IJMMS_2024v14n4

International Journal of Molecular Medical Science, 2024, Vol.14, No.4, 239-251 http://medscipublisher.com/index.php/ijmms 240 The ability to analyze tumors at single-cell resolution has profound implications for precision medicine. By identifying specific genetic and phenotypic subclones, researchers can better understand the mechanisms driving tumor evolution, metastasis, and drug resistance. This knowledge can inform the development of targeted therapies and improve patient outcomes. For example, single-cell sequencing has been used to identify rare cancer stem cells and their role in metastasis and recurrence, providing potential targets for therapeutic intervention (Min et al., 2017). High tumor heterogeneity of CRC poses significant challenges for effective treatment and prognosis. Single-cell sequencing allows for the detailed analysis of genetic and protein variations between individual cancer cells, offering a more comprehensive understanding of the tumor microenvironment and the mechanisms driving cancer progression and metastasis (Wen et al., 2023). This technology has also been applied to other cancers, such as liver cancer and high-grade serous ovarian cancer, revealing the complex interplay between cancer cells and their microenvironment (Tian and Li, 2022; Xu et al., 2022). This study explores the application of single-cell sequencing in uncovering cellular heterogeneity in CRC. It summarizes advancements in single-cell sequencing technologies, highlighting their role in understanding intratumor heterogeneity and tumor evolution. It analyzes the tumor microenvironment (TME), discussing how single-cell sequencing characterizes the TME in CRC and identifies key cellular interactions and signaling pathways that contribute to cancer progression and treatment resistance. Additionally, this study reviews findings that identify novel biomarkers and therapeutic targets for more precise and effective CRC treatments. It also addresses current challenges and proposes future research directions to enhance the understanding of cellular heterogeneity in CRC. 2 Colorectal Cancer and Cellular Heterogeneity 2.1 Definition and significance of cellular heterogeneity Cellular heterogeneity refers to the existence of diverse cell populations within a tumor, each with distinct genetic, epigenetic, and phenotypic profiles. This heterogeneity can be observed at multiple levels, including genetic mutations, gene expression patterns, and epigenetic modifications (Li et al., 2017; Bian et al., 2018; Min et al., 2020). In the context of colon cancer, cellular heterogeneity is a critical factor that contributes to tumor evolution, metastasis, and resistance to therapy. Single-cell sequencing technologies have been instrumental in uncovering the extent of this heterogeneity, revealing the presence of various subclones within tumors that may have different roles in cancer progression and response to treatment (Dalerba et al., 2011; Li et al., 2017; Min et al., 2020). 2.2 Impact of heterogeneity on cancer progression and treatment The heterogeneity within colon cancer tumors has profound implications for disease progression and treatment outcomes. Different subclones within a tumor can exhibit varying degrees of aggressiveness, metastatic potential, and sensitivity to therapies. For instance, cancer stem cells (CSCs), which are a rare population within tumors, have been shown to drive metastasis and recurrence due to their ability to self-renew and differentiate into multiple cell types (Min et al., 2020). Additionally, the presence of distinct genetic and epigenetic profiles among tumor cells can lead to differential responses to chemotherapy and targeted therapies, making it challenging to achieve complete eradication of the tumor (Bian et al., 2018; Hamid et al., 2020; Xiang et al., 2021). Single-cell sequencing studies have provided valuable insights into the mechanisms underlying tumor heterogeneity and its impact on treatment resistance. For example, the identification of specific genetic mutations and epigenetic modifications in single circulating tumor cells (CTCs) has highlighted the genetic diversity among these cells and their potential role in disease progression (Hamid et al., 2020). Furthermore, the development of prognostic gene signatures based on cell differentiation trajectories has shown promise in predicting patient outcomes and guiding personalized treatment strategies (Xiang et al., 2021). In summary, understanding and addressing cellular heterogeneity in colon cancer is crucial for improving therapeutic strategies and achieving better clinical outcomes. The integration of single-cell sequencing technologies into clinical practice holds great potential for advancing precision medicine and tailoring treatments to the unique genetic and epigenetic landscape of each patient's tumor.

RkJQdWJsaXNoZXIy MjQ4ODYzNQ==