February 19, 2024

Comprehensive Guide to Cancer Types Analyzed by NGS Testing

This comprehensive guide delves into the various cancer types analyzed through Next Generation Sequencing (NGS) testing, highlighting its pivotal role in revolutionizing cancer diagnosis and treatment. Explore how NGS contributes to personalized medicine, offering detailed insights into breast, lung, colorectal cancers, and more, shaping the future of oncology research and patient care.

In the realm of modern oncology, Next Generation Sequencing (NGS) stands as a beacon of hope and innovation. With its unparalleled ability to unravel the complex genetic underpinnings of various cancer types, NGS is reshaping our approach to cancer diagnosis and treatment. This comprehensive guide embarks on an explorative journey into the diverse world of cancer types analyzed by NGS. From breast to lung and colorectal cancers, we delve into how NGS testing provides deeper insights, aiding in the development of more effective, personalized treatment strategies. As we navigate through the intricacies of NGS and its applications in oncology, we aim to shed light on the transformative impact of this technology, ushering in a new era of precision medicine that promises a better future for cancer patients worldwide.

Introduction to NGS in Cancer Diagnostics

The dawn of Next Generation Sequencing (NGS) in cancer diagnostics has marked a transformative shift in how we understand and treat this complex disease. NGS, with its advanced capabilities, allows for an in-depth analysis of cancerous cells, identifying specific genetic mutations and alterations that drive tumor growth. This level of precision not only aids in accurate cancer diagnosis but also helps in crafting targeted treatment plans tailored to the genetic profile of an individual's tumor. The introduction of NGS into the diagnostic landscape has thus opened up new avenues in personalized medicine, making it possible to approach cancer treatment with a precision previously unimaginable. As we delve into the nuances of NGS, it becomes evident that this technology is not just an advancement in diagnostics; it's a paradigm shift in our fight against cancer.

The Impact of NGS on Oncology

The impact of Next Generation Sequencing (NGS) on oncology has been nothing short of revolutionary. By providing a detailed genomic landscape of cancer, NGS has significantly enhanced the precision of cancer diagnostics and prognosis. This technology facilitates the identification of novel genetic markers and mutations specific to different cancer types, leading to a better understanding of tumor biology. The insights gained from NGS have been instrumental in the development of targeted therapies, allowing oncologists to move beyond one-size-fits-all treatments to more personalized approaches. Moreover, NGS has been pivotal in monitoring treatment efficacy and resistance, enabling more dynamic and responsive cancer management strategies. The integration of NGS into oncological practices signifies a major leap forward, not only in treating cancer but also in improving survival rates and quality of life for patients.

Types of Cancers Analyzed by NGS

Next Generation Sequencing (NGS) has revolutionized our approach to understanding and treating various types of cancer. This technology has the capability to analyze a wide array of cancers with high precision, offering insights that were previously unattainable. NGS is particularly effective in identifying and understanding cancers such as breast cancer, where it can pinpoint specific genetic mutations that might influence treatment choices. In lung cancer, NGS aids in detecting mutations that could be targeted by specific drugs. For colorectal cancer, it helps in identifying both hereditary risks and treatment responses. Similarly, in melanoma and hematological cancers, NGS plays a crucial role in determining genetic alterations that can guide targeted therapy decisions. The versatility of NGS in analyzing these and many other cancer types underscores its invaluable role in the ongoing fight against cancer, paving the way for more personalized and effective treatment strategies.

Breast Cancer

Breast cancer, one of the most common and extensively studied cancer types, has seen significant advancements in diagnosis and treatment due to Next Generation Sequencing (NGS). NGS offers a profound understanding of the genetic alterations specific to breast cancer, including BRCA1 and BRCA2 mutations, which are crucial for determining both risk and effective treatment strategies. This technology enables oncologists to identify subtypes of breast cancer more accurately, leading to more personalized treatment plans. By analyzing tumor genetics, NGS guides the use of targeted therapies, hormone treatments, and helps in predicting response to chemotherapy. The application of NGS in breast cancer not only enhances the precision of treatments but also contributes to ongoing research, ultimately aiming to improve patient outcomes and survival rates.

Lung Cancer

In the realm of lung cancer, Next Generation Sequencing (NGS) has emerged as a game-changer, dramatically altering the landscape of diagnosis and treatment. NGS enables a detailed genetic analysis of lung tumors, uncovering mutations such as EGFR, ALK, and KRAS, which are critical in tailoring patient-specific treatment plans. This level of genetic insight allows for the identification of targetable mutations, guiding the use of targeted therapies and immunotherapies that can significantly improve patient outcomes. Furthermore, NGS facilitates the detection of rare genetic alterations, offering opportunities for patients to participate in clinical trials for new and emerging therapies. The implementation of NGS in lung cancer not only personalizes patient care but also contributes to a deeper understanding of the disease, ultimately guiding more effective and individualized treatment strategies.

Colorectal Cancer

Colorectal cancer treatment and research have been significantly enhanced by the advent of Next Generation Sequencing (NGS). NGS offers a comprehensive genetic analysis, identifying key mutations such as KRAS, NRAS, and BRAF, which are pivotal in determining the most effective treatment approaches. This technology allows for the detection of specific genetic changes that may influence response to certain chemotherapies and targeted drugs, enabling a more personalized treatment strategy. Additionally, NGS plays a crucial role in identifying hereditary colorectal cancer syndromes, aiding in early detection and prevention strategies. The use of NGS in colorectal cancer not only aids in the precise categorization of the disease but also paves the way for the development of new therapeutic options, ultimately contributing to improved patient outcomes and personalized care.

NGS in Genetic Mutation Analysis

Next Generation Sequencing (NGS) has revolutionized the field of genetic mutation analysis, offering a high-resolution view into the genome's intricacies. NGS's comprehensive approach enables the detection of a wide array of mutations, from single nucleotide variants to larger genomic rearrangements, critical in understanding various diseases, including cancer. This detailed analysis allows for the identification of both known and novel mutations, facilitating the development of targeted therapies and personalized treatment plans. In oncology, NGS aids in pinpointing driver mutations, understanding tumor heterogeneity, and tracking disease progression or resistance to treatment. The application of NGS in genetic mutation analysis is not just enhancing diagnostic accuracy; it is also shaping the future of precision medicine by providing deeper insights into the molecular basis of diseases.

Advancements in NGS Technology

The field of Next Generation Sequencing (NGS) has witnessed remarkable advancements, continuously pushing the boundaries of genetic analysis. These technological leaps have resulted in faster, more accurate, and cost-effective sequencing methods. Key improvements include enhanced sequencing throughput, increased sensitivity in detecting low-frequency genetic variants, and the ability to sequence whole genomes or targeted regions with greater precision. Additionally, advances in bioinformatics have greatly improved data analysis, enabling the processing and interpretation of vast amounts of genetic data more efficiently. These advancements in NGS technology are not only accelerating research in various fields, including oncology, infectious diseases, and inherited disorders but are also paving the way for more widespread clinical application, ultimately contributing to personalized medicine and better patient outcomes.

Case Studies and Success Stories

Next Generation Sequencing (NGS) has been central to numerous success stories and breakthroughs in medical research, as illustrated by compelling case studies. For instance, in oncology, NGS has enabled the identification of rare mutations in cancer patients, leading to tailored treatments that significantly improved outcomes. Inherited genetic disorders, once undiagnosed or misdiagnosed, are now being accurately identified, allowing for early intervention and better management. Additionally, NGS has played a crucial role in understanding infectious diseases, aiding in the rapid identification of pathogens and informing public health responses. These success stories not only highlight the transformative power of NGS technology but also reflect its potential to drive future innovations in healthcare, offering hope and new treatment possibilities for patients worldwide.

Future Directions in NGS and Cancer Research

As Next Generation Sequencing (NGS) continues to evolve, its future in cancer research is poised for groundbreaking developments. Emerging trends include the integration of NGS with artificial intelligence and machine learning to enhance genetic data analysis and interpretation. This integration is expected to lead to more precise predictive models for cancer progression and treatment response. Additionally, the expanding field of liquid biopsies, which utilizes NGS for non-invasive cancer detection and monitoring, is set to transform cancer diagnostics and patient monitoring. The ongoing refinement of NGS technology will also enable more comprehensive genomic profiling, potentially uncovering new therapeutic targets and personalized treatment options. These future directions not only underscore the continual innovation within NGS technology but also highlight its growing impact in advancing cancer research, promising improved outcomes and personalized approaches in oncology.

Conclusion: The Transformative Power of NGS in Cancer Fight

In conclusion, the transformative power of Next Generation Sequencing (NGS) in the fight against cancer cannot be overstated. NGS has not only redefined the landscape of cancer diagnostics and treatment but has also played a pivotal role in advancing our understanding of this complex disease. By enabling precise genetic profiling and identification of specific mutations, NGS has paved the way for personalized medicine, offering hope for more effective treatments and improved patient outcomes. The continual advancements in NGS technology promise to further refine our approach to cancer care, making it more targeted, effective, and adaptable to individual patient needs. As we look to the future, NGS stands as a beacon of innovation in oncology, symbolizing our ongoing commitment to conquering cancer and improving lives.

Getting Started with NGS Testing

Embarking on the journey of NGS Testing with Oncogena means stepping into the future of cancer diagnostics and genetic analysis. We offer advanced NGS solutions tailored to meet your specific needs, whether you're a healthcare professional, researcher, or seeking personal genetic insights.

Schedule a Call with Us to Explore How We Can Help: Our team of experts is ready to guide you through the process and answer any questions you might have. Discover how our cutting-edge NGS technology can benefit you or your patients by scheduling a call with us. We are committed to providing personalized support every step of the way.

Contact Information:

  • Email: info@oncogena.com
  • Phone: +90 (533) 779 82 62
  • Address: Acıbadem, Elysium Elit, Şeyh Galip Sk. 2/2 A Blok, 34718 Kadıköy/İstanbul
  • For more information, visit: oncogena.com/contact

Learn More About Our NGS Test: To gain a deeper understanding of what our NGS Test entails and how it can be pivotal in cancer diagnostics and beyond, please visit oncogena.com/oncogena-comprehensive-assay-plus. Here, you'll find detailed information about the test, its applications, and the benefits it offers.

At Oncogena, we believe in empowering our clients with the most advanced genetic analysis tools. Our NGS testing is more than just a service; it's a pathway to unlocking vital genetic insights that can transform healthcare outcomes. By choosing Oncogena, you're not only accessing state-of-the-art technology but also joining a community dedicated to advancing medical science and improving lives.

Start your journey with us today, and take a step towards the future of personalized medicine and genetic understanding.

Authorship and Disclaimer

This blog content has been meticulously prepared by our team of oncologists at Oncogena, leveraging their expertise and experience in the field of genetics and cancer treatment. Our aim is to provide informative and up-to-date insights on Next Generation Sequencing (NGS) and its applications.

Disclaimer: While this content is written by healthcare professionals, it is intended for informational purposes only and should not be considered as medical advice. Oncogena does not assume any liability for the accuracy or completeness of the information provided. We strongly recommend consulting with qualified healthcare professionals for specific medical advice, diagnoses, and treatment decisions.

Further Reading and References: For those interested in exploring more about NGS Testing and its applications, a wealth of resources and scientific literature is available. We encourage you to delve into these references for a more comprehensive understanding of the topic. These can be found in academic journals, medical publications, and on our website's resource section.

References

  1. Pavlidis N, Pentheroudakis G.. Cancer of unknown primary site. Lancet. 2012;379(9824):1428-1435. [PubMed] [Google Scholar]
  2. Siegel RL, Miller KD, Fuchs HE, Jemal A.. Cancer statistics, 2021. CA Cancer J Clin. 2021;71(1):7-33. [PubMed] [Google Scholar]
  3. National Comprehensive Cancer Network. N.C.C.N. Guidelines. Occult primary. Version 2.2021.
  4. Golfinopoulos V, Pentheroudakis G, Salanti G, Nearchou AD, Ioannidis JP, Pavlidis N.. Comparative survival with diverse chemotherapy regimens for cancer of unknown primary site: multiple-treatments meta-analysis. Cancer Treat Rev. 2009;35(7):570-573. [PubMed] [Google Scholar]
  5. Fizazi K, Greco FA, Pavlidis N, Daugaard G, Oien K, Pentheroudakis G; ESMO Guidelines Committee . Cancers of unknown primary site: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26 Suppl 5:v133-v138. [PubMed] [Google Scholar]
  6. Petrakis D, Pentheroudakis G, Voulgaris E, Pavlidis N.. Prognostication in cancer of unknown primary (CUP): development of a prognostic algorithm in 311 cases and review of the literature. Cancer Treat Rev. 2013;39(7):701-708. [PubMed] [Google Scholar]
  7. U.S. Food and Drug Administration (FDA). Oncology (cancer)/hematologic malignancies approval notifications. Accessed July 1, 2021. https://www.Fda.Gov/drugs/resources-information-approved-drugs/oncology-cancer-hematologic-malignancies-approval-notifications.
  8. Vela CM, Knepper TC, Gillis NK, Walko CM, McLeod HL, Hicks JK.. Quantitation of targetable somatic mutations among patients evaluated by a personalized medicine clinical service: considerations for off-label drug use. Pharmacotherapy. 2017;37(9):1043-1051. [PubMed] [Google Scholar]
  9. Zhao P, Peng L, Wu W, et al.. Carcinoma of unknown primary with EML4-ALK fusion response to ALK inhibitors. The Oncologist. 2019;24(4):449-454. [PMC free article] [PubMed] [Google Scholar]
  10. Genentech. Rozyltrek (entrectinib). U.S. Food and drug administration website. Accessed October 19, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2019/212725Orig1s000,%20212726Orig1s000Approv.pdf.
  11. Loxo oncology. Vitrakvi (larotrectinib). U.S. Food and drug administration website. Accessed October 19, 2020.https://www.accessdata.fda.gov/drugsatfda_docs/nda/2018/210861Orig1s000_211710Orig1s000MultidisciplineR.pdf.
  12. Merck co. Keytruda (pembrolizumab). U.S. Food and drug administration website. Accessed October 19, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/125514Orig1s054lbl.pdf.
  13. Mosele F, Remon J, Mateo J, et al.. Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO Precision Medicine Working Group. Ann Oncol. 2020;31(11):1491-1505. [PubMed] [Google Scholar]
  14. Knepper TC, Bell GC, Hicks JK, et al.. Key lessons learned from Moffitt's molecular tumor board: the clinical genomics action committee experience. The Oncologist. 2017;22(2):144-151. [PMC free article] [PubMed] [Google Scholar]
  15. Frampton GM, Fichtenholtz A, Otto GA, et al.. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. 2013;31(11):1023-1031. [PMC free article] [PubMed] [Google Scholar]
  16. He J, Abdel-Wahab O, Nahas MK, et al.. Integrated genomic DNA/RNA profiling of hematologic malignancies in the clinical setting. Blood 2016;127(24):3004-3014. [PMC free article] [PubMed] [Google Scholar]
  17. Odegaard JI, Vincent JJ, Mortimer S, et al.. Validation of a plasma-based comprehensive cancer genotyping assay utilizing orthogonal tissue- and plasma-based methodologies. Clin Cancer Res. 2018;24(15):3539. [PubMed] [Google Scholar]
  18. Chakravarty D, Gao J, Phillips S, et al.. OncoKB: a precision oncology knowledge base. JCO Precis Oncol. 2017;1:1-16. [PMC free article] [PubMed] [Google Scholar]
  19. Marcus L, Fashoyin-Aje LA, Donoghue M, et al.. Fda approval summary: Pembrolizumab for the treatment of tumor mutational burden-high solid tumors. Clin Cancer Res. 2021; 27(17):4685-4689. [PMC free article] [PubMed] [Google Scholar]
  20. Tothill RW, Li J, Mileshkin L, et al.. Massively-parallel sequencing assists the diagnosis and guided treatment of cancers of unknown primary. J Pathol. 2013;231(4):413-423. [PubMed] [Google Scholar]
  21. Von Hoff DD, Stephenson JJ Jr, Rosen P, et al.. Pilot study using molecular profiling of patients’ tumors to find potential targets and select treatments for their refractory cancers. J Clin Oncol. 2010;28(33):4877-4883. [PubMed] [Google Scholar]
  22. Rodon J, Soria JC, Berger R, et al.. Genomic and transcriptomic profiling expands precision cancer medicine: the WINTHER trial. Nat Med. 2019;25(5):751-758. [PMC free article] [PubMed] [Google Scholar]
  23. Sicklick JK, Kato S, Okamura R, et al.. Molecular profiling of cancer patients enables personalized combination therapy: the I-PREDICT study. Nat Med. 2019;25(5):744-750. [PMC free article] [PubMed] [Google Scholar]
  24. Amgen inc. Lumakras (sotorasib). U.S. Food and drug administration website. Accessed July 1, 2021. https://www.Accessdata.Fda.Gov/drugsatfda_docs/label/2021/214665s000lbl.Pdf.
  25. Varghese AM, Arora A, Capanu M, et al.. Clinical and molecular characterization of patients with cancer of unknown primary in the modern era. Ann Oncol. 2017;28(12):3015-3021. [PMC free article] [PubMed] [Google Scholar]
  26. AACR Project GENIE Consortium. AACR Project Genie: powering precision medicine through an international consortium. Cancer Discovery.2017;7(8):818-831. [PMC free article] [PubMed] [Google Scholar]
  27. Marabelle A, Fakih M, Lopez J, et al.. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 2020;21(10):1353-1365. [PubMed] [Google Scholar]
  28. Pilié PG, Tang C, Mills GB, Yap TA.. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat Rev Clin Oncol. 2019;16(2):81-104. [PMC free article] [PubMed] [Google Scholar]
  29. Konstantinopoulos PA, Ceccaldi R, Shapiro GI, D’Andrea AD.. Homologous recombination deficiency: exploiting the fundamental vulnerability of ovarian cancer. Cancer Discov. 2015;5(11):1137. [PMC free article] [PubMed] [Google Scholar]
  30. Golan T, Hammel P, Reni M, et al.. Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N Engl J Med. 2019;381(4):317-327. [PMC free article] [PubMed] [Google Scholar]
  31. Coleman RL, Oza AM, Lorusso D, et al.. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ariel3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;390:1949-1961. [PMC free article] [PubMed] [Google Scholar]
  32. Ross JS, Wang K, Gay L, et al.. Comprehensive genomic profiling of carcinoma of unknown primary site: new routes to targeted therapies. JAMA Oncol. 2015;1(1):40-49. [PubMed] [Google Scholar]
  33. Ross JS, Sokol ES, Moch H, et al.. Comprehensive genomic profiling of carcinoma of unknown primary origin: retrospective molecular classification considering the CUPISCO study design. The Oncologist. 2020. [PMC free article] [PubMed] [Google Scholar]
  34. Another KRAS inhibitor holds its own. Cancer Discov. 2020;10:OF2. PMID: 33115812. [PubMed] [Google Scholar]
  35. Hong DS, Fakih MG, Strickler JH, et al.. KRASG12C Inhibition with sotorasib in advanced solid tumors. N Engl J Med. 2020;383(13):1207-1217. [PMC free article] [PubMed] [Google Scholar]
  36. Sabari JK, Park H, Tolcher AW, et al.. Krystal-2: a phase i/ii trial of adagrasib (mrtx849) in combination with tno155 in patients with advanced solid tumors with Kras g12c mutation. J Clin Oncol 2021;39:TPS146. [Google Scholar]
  37. Patelli G, Tosi F, Amatu A, et al.. Strategies to tackle RAS-mutated metastatic colorectal cancer. ESMO Open. 2021;6(3). [PMC free article] [PubMed] [Google Scholar]
  38. Stadler ZK, Maio A, Chakravarty D, et al.. Therapeutic implications of germline testing in patients with advanced cancers. J Clin Oncol. 2021;0:JCO.20.03661. [PMC free article] [PubMed] [Google Scholar]
  39. Bochtler T, Endris V, Leichsenring J, et al.. Comparative genetic profiling aids diagnosis and clinical decision making in challenging cases of CUP syndrome. Int J Cancer. 2019;145(11):2963-2973. [PubMed] [Google Scholar]
  40. Cerami E, Gao J, Dogrusoz U, et al.. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401. [PMC free article] [PubMed] [Google Scholar]
  41. Zehir A, Benayed R, Shah RH, et al.. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med. 2017;23(6):703-713. [PMC free article] [PubMed] [Google Scholar]
  42. Hayashi H, Kurata T, Takiguchi Y, et al.. Randomized phase II trial comparing site-specific treatment based on gene expression profiling with carboplatin and paclitaxel for patients with cancer of unknown primary site. J Clin Oncol. 2019;37(7):570-579. [PubMed] [Google Scholar]
  43. Fizazi K, Maillard A, Penel N, et al.. A phase iii trial of empiric chemotherapy with cisplatin and gemcitabine or systemic treatment tailored by molecular gene expression analysis in patients with carcinomas of an unknown primary (cup) site (gefcapi 04). Ann Oncol. 2019;30:v851. [Google Scholar]
  44. Hemminki K, Riihimäki M, Sundquist K, Hemminki A.. Site-specific survival rates for cancer of unknown primary according to location of metastases. Int J Cancer. 2013;133(1):182-189. [PubMed] [Google Scholar]
  45. Pauli C, Bochtler T, Mileshkin L, et al.. A challenging task: identifying patients with cancer of unknown primary (CUP) according to ESMO guidelines: the CUPISCO trial experience. The Oncologist. 2021;26(5):e769-e779. [PMC free article] [PubMed] [Google Scholar]
  46. Kato S, Kim KH, Lim HJ, et al.. Real-world data from a molecular tumor board demonstrates improved outcomes with a precision N-of-One strategy. Nat Commun. 2020;11(1):4965. [PMC free article] [PubMed] [Google Scholar]

Articles from The Oncologist are provided here courtesy of Oxford University Press

More news, more insights

see all

Schedule a Call with Us to Explore How We Can Help

Discover the future of cancer diagnostics with our advanced NGS solutions. Contact us now to explore how we can tailor our technology to meet your specific needs.