1
|
Yang J, Zhu Y, Zhou Y, Zhang J, Wei Y, Liu Y, Zhang B, Xie J, An X, Qi X, Yue Y, Zhang L, Zhang X, Fu Z, Liu K. Potential biomarkers develop for predicting the prognosis of patients with esophageal squamous cell carcinoma after optimized chemoradiotherapy using serum metabolomics. BMC Cancer 2025; 25:438. [PMID: 40069698 PMCID: PMC11900641 DOI: 10.1186/s12885-025-13866-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC), the most common type of esophageal cancer, characterized by low five-year survival rate, and concurrent chemoradiotherapy (CCRT) has been proposed to treat ESCC, while potential biomarkers for prognostic monitoring after optimized CCRT remains unknown. METHODS Serum samples from 45 patients with ESCC were collected and categorized into three groups: Control (pre-CCRT), CCRT (during CCRT), and CCRT-1 M (one-month post-CCRT). The therapeutic effect was evaluated using CT imaging and established evaluation criteria. Untargeted metabolomic analysis was performed on the serum samples to identify differential metabolites caused by CCRT treatment, assessing their potential for prognostic monitoring. RESULTS CCRT had significant therapeutic efficacy in patients with ESCC, as indicated by CT imaging and RECIST 1.1 solid tumor evaluation criteria. Notably, several metabolic markers were identified through non-targeted metabolomic analysis, highlighting changes following CCRT treatment. These differential metabolites are involved in the dysregulation of phenylalanine, tyrosine, and tryptophan biosynthesis, as well as histidine, arginine, and proline metabolism, and glycine, serine, and threonine metabolism, suggesting a reduction in glucose metabolism in patients with ESCC after CCRT. Additionally, ROC analysis indicated that the AUC of these metabolites exceeded 0.661, underscoring their diagnostic value for assessing CCRT efficacy and their potential use in prognostic monitoring. Comparative metabolomic analysis identified L-phenylalanine and lysine as promising serum biomarkers for predicting therapeutic outcomes. CONCLUSIONS CCRT shows considerable therapeutic benefit in patients with ESCC, with observed reductions in glucose metabolism post-treatment. L-phenylalanine and lysine may serve as potential serum biomarkers to predict CCRT efficacy.
Collapse
Affiliation(s)
- Jie Yang
- Central Laboratory, Danyang People's Hospital of Jiangsu Province, Danyang, Jiangsu, 212300, P.R. China
| | - Yunyun Zhu
- Department of Radiotherapy, 900 Hospital of the Joint Logistics Team, (Dongfang Hospital, Xiamen University), Fuzhou, Fujian, 350025, P.R. China
| | - Yijian Zhou
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Jiaying Zhang
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Yuxuan Wei
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Yongpan Liu
- School of Life Science, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Bo Zhang
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Jialing Xie
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Xiaolu An
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Xianhua Qi
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Yuting Yue
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Lijia Zhang
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Xiajun Zhang
- Central Laboratory, Danyang People's Hospital of Jiangsu Province, Danyang, Jiangsu, 212300, P.R. China.
| | - Zhichao Fu
- Department of Radiotherapy, 900 Hospital of the Joint Logistics Team, (Dongfang Hospital, Xiamen University), Fuzhou, Fujian, 350025, P.R. China.
| | - Kuancan Liu
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China.
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China.
| |
Collapse
|
2
|
Zhu E, Xie Q, Huang X, Zhang Z. Application of spatial omics in gastric cancer. Pathol Res Pract 2024; 262:155503. [PMID: 39128411 DOI: 10.1016/j.prp.2024.155503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/13/2024]
Abstract
Gastric cancer (GC), a globally prevalent and lethal malignancy, continues to be a key research focus. However, due to its considerable heterogeneity and complex pathogenesis, the treatment and diagnosis of gastric cancer still face significant challenges. With the rapid development of spatial omics technology, which provides insights into the spatial information within tumor tissues, it has emerged as a significant tool in gastric cancer research. This technology affords new insights into the pathology and molecular biology of gastric cancer for scientists. This review discusses recent advances in spatial omics technology for gastric cancer research, highlighting its applications in the tumor microenvironment (TME), tumor heterogeneity, tumor genesis and development mechanisms, and the identification of potential biomarkers and therapeutic targets. Moreover, this article highlights spatial omics' potential in precision medicine and summarizes existing challenges and future directions. It anticipates spatial omics' continuing impact on gastric cancer research, aiming to improve diagnostic and therapeutic approaches for patients. With this review, we aim to offer a comprehensive overview to scientists and clinicians in gastric cancer research, motivating further exploration and utilization of spatial omics technology. Our goal is to improve patient outcomes, including survival rates and quality of life.
Collapse
Affiliation(s)
- Erran Zhu
- Department of Clinical Medicine, Grade 20, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Qi Xie
- Department of Clinical Medicine, Grade 20, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Xinqi Huang
- Excellent Class, Clinical Medicine, Grade 20, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Zhiwei Zhang
- Cancer Research Institute of Hengyang Medical College, University of South China; Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan; Department of Pathology, Department of Pathology of Hengyang Medical College, University of South China; The First Affiliated Hospital of University of South China, China.
| |
Collapse
|
3
|
Silva AAR, Cardoso MR, de Oliveira DC, Godoy P, Talarico MCR, Gutiérrez JM, Rodrigues Peres RM, de Carvalho LM, Miyaguti NADS, Sarian LO, Tata A, Derchain SFM, Porcari AM. Plasma Metabolome Signatures to Predict Responsiveness to Neoadjuvant Chemotherapy in Breast Cancer. Cancers (Basel) 2024; 16:2473. [PMID: 39001535 PMCID: PMC11240312 DOI: 10.3390/cancers16132473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 06/27/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy (NACT) has arisen as a treatment option for breast cancer (BC). However, the response to NACT is still unpredictable and dependent on cancer subtype. Metabolomics is a tool for predicting biomarkers and chemotherapy response. We used plasma to verify metabolomic alterations in BC before NACT, relating to clinical data. METHODS Liquid chromatography coupled to mass spectrometry (LC-MS) was performed on pre-NACT plasma from patients with BC (n = 75). After data filtering, an SVM model for classification was built and validated with 75%/25% of the data, respectively. RESULTS The model composed of 19 identified metabolites effectively predicted NACT response for training/validation sets with high sensitivity (95.4%/93.3%), specificity (91.6%/100.0%), and accuracy (94.6%/94.7%). In both sets, the panel correctly classified 95% of resistant and 94% of sensitive females. Most compounds identified by the model were lipids and amino acids and revealed pathway alterations related to chemoresistance. CONCLUSION We developed a model for predicting patient response to NACT. These metabolite panels allow clinical gain by building precision medicine strategies based on tumor stratification.
Collapse
Affiliation(s)
- Alex Ap. Rosini Silva
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Sala 211, Prédio 5, Bragança Paulista 12916900, São Paulo, Brazil; (A.A.R.S.); (D.C.d.O.)
| | - Marcella R. Cardoso
- Department of Obstetrics and Gynecology, Division of Gynecologic and Breast Oncology, Faculty of Medical Sciences, University of Campinas (UNICAMP—Universidade Estadual de Campinas), Campinas 13083881, São Paulo, Brazil
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Danilo Cardoso de Oliveira
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Sala 211, Prédio 5, Bragança Paulista 12916900, São Paulo, Brazil; (A.A.R.S.); (D.C.d.O.)
| | - Pedro Godoy
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Sala 211, Prédio 5, Bragança Paulista 12916900, São Paulo, Brazil; (A.A.R.S.); (D.C.d.O.)
| | - Maria Cecília R. Talarico
- Department of Obstetrics and Gynecology, Division of Gynecologic and Breast Oncology, Faculty of Medical Sciences, University of Campinas (UNICAMP—Universidade Estadual de Campinas), Campinas 13083881, São Paulo, Brazil
| | - Junier Marrero Gutiérrez
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Sala 211, Prédio 5, Bragança Paulista 12916900, São Paulo, Brazil; (A.A.R.S.); (D.C.d.O.)
| | - Raquel M. Rodrigues Peres
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Sala 211, Prédio 5, Bragança Paulista 12916900, São Paulo, Brazil; (A.A.R.S.); (D.C.d.O.)
| | - Lucas M. de Carvalho
- Post Graduate Program in Health Sciences, São Francisco University, Bragança Paulista 12916900, São Paulo, Brazil
| | - Natália Angelo da Silva Miyaguti
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Sala 211, Prédio 5, Bragança Paulista 12916900, São Paulo, Brazil; (A.A.R.S.); (D.C.d.O.)
| | - Luis O. Sarian
- Department of Obstetrics and Gynecology, Division of Gynecologic and Breast Oncology, Faculty of Medical Sciences, University of Campinas (UNICAMP—Universidade Estadual de Campinas), Campinas 13083881, São Paulo, Brazil
| | - Alessandra Tata
- Laboratory of Experimental Chemistry, Istituto Zooprofilattico Sperimentale delle Venezie (IZSVe), Viale Fiume 78, 36100 Vicenza, Italy;
| | - Sophie F. M. Derchain
- Department of Obstetrics and Gynecology, Division of Gynecologic and Breast Oncology, Faculty of Medical Sciences, University of Campinas (UNICAMP—Universidade Estadual de Campinas), Campinas 13083881, São Paulo, Brazil
| | - Andreia M. Porcari
- MSLife Laboratory of Mass Spectrometry, Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Sala 211, Prédio 5, Bragança Paulista 12916900, São Paulo, Brazil; (A.A.R.S.); (D.C.d.O.)
| |
Collapse
|
4
|
Stillger MN, Li MJ, Hönscheid P, von Neubeck C, Föll MC. Advancing rare cancer research by MALDI mass spectrometry imaging: Applications, challenges, and future perspectives in sarcoma. Proteomics 2024; 24:e2300001. [PMID: 38402423 DOI: 10.1002/pmic.202300001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 02/26/2024]
Abstract
MALDI mass spectrometry imaging (MALDI imaging) uniquely advances cancer research, by measuring spatial distribution of endogenous and exogenous molecules directly from tissue sections. These molecular maps provide valuable insights into basic and translational cancer research, including tumor biology, tumor microenvironment, biomarker identification, drug treatment, and patient stratification. Despite its advantages, MALDI imaging is underutilized in studying rare cancers. Sarcomas, a group of malignant mesenchymal tumors, pose unique challenges in medical research due to their complex heterogeneity and low incidence, resulting in understudied subtypes with suboptimal management and outcomes. In this review, we explore the applicability of MALDI imaging in sarcoma research, showcasing its value in understanding this highly heterogeneous and challenging rare cancer. We summarize all MALDI imaging studies in sarcoma to date, highlight their impact on key research fields, including molecular signatures, cancer heterogeneity, and drug studies. We address specific challenges encountered when employing MALDI imaging for sarcomas, and propose solutions, such as using formalin-fixed paraffin-embedded tissues, and multiplexed experiments, and considerations for multi-site studies and digital data sharing practices. Through this review, we aim to spark collaboration between MALDI imaging researchers and clinical colleagues, to deploy the unique capabilities of MALDI imaging in the context of sarcoma.
Collapse
Affiliation(s)
- Maren Nicole Stillger
- Institute for Surgical Pathology, Faculty of Medicine, University Medical Center, Freiburg, Germany
- Bioinformatics Group, Department of Computer Science, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Mujia Jenny Li
- Institute for Surgical Pathology, Faculty of Medicine, University Medical Center, Freiburg, Germany
- Institute for Pharmaceutical Sciences, University of Freiburg, Freiburg, Germany
| | - Pia Hönscheid
- Institute of Pathology, Faculty of Medicine, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases, Partner Site Dresden, German Cancer Research Center Heidelberg, Dresden, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cläre von Neubeck
- Department of Particle Therapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Melanie Christine Föll
- Institute for Surgical Pathology, Faculty of Medicine, University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Khoury College of Computer Sciences, Northeastern University, Boston, USA
| |
Collapse
|
5
|
Yang Z, Guan F, Bronk L, Zhao L. Multi-omics approaches for biomarker discovery in predicting the response of esophageal cancer to neoadjuvant therapy: A multidimensional perspective. Pharmacol Ther 2024; 254:108591. [PMID: 38286161 DOI: 10.1016/j.pharmthera.2024.108591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/02/2023] [Accepted: 01/04/2024] [Indexed: 01/31/2024]
Abstract
Neoadjuvant chemoradiotherapy (NCRT) followed by surgery has been established as the standard treatment strategy for operable locally advanced esophageal cancer (EC). However, achieving pathologic complete response (pCR) or near pCR to NCRT is significantly associated with a considerable improvement in survival outcomes, while pCR patients may help organ preservation for patients by active surveillance to avoid planned surgery. Thus, there is an urgent need for improved biomarkers to predict EC chemoradiation response in research and clinical settings. Advances in multiple high-throughput technologies such as next-generation sequencing have facilitated the discovery of novel predictive biomarkers, specifically based on multi-omics data, including genomic/transcriptomic sequencings and proteomic/metabolomic mass spectra. The application of multi-omics data has shown the benefits in improving the understanding of underlying mechanisms of NCRT sensitivity/resistance in EC. Particularly, the prominent development of artificial intelligence (AI) has introduced a new direction in cancer research. The integration of multi-omics data has significantly advanced our knowledge of the disease and enabled the identification of valuable biomarkers for predicting treatment response from diverse dimension levels, especially with rapid advances in biotechnological and AI methodologies. Herein, we summarize the current status of research on the use of multi-omics technologies in predicting NCRT response for EC patients. Current limitations, challenges, and future perspectives of these multi-omics platforms will be addressed to assist in experimental designs and clinical use for further integrated analysis.
Collapse
Affiliation(s)
- Zhi Yang
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, 15 West Changle Road, Xi'an, China
| | - Fada Guan
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510, United States of America
| | - Lawrence Bronk
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States of America
| | - Lina Zhao
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, 15 West Changle Road, Xi'an, China.
| |
Collapse
|
6
|
Jiao R, Jiang W, Xu K, Luo Q, Wang L, Zhao C. Lipid metabolism analysis in esophageal cancer and associated drug discovery. J Pharm Anal 2024; 14:1-15. [PMID: 38352954 PMCID: PMC10859535 DOI: 10.1016/j.jpha.2023.08.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/27/2023] [Accepted: 08/29/2023] [Indexed: 02/16/2024] Open
Abstract
Esophageal cancer is an upper gastrointestinal malignancy with a bleak prognosis. It is still being explored in depth due to its complex molecular mechanisms of occurrence and development. Lipids play a crucial role in cells by participating in energy supply, biofilm formation, and signal transduction processes, and lipid metabolic reprogramming also constitutes a significant characteristic of malignant tumors. More and more studies have found esophageal cancer has obvious lipid metabolism abnormalities throughout its beginning, progress, and treatment resistance. The inhibition of tumor growth and the enhancement of antitumor therapy efficacy can be achieved through the regulation of lipid metabolism. Therefore, we reviewed and analyzed the research results and latest findings for lipid metabolism and associated analysis techniques in esophageal cancer, and comprehensively proved the value of lipid metabolic reprogramming in the evolution and treatment resistance of esophageal cancer, as well as its significance in exploring potential therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Ruidi Jiao
- Bionic Sensing and Intelligence Center, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518000, China
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, 518116, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518000, China
| | - Wei Jiang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, 518116, China
| | - Kunpeng Xu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, 518116, China
| | - Qian Luo
- Bionic Sensing and Intelligence Center, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Luhua Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, 518116, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518000, China
| | - Chao Zhao
- Bionic Sensing and Intelligence Center, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518000, China
| |
Collapse
|
7
|
Krestensen KK, Heeren RMA, Balluff B. State-of-the-art mass spectrometry imaging applications in biomedical research. Analyst 2023; 148:6161-6187. [PMID: 37947390 DOI: 10.1039/d3an01495a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Mass spectrometry imaging has advanced from a niche technique to a widely applied spatial biology tool operating at the forefront of numerous fields, most notably making a significant impact in biomedical pharmacological research. The growth of the field has gone hand in hand with an increase in publications and usage of the technique by new laboratories, and consequently this has led to a shift from general MSI reviews to topic-specific reviews. Given this development, we see the need to recapitulate the strengths of MSI by providing a more holistic overview of state-of-the-art MSI studies to provide the new generation of researchers with an up-to-date reference framework. Here we review scientific advances for the six largest biomedical fields of MSI application (oncology, pharmacology, neurology, cardiovascular diseases, endocrinology, and rheumatology). These publications thereby give examples for at least one of the following categories: they provide novel mechanistic insights, use an exceptionally large cohort size, establish a workflow that has the potential to become a high-impact methodology, or are highly cited in their field. We finally have a look into new emerging fields and trends in MSI (immunology, microbiology, infectious diseases, and aging), as applied MSI is continuously broadening as a result of technological breakthroughs.
Collapse
Affiliation(s)
- Kasper K Krestensen
- The Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, 6229 ER Maastricht, The Netherlands.
| | - Ron M A Heeren
- The Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, 6229 ER Maastricht, The Netherlands.
| | - Benjamin Balluff
- The Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, 6229 ER Maastricht, The Netherlands.
| |
Collapse
|
8
|
Cheng D, Zhao W, Chen R, Li D, Tang S, Fang C, Ji M. Neoadjuvant PD-1 blockade combined with chemotherapy is not superior to neoadjuvant chemotherapy alone in resectable locally advanced esophageal carcinoma. World J Surg Oncol 2023; 21:33. [PMID: 36737768 PMCID: PMC9896760 DOI: 10.1186/s12957-023-02915-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy (nCT) or neoadjuvant chemoradiotherapy followed by surgery has been recommended as standard treatment in patients with locally advanced esophageal cancer (LAEC). But the risk of tumor recurrence still remained, and many patients refused or abandoned radiotherapy because of the intolerable adverse effects in China. Neoadjuvant immunochemotherapy (nICT) followed by surgery has become an emerging treatment in patients with esophageal cancer. There was still no consensus on whether nICT was superior to nCT alone in patients with esophageal cancer. METHODS In this retrospective study, patients with resectable esophageal cancer who received surgery after nICT (n=26, 40%) or nCT alone (n=39, 60%) were included. The patients were classified as nICT or nCT arm. The primary endpoints were pathological tumor response (PTR) and event-free survival (EFS). The different clinic-pathological features were compared by the Kruskal-Wallis test for continuous variables and the Chi-square (χ2) test for categorical variables. Kaplan-Meier curves were used to estimate EFS from the date of treatment to recurrence or death. All tests were 2-sided with a significative P-value defined <.05. RESULTS Three (11.5%) of the 26 patients achieved pathological complete remission (pCR) in the nICT group, and four (10.3%) of the 39 patients achieved pCR in the nCT group, respectively (P=1.000). Six (23.1%) of the 26 patients achieved major pathological response (MPR) in the nICT group, and 11 (28.2%) of the 39 patients achieved MPR in the nCT group, respectively (P=0.645). Downstaging was achieved in 13 (44.8%) patients in the nICT group and 16 (55.2%) patients in the nCT group, respectively (P=0.732). To verify the tumor regression grade (TRG) results, we compared them with MPR and pCR, which showed a significant dependency (P< 0.001). Patients who achieved downgrading showed better MPR and pCR rates (P<0.001 and P =0.010). There was no significant difference in EFS between the nICT and nCT groups (HR=1.011, 95% CI: 0.421-2.425, P = 0.981). CONCLUSIONS Neoadjuvant PD-1 blockade combined with chemotherapy was not superior to chemotherapy alone for patients with resectable locally advanced esophageal carcinoma. However, more studies with long-term follow-up were needed to confirm this result.
Collapse
Affiliation(s)
- Daoan Cheng
- Departments of Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213004, China
| | - Weiqing Zhao
- Departments of Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213004, China
| | - Rui Chen
- Departments of Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213004, China
| | - Dong Li
- Departments of Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213004, China
| | - Shuxian Tang
- Departments of Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213004, China
| | - Cheng Fang
- Departments of Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213004, China.
| | - Mei Ji
- Departments of Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213004, China.
| |
Collapse
|
9
|
C3d(g), iron nanoparticles, hemin and cytochrome c may induce oxidative cytotoxicity in tumors and reduce tumor-associated myeloid cells-mediated immunosuppression. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
10
|
Yang XL, Wang P, Ye H, Jiang M, Su YB, Peng XX, Li H, Zhang JY. Untargeted serum metabolomics reveals potential biomarkers and metabolic pathways associated with esophageal cancer. Front Oncol 2022; 12:938234. [PMID: 36176418 PMCID: PMC9513043 DOI: 10.3389/fonc.2022.938234] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 08/16/2022] [Indexed: 12/24/2022] Open
Abstract
Metabolomics has been reported as an efficient tool to screen biomarkers that are related to esophageal cancer. However, the metabolic biomarkers identifying malignant degrees and therapeutic efficacy are still largely unknown in the disease. Here, GC-MS-based metabolomics was used to understand metabolic alteration in 137 serum specimens from patients with esophageal cancer, which is approximately two- to fivefold as many plasma specimens as the previous reports. The elevated amino acid metabolism is in sharp contrast to the reduced carbohydrate as a characteristic feature of esophageal cancer. Comparative metabolomics showed that most metabolic differences were determined between the early stage (0–II) and the late stage (III and IV) among the 0–IV stages of esophageal cancer and between patients who received treatment and those who did not receive treatment. Glycine, serine, and threonine metabolism and glycine were identified as the potentially overlapped metabolic pathway and metabolite, respectively, in both disease progress and treatment effect. Glycine, fructose, ornithine, and threonine can be a potential array for the evaluation of disease prognosis and therapy in esophageal cancer. These results highlight the means of identifying previously unknown biomarkers related to esophageal cancer by a metabolomics approach.
Collapse
Affiliation(s)
- Xiao-li Yang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
| | - Peng Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology and College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Hua Ye
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology and College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Ming Jiang
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
| | - Yu-bin Su
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xuan-xian Peng
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
| | - Hui Li
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
- *Correspondence: Jian-ying Zhang, ; Hui Li,
| | - Jian-ying Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Jian-ying Zhang, ; Hui Li,
| |
Collapse
|
11
|
Shen J, Sun N, Zens P, Kunzke T, Buck A, Prade VM, Wang J, Wang Q, Hu R, Feuchtinger A, Berezowska S, Walch A. Spatial metabolomics for evaluating response to neoadjuvant therapy in non-small cell lung cancer patients. Cancer Commun (Lond) 2022; 42:517-535. [PMID: 35593195 PMCID: PMC9198346 DOI: 10.1002/cac2.12310] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/18/2022] [Accepted: 05/10/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The response to neoadjuvant chemotherapy (NAC) differs substantially among individual patients with non-small cell lung cancer (NSCLC). Major pathological response (MPR) is a histomorphological read-out used to assess treatment response and prognosis in patients NSCLC after NAC. Although spatial metabolomics is a promising tool for evaluating metabolic phenotypes, it has not yet been utilized to assess therapy responses in patients with NSCLC. We evaluated the potential application of spatial metabolomics in cancer tissues to assess the response to NAC, using a metabolic classifier that utilizes mass spectrometry imaging combined with machine learning. METHODS Resected NSCLC tissue specimens obtained after NAC (n = 88) were subjected to high-resolution mass spectrometry, and these data were used to develop an approach for assessing the response to NAC in patients with NSCLC. The specificities of the generated tumor cell and stroma classifiers were validated by applying this approach to a cohort of biologically matched chemotherapy-naïve patients with NSCLC (n = 85). RESULTS The developed tumor cell metabolic classifier stratified patients into different prognostic groups with 81.6% accuracy, whereas the stroma metabolic classifier displayed 78.4% accuracy. By contrast, the accuracies of MPR and TNM staging for stratification were 62.5% and 54.1%, respectively. The combination of metabolic and MPR classifiers showed slightly lower accuracy than either individual metabolic classifier. In multivariate analysis, metabolic classifiers were the only independent prognostic factors identified (tumor: P = 0.001, hazards ratio [HR] = 3.823, 95% confidence interval [CI] = 1.716-8.514; stroma: P = 0.049, HR = 2.180, 95% CI = 1.004-4.737), whereas MPR (P = 0.804; HR = 0.913; 95% CI = 0.445-1.874) and TNM staging (P = 0.078; HR = 1.223; 95% CI = 0.977-1.550) were not independent prognostic factors. Using Kaplan-Meier survival analyses, both tumor and stroma metabolic classifiers were able to further stratify patients as NAC responders (P < 0.001) and non-responders (P < 0.001). CONCLUSIONS Our findings indicate that the metabolic constitutions of both tumor cells and the stroma are valuable additions to the classical histomorphology-based assessment of tumor response.
Collapse
Affiliation(s)
- Jian Shen
- Research Unit Analytical PathologyHelmholtz Zentrum München – German Research Center for Environmental HealthNeuherberg85764Germany
| | - Na Sun
- Research Unit Analytical PathologyHelmholtz Zentrum München – German Research Center for Environmental HealthNeuherberg85764Germany
| | - Philipp Zens
- Institute of PathologyUniversity of BernBern3008Switzerland
- Graduate School for Health SciencesUniversity of BernMittelstrasse 43Bern3012Switzerland
| | - Thomas Kunzke
- Research Unit Analytical PathologyHelmholtz Zentrum München – German Research Center for Environmental HealthNeuherberg85764Germany
| | - Achim Buck
- Research Unit Analytical PathologyHelmholtz Zentrum München – German Research Center for Environmental HealthNeuherberg85764Germany
| | - Verena M. Prade
- Research Unit Analytical PathologyHelmholtz Zentrum München – German Research Center for Environmental HealthNeuherberg85764Germany
| | - Jun Wang
- Research Unit Analytical PathologyHelmholtz Zentrum München – German Research Center for Environmental HealthNeuherberg85764Germany
| | - Qian Wang
- Research Unit Analytical PathologyHelmholtz Zentrum München – German Research Center for Environmental HealthNeuherberg85764Germany
| | - Ronggui Hu
- Center for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghai200031P. R. China
| | - Annette Feuchtinger
- Research Unit Analytical PathologyHelmholtz Zentrum München – German Research Center for Environmental HealthNeuherberg85764Germany
| | - Sabina Berezowska
- Institute of PathologyUniversity of BernBern3008Switzerland
- Department of Laboratory Medicine and PathologyInstitute of PathologyLausanne University Hospital and University of LausanneLausanne1011Switzerland
| | - Axel Walch
- Research Unit Analytical PathologyHelmholtz Zentrum München – German Research Center for Environmental HealthNeuherberg85764Germany
| |
Collapse
|
12
|
Dent A, Diamandis P. Integrating computational pathology and proteomics to address tumor heterogeneity. J Pathol 2022; 257:445-453. [DOI: 10.1002/path.5905] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/20/2022] [Accepted: 03/30/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Anglin Dent
- Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario M5S 1A8 Canada
- Princess Margaret Cancer Center University Health Network, Toronto, Ontario, 610 University Avenue, M5G 2C1 Canada
| | - Phedias Diamandis
- Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario M5S 1A8 Canada
- Princess Margaret Cancer Center University Health Network, Toronto, Ontario, 610 University Avenue, M5G 2C1 Canada
- Laboratory Medicine Program University Health Network, 200 Elizabeth Street, Toronto, ON Toronto Ontario M5G 2C4 Canada
- Department of Medical Biophysics University of Toronto Toronto Ontario M5S 1A8 Canada
| |
Collapse
|