胃肠道肿瘤的诊断和治疗的部分进展

Mingguang Ju, Ziming Gao, Kai Li, Zhenning Wang
{"title":"胃肠道肿瘤的诊断和治疗的部分进展","authors":"Mingguang Ju,&nbsp;Ziming Gao,&nbsp;Kai Li,&nbsp;Zhenning Wang","doi":"10.1002/cdt3.36","DOIUrl":null,"url":null,"abstract":"<p>Gastrointestinal cancers are difficult to be cured with a high recurrence rate accounting for more than 50% of global cancer-related morbidity and mortality.<span><sup>1</sup></span> Many patients with gastrointestinal cancer are diagnosed at a late stage. Over the past few decades, basic and clinical research with new technologies have made significant progress in the diagnosis and treatment of gastrointestinal cancers, which significantly improved the quality of life and prolonged the survival of patients. Here, we briefly highlight several advances in diagnosing and treating gastrointestinal tumors, mainly gastric cancer and colorectal cancer from multiple perspectives.</p><p>Early diagnosis is crucial for the treatment of gastrointestinal cancers. With the continuous advances in molecular biology, genomics, and epigenetics, individualized diagnosis of gastric cancer and colorectal cancer holds promise for basic research and clinical applications. Liquid biopsy, including detection of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), tumor-related extracellular vesicles (exosomes and microvesicles), tumor-educated platelets, proteins as well as metabolites in a range of bodily fluids offers a cost-efficient and noninvasive approach to screening tumor and monitor relapse and response to treatment.<span><sup>2</sup></span> CTCs are tumor cells in peripheral blood, falling off from the solid tumor focus (primary focus or metastatic focus) due to spontaneous or diagnostic and treatment procedures.<span><sup>3</sup></span> Most of the CTCs undergo apoptosis or phagocytosis and are engulfed by immune cells after entering the peripheral blood; only a few can escape and invade a distant organ to form metastatic foci, increasing the risk of death in patients with gastrointestinal cancers.<span><sup>3</sup></span> Recent studies have shown that CTCs are heterogenic and can proliferate <i>in vivo</i> and <i>in vitro</i>. These, together with the findings from the single-cell molecular analysis, have provided unique insights into the biology of cancer metastasis and therapeutic response.</p><p>ctDNA may reflect tumor-specific abnormalities, and analysis of ctDNA can be applied in the diagnosis, therapeutic response, and prognosis of cancer patients. The mutations in specific genes have been detected in the plasma of patients with several types of gastrointestinal cancers, suggesting that ctDNA may be a possible biomarker of gastrointestinal cancers. The minimal residual disease (MRD) is a microscopic focus of treatment-insensitive tumor cells or the early recurrence of tumors.<span><sup>4</sup></span> Hence, precise evaluation of MRD is especially significant during or after the treatment of gastrointestinal tumors.<span><sup>5</sup></span> The detection of ctDNA and CTCs can help identify and explain the nature of MRD and may provide a new strategy for the prevention and treatment of tumor metastasis.<span><sup>3, 5</sup></span> Nevertheless, ctDNA fragments usually have a short half-life, and currently, there are no convincing cut-off values to discriminate the boundary between high and low ctDNA concentrations. Therefore, the clinical value in the detection of ctDNA has not been gained wide recognition yet.<span><sup>6</sup></span> Another attractive lipid biopsy item is exosomes derived from cancer cells which can promote tumor cell invasion and metastasis by mediating the interaction between tumor cells and the intercellular matrix; exosomes are also closely related to epithelial-mesenchymal transformation (EMT), tumor angiogenesis, and chemotherapeutic drug resistance.<span><sup>7</sup></span> The Food and Drug Administration (FDA) has approved several single-gene and multigene tests as complementary diagnoses for cancer-specific molecular targeted therapy, marking the widespread clinical use of liquid biopsies, especially in patients with advanced cancer.<span><sup>2, 7</sup></span></p><p>AI, especially machine learning and deep learning, has opened up new avenues for scientific and clinical research. With increasingly multidimensional data generated in daily treatment, AI can aid clinicians to form a personalized perspective on patients' treatment paths and ultimately guide clinical decision-making. These decisions rely on integrating different and complex data streams, including clinical manifestations, patient history, pathological results, genomics, medical imaging, and matching these data with the conclusions of the growing scientific literature. Early data and computational limitations often require the simplification of unstructured patient data, such as medical images and biopsies, to a set of discrete observations of the degree of disease that humans can understand. A prominent example of this simplification is the cancer staging system, most notably the TNM classification of the American Joint Commission on Cancer (AJCC).<span><sup>8</sup></span> AI has been used to improve the accuracy of liquid biopsy analysis of the data from CTC and ctDNA tests, which may facilitate its integration into clinical workflows in the future. For example, machine learning has been applied to identify CTCs in cancer patients,<span><sup>9</sup></span> analyze ctDNA for cancer detection and location, comprehensive multigroup analysis, liquid biopsy tests and other clinical genetics, metabolomics, immunology, microbiology, and steady-state data to guide treatment decisions. Currently, the FDA has approved a few AI applications for oncology indications and many applications are underway. There is a great interest in simplifying the gap between the development of AI and clinical transformation. Consequently, the FDA is designing specific guidelines for AI and machine learning for the approved clinical applications.<span><sup>10</sup></span> The prospect of AI is bright, but there are still many challenges in successfully integrating AI into clinical oncology, it may be only suitable for some particular cancer problems and its specific value may stem from specific cases in clinics.<span><sup>11</sup></span> Future studies should analyze the prospects, successes, and failures of AI as well as translate them clinically on a case-by-case basis. The further development of AI applications in cancer diagnosis and treatment may benefit the clinical management of gastrointestinal cancers.</p><p>In 2018, the European Society for Medical Oncology (ESMO) and oncologists from Asian countries revised the guidelines 2016 version, based on scientific evidence, for better management of gastric cancer. The main change in the 2019 ESMO metastatic gastric cancer guidelines updated immunotherapy as a treatment for gastric cancer.<span><sup>12</sup></span> According to the tumor immunoediting theory, immune checkpoint inhibitors (ICIs) have become effective strategies for the treatment of gastrointestinal tumors. The most commonly used ICIs include monoclonal antibodies programmed cell death protein-1 (PD-1)/programmed death protein ligand-1 (PD-L1) and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4). In addition, other immunotherapies are available, including adoptive T cell therapies (ACT), such as adoptive transfer of chimeric antigen receptor (CAR)- and T cell receptor (TCR)-T cells, as well as bulk tumor-infiltrating lymphocyte (TIL) therapy. Unfortunately, the response rates of immunotherapy are not satisfactory, and some patients may lose their therapeutic effects after a period of treatment. Moreover, gastrointestinal cancers usually have a low response rate to immunotherapies because cancer cells may evade the immune response, contributing to immune tolerance.<span><sup>13</sup></span> Further exploration of the mechanisms by which cancer cells are resistant to immunotherapies may provide a basis for the development of new promising immunotherapies for gastrointestinal cancers.<span><sup>14</sup></span> A previous study has revealed that the gut microbiota, a complex community of trillions of microorganisms, plays a crucial role in immunotherapeutic responses in patients with gastrointestinal cancer.<span><sup>15</sup></span> Besides physiological functions, the gut microbiota can regulate oncogenesis and resistance to antitumor therapies. During antitumor therapies, many medicines are commonly used for reducing adverse events and these medicines include antidiabetics, aspirin, proton pump inhibitors, psychotropic drugs, and nonsteroidal anti-inflammatory drugs, and analgesics such as opioids. Interestingly, treatment with these drugs can significantly modulate the composition of gut microbiota. Hence, understanding the regulatory effect of gut microbiota on antitumor immunotherapies may help improve their therapeutic efficacy in gastrointestinal cancers. Nanoparticles represent a promising strategy to enhance efficacy and reduce the toxicity of immunotherapy. In the context of treatment of colorectal cancer, nanoparticles can be used to transmit various substances with the potential to affect immune cells and their immunoreactivity within the tumor immune microenvironment, targeting the immune system, and activating multiple immune cells including, but not limited to, T cells.<span><sup>16</sup></span></p><p>Besides tumor cells, there are several types of immune cells, endothelial cells, fibroblasts, extracellular matrix as well as others in the TME of gastrointestinal tumors. These components in TME are the key factors affecting tumor initiation, progression, and immune responses. The balance between cellular and humoral components as well as various inflammatory responses in the TME is critical for tumor growth. Cellular immunotherapy and pharmacological immunotherapy can modulate the TME to enhance tumor immunogenicity and antitumor immune responses to limit tumor growth. However, the efficacy of immunotherapies, such as ICIs, is impaired by the dynamic changes in the TME, which may induce acquired resistance in gastrointestinal cancers. Thus, further understanding of the dynamic changes in the TME may help develop new therapeutic strategies for improvement of immunotherapies and prevention of therapeutic resistance in gastrointestinal cancers. To determine the interaction between TME and tumor cells, further exploration of the mechanism underlying immune escape and immune tolerance, and effectively reshaping the TME are the leading solutions for cancer immunotherapies. Bioprinting of 3D cell culture models can help monitor complex cell behaviors under physical simulation conditions, modulate multiple types of cells in an easy-to-handle system, and study cell interaction in real-time.<span><sup>17, 18</sup></span> With improved access to clinical tissues and better technologies to visualize cells <i>in situ</i> and dissect heterocellular signaling, the use of bioprinting to understand the TME holds great promise to address translational research questions. Furthermore, the 3D cell culture models are valuable for the development of anticancer drugs. Personalized cancer models can also be used to screen personalized antitumor drugs and promote basic cancer research, drug development, and precise medicines. Despite the attractiveness of 3D bioprinting, there are several technical challenges in the printing platforms, cells, and materials used to build 3D models because of the inherent challenge of comprehensively replicating the cellular behaviors and structural complexity of natural tissues.</p><p>Emerging biomarkers, particularly ctDNA, CTCs, and exosomes, are increasingly driving diagnosis advances, allowing for the early identification and dynamic monitoring of gastrointestinal cancers. Apart from the other useful treatment modalities (such as endoscopic therapy, surgery, radiotherapy, chemotherapy, and targeted therapy), immunotherapy, with its unique and valuable benefits, has made significant progress in the management of gastrointestinal cancers, which is our primary focus area. Concomitant nano-based therapeutics represent a promising strategy to improve the efficacy of immunotherapy. Fortunately, the collaboration in nonmedical disciplines, such as AI, 3D bioprinting, and materials science, can enable in-depth characterization of the TME, improving the diagnosis and treatment of gastrointestinal cancers and fostering multidisciplinary treatment strategies in the future. However, limitations are yet undeniable, which implies that new advances are often accompanied by formidable challenges to overcome.</p><p>Kai Li and Zhenning Wang conceptualized and designed the study. Mingguang Ju drafted the first version of the manuscript. All authors read and edited the manuscript.</p><p>The authors declare no conflict of interest. Professor Kai Li and Zhenning Wang are members of Chronic Diseases and Translational Medicine editorial board and are not involved in the peer review process of this article.</p><p>None.</p>","PeriodicalId":32096,"journal":{"name":"Chronic Diseases and Translational Medicine","volume":"9 1","pages":"1-4"},"PeriodicalIF":0.0000,"publicationDate":"2022-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/99/ca/CDT3-9-1.PMC10011665.pdf","citationCount":"0","resultStr":"{\"title\":\"Partial advances in the diagnosis and treatment of gastrointestinal cancer\",\"authors\":\"Mingguang Ju,&nbsp;Ziming Gao,&nbsp;Kai Li,&nbsp;Zhenning Wang\",\"doi\":\"10.1002/cdt3.36\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Gastrointestinal cancers are difficult to be cured with a high recurrence rate accounting for more than 50% of global cancer-related morbidity and mortality.<span><sup>1</sup></span> Many patients with gastrointestinal cancer are diagnosed at a late stage. Over the past few decades, basic and clinical research with new technologies have made significant progress in the diagnosis and treatment of gastrointestinal cancers, which significantly improved the quality of life and prolonged the survival of patients. Here, we briefly highlight several advances in diagnosing and treating gastrointestinal tumors, mainly gastric cancer and colorectal cancer from multiple perspectives.</p><p>Early diagnosis is crucial for the treatment of gastrointestinal cancers. With the continuous advances in molecular biology, genomics, and epigenetics, individualized diagnosis of gastric cancer and colorectal cancer holds promise for basic research and clinical applications. Liquid biopsy, including detection of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), tumor-related extracellular vesicles (exosomes and microvesicles), tumor-educated platelets, proteins as well as metabolites in a range of bodily fluids offers a cost-efficient and noninvasive approach to screening tumor and monitor relapse and response to treatment.<span><sup>2</sup></span> CTCs are tumor cells in peripheral blood, falling off from the solid tumor focus (primary focus or metastatic focus) due to spontaneous or diagnostic and treatment procedures.<span><sup>3</sup></span> Most of the CTCs undergo apoptosis or phagocytosis and are engulfed by immune cells after entering the peripheral blood; only a few can escape and invade a distant organ to form metastatic foci, increasing the risk of death in patients with gastrointestinal cancers.<span><sup>3</sup></span> Recent studies have shown that CTCs are heterogenic and can proliferate <i>in vivo</i> and <i>in vitro</i>. These, together with the findings from the single-cell molecular analysis, have provided unique insights into the biology of cancer metastasis and therapeutic response.</p><p>ctDNA may reflect tumor-specific abnormalities, and analysis of ctDNA can be applied in the diagnosis, therapeutic response, and prognosis of cancer patients. The mutations in specific genes have been detected in the plasma of patients with several types of gastrointestinal cancers, suggesting that ctDNA may be a possible biomarker of gastrointestinal cancers. The minimal residual disease (MRD) is a microscopic focus of treatment-insensitive tumor cells or the early recurrence of tumors.<span><sup>4</sup></span> Hence, precise evaluation of MRD is especially significant during or after the treatment of gastrointestinal tumors.<span><sup>5</sup></span> The detection of ctDNA and CTCs can help identify and explain the nature of MRD and may provide a new strategy for the prevention and treatment of tumor metastasis.<span><sup>3, 5</sup></span> Nevertheless, ctDNA fragments usually have a short half-life, and currently, there are no convincing cut-off values to discriminate the boundary between high and low ctDNA concentrations. Therefore, the clinical value in the detection of ctDNA has not been gained wide recognition yet.<span><sup>6</sup></span> Another attractive lipid biopsy item is exosomes derived from cancer cells which can promote tumor cell invasion and metastasis by mediating the interaction between tumor cells and the intercellular matrix; exosomes are also closely related to epithelial-mesenchymal transformation (EMT), tumor angiogenesis, and chemotherapeutic drug resistance.<span><sup>7</sup></span> The Food and Drug Administration (FDA) has approved several single-gene and multigene tests as complementary diagnoses for cancer-specific molecular targeted therapy, marking the widespread clinical use of liquid biopsies, especially in patients with advanced cancer.<span><sup>2, 7</sup></span></p><p>AI, especially machine learning and deep learning, has opened up new avenues for scientific and clinical research. With increasingly multidimensional data generated in daily treatment, AI can aid clinicians to form a personalized perspective on patients' treatment paths and ultimately guide clinical decision-making. These decisions rely on integrating different and complex data streams, including clinical manifestations, patient history, pathological results, genomics, medical imaging, and matching these data with the conclusions of the growing scientific literature. Early data and computational limitations often require the simplification of unstructured patient data, such as medical images and biopsies, to a set of discrete observations of the degree of disease that humans can understand. A prominent example of this simplification is the cancer staging system, most notably the TNM classification of the American Joint Commission on Cancer (AJCC).<span><sup>8</sup></span> AI has been used to improve the accuracy of liquid biopsy analysis of the data from CTC and ctDNA tests, which may facilitate its integration into clinical workflows in the future. For example, machine learning has been applied to identify CTCs in cancer patients,<span><sup>9</sup></span> analyze ctDNA for cancer detection and location, comprehensive multigroup analysis, liquid biopsy tests and other clinical genetics, metabolomics, immunology, microbiology, and steady-state data to guide treatment decisions. Currently, the FDA has approved a few AI applications for oncology indications and many applications are underway. There is a great interest in simplifying the gap between the development of AI and clinical transformation. Consequently, the FDA is designing specific guidelines for AI and machine learning for the approved clinical applications.<span><sup>10</sup></span> The prospect of AI is bright, but there are still many challenges in successfully integrating AI into clinical oncology, it may be only suitable for some particular cancer problems and its specific value may stem from specific cases in clinics.<span><sup>11</sup></span> Future studies should analyze the prospects, successes, and failures of AI as well as translate them clinically on a case-by-case basis. The further development of AI applications in cancer diagnosis and treatment may benefit the clinical management of gastrointestinal cancers.</p><p>In 2018, the European Society for Medical Oncology (ESMO) and oncologists from Asian countries revised the guidelines 2016 version, based on scientific evidence, for better management of gastric cancer. The main change in the 2019 ESMO metastatic gastric cancer guidelines updated immunotherapy as a treatment for gastric cancer.<span><sup>12</sup></span> According to the tumor immunoediting theory, immune checkpoint inhibitors (ICIs) have become effective strategies for the treatment of gastrointestinal tumors. The most commonly used ICIs include monoclonal antibodies programmed cell death protein-1 (PD-1)/programmed death protein ligand-1 (PD-L1) and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4). In addition, other immunotherapies are available, including adoptive T cell therapies (ACT), such as adoptive transfer of chimeric antigen receptor (CAR)- and T cell receptor (TCR)-T cells, as well as bulk tumor-infiltrating lymphocyte (TIL) therapy. Unfortunately, the response rates of immunotherapy are not satisfactory, and some patients may lose their therapeutic effects after a period of treatment. Moreover, gastrointestinal cancers usually have a low response rate to immunotherapies because cancer cells may evade the immune response, contributing to immune tolerance.<span><sup>13</sup></span> Further exploration of the mechanisms by which cancer cells are resistant to immunotherapies may provide a basis for the development of new promising immunotherapies for gastrointestinal cancers.<span><sup>14</sup></span> A previous study has revealed that the gut microbiota, a complex community of trillions of microorganisms, plays a crucial role in immunotherapeutic responses in patients with gastrointestinal cancer.<span><sup>15</sup></span> Besides physiological functions, the gut microbiota can regulate oncogenesis and resistance to antitumor therapies. During antitumor therapies, many medicines are commonly used for reducing adverse events and these medicines include antidiabetics, aspirin, proton pump inhibitors, psychotropic drugs, and nonsteroidal anti-inflammatory drugs, and analgesics such as opioids. Interestingly, treatment with these drugs can significantly modulate the composition of gut microbiota. Hence, understanding the regulatory effect of gut microbiota on antitumor immunotherapies may help improve their therapeutic efficacy in gastrointestinal cancers. Nanoparticles represent a promising strategy to enhance efficacy and reduce the toxicity of immunotherapy. In the context of treatment of colorectal cancer, nanoparticles can be used to transmit various substances with the potential to affect immune cells and their immunoreactivity within the tumor immune microenvironment, targeting the immune system, and activating multiple immune cells including, but not limited to, T cells.<span><sup>16</sup></span></p><p>Besides tumor cells, there are several types of immune cells, endothelial cells, fibroblasts, extracellular matrix as well as others in the TME of gastrointestinal tumors. These components in TME are the key factors affecting tumor initiation, progression, and immune responses. The balance between cellular and humoral components as well as various inflammatory responses in the TME is critical for tumor growth. Cellular immunotherapy and pharmacological immunotherapy can modulate the TME to enhance tumor immunogenicity and antitumor immune responses to limit tumor growth. However, the efficacy of immunotherapies, such as ICIs, is impaired by the dynamic changes in the TME, which may induce acquired resistance in gastrointestinal cancers. Thus, further understanding of the dynamic changes in the TME may help develop new therapeutic strategies for improvement of immunotherapies and prevention of therapeutic resistance in gastrointestinal cancers. To determine the interaction between TME and tumor cells, further exploration of the mechanism underlying immune escape and immune tolerance, and effectively reshaping the TME are the leading solutions for cancer immunotherapies. Bioprinting of 3D cell culture models can help monitor complex cell behaviors under physical simulation conditions, modulate multiple types of cells in an easy-to-handle system, and study cell interaction in real-time.<span><sup>17, 18</sup></span> With improved access to clinical tissues and better technologies to visualize cells <i>in situ</i> and dissect heterocellular signaling, the use of bioprinting to understand the TME holds great promise to address translational research questions. Furthermore, the 3D cell culture models are valuable for the development of anticancer drugs. Personalized cancer models can also be used to screen personalized antitumor drugs and promote basic cancer research, drug development, and precise medicines. Despite the attractiveness of 3D bioprinting, there are several technical challenges in the printing platforms, cells, and materials used to build 3D models because of the inherent challenge of comprehensively replicating the cellular behaviors and structural complexity of natural tissues.</p><p>Emerging biomarkers, particularly ctDNA, CTCs, and exosomes, are increasingly driving diagnosis advances, allowing for the early identification and dynamic monitoring of gastrointestinal cancers. Apart from the other useful treatment modalities (such as endoscopic therapy, surgery, radiotherapy, chemotherapy, and targeted therapy), immunotherapy, with its unique and valuable benefits, has made significant progress in the management of gastrointestinal cancers, which is our primary focus area. Concomitant nano-based therapeutics represent a promising strategy to improve the efficacy of immunotherapy. Fortunately, the collaboration in nonmedical disciplines, such as AI, 3D bioprinting, and materials science, can enable in-depth characterization of the TME, improving the diagnosis and treatment of gastrointestinal cancers and fostering multidisciplinary treatment strategies in the future. However, limitations are yet undeniable, which implies that new advances are often accompanied by formidable challenges to overcome.</p><p>Kai Li and Zhenning Wang conceptualized and designed the study. Mingguang Ju drafted the first version of the manuscript. All authors read and edited the manuscript.</p><p>The authors declare no conflict of interest. Professor Kai Li and Zhenning Wang are members of Chronic Diseases and Translational Medicine editorial board and are not involved in the peer review process of this article.</p><p>None.</p>\",\"PeriodicalId\":32096,\"journal\":{\"name\":\"Chronic Diseases and Translational Medicine\",\"volume\":\"9 1\",\"pages\":\"1-4\"},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2022-07-04\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/99/ca/CDT3-9-1.PMC10011665.pdf\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Chronic Diseases and Translational Medicine\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/cdt3.36\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"Medicine\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Chronic Diseases and Translational Medicine","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cdt3.36","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"Medicine","Score":null,"Total":0}
引用次数: 0

摘要

胃肠道肿瘤难以治愈,复发率高,占全球癌症相关发病率和死亡率的50%以上许多胃肠道癌症患者在晚期才被诊断出来。近几十年来,新技术的基础和临床研究使胃肠道肿瘤的诊断和治疗取得了重大进展,显著提高了患者的生活质量,延长了患者的生存期。在此,我们从多个角度简要介绍几种胃肠道肿瘤的诊断和治疗进展,主要是胃癌和结直肠癌。早期诊断对胃肠道癌症的治疗至关重要。随着分子生物学、基因组学和表观遗传学的不断进步,胃癌和结直肠癌的个体化诊断具有广阔的基础研究和临床应用前景。液体活检,包括循环肿瘤细胞(CTCs)、循环肿瘤DNA (ctDNA)、肿瘤相关细胞外囊泡(外泌体和微囊泡)、肿瘤诱导血小板、蛋白质以及一系列体液中的代谢物的检测,为肿瘤筛查和监测复发和治疗反应提供了一种经济有效的非侵入性方法ctc是外周血中的肿瘤细胞,由于自发或诊断和治疗过程而从实体瘤病灶(原发病灶或转移灶)脱落大部分ctc进入外周血后发生凋亡或吞噬,被免疫细胞吞噬;只有少数能逃逸并侵入远端器官形成转移灶,增加了胃肠道癌患者的死亡风险最近的研究表明,ctc具有异质性,可以在体内和体外增殖。这些与单细胞分子分析的发现一起,为癌症转移和治疗反应的生物学提供了独特的见解。ctDNA可能反映肿瘤特异性异常,分析ctDNA可用于癌症患者的诊断、治疗反应和预后。在几种胃肠道癌症患者的血浆中检测到特定基因的突变,这表明ctDNA可能是胃肠道癌症的一种可能的生物标志物。微小残留病(MRD)是治疗不敏感的肿瘤细胞或肿瘤早期复发的显微病灶因此,在胃肠道肿瘤治疗期间或之后,精确评估MRD尤为重要ctDNA和CTCs的检测有助于识别和解释MRD的性质,并可能为预防和治疗肿瘤转移提供新的策略。然而,ctDNA片段通常具有较短的半衰期,目前,没有令人信服的临界值来区分高和低ctDNA浓度之间的界限。因此,ctDNA检测的临床价值尚未得到广泛认可另一个吸引人的脂质活检项目是来自癌细胞的外泌体,它可以通过介导肿瘤细胞与细胞间基质的相互作用来促进肿瘤细胞的侵袭和转移;外泌体还与上皮-间质转化(EMT)、肿瘤血管生成和化疗耐药密切相关美国食品和药物管理局(FDA)已经批准了几种单基因和多基因检测作为癌症特异性分子靶向治疗的补充诊断,标志着液体活检的广泛临床应用,特别是在晚期癌症患者中。人工智能,特别是机器学习和深度学习,为科学和临床研究开辟了新的途径。随着日常治疗中产生的多维数据越来越多,人工智能可以帮助临床医生对患者的治疗路径形成个性化的视角,最终指导临床决策。这些决策依赖于整合不同且复杂的数据流,包括临床表现、患者病史、病理结果、基因组学、医学影像,并将这些数据与越来越多的科学文献的结论相匹配。早期数据和计算限制往往需要将非结构化的患者数据(如医学图像和活检)简化为一组人类能够理解的疾病程度的离散观察结果。这种简化的一个突出例子是癌症分期系统,最引人注目的是美国癌症联合委员会(AJCC)的TNM分类人工智能已被用于提高CTC和ctDNA检测数据的液体活检分析的准确性,这可能有助于其在未来融入临床工作流程。 例如,机器学习已被应用于识别癌症患者的CTCs,9分析ctDNA用于癌症检测和定位,综合多组分析,液体活检测试和其他临床遗传学,代谢组学,免疫学,微生物学和稳态数据,以指导治疗决策。目前,FDA已经批准了一些用于肿瘤适应症的人工智能应用,许多应用正在进行中。人们对缩小人工智能发展与临床转化之间的差距非常感兴趣。因此,FDA正在为已批准的临床应用设计人工智能和机器学习的具体指导方针人工智能的前景是光明的,但将人工智能成功地融入临床肿瘤学仍有许多挑战,它可能只适用于某些特定的癌症问题,其具体价值可能源于临床的具体病例未来的研究应该分析人工智能的前景、成功和失败,并根据具体情况将其转化为临床。人工智能在癌症诊断和治疗中的应用的进一步发展可能有利于胃肠道癌症的临床管理。2018年,欧洲肿瘤医学学会(ESMO)和来自亚洲国家的肿瘤学家根据科学证据修订了2016年版指南,以更好地管理胃癌。2019年ESMO转移性胃癌指南的主要变化是更新了免疫疗法作为胃癌的治疗方法根据肿瘤免疫编辑理论,免疫检查点抑制剂(immune checkpoint inhibitors, ICIs)已成为治疗胃肠道肿瘤的有效策略。最常用的ICIs包括单克隆抗体程序性细胞死亡蛋白-1 (PD-1)/程序性死亡蛋白配体-1 (PD-L1)和细胞毒性t淋巴细胞相关蛋白-4 (CTLA-4)。此外,其他免疫疗法是可用的,包括过继T细胞疗法(ACT),如嵌合抗原受体(CAR)和T细胞受体(TCR)-T细胞的过继转移,以及大量肿瘤浸润淋巴细胞(TIL)治疗。不幸的是,免疫治疗的反应率并不令人满意,有些患者在治疗一段时间后可能失去治疗效果。此外,胃肠道癌症对免疫治疗的应答率通常较低,因为癌细胞可能逃避免疫反应,导致免疫耐受进一步探索癌细胞对免疫疗法产生耐药性的机制可能为开发新的有前景的胃肠道癌症免疫疗法提供基础先前的一项研究表明,肠道微生物群是一个由数万亿微生物组成的复杂群落,在胃肠道癌症患者的免疫治疗反应中起着至关重要的作用除了生理功能外,肠道微生物群还可以调节肿瘤的发生和抗肿瘤治疗的耐药性。在抗肿瘤治疗中,通常使用许多药物来减少不良事件,这些药物包括抗糖尿病药、阿司匹林、质子泵抑制剂、精神药物、非甾体抗炎药和镇痛药,如阿片类药物。有趣的是,用这些药物治疗可以显著调节肠道微生物群的组成。因此,了解肠道菌群对抗肿瘤免疫疗法的调控作用可能有助于提高其对胃肠道肿瘤的治疗效果。纳米颗粒代表了一种有前途的策略,以提高疗效和减少免疫治疗的毒性。在结直肠癌的治疗中,纳米颗粒可用于在肿瘤免疫微环境中传递各种有可能影响免疫细胞及其免疫反应性的物质,靶向免疫系统,激活多种免疫细胞,包括但不限于T细胞。16胃肠道肿瘤TME除肿瘤细胞外,还包括免疫细胞、内皮细胞、成纤维细胞、细胞外基质等多种类型。TME中的这些成分是影响肿瘤发生、进展和免疫反应的关键因素。细胞和体液成分之间的平衡以及TME中各种炎症反应对肿瘤生长至关重要。细胞免疫治疗和药物免疫治疗可调节TME增强肿瘤免疫原性和抗肿瘤免疫反应,限制肿瘤生长。然而,免疫疗法(如ICIs)的疗效受到TME动态变化的影响,这可能导致胃肠道癌症的获得性耐药。因此,进一步了解TME的动态变化可能有助于开发新的治疗策略,以改善胃肠道癌症的免疫治疗和预防治疗耐药。 确定TME与肿瘤细胞的相互作用,进一步探索免疫逃逸和免疫耐受的机制,有效重塑TME是癌症免疫治疗的主要解决方案。3D细胞培养模型的生物打印可以帮助在物理模拟条件下监测复杂的细胞行为,在一个易于处理的系统中调节多种类型的细胞,并实时研究细胞相互作用。17,18随着临床组织的改善和更好的技术来观察原位细胞和解剖异细胞信号,使用生物打印来理解TME在解决转化研究问题方面具有很大的希望。此外,三维细胞培养模型对抗癌药物的开发具有重要价值。个性化癌症模型还可用于筛选个性化抗肿瘤药物,促进基础癌症研究、药物开发和精准用药。尽管3D生物打印具有吸引力,但由于全面复制自然组织的细胞行为和结构复杂性的固有挑战,因此在用于构建3D模型的打印平台,细胞和材料方面存在一些技术挑战。新兴的生物标志物,特别是ctDNA、ctc和外泌体,正日益推动着诊断的进步,使胃肠道癌症的早期识别和动态监测成为可能。除了其他有用的治疗方式(如内镜治疗、手术、放疗、化疗和靶向治疗)外,免疫治疗以其独特而宝贵的益处,在胃肠道癌症的治疗方面取得了重大进展,这是我们的主要关注领域。同时使用纳米疗法是提高免疫治疗疗效的一种很有前途的策略。幸运的是,在人工智能、3D生物打印和材料科学等非医学学科的合作,可以深入表征TME,改善胃肠道癌症的诊断和治疗,并在未来促进多学科治疗策略。然而,局限性也是不可否认的,这意味着新的进步往往伴随着需要克服的巨大挑战。李凯和王振宁构思并设计了这项研究。居明光起草了第一版手稿。所有作者都阅读并编辑了手稿。作者声明无利益冲突。李凯教授和王振宁教授为《慢性疾病与转化医学》编委会成员,未参与本文的同行评议过程。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Partial advances in the diagnosis and treatment of gastrointestinal cancer

Gastrointestinal cancers are difficult to be cured with a high recurrence rate accounting for more than 50% of global cancer-related morbidity and mortality.1 Many patients with gastrointestinal cancer are diagnosed at a late stage. Over the past few decades, basic and clinical research with new technologies have made significant progress in the diagnosis and treatment of gastrointestinal cancers, which significantly improved the quality of life and prolonged the survival of patients. Here, we briefly highlight several advances in diagnosing and treating gastrointestinal tumors, mainly gastric cancer and colorectal cancer from multiple perspectives.

Early diagnosis is crucial for the treatment of gastrointestinal cancers. With the continuous advances in molecular biology, genomics, and epigenetics, individualized diagnosis of gastric cancer and colorectal cancer holds promise for basic research and clinical applications. Liquid biopsy, including detection of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), tumor-related extracellular vesicles (exosomes and microvesicles), tumor-educated platelets, proteins as well as metabolites in a range of bodily fluids offers a cost-efficient and noninvasive approach to screening tumor and monitor relapse and response to treatment.2 CTCs are tumor cells in peripheral blood, falling off from the solid tumor focus (primary focus or metastatic focus) due to spontaneous or diagnostic and treatment procedures.3 Most of the CTCs undergo apoptosis or phagocytosis and are engulfed by immune cells after entering the peripheral blood; only a few can escape and invade a distant organ to form metastatic foci, increasing the risk of death in patients with gastrointestinal cancers.3 Recent studies have shown that CTCs are heterogenic and can proliferate in vivo and in vitro. These, together with the findings from the single-cell molecular analysis, have provided unique insights into the biology of cancer metastasis and therapeutic response.

ctDNA may reflect tumor-specific abnormalities, and analysis of ctDNA can be applied in the diagnosis, therapeutic response, and prognosis of cancer patients. The mutations in specific genes have been detected in the plasma of patients with several types of gastrointestinal cancers, suggesting that ctDNA may be a possible biomarker of gastrointestinal cancers. The minimal residual disease (MRD) is a microscopic focus of treatment-insensitive tumor cells or the early recurrence of tumors.4 Hence, precise evaluation of MRD is especially significant during or after the treatment of gastrointestinal tumors.5 The detection of ctDNA and CTCs can help identify and explain the nature of MRD and may provide a new strategy for the prevention and treatment of tumor metastasis.3, 5 Nevertheless, ctDNA fragments usually have a short half-life, and currently, there are no convincing cut-off values to discriminate the boundary between high and low ctDNA concentrations. Therefore, the clinical value in the detection of ctDNA has not been gained wide recognition yet.6 Another attractive lipid biopsy item is exosomes derived from cancer cells which can promote tumor cell invasion and metastasis by mediating the interaction between tumor cells and the intercellular matrix; exosomes are also closely related to epithelial-mesenchymal transformation (EMT), tumor angiogenesis, and chemotherapeutic drug resistance.7 The Food and Drug Administration (FDA) has approved several single-gene and multigene tests as complementary diagnoses for cancer-specific molecular targeted therapy, marking the widespread clinical use of liquid biopsies, especially in patients with advanced cancer.2, 7

AI, especially machine learning and deep learning, has opened up new avenues for scientific and clinical research. With increasingly multidimensional data generated in daily treatment, AI can aid clinicians to form a personalized perspective on patients' treatment paths and ultimately guide clinical decision-making. These decisions rely on integrating different and complex data streams, including clinical manifestations, patient history, pathological results, genomics, medical imaging, and matching these data with the conclusions of the growing scientific literature. Early data and computational limitations often require the simplification of unstructured patient data, such as medical images and biopsies, to a set of discrete observations of the degree of disease that humans can understand. A prominent example of this simplification is the cancer staging system, most notably the TNM classification of the American Joint Commission on Cancer (AJCC).8 AI has been used to improve the accuracy of liquid biopsy analysis of the data from CTC and ctDNA tests, which may facilitate its integration into clinical workflows in the future. For example, machine learning has been applied to identify CTCs in cancer patients,9 analyze ctDNA for cancer detection and location, comprehensive multigroup analysis, liquid biopsy tests and other clinical genetics, metabolomics, immunology, microbiology, and steady-state data to guide treatment decisions. Currently, the FDA has approved a few AI applications for oncology indications and many applications are underway. There is a great interest in simplifying the gap between the development of AI and clinical transformation. Consequently, the FDA is designing specific guidelines for AI and machine learning for the approved clinical applications.10 The prospect of AI is bright, but there are still many challenges in successfully integrating AI into clinical oncology, it may be only suitable for some particular cancer problems and its specific value may stem from specific cases in clinics.11 Future studies should analyze the prospects, successes, and failures of AI as well as translate them clinically on a case-by-case basis. The further development of AI applications in cancer diagnosis and treatment may benefit the clinical management of gastrointestinal cancers.

In 2018, the European Society for Medical Oncology (ESMO) and oncologists from Asian countries revised the guidelines 2016 version, based on scientific evidence, for better management of gastric cancer. The main change in the 2019 ESMO metastatic gastric cancer guidelines updated immunotherapy as a treatment for gastric cancer.12 According to the tumor immunoediting theory, immune checkpoint inhibitors (ICIs) have become effective strategies for the treatment of gastrointestinal tumors. The most commonly used ICIs include monoclonal antibodies programmed cell death protein-1 (PD-1)/programmed death protein ligand-1 (PD-L1) and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4). In addition, other immunotherapies are available, including adoptive T cell therapies (ACT), such as adoptive transfer of chimeric antigen receptor (CAR)- and T cell receptor (TCR)-T cells, as well as bulk tumor-infiltrating lymphocyte (TIL) therapy. Unfortunately, the response rates of immunotherapy are not satisfactory, and some patients may lose their therapeutic effects after a period of treatment. Moreover, gastrointestinal cancers usually have a low response rate to immunotherapies because cancer cells may evade the immune response, contributing to immune tolerance.13 Further exploration of the mechanisms by which cancer cells are resistant to immunotherapies may provide a basis for the development of new promising immunotherapies for gastrointestinal cancers.14 A previous study has revealed that the gut microbiota, a complex community of trillions of microorganisms, plays a crucial role in immunotherapeutic responses in patients with gastrointestinal cancer.15 Besides physiological functions, the gut microbiota can regulate oncogenesis and resistance to antitumor therapies. During antitumor therapies, many medicines are commonly used for reducing adverse events and these medicines include antidiabetics, aspirin, proton pump inhibitors, psychotropic drugs, and nonsteroidal anti-inflammatory drugs, and analgesics such as opioids. Interestingly, treatment with these drugs can significantly modulate the composition of gut microbiota. Hence, understanding the regulatory effect of gut microbiota on antitumor immunotherapies may help improve their therapeutic efficacy in gastrointestinal cancers. Nanoparticles represent a promising strategy to enhance efficacy and reduce the toxicity of immunotherapy. In the context of treatment of colorectal cancer, nanoparticles can be used to transmit various substances with the potential to affect immune cells and their immunoreactivity within the tumor immune microenvironment, targeting the immune system, and activating multiple immune cells including, but not limited to, T cells.16

Besides tumor cells, there are several types of immune cells, endothelial cells, fibroblasts, extracellular matrix as well as others in the TME of gastrointestinal tumors. These components in TME are the key factors affecting tumor initiation, progression, and immune responses. The balance between cellular and humoral components as well as various inflammatory responses in the TME is critical for tumor growth. Cellular immunotherapy and pharmacological immunotherapy can modulate the TME to enhance tumor immunogenicity and antitumor immune responses to limit tumor growth. However, the efficacy of immunotherapies, such as ICIs, is impaired by the dynamic changes in the TME, which may induce acquired resistance in gastrointestinal cancers. Thus, further understanding of the dynamic changes in the TME may help develop new therapeutic strategies for improvement of immunotherapies and prevention of therapeutic resistance in gastrointestinal cancers. To determine the interaction between TME and tumor cells, further exploration of the mechanism underlying immune escape and immune tolerance, and effectively reshaping the TME are the leading solutions for cancer immunotherapies. Bioprinting of 3D cell culture models can help monitor complex cell behaviors under physical simulation conditions, modulate multiple types of cells in an easy-to-handle system, and study cell interaction in real-time.17, 18 With improved access to clinical tissues and better technologies to visualize cells in situ and dissect heterocellular signaling, the use of bioprinting to understand the TME holds great promise to address translational research questions. Furthermore, the 3D cell culture models are valuable for the development of anticancer drugs. Personalized cancer models can also be used to screen personalized antitumor drugs and promote basic cancer research, drug development, and precise medicines. Despite the attractiveness of 3D bioprinting, there are several technical challenges in the printing platforms, cells, and materials used to build 3D models because of the inherent challenge of comprehensively replicating the cellular behaviors and structural complexity of natural tissues.

Emerging biomarkers, particularly ctDNA, CTCs, and exosomes, are increasingly driving diagnosis advances, allowing for the early identification and dynamic monitoring of gastrointestinal cancers. Apart from the other useful treatment modalities (such as endoscopic therapy, surgery, radiotherapy, chemotherapy, and targeted therapy), immunotherapy, with its unique and valuable benefits, has made significant progress in the management of gastrointestinal cancers, which is our primary focus area. Concomitant nano-based therapeutics represent a promising strategy to improve the efficacy of immunotherapy. Fortunately, the collaboration in nonmedical disciplines, such as AI, 3D bioprinting, and materials science, can enable in-depth characterization of the TME, improving the diagnosis and treatment of gastrointestinal cancers and fostering multidisciplinary treatment strategies in the future. However, limitations are yet undeniable, which implies that new advances are often accompanied by formidable challenges to overcome.

Kai Li and Zhenning Wang conceptualized and designed the study. Mingguang Ju drafted the first version of the manuscript. All authors read and edited the manuscript.

The authors declare no conflict of interest. Professor Kai Li and Zhenning Wang are members of Chronic Diseases and Translational Medicine editorial board and are not involved in the peer review process of this article.

None.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
6.70
自引率
0.00%
发文量
195
审稿时长
35 weeks
期刊介绍: This journal aims to promote progress from basic research to clinical practice and to provide a forum for communication among basic, translational, and clinical research practitioners and physicians from all relevant disciplines. Chronic diseases such as cardiovascular diseases, cancer, diabetes, stroke, chronic respiratory diseases (such as asthma and COPD), chronic kidney diseases, and related translational research. Topics of interest for Chronic Diseases and Translational Medicine include Research and commentary on models of chronic diseases with significant implications for disease diagnosis and treatment Investigative studies of human biology with an emphasis on disease Perspectives and reviews on research topics that discuss the implications of findings from the viewpoints of basic science and clinical practic.
期刊最新文献
Table of Contents Guide for Authors Association of cardiorenal biomarkers with mortality in metabolic syndrome patients: A prospective cohort study from NHANES Current status and perspectives in environmental oncology S-acylation of Ca2+ transport proteins in cancer
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1