Unlocking the Future: Cancer Treatment through Personalized Medicine


Cancer patient treatment has been fundamentally transformed in recent years, as various molecular changes have been identified as the cause of cancer formation and progression. One of the most significant advancements in modern oncology is the shift from an organ-centric philosophy guiding therapy choice to thorough molecular analysis driving a personalized strategy (Gambardella et al., 2020). The term personalized medicine was used ten years ago for the first time, the right drug for the right patient explains best. ‘Personalized medicine' refers to the adaption of medical treatment to each patient's unique traits. It doesn’t mean the development of patient–specific medications or medical devices, but rather the ability to divide individuals into subpopulations that differ in their susceptibility to a given disease or their reaction to a specific treatment (Bates, 2010). Cancer therapy response rates are among the lowest of any major disease, however, now a well-established genomic basis of cancer pathology, has long put cancer research at the cutting edge of personalized medicine.  Herceptin in breast cancer and Gleevec in chronic myeloid leukemia (CML) are successful examples in the oncology world of personalized medicine (Bates, 2010).

Cancer rates are rapidly increasing, but treatment progress has been slow, yielding only modest benefits lasting weeks to months. Traditionally, doctors rely on pathology, symptoms, and medical history. Genetic changes, either hereditary or acquired, can trigger cancer. Hereditary cases, a key aspect of medical genetics, are understood through cancer genetics. Most cancers (85-90%) result from environmental factors, infections, and lifestyle, with familial cases accounting for 10-15%. These insights help predict an individual's cancer risk. Despite DNA similarity, genes function uniquely in different organs, impacting cancer behavior. Even with shared DNA, tumors differ in gene expression. Technologies like gene-expression microarrays reveal cancer-associated profiles. Modern personalized medicine considers genetics and medical history, unlike traditional methods based on family, environment, and lifestyle (Verma, 2012).

As advanced genetic and genomic technologies completely transform our methods for prognosis, screening, and precise therapeutic interventions, the era of personalized and predictive medicine has not only shaped the current landscape of clinical practice but also anticipates its practice in the future. The foundation of personalized medicine leans on the complex analysis of clinical molecular markers which can be prognostic, predictive, pharmacodynamic, and diagnostic (Ong et al., 2012).

While genomics provides a strong foundation for creating precise strategies in treating cancer patients, it's obvious that we require detailed insights into the molecular traits and features of tumors. This is critical for enhancing our approach to precision medicine. In certain circumstances, the use of proteomics, which examines proteins, becomes beneficial, especially when there are multiple molecular changes present, making it tricky to pinpoint the most important one to target. In this context, the Human Proteome Project aimed to create profiles of peptides and proteins in healthy individuals and compare them with cancer patients' profiles. Genes hold function potential, but proteins carry out actual functions. Thus, grasping protein profiles and their changes in normal and cancerous conditions is vital. Many FDA-approved drugs target proteins. While mutations trigger cancer, proteins and enzyme-guided signals drive its progression (Verma, 2012). Metabolomics, a recent inclusion in personalized medicine, explores small molecules or metabolites in cells and biological systems. The metabolome reflects the outcomes of biological processes, making it a better indicator of a cell's function compared to other "omics" like genomics or proteomics (Verma, 2012).

Due to advanced technologies, different potential targets are being uncovered for new medications. Some of these compounds have recently gained approval or are actively being studied for their effectiveness (Gambardella et al., 2020). A better understanding of cancer's molecular causes, developments in molecular diagnostics, and proteomic technologies, which are being enhanced by the availability of nanobiotechnologies, all contribute to a more personalized approach to cancer management. The most significant influence of nanobiotechnology is on molecular diagnostics for cancer, often known as nanodiagnostics (Jain & treatment, 2005).

For a while now, Oncologists have understood that each individual with cancer is different in terms of how the disease shows up, its progression, tumor’s reaction to treatment, and how well they endure it. This uniqueness also extends to varying risks of cancer returning, developing another cancer, and facing long-term treatment effects. Each form of cancer is essentially made up of different biological categories that contrast how they act clinically and their reaction to therapies (Schilsky, 2010).

Another important aspect in the progress of personalized medicine is the regulatory consequences. A huge number of exploratory biomarkers can be studied during clinical trials, however, to make therapeutic decisions such as stratification for randomized studies, the used test should be done in a Clinical Laboratory Improvement Amendments–certified environment. Moreover, new clinical trial designs to identify and authenticate biomarkers and targeted therapeutics require instructions from regulatory committees in institutions and at the US Food and Drug Administration to ensure that effective approaches reach patients efficiently without undermining their safety (Gonzalez-Angulo, Hennessy, & Mills, 2010).

Personalized treatment in Phase I clinical trials has resulted in improved patient outcomes. Patients selected based on a specific biomarker exhibit better response rates and longer progression-free survival compared to those without the biomarker (Krzyszczyk et al., 2018). Personalized cancer care is promptly becoming a practical aspect of assessing and treating patients in clinical settings. Consequently, it will enhance treatment effectiveness, decrease side effects, and lower expenses (Schilsky, 2010).

 By; Khadeeja Qadeer

 

References:                                   

 

Bates, S. J. D. d. t. (2010). Progress towards personalized medicine. 15(3-4), 115-120.

Gambardella, V., Tarazona, N., Cejalvo, J. M., Lombardi, P., Huerta, M., Roselló, S., . . . Cervantes, A. J. C. (2020). Personalized medicine: recent progress in cancer therapy. 12(4), 1009.

Gonzalez-Angulo, A. M., Hennessy, B. T., & Mills, G. B. J. J. o. c. o. (2010). Future of personalized medicine in oncology: a systems biology approach. 28(16), 2777.

Jain, K. J. T. i. c. r., & treatment. (2005). Role of nanobiotechnology in developing personalized medicine for cancer. 4(6), 645-650.

Krzyszczyk, P., Acevedo, A., Davidoff, E. J., Timmins, L. M., Marrero-Berrios, I., Patel, M., . . . Hartmanshenn, C. J. T. (2018). The growing role of precision and personalized medicine for cancer treatment. 6(03n04), 79-100.

Ong, F. S., Das, K., Wang, J., Vakil, H., Kuo, J. Z., Blackwell, W.-L. B., . . . Rotter, J. I. J. E. r. o. m. d. (2012). Personalized medicine and pharmacogenetic biomarkers: progress in molecular oncology testing. 12(6), 593-602.

Schilsky, R. L. J. N. r. D. d. (2010). Personalized medicine in oncology: The future is now. 9(5), 363-366.

Verma, M. J. J. o. p. m. (2012). Personalized medicine and cancer. 2(1), 1-14.

 

 

Comments

Popular posts from this blog

Characteristics and therapeutic properties of lactoferrin protein

Hydrogels

Registration Form for Volunteer Batch 2k22