Blood-based biomarkers in biopharmaceutical applications

Blood-based biomarkers are helpful in biopharmaceutical applications such as drug research, development, and personalized medicine. A cancer biomarker refers to a tumor characteristic or a response of the body in the presence of cancer that can be objectively measured and evaluated. This most commonly involves the assessment of a genomic alteration or a difference in protein expression levels1. Pharmacokinetic, pharmacodynamic, and pharmacogenomic biomarkers are for evaluation of the interaction between the drug and patient2.

One main role that biomarkers have is stratifying individuals in clinical studies. Stratification ensures that individuals have similar biomarker profiles, and the process assists healthcare providers in identifying categories of patients more likely to react to a particular medication – thus resulting in more efficient and targeted clinical trials.

Additionally, blood-based biomarkers allow for the early diagnosis of illnesses or ailments, frequently before symptoms appear. Early diagnosis allows for prompt action, which may improve prognosis and treatment results. Measurable markers in the blood, tissue, or other biological samples can prove the existence of a specific disease or condition, which helps with differential diagnosis.

In this article, we will explore in depth the roles that blood-based biomarkers play in biopharmaceutical research and study – in particular, how thymidine kinase aids in cancer monitoring and treatment, and how biomarkers can be integrated into clinical trials.

Blood-based biomarkers in pharmaceutical applications

Different types of blood-based biomarkers are essential in a variety of pharmaceutical applications, as they identify helpful information for drug research, development, and patient care.

Below, we explore how they provide insights into the physiological or pathological processes connected with illnesses by reviewing some common pharmaceutical applications of blood-based biomarkers.

Disease detection and early-stage monitoring

Blood-based biomarkers are frequently utilized for illness identification in the early stages. This allows for timely intervention and treatment start while the condition is more manageable.

Biomarkers also identify illnesses with identical symptoms allowing for more accurate and tailored treatment strategies. Blood tests are a less intrusive technique to gather diagnostic information than more invasive treatments, which improves patient comfort and lowers the risk of contamination of other health issues.

In disease detection and early-stage monitoring, specific blood biomarkers act as predictive indicators, assisting in predicting the expected course of a disease. Serial blood biomarker measures also enable doctors to track illness development over time and provide insight into the condition’s dynamic.

Treatment response tracking

It is now widely accepted that compounds that target driver genomic alterations involved in cancer initiation and progression can be successfully pursued as rational therapeutic strategies2. Nevertheless, there are still only a limited number of effective targeted therapies associated with robust predictive biomarkers of response for the treatment of patients with solid tumors. In all stages of disease progression, changes in blood biomarker levels can be used as early markers of therapy response which enables fast modifications to improve therapy effects. They support adaptive treatment techniques and doctors modifying treatment regimens based on real-time patient response data.

Customization of treatment strategies

Personalized cancer medicine is based on increased knowledge of the cancer mutation repertoire and the availability of agents that target altered genes or pathways3. Going hand in hand with treatment response tracking, blood-based biomarkers allow for the customization of treatment strategies. Amplification of the human epidermal growth factor receptor 2 (HER2) gene and attendant protein overexpression are present in 10% to 20% of primary breast cancers2. HER2 status predicts sensitivity to anthracycline-based chemotherapy regimens4 as well as relative resistance to cyclophosphamide-based regimens and tamoxifen-based therapies in the setting of estrogen receptor–positive breast cancers.

Breast cancers with HER2 alterations are now treated with targeted therapies such as trastuzumab and lapatinib, which have been shown to markedly improve response rate and survival when added to chemotherapy or as monotherapy5.

These biomarkers make interventions that match the unique characteristics of each patient and condition. Early diagnosis and pursuit of blood-based biomarkers allow for prompt treatments and alteration of disease development.

Accurate therapeutic development

For medical researchers and practitioners, blood-based biomarkers help to identify patient subpopulations with comparable molecular characteristics creating room for more exact segmentation of clinical trials. This ensures that individuals more likely to react to a particular medication are included.

For example, in biomarker testing for cancer treatment, patients with breast cancer are grouped according to the expression of these biomarkers. HER2-positive individuals may respond to HER2-targeted medicines such as trastuzumab, but ER-positive patients may benefit from endocrine therapy such as tamoxifen.

Identification of prospective therapeutic targets

Blood-based biomarkers are essential in the identification of prospective therapeutic targets. Blood samples from individuals with a particular ailment are analyzed to discover molecular targets linked with the condition. They are used to confirm the efficacy of specified medication targets. Confirmation that changes in these indicators are associated with illness development enhances the argument for a specific target.

For example, in the broader context of medicine, elevated levels of beta-amyloid or tau proteins in the blood can suggest Alzheimer’s disease. These indicators are important in identifying possible therapeutic targets that reduce the accumulation of these proteins and decrease disease progression.

Real-time analysis of patient responses

Finally, blood-based biomarkers are used to analyze a medication’s pharmacodynamics providing information about how the medicine impacts various pathways or processes in the body. Changes in blood biomarker levels indicate therapy response and can provide real-time input for researchers and physicians to assess the efficacy of the medicine.

Again, in the broader context of medicine, blood indicators such as CRP and ESR measure inflammation levels in rheumatoid arthritis patients. A reduction in these indicators after therapy with anti-inflammatory medicines implies that the treatment was effective.

The benefits of biomarker testing in pharma

The multitude of benefits that biomarker testing offers in pharmaceutical research and development can all lead to eventual, increased precision in the medical approach.

Since pharmacogenomic techniques are guided by genetic indicators in the blood, they can assist in identifying patients who are more likely to react positively to specific treatments.

Patient stratification and blood-based biomarkers in early-stage studies can also enable more efficient and focused enrollment, resulting in shorter development schedules focusing on those most likely to benefit.

Thymidine kinase activity as a blood-based biomarker

Thymidine kinase, a promising biomarker, is an enzyme critical for DNA synthesis and, essentially, a marker of cell proliferation6. Studies have shown that patients with metastatic breast cancer have higher levels of TK compared with healthy controls7 and that higher serum TK1 activity is associated with poorer breast cancer outcomes.

Measuring TK activity in the blood can aid in detecting and surveillance as a breast cancer biomarker to determine the efficacy of breast cancer treatments. Typically, a reduction in TK activity after medication indicates a good response to treatment, whereas consistently high levels may signal resistance or disease progression.

Benefits of thymidine kinase biomarker testing

Some unique benefits come with measuring TK activity in the bloodstream of breast cancer patients. Serum TK1 activity exhibits several advantages as a biomarker, such as analytical and clinical validity7 as well as ease to obtain serial samples compared with Ki67, which is a tissue-based marker.

TK activity changes in response to therapy. Surveillance of TK activity enables real-time evaluation of therapeutic effectiveness. For example, a reduction in TK activity may signal a favorable response to chemotherapy in leukemia treatment.

Additionally, the trace of TK activity in the blood of HSV-infected individuals can be used as a pharmacodynamic measure in medication development. A drop in TK activity after antiviral medication administration might imply an effective reduction in viral reproduction.

Integrating blood-based biomarkers into clinical trials and research

In the past 10 to 15 years, cancer clinical trials have experienced some important paradigm changes to embrace the era of precision oncology as defined by various biomarkers8.

With the advent of the targeted therapy era, molecular biomarkers in particular are becoming increasingly important within both clinical research and clinical practice9. It not only allows for a more individualized and precise approach to diagnoses and therapy but provides vital information that can be used for further pharmaceutical development of treatment plans and innovations.

Biomarker research divides patients into subgroups with comparable molecular traits creating better chances for more homogenous research populations. This has the potential to improve the capacity to detect treatment effects and the overall efficiency of clinical studies, such as in the creation of breast cancer treatment innovations.

Pharmacogenomic techniques are also guided by genomic biomarker blood tests, which enable medication selection based on individual genetic variances – this can promote the development of customized medicine tactics in clinical trials and breast cancer research.

Challenges in biomarker-research integration

There is currently an overwhelming interest in biomarker research within both the natural and clinical sciences. Evidence of this effort is clear, as the volume of literature devoted to biomarker discovery, characterization, and validation continues to trend with an annual expansion. However, integrating biomarker research poses several challenges due to the complexity of biological systems and the multifactorial nature of disease.

Even though a lot of sophisticated bioinformatic techniques can assist distinguish between passenger alterations and cancer causes, intratumoral heterogeneity continues to be a major barrier to the identification and development of new biomarkers. Population-level genetic changes are often identified as cancer drivers, but individual patient variability, including tumor immunology, hinders replication of findings10.

There are also challenges in the implementation of biomarkers in clinical drug development. Future clinical trials must consider co-alterations driving resistance to single-agent therapies, integrating anticancer drugs based on tumor biology and pharmacology11.

Differences in laboratory procedures and test platforms might result in strange findings, and many biomarkers blood tests, still in stages of development, need more thorough validation before they can be trusted in their reliability and repeatability in usage. Individual biomarker levels may also be influenced by individuals’ inherent biological diversity, which may obfuscate the treatment process.

Considerations in navigating these challenges

Some considerations – such as the standardization of techniques, reference materials, and quality control procedures – can solve some of these problems, and many medical researchers have put them in place.

Collaboration and adherence to established principles can also reduce test variability. Thorough validation studies are conducted before blood-based biomarkers are widely used. This includes large-scale multicenter trials that are undertaken to verify their reliability and repeatability, and correct interpretations of biomarker data require patient stratification based on standardized clinical and molecular parameters.

Ensuring accurate and ethical biomarker data

Integrating blood-based biomarkers into clinical practice and research entails addressing several standards to assure the reliability, repeatability, and ethical use of biomarker data. These methods include standardized protocols, peer validation, and EU regulations.

Standardized protocols

Standardization reduces variability in biomarker tests. Protocols that researchers take must be – and are – thorough, with information on sample collection techniques, storage conditions, and analysis methods. Consensus recommendations are also followed, such as those issued by relevant scientific institutions or international organizations.

Peer validation

Biomarkers are validated separately by various research groups. Peer-reviewed papers, particularly in respected journals, demonstrate the biomarker’s validity and reliability, and individual, concurring research strengthens the credibility of biomarker data and findings.

Compliance with EU regulations

Biomarker development and use in the European Union are governed by rules, particularly by the European Medicines Agency (EMA). Compliance with Good Clinical Practice (GCP) guidelines, applicable directives, and legislation is vital.

Final words

Blood-based biomarkers play a multifaceted role in biopharmaceutical applications, from early drug discovery to patient stratification, surveillance, and the development of personalized therapies. Their use contributes significantly to the advancement of precision medicine and the overall effectiveness of pharmaceutical interventions.

Collaborate with DiviTum to improve cancer research and treatment

DiviTum is at the forefront of biomarker testing innovations with our DiviTum® TKa test, which helps oncologists make confident treatment decisions by providing insights into MBC patient response. TKa testing also provides opportunities for clinical innovations.

If you are a healthcare provider who wishes to learn more about the science behind the testing tool or details of test procurement, you can read more here. Pharma and biotechnology companies who wish to collaborate in pre-clinical, clinical, and companion diagnostic (CDx) initiatives with DiviTum can also learn more about our supporting research and agreement types.

Get in touch with us to improve breast cancer research and treatment today.

References

  1. Atkinson, A.J., et al. “Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework.” Clinical Pharmacology & Therapeutics, 69(13), pp. 89-95. Published March 1, 2001. https://www.webofscience.com/wos/woscc/full-record/WOS:000167641800001?SID=EUW1ED0F51KBOambl6ufIYusZGhVL
  2. Dancey, J. E. et al. “The Genetic Basis for Cancer Treatment Decisions.” CELL, 148(3), pp. 409-420. Published February 3, 2012. https://www.webofscience.com/wos/woscc/full-record/WOS:000300225000011?SID=EUW1ED0F51KBOambl6ufIYusZGhVL
  3. Dumbrava, E. I., Meric-Bernstam, F. and Yap, T. A. “Challenges with biomarkers in cancer drug discovery and development.” Expert opinion on drug discovery, 13(8), pp. 685–690. Published May 24, 2018. https://www.tandfonline.com/doi/full/10.1080/17460441.2018.1479740
  4. Lal, P. et al. “HER-2 testing in breast cancer using immunohistochemical analysis and fluorescence in situ hybridization: A single-institution experience of 2,279 cases and comparison of dual-color and single-color scoring”. American Journal of Clinical Pathology, 121(5), pp. 631–636. Published May 2004. https://academic.oup.com/ajcp/article/121/5/631/1759222
  5. Villman, K. et al. “TOP2A and HER2 gene amplification as predictors of response to anthracycline treatment in breast cancer.” Acta Oncologica (Stockholm, Sweden), 45(5), pp. 590–596. Published July 8, 2009. https://www.tandfonline.com/doi/full/10.1080/02841860500543182
  6. Zhou, X. et al. “Lapatinib plus capecitabine versus capecitabine alone for HER2+ (ErbB2+) metastatic breast cancer: quality-of-life assessment.” Breast cancer research and treatment, 117(3), pp. 577–589. Published January 20, 2009. https://link.springer.com/article/10.1007/s10549-009-0310-8
  7. McCartney, A. et al. “Prognostic role of serum thymidine kinase 1 activity in patients with hormone receptor–positive metastatic breast cancer: Analysis of the randomised phase III Evaluation of Faslodex versus Exemestane Clinical Trial (EFECT).” European Journal of Cancer, 114, pp. 55–66. E-published May 3, 2019. https://pubmed.ncbi.nlm.nih.gov/31059974/
  8. Hu, C. and Dignam, J. J. “Biomarker-driven oncology clinical trials: Key design elements, types, features, and practical considerations.” JCO Precision Oncology, 3(3), pp. 1–12. Published October 24, 2019. https://ascopubs.org/doi/full/10.1200/PO.19.00086
  9. Buyse, M. et al. “Integrating biomarkers in clinical trials.” Expert Review of Molecular Diagnostics, 11(2), pp. 171–182. Published March 11, 2011. https://pubmed.ncbi.nlm.nih.gov/21405968/
  10. Andre, F. et al. “Prioritizing targets for precision cancer medicine.” Annals of Oncology, 25(12), pp. 2295–2303. Published December 2014. https://pubmed.ncbi.nlm.nih.gov/25344359/
  11. Yap, T. A., Omlin, A. and de Bono, J. S. “Development of therapeutic combinations targeting major cancer signaling pathways.” Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology, 31(12), pp. 1592–1605. Published April 20, 2013. https://pubmed.ncbi.nlm.nih.gov/23509311/

PLEASE SELECT ONE

Please specify your home location