. A decrease in ctDNA levels might suggest that the treatment is effective, while an increase could indicate progression or recurrence of the disease. Furthermore, analysis of ctDNA can help identify specific mutations within the tumor. This information can be critical for selecting targeted therapies that are more likely to be effective against cancers with certain genetic profiles. However, detecting ctDNA can be challenging, especially in early-stage cancers where the quantity of ctDNA is very low. Improvements in technology and methods are continually being made to enhance the sensitivity of ctDNA detection. As research advances, the potential applications of ctDNA in personalized medicine are expanding. This includes not only cancer detection and monitoring but also the possibility of using ctDNA profiles to guide therapy choices and predict patient outcomes. However, further studies and clinical trials are needed to fully integrate ctDNA analysis into standard oncology practice. The development of more sensitive and specific ctDNA assays will also be key in realizing the full potential of this technology in cancer care.

Liquid biopsy, a rapidly advancing technology, has revolutionized the approach to cancer diagnostics. Traditionally, tissue biopsies have been the gold standard for tumor analysis. However, they are invasive, often painful, and sometimes impossible if the tumor location is unknown or inaccessible. This limitation has driven the development of liquid biopsies, which use a simple blood draw to detect ctDNA.   The core issue with liquid biopsies is the scarcity of circulating tumor DNA in blood, which is often too limited for effective detection, especially in early-stage cancers or small tumors. Martin-Alonso et al. tackled this challenge by introducing two types of priming agents that transiently attenuate the natural clearance of cfDNA in blood. This approach resulted in a significant increase in the recovery of ctDNA from blood draws, thereby enhancing the sensitivity of liquid biopsies.

In a new study published in Science Journal and led by Dr. Viktor Adalsteinsson from the Gerstner Center for Cancer Diagnostics at Broad Institute of MIT and Harvard,  researchers focused on enhancing the sensitivity and reliability of liquid biopsies for tumor detection. Liquid biopsies, which involve analyzing cfDNA in blood samples, offer a non-invasive method to detect ctDNA. However, the inherent challenge with this approach has been the low abundance of ctDNA in blood, particularly in cases of early-stage cancers or small tumors. To address this challenge, the researchers developed two distinct types of priming agents that temporarily inhibit the natural clearance of cfDNA from the bloodstream. This strategy aimed to increase the concentration and therefore the detectability of ctDNA in a standard blood draw. Here’s an overview of their approach:

The first priming agent is based on nanoparticles that target the cells responsible for cfDNA clearance, primarily the liver-resident macrophages. The research demonstrated that these nanoparticles, by saturating the uptake capacity of these macrophages, significantly prolonged the half-life of cfDNA in circulation. The second type of agent involved DNA-binding monoclonal antibodies (mAbs). These mAbs bind to cfDNA, shielding it from enzymatic digestion, and thus prolonging its presence in the bloodstream. The effectiveness of these agents was validated in mouse models of cancer. The results were striking – a more than tenfold increase in ctDNA recovery, enabling more accurate tumor molecular profiling from blood samples. Notably, the sensitivity for detecting small tumors increased dramatically from under 10% to over 75%. This finding is particularly significant as early detection is crucial for effective cancer treatment. The implications of this study for clinical practice are profound. Priming agents could revolutionize liquid biopsies, making them a more reliable tool for early cancer detection, monitoring disease progression, and tailoring personalized treatment strategies. This advance could also extend the utility of liquid biopsies to other applications, such as prenatal testing, infectious diseases, and organ transplant monitoring. While the study presents promising results, several challenges must be addressed before these priming agents can be used clinically. First, the safety and efficacy of these agents need to be established in human trials. The interaction of these agents with human physiology, potential side effects, and long-term implications must be thoroughly investigated. Moreover, the integration of these priming agents into existing clinical workflows will require careful consideration. For instance, the timing of blood draws relative to the administration of priming agents will be crucial to maximize ctDNA recovery. Additionally, the cost-effectiveness of incorporating these agents into routine clinical practice must be evaluated. In conclusion, the study led by Dr. Adalsteinsson marks a significant advancement in the field of liquid biopsies. By enhancing the sensitivity and robustness of ctDNA testing, these novel priming agents hold the promise of transforming cancer diagnostics and management. As with any groundbreaking technology, rigorous clinical testing and thoughtful integration into healthcare systems are essential to realize their full potential. The future of cancer diagnostics looks brighter with such innovations paving the way for more accurate, non-invasive, and patient-friendly testing methods.

Advancement in Cancer Detection: A Leap Forward with Novel Priming Agents in Liquid Biopsies - Medicine Innovates

About the author

Viktor Adalsteinsson, Ph.D.
Director, Gerstner Center for Cancer Diagnostics

Viktor Adalsteinsson is the director of the Gerstner Center for Cancer Diagnostics at the Broad Institute of MIT and Harvard. He also leads the Blood Biopsy Team, a multi-institutional collaboration to profile cancer genomes directly from blood samples. The Blood Biopsy Team includes scientists, engineers, oncologists, and computational biologists spanning numerous investigators and labs at the Broad Institute, MIT, Dana-Farber Cancer Institute, Massachusetts General Hospital, and others. The goal of their research is to develop impactful new diagnostic methods that stand to benefit millions of cancer patients, such as novel approaches for cancer detection and monitoring using blood biopsies.

Adalsteinsson holds a Ph.D. in chemical engineering from MIT (J. Christopher Love lab), where he developed novel approaches for functional and genomic profiling of single cells in cancer such as circulating tumor cells. He has been an affiliate of the Broad Institute since 2011, has run his own research lab at the Broad since 2015, and established the Gerstner Center for Cancer Diagnostics with Todd Golub in 2019. Adalsteinsson has analyzed over 15,000 blood biopsies and contributed to more than 45 publications and 12 patents with 4,000 citations in the fields of cancer genomics, cancer diagnostics, and biotechnology. Adalsteinsson was honored by MIT Technology Review in 2017 as a visionary member of its 35 Innovators Under 35 and by Clinical OMICs in 2021 as one of its Pioneers Under 40.

Reference

Martin-Alonso C, Tabrizi S, Xiong K, Blewett T, Sridhar S, Crnjac A, Patel S, An Z, Bekdemir A, Shea D, Wang ST, Rodriguez-Aponte S, Naranjo CA, Rhoades J, Kirkpatrick JD, Fleming HE, Amini AP, Golub TR, Love JC, Bhatia SN, Adalsteinsson VA. Priming agents transiently reduce the clearance of cell-free DNA to improve liquid biopsies. Science. 2024 Jan 19;383(6680):eadf2341. doi: 10.1126/science.adf2341.

Go To Science.