Blogs

Genomic Profiling's Role in Gynecological Cancer Immunotherapy

In the rapidly evolving landscape of gynecological cancer treatment, genomic profiling is emerging as a powerful tool that uncovers the unique genetic makeup of each patient's tumor, enabling personalized treatment strategies that combine immunotherapy and targeted therapies. This approach holds immense promise for a future of tailored cancer treatments, optimizing treatment efficacy while minimizing off-target effects.

The Role of Genomic Profiling

Genomic profiling involves analyzing the entire genome or targeted genomic regions derived from tumor specimens. This comprehensive analysis aims to uncover a wide array of genetic aberrations, including mutations and chromosomal rearrangements, that underpin the onset and progression of gynecological malignancies. By showing the genetic landscape of tumors, genomic profiling not only facilitates the identification of therapeutic targets but also paves the way for the development of personalized treatment regimens tailored to the unique genetic makeup of an individual patient's tumors.

Genomic profiling can reveal:

  • Somatic Mutations: Can activate oncogenes, leading to uncontrolled cellular proliferation, or inactivate tumor suppressor genes, resulting in unchecked growth.

  • Gene Expression Profiling: Quantifying aberrant over or underexpression of genes, unveiling key molecules that can influence gynecological cancer development and progression.

  • Genomic Rearrangements: Large-scale chromosomal changes disrupt normal gene function, causing gene fusions, deletions, or duplications, which contribute to cancer.

Genetic profiling makes it possible to:

  • Identify oncogenic drivers: These key genes act as the engine of the tumor's growth. Once identified, these oncogenic drivers become prime targets for specific therapeutic interventions, such as targeted therapies or gene editing approaches.

  • Select targeted therapies: Understanding the specific mutational landscape of a patient's cancer helps clinicians choose targeted therapies that are likely to be effective against that particular tumor. This personalized approach optimizes treatment efficacy while minimizing off-target effects.

By uncovering the unique genetic makeup of each patient's cancer, we can move beyond a "one-size-fits-all" approach and tailor treatment strategies to maximize therapeutic benefit for individual patients and potentially minimize unnecessary toxicities.

POLE Mutations and Checkpoint Inhibitors in Lynch-Associated Gynecological Cancers

DNA polymerase epsilon (POLE) mutations and microsatellite instability (MSI) can influence the response to immune checkpoint inhibitors in gynecological cancers, particularly in endometrial cancer:

  • POLE is a DNA replication and proofreading gene. Mutations in POLE can lead to a very high tumor mutational burden, resulting in "POLE-ultra mutated" endometrial cancers.

  • POLE-ultramutated endometrial cancers have many tumor-infiltrating lymphocytes and increased expression of immune checkpoint proteins like PD-1 and PD-L1, suggesting they may be responsive to immune checkpoint blockade.

Lynch syndrome, also known as hereditary non-polyposis colorectal cancer (HNPCC), is an inherited condition that increases the risk of various cancers, including endometrial and ovarian cancers. It is characterized by defects in DNA mismatch repair genes (MLH1, MSH2, MSH6, PMS2), leading to microsatellite instability (MSI). MSI-high (MSI-H) tumors, including those associated with Lynch syndrome, have a high mutational burden and increased neoantigen expression, making them potentially responsive to immune checkpoint inhibitors.

Cancer Biomarkers and Immunotherapy Efficacy

The effectiveness of immunotherapy in gynecological cancers is linked to the detection of specific cancer biomarkers that indicate the likelihood of response to immunotherapy. Key biomarkers include:

  • Tumor mutational burden (TMB): The number of mutations in a tumor. This number is correlated with the susceptibility to checkpoint inhibitor immunotherapy.

  • DNA repair gene mutations: Mutations can influence response to immunotherapy.

  • Immune profiling of the tumor microenvironment: Analysis assessing the immune landscape of the tumor and providing valuable insights into potential immunotherapy options.

Biomarkers, identified through genomic profiling, play an essential role in determining the suitability of immunotherapeutic treatments.

The Intersection of Genomic Profiling and Immunotherapy

Genomic profiling with immunotherapy facilitates the customization of immunotherapy treatments to the individual's tumor genetic profile, significantly improving treatment efficacy.

Genomic Profiling

Genomic profiling uses advanced technologies like next-generation sequencing (NGS) to comprehensively picture a tumor's genetic makeup. NGS is a high-throughput sequencing method that allows for the rapid and parallel sequencing of millions of DNA fragments, enabling detailed analysis of a cancer cell's genome.

This analysis identifies:

  • Mutations: Somatic alterations driving tumorigenesis and progression, including single nucleotide variants, insertions, deletions, and gene fusions. These mutations can activate oncogenes, inactivate tumor suppressor genes, or alter protein function, ultimately leading to uncontrolled cell growth.

  • Copy number variations (CNVs): Gains or losses of genomic regions can impact gene expression. CNVs can lead to amplifications of oncogenes or deletions of tumor suppressor genes, further contributing to tumor development.

  • Tumor mutational burden (TMB): The total number of mutations within the tumor genome, potentially influencing immunotherapy response. High TMB tumors may express neoantigens, making them more recognizable by the immune system and potentially more responsive to checkpoint inhibitor therapy.

Immunotherapy

Immunotherapy has emerged as a revolutionary approach in gynecologic oncology. Different types of immunotherapy exist, including:

  • Checkpoint inhibitors: Target-specific molecules (like PD-1/PD-L1) block immune checkpoints, allowing T cell-mediated tumor cytotoxicity.

  • Antibody-drug conjugates (ADCs): These types of immunotherapy combine a monoclonal antibody with a cytotoxic drug. The antibody specifically targets a protein on the surface of cancer cells, delivering the toxic payload directly to the tumor cells. Tisotumab vedotin is an example of an ADC approved for advanced cervical cancer.

Personalized Immunotherapy Strategies

Genomic profiling plays an important role in tailoring immunotherapy regimens to achieve optimal effectiveness in gynecological cancers.

Methods include:

  • MSI/dMMR biomarkers: Tumors with microsatellite instability (MSI) or mismatch repair deficiency (dMMR) often have high mutation burdens, making them more susceptible to checkpoint inhibitors.

  • Identifying immune checkpoint targets: Profiling can uncover mutations that activate specific immune checkpoints. This allows for the selection of targeted checkpoint inhibitors for a more precise attack on the tumor's immune escape mechanisms.

  • Optimizing combination therapies: Genomic profiling may involve using immunotherapy in combination with other targeted therapies or chemotherapy, potentially leading to synergistic effects and improved outcomes.

This personalized approach based on a tumor's unique genetic and immune profile holds immense promise for gynecological cancers, including ovarian, endometrial, and cervical cancers.

The Power of Genomic Profiling in Immunotherapy

Beyond identifying tumor type and stage, genomic profiling has become a powerful tool for predicting response to specific immunotherapies in gynecological malignancies. This analysis can determine a tumor's unique genetic makeup and immune microenvironment. By understanding these characteristics, we can gain valuable insights into a patient's potential response to different immunotherapy agents.

Genomic profiling goes beyond diagnosis to predict immunotherapy response:

  • High tumor mutational burden (TMB): Profiling identifies tumors with a high number of mutations (TMB), making them prime candidates for checkpoint inhibitor immunotherapy, which boosts the immune system's attack on cancer cells with many mutations.

  • Immune checkpoint dysregulation: Profiling reveals mutations that overactivate immune checkpoints, which normally prevent T cell attacks on healthy tissue. This allows the selection of specific checkpoint inhibitors targeting the relevant pathway.

  • Predicting response: Profiling can predict a patient's likelihood of responding to immunotherapy given before surgery, guiding treatment decisions and potentially improving surgical outcomes.

Future Directions

The integration of genomic profiling with immunotherapy promises to further refine and enhance the precision of gynecological cancer treatments. Ongoing research and clinical trials will need to focus on identifying novel biomarkers and developing advanced genomic tools that can provide deeper insights into tumor biology.

Genomic profiling is revolutionizing gynecological oncology by allowing us to tailor treatments to each patient's unique cancer. This is the essence of personalized medicine. By understanding a tumor's genetic makeup and immune system, we can choose drugs that target its specific weaknesses.

References

1. Previs, R. A., Sood, A. K., Mills, G. B., & Westin, S. N. (2016). The rise of genomic profiling in ovarian cancer. Expert Review of Molecular Diagnostics, 16(12), 1337-1351. https://doi.org/10.1080/14737159.2016.1259069

2. International Journal of Molecular and Immuno Oncology. (n.d.). Driver Mutations in Oncogenesis. Retrieved fromhttps://ijmio.com/driver-mutations-in-oncogenesis/

3. Sha, D., Jin, Z., Budczies, J., Kluck, K., Stenzinger, A., & Sinicrope, F. A. (2020). Tumor mutational burden as a predictive biomarker in solid tumors. Cancer Discovery, 10(12), 1808-1825. https://doi.org/10.1158/2159-8290.CD-20-0522

4. Hellmann, M. D., Nathanson, T., Rizvi, H., Creelan, B. C., Sanchez-Vega, F., Ahuja, A., ... Wolchok, J. D. (2018). Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell, 33(5), 843-852.e4. https://doi.org/10.1016/j.ccell.2018.03.018

5. Di Resta C, Galbiati S, Carrera P, Ferrari M. Next-generation sequencing approach for the diagnosis of human diseases: open challenges and new opportunities. EJIFCC. 2018 Apr 30;29(1):4-14. PMID: 29765282; PMCID: PMC5949614. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5949614/

6. Pös, O., Radvanszky, J., Buglyó, G., Pös, Z., Rusnakova, D., Nagy, B., & Szemes, T. (2021). DNA copy number variation: Main characteristics, evolutionary significance, and pathological aspects. Biomedical Journal, 44(5), 548-559. https://doi.org/10.1016/j.bj.2021.02.003

7. Qin, S., Xu, L., Yi, M., & et al. (2019). Novel immune checkpoint targets: moving beyond PD-1 and CTLA-4. Molecular Cancer, 18, 155. https://doi.org/10.1186/s12943-019-1091-2

8. McNamara B, Chang Y, Goreshnik A, Santin AD. Value of Antibody Drug Conjugates for Gynecological Cancers: A Modern Appraisal Following Recent FDA Approvals. Int J Womens Health. 2023 Aug 28;15:1353-1365. doi: 10.2147/IJWH.S400537. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10474218/

9. National Cancer Institute. (n.d.). CAR T Cells: Engineering Patients' Immune Cells to Treat Their Cancers. Retrieved fromhttps://www.cancer.gov/about-cancer/treatment/research/car-t-cells

10. Sathyanarayanan V, Neelapu SS. Cancer immunotherapy: Strategies for personalization and combinatorial approaches. Mol Oncol. 2015 Dec;9(10):2043-53. doi: 10.1016/j.molonc.2015.10.009. Epub 2015 Oct 23. PMID: 26548534; PMCID: PMC4684975. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4684975/

11. Kavun, A., Veselovsky, E., Lebedeva, A., Belova, E., Kuznetsova, O., Yakushina, V., Ivanov, M. (2023). Microsatellite instability: A review of molecular epidemiology and implications for immune checkpoint inhibitor therapy. Cancers (Basel), 15(8), 2288. https://doi.org/10.3390/cancers15082288

12. Mills, A. M., Bullock, T. N., & Ring, K. L. (2022). Targeting immune checkpoints in gynecologic cancer: updates & perspectives for pathologists. Modern Pathology, 35(2), 142-151. https://www.sciencedirect.com/science/article/pii/S0893395222003258

13. Rubinstein, M. M., & Makker, V. (2020). Optimizing immunotherapy for gynecologic cancers. Current Opinion in Obstetrics and Gynecology, 32(1), 1-8. https://doi.org/10.1097/GCO.0000000000000603

Qian, N., Gregory, O., Harshpreet, C., Matthew, P., Meng-Chang, H., Bridgette, M., et al. (2020). Molecular profiling of gynecologic cancers for treatment and management of disease – demonstrating clinical significance using the AMP/ASCO/CAP guidelines for interpretation and reporting of somatic variants. Cancer Genetics, 242, 25-34.https://www.sciencedirect.com/science/article/abs/pii/S2210776219303795?via%3Dihub
Blog