What is Cancer Functional Genomics?

Functional genomics in cancer research delves into the intricate interplay of genetic, epigenetic, and transcriptional modifications and their impact on cancer phenotypes. This field employs advanced sequencing techniques to scrutinize the dynamic alterations shaping cancer development and its responsiveness to therapeutic interventions.

Traditionally, our comprehension of the correlation between cancer DNA and its biological manifestations relied on a limited set of pivotal cancer driver genes. Mutations in these genes trigger the activation of pathways promoting tumor growth. The identification of these potent drivers has been pivotal in devising targeted therapies that counteract their tumor-promoting effects. Notable successes include imatinib, addressing the BCR-ABL fusion protein in chronic granulocytic leukemia, and trastuzumab, targeting HER-2 overexpression in breast cancer.

While these achievements underscore the effectiveness of targeting specific genes, the paradigm of single drug targets is approaching its limits. Breakthroughs in pinpointing individual cancer genes are challenging due to the intricate nature of most cancer genomes. The complexity lies in the synchronized impact of multiple mutations under genetic conditions, yielding diverse cancer phenotypes throughout disease progression. Researchers are compelled to adopt a holistic approach, scrutinizing the entire genome to unravel how these mutations collectively shape cancer biology.

Decoding Cancer Genomic Landscapes

In traditional cancer research, emphasis has long been on identifying mutational patterns, linking them to factors like senescence, homologous recombination defects, and environmental exposures. These mutations in genomic stability drivers can trigger downstream mutations in multiple cancer genes.

Recent studies build on these concepts, revealing how complex mutational landscapes in cancer stem from a single initiating event. These landscapes include rearrangement events, signatures, and duplications, involving classical oncogenes and tumor suppressors cooperating in tumorigenesis.

Adding to the complexity is human germline variation, influencing cancer risk. While specific susceptibility genes play a role, genome-wide association studies highlight SNPs tied to increased cancer risk. Polygenic risk scores quantify cumulative allele effects but lack the gene specificity seen in other approaches, limiting their ability to assess increased cancer risk with precision.

Systems Biology Unravel the Cancer Genome-Transcriptome-Phenotype

Recent insights underscore that cancer phenotypes seldom hinge on individual gene mutations; rather, they predominantly emerge from a mosaic of genomic perturbations. The intricacy of these combinations stems not just from their quantity but also from the diversity of genetic and transcriptional disturbances—ranging from activating mutations, chimeric fusions, and expression changes to splicing variants, regulatory region mutations, and proteomic alterations. The challenge at hand is to seamlessly integrate this wealth of mutational data into robust, mechanism-driven models capable of predicting biological vulnerabilities and therapeutic avenues.

Addressing these combinatorial intricacies, systems biologists are pioneering novel methods to map and model protein and gene interaction networks. The efficacy of these models hinges on access to extensive datasets encompassing genomic and functional associations. Recently, the synergy of systems biology with machine learning has proven successful in accurately predicting treatment responses in cancer patients. Moreover, this integrated approach has identified immunotherapy response biomarkers in actual patients and proposed potential mechanisms underlying drug resistance.

The Prospects for Functional Genomics in Tumor Research

As elucidated earlier, the landscape of cancer functional genomics hinges on (1) the intricate interplay of mutations in relevant coding genes, shaping the cancer phenotype, (2) transcriptional signatures influencing key pathways, (3) genome-wide mutational signatures determining the cancer's origin, and (4) disruptions across the genome activating immune responses through increased antigenic load.

Moreover, the tumor's evolution over time and the robustness of each cancer progeny plays pivotal roles in determining the potential for cure. Precise computational assessments are essential to decode the full cancer life cycle, though the field is in its early stages, requiring more reliable methods. Notably, monitoring the dynamics of tumor evolution to determine the on-off time of anti-androgen treatment in prostate cancer has yielded promising clinical results, hinting at a positive future trajectory.

Envisioning a future where all regulatory and structural mutations in the cancer genome can be detected and their functions understood, coupled with the identification of measures of genomic instability, opens the door to more refined computational methods. These advancements hold promise for predicting cancer responsiveness and potential curability, even in complex genomes. Additionally, exploring mechanisms that target the maintenance of high genomic instability may curb tumor heterogeneity and constrain the emergence of new mutations. This tantalizing prospect could inspire the development of novel adjuvant cancer therapies, ultimately enhancing the curative potential for complex malignancies.

For Research Use Only. Not for use in diagnostic procedures.
Related Services
Speak to Our Scientists
What would you like to discuss?
With whom will we be speaking?

* is a required item.

Contact CD Genomics
Terms & Conditions | Privacy Policy | Feedback   Copyright © CD Genomics. All rights reserved.
Top