1
|
de Souza Leão LQ, de Andrade JC, Marques GM, Guimarães CC, Vieira de Albuquerque RDF, E Silva AS, de Araujo KP, de Oliveira MP, Gonçalves AF, Figueiredo HF, Lira DL, Alves MA, Conte-Junior CA, de Aquino PF. Rapid prediction of cervical cancer and high-grade precursor lesions: An integrated approach using low-field 1H NMR and chemometric analysis. Clin Chim Acta 2025; 574:120346. [PMID: 40334834 DOI: 10.1016/j.cca.2025.120346] [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: 03/07/2025] [Revised: 04/11/2025] [Accepted: 05/04/2025] [Indexed: 05/09/2025]
Abstract
Cervical cancer (CC) is a significant cause of morbidity and mortality in women, often preceded by high-grade cervical intraepithelial lesions (HSIL). Although conventional cytology (Pap smear) is widely used for screening, its sensitivity limitations and high false-positive rate reinforce the need for complementary methods. This study investigated the feasibility of low-field 1H NMR spectroscopy combined with chemometric modeling to differentiate healthy individuals (CON) from patients with HSIL and CC. Principal Component Analysis (PCA) was applied to explore metabolic patterns and identify relevant spectral variables in group differentiation. PCA1 highlighted the separation between CC and the other groups, while PCA2 and PCA3 evidenced intermediate metabolic characteristics in HSIL, reinforcing its role as a transition stage. Three classification scenarios were evaluated using Data-Driven Soft Independent Modeling of Class Analogy (DD-SIMCA): (1) CON as target class, HSIL/CC as outclass classes; (2) HSIL as target class, CON as outclass class; and (3) CC as target class, CON as outclass class. Calibration was optimal (100 % SEN, SPE, ACC, MCC), and prediction showed higher efficacy in detecting CC (SPE = 100 %, MCC = 70 %), indicating that the model was more efficient in screening cervical cancer cases. Furthermore, low-field 1H NMR has demonstrated potential as a metabolomic screening tool. It is a promising alternative due to its greater accessibility, lower operational cost, and non-invasive nature, complementing traditional methods of early detection of tumors and cervical lesions.
Collapse
Affiliation(s)
| | - Jelmir Craveiro de Andrade
- Analytical and Molecular Laboratorial Center (CLAn), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, RJ 21.941-909, Brazil; Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909 RJ, Brazil.
| | - Giovanna Melo Marques
- Leonidas and Maria Deane Institute/Fiocruz Amazônia (ILMD/Fiocruz Amazônia), Manaus, AM, Brazil
| | | | | | | | | | | | | | | | | | - Marina Amaral Alves
- Laboratory of metabolomics applied to systems medicine (Meta2MS), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909 RJ, Brazil
| | - Carlos Adam Conte-Junior
- Analytical and Molecular Laboratorial Center (CLAn), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro, RJ 21.941-909, Brazil; Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909 RJ, Brazil.
| | | |
Collapse
|
2
|
Khan G, Hussain MS, Ahmad S, Alam N, Ali MS, Alam P. Metabolomics as a tool for understanding and treating triple-negative breast cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04234-4. [PMID: 40314763 DOI: 10.1007/s00210-025-04234-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and heterogeneous variant of breast cancer distinguished by a lack of targeted therapies, posing significant challenges in diagnosis and treatment. Metabolomics, the comprehensive study of small compounds in biological systems, has been identified as an instrument for revealing the metabolic underpinnings of TNBC. This review highlights recent advancements in metabolomic approaches, such as mass spectrometry and nuclear magnetic resonance, which have identified metabolic vulnerabilities, resistance mechanisms, and potential therapeutic targets. Key findings include alterations in fatty acid, amino acid, and glutathione metabolism, along with hypoxia-driven metabolic reprogramming that contributes to disease progression. The combination of metabolomics with multi-omics techniques, supported by advanced computational methods such as machine learning, offers a pathway to overcome challenges in data standardization and biological complexity. Emerging strategies, including the use of artificial intelligence and multidimensional omics approaches, are paving the way for personalized medicine by enabling the discovery of novel biomarkers and targeted therapies. Despite these advances, significant hurdles remain, including the need for robust data standardization, validation of findings in diverse patient cohorts, and seamless integration with clinical workflows. By addressing these challenges, metabolomics has the potential to revolutionize TNBC management, offering tools for early detection, precision therapy, and improved patient outcomes. This review underscores the importance of interdisciplinary collaboration to translate metabolomic insights into actionable clinical applications.
Collapse
Affiliation(s)
- Gyas Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Md Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Prem Nagar, Dehradun, Uttarakhand, 248007, India.
| | - Sarfaraz Ahmad
- Department of Clinical Practice, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Nawazish Alam
- Department of Clinical Practice, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Md Sajid Ali
- Department of Pharmaceutics, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Prawez Alam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| |
Collapse
|
3
|
Chiu A, Rahimi M, Lee W. A-SIMA/A-MAP: a comprehensive toolkit for NMR-based metabolomics analysis. Metabolomics 2024; 21:10. [PMID: 39702618 DOI: 10.1007/s11306-024-02208-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 12/07/2024] [Indexed: 12/21/2024]
Abstract
INTRODUCTION Metabolomics is the comprehensive study of small molecules in biological systems. It has recently garnered attention for its wide variety of applications such as diseases, drug treatments, agriculture, and more. As the interest in metabolomics grow, meeting the demands of cutting-edge research requires software tools that not only advance analytical capabilities, but also prioritize user-friendly features. OBJECTIVES In response to this need, we present two new computer programs, A-SIMA: Advanced-Software for Interactive Metabolite Analysis and A-MAP: A Multivariate Analysis Program. These tools aim to introduce new capabilities for metabolite identification and data analysis, and thereby advancing the computational methodology in NMR-based metabolomics. METHODS A-SIMA is designed with an easy-to-use graphical user interface which allows users to perform metabolite identification on 1D and 2D NMR data effortlessly with complete control over the identification process. Similarly, A-MAP facilitates multivariate statistical analysis of metabolite data through a straightforward process. It offers analysis options such as Principal Component Analysis and Orthogonal Partial Least Squares-Discriminant Analysis using regions of interests as inputs. RESULTS Both A-SIMA and A-MAP are pre-built in the POKY suite, available at https://poky.clas.ucdenver.edu , with tutorial videos on YouTube for guidance on not only the programs, but also installation. The POKY suite is a software program for NMR biomolecular analysis. With the addition of these programs in POKY, researchers and professionals can experience a fully integrated process for every step of their metabolite analysis. Data can also be easily exported from these programs to be applied elsewhere. CONCLUSION The introduction of A-SIMA and A-MAP can be promising tools that can lead significant advancements in metabolomics research. These tools offer enhanced capabilities for metabolite analysis and statistical modelling in a user-friendly manner. Their integration into the POKY suite ensures accessibility, usability, and efficiency.
Collapse
Affiliation(s)
- Abigail Chiu
- Department of Chemistry, University of Colorado Denver, Denver, CO, 80204, USA
| | - Mehdi Rahimi
- Department of Chemistry, University of Colorado Denver, Denver, CO, 80204, USA
| | - Woonghee Lee
- Department of Chemistry, University of Colorado Denver, Denver, CO, 80204, USA.
| |
Collapse
|
4
|
Serrano García L, Jávega B, Llombart Cussac A, Gión M, Pérez-García JM, Cortés J, Fernández-Murga ML. Patterns of immune evasion in triple-negative breast cancer and new potential therapeutic targets: a review. Front Immunol 2024; 15:1513421. [PMID: 39735530 PMCID: PMC11671371 DOI: 10.3389/fimmu.2024.1513421] [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: 10/18/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer characterized by the absence of progesterone and estrogen receptors and low (or absent) HER2 expression. TNBC accounts for 15-20% of all breast cancers. It is associated with younger age, a higher mutational burden, and an increased risk of recurrence and mortality. Standard treatment for TNBC primarily relies on cytotoxic agents, such as taxanes, anthracyclines, and platinum compounds for both early and advanced stages of the disease. Several targeted therapies, including bevacizumab and sunitinib, have failed to demonstrate significant clinical benefit in TNBC. The emergence of immune checkpoint inhibitors (ICI) has revolutionized cancer treatment. By stimulating the immune system, ICIs induce a durable anti-tumor response across various solid tumors. TNBC is a particularly promising target for treatment with ICIs due to the higher levels of tumor-infiltrating lymphocytes (TIL), increased PD-L1 expression, and higher mutational burden, which generates tumor-specific neoantigens that activate immune cells. ICIs administered as monotherapy in advanced TNBC yields only a modest response; however, response rates significantly improve when ICIs are combined with cytotoxic agents, particularly in tumors expressing PD-L1. Pembrolizumab is approved for use in both early and advanced TNBC in combination with standard chemotherapy. However, more research is needed to identify more potent biomarkers, and to better elucidate the synergism of ICIs with other targeted agents. In this review, we explore the challenges of immunotherapy in TNBC, examining the mechanisms of tumor progression mediated by immune cells within the tumor microenvironment, and the signaling pathways involved in both primary and acquired resistance. Finally, we provide a comprehensive overview of ongoing clinical trials underway to investigate novel immune-targeted therapies for TNBC.
Collapse
Affiliation(s)
- Lucía Serrano García
- Medical Oncology Department, Hospital Arnau de Vilanova, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Valencia, Spain
| | - Beatriz Jávega
- Medical Oncology Department, Hospital Arnau de Vilanova, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Valencia, Spain
| | - Antonio Llombart Cussac
- Medical Oncology Department, Hospital Arnau de Vilanova, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Valencia, Spain
- Grupo Oncología Traslacional, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-Centro de Estudios Universitarios (CEU), Alfara del Patriarca, Spain
- Medica Scientia Innovation Research (MEDSIR), Oncoclínicas & Co., Jersey City, NJ, United States
| | - María Gión
- Medical Oncology Department, Hospital Ramon y Cajal, Madrid, Spain
| | - José Manuel Pérez-García
- Medica Scientia Innovation Research (MEDSIR), Oncoclínicas & Co., Jersey City, NJ, United States
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain
| | - Javier Cortés
- Medica Scientia Innovation Research (MEDSIR), Oncoclínicas & Co., Jersey City, NJ, United States
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain
- Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, Spain
| | - María Leonor Fernández-Murga
- Medical Oncology Department, Hospital Arnau de Vilanova, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Valencia, Spain
| |
Collapse
|
5
|
Yamada M, Jinno H, Naruse S, Isono Y, Maeda Y, Sato A, Matsumoto A, Ikeda T, Sugimoto M. Predictive analysis of breast cancer response to neoadjuvant chemotherapy through plasma metabolomics. Breast Cancer Res Treat 2024; 207:393-404. [PMID: 38740665 DOI: 10.1007/s10549-024-07370-2] [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: 02/01/2024] [Accepted: 04/25/2024] [Indexed: 05/16/2024]
Abstract
PURPOSE Preoperative chemotherapy is a critical component of breast cancer management, yet its effectiveness is not uniform. Moreover, the adverse effects associated with chemotherapy necessitate the identification of a patient subgroup that would derive the maximum benefit from this treatment. This study aimed to establish a method for predicting the response to neoadjuvant chemotherapy in breast cancer patients utilizing a metabolomic approach. METHODS Plasma samples were obtained from 87 breast cancer patients undergoing neoadjuvant chemotherapy at our facility, collected both before the commencement of the treatment and before the second treatment cycle. Metabolite analysis was conducted using capillary electrophoresis-mass spectrometry (CE-MS) and liquid chromatography-mass spectrometry (LC-MS). We performed comparative profiling of metabolite concentrations by assessing the metabolite profiles of patients who achieved a pathological complete response (pCR) against those who did not, both in initial and subsequent treatment cycles. RESULTS Significant variances were observed in the metabolite profiles between pCR and non-pCR cases, both at the onset of preoperative chemotherapy and before the second cycle. Noteworthy distinctions were also evident between the metabolite profiles from the initial and the second neoadjuvant chemotherapy courses. Furthermore, metabolite profiles exhibited variations associated with intrinsic subtypes at all assessed time points. CONCLUSION The application of plasma metabolomics, utilizing CE-MS and LC-MS, may serve as a tool for predicting the efficacy of neoadjuvant chemotherapy in breast cancer in the future after all necessary validations have been completed.
Collapse
Affiliation(s)
- Miki Yamada
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Hiromitsu Jinno
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan.
| | - Saki Naruse
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Yuka Isono
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Yuka Maeda
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Ayana Sato
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Akiko Matsumoto
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Tatsuhiko Ikeda
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Masahiro Sugimoto
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata, 997-0052, Japan
- Institute of Medical Science, Tokyo Medical University, Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| |
Collapse
|
6
|
Bhardwaj JK, Siwach A, Sachdeva SN. Metabolomics and cellular altered pathways in cancer biology: A review. J Biochem Mol Toxicol 2024; 38:e23807. [PMID: 39148273 DOI: 10.1002/jbt.23807] [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: 03/05/2024] [Revised: 07/16/2024] [Accepted: 08/01/2024] [Indexed: 08/17/2024]
Abstract
Cancer is a deadly disease that affects a cell's metabolism and surrounding tissues. Understanding the fundamental mechanisms of metabolic alterations in cancer cells would assist in developing cancer treatment targets and approaches. From this perspective, metabolomics is a great analytical tool to clarify the mechanisms of cancer therapy as well as a useful tool to investigate cancer from a distinct viewpoint. It is a powerful emerging technology that detects up to thousands of molecules in tissues and biofluids. Like other "-omics" technologies, metabolomics involves the comprehensive investigation of micromolecule metabolites and can reveal important details about the cancer state that is otherwise not apparent. Recent developments in metabolomics technologies have made it possible to investigate cancer metabolism in greater depth and comprehend how cancer cells utilize metabolic pathways to make the amino acids, nucleotides, and lipids required for tumorigenesis. These new technologies have made it possible to learn more about cancer metabolism. Here, we review the cellular and systemic effects of cancer and cancer treatments on metabolism. The current study provides an overview of metabolomics, emphasizing the current technologies and their use in clinical and translational research settings.
Collapse
Affiliation(s)
- Jitender Kumar Bhardwaj
- Reproductive Physiology Laboratory, Department of Zoology, Kurukshetra University, Kurukshetra, Haryana, India
| | - Anshu Siwach
- Reproductive Physiology Laboratory, Department of Zoology, Kurukshetra University, Kurukshetra, Haryana, India
| | - Som Nath Sachdeva
- Department of Civil Engineering, National Institute of Technology, Kurukshetra and Kurukshetra University, Kurukshetra, Haryana, India
| |
Collapse
|
7
|
Talarico MCR, Derchain S, da Silva LF, Sforça ML, Rocco SA, Cardoso MR, Sarian LO. Metabolomic Profiling of Breast Cancer Patients Undergoing Neoadjuvant Chemotherapy for Predicting Disease-Free and Overall Survival. Int J Mol Sci 2024; 25:8639. [PMID: 39201325 PMCID: PMC11354796 DOI: 10.3390/ijms25168639] [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: 06/27/2024] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Breast cancer (BC) remains a significant global health concern, with neoadjuvant chemotherapy (NACT) offering preoperative benefits like tumor downstaging and treatment response assessment. However, identifying factors influencing post-NACT treatment response and survival outcomes is challenging. Metabolomic approaches offer promising insights into understanding these outcomes. This study analyzed the serum of 80 BC patients before and after NACT, followed for up to five years, correlating with disease-free survival (DFS) and overall survival (OS). Using untargeted nuclear magnetic resonance (NMR) spectroscopy and a novel statistical model that avoids collinearity issues, we identified metabolic changes associated with survival outcomes. Four metabolites (histidine, lactate, serine, and taurine) were significantly associated with DFS. We developed a metabolite-related survival score (MRSS) from these metabolites, stratifying patients into low- and high-risk relapse groups, independent of classical prognostic factors. High-risk patients had a hazard ratio (HR) for DFS of 3.42 (95% CI 1.51-7.74; p = 0.003) after adjustment for disease stage and age. A similar trend was observed for OS (HR of 3.34, 95% CI 1.64-6.80; p < 0.001). Multivariate Cox proportional hazards analysis confirmed the independent prognostic value of the MRSS. Our findings suggest the potential of metabolomic data, alongside traditional markers, in guiding personalized treatment decisions and risk stratification in BC patients undergoing NACT. This study provides a methodological framework for leveraging metabolomics in survival analyses.
Collapse
Affiliation(s)
- Maria Cecília Ramiro Talarico
- Department of Obstetrics and Gynecology, Division of Gynecologic and Breast Oncology, School of Medical Sciences, University of Campinas (UNICAMP-Universidade Estadual de Campinas), Campinas 13083-881, SP, Brazil
| | - Sophie Derchain
- Department of Obstetrics and Gynecology, Division of Gynecologic and Breast Oncology, School of Medical Sciences, University of Campinas (UNICAMP-Universidade Estadual de Campinas), Campinas 13083-881, SP, Brazil
| | | | - Maurício L. Sforça
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, SP, Brazil
| | - Silvana A. Rocco
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, SP, Brazil
| | - Marcella R. Cardoso
- Division of Gynecologic Oncology-MGH Global Disaster Response, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Center for Global Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Luís Otávio Sarian
- Department of Obstetrics and Gynecology, Division of Gynecologic and Breast Oncology, School of Medical Sciences, University of Campinas (UNICAMP-Universidade Estadual de Campinas), Campinas 13083-881, SP, Brazil
| |
Collapse
|
8
|
Espelage L, Wagner N, Placke JM, Ugurel S, Tasdogan A. The Interplay between Metabolic Adaptations and Diet in Cancer Immunotherapy. Clin Cancer Res 2024; 30:3117-3127. [PMID: 38771898 DOI: 10.1158/1078-0432.ccr-22-3468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/07/2023] [Accepted: 04/15/2024] [Indexed: 05/23/2024]
Abstract
Over the past decade, cancer immunotherapy has significantly advanced through the introduction of immune checkpoint inhibitors and the augmentation of adoptive cell transfer to enhance the innate cancer defense mechanisms. Despite these remarkable achievements, some cancers exhibit resistance to immunotherapy, with limited patient responsiveness and development of therapy resistance. Metabolic adaptations in both immune cells and cancer cells have emerged as central contributors to immunotherapy resistance. In the last few years, new insights emphasized the critical role of cancer and immune cell metabolism in animal models and patients. During therapy, immune cells undergo important metabolic shifts crucial for their acquired effector function against cancer cells. However, cancer cell metabolic rewiring and nutrient competition within tumor microenvironment (TME) alters many immune functions, affecting their fitness, polarization, recruitment, and survival. These interactions have initiated the development of novel therapies targeting tumor cell metabolism and favoring antitumor immunity within the TME. Furthermore, there has been increasing interest in comprehending how diet impacts the response to immunotherapy, given the demonstrated immunomodulatory and antitumor activity of various nutrients. In conclusion, recent advances in preclinical and clinical studies have highlighted the capacity of immune-based cancer therapies. Therefore, further exploration into the metabolic requirements of immune cells within the TME holds significant promise for the development of innovative therapeutic approaches that can effectively combat cancer in patients.
Collapse
Affiliation(s)
- Lena Espelage
- Department of Dermatology, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
| | - Natalie Wagner
- Department of Dermatology, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
| | - Jan-Malte Placke
- Department of Dermatology, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
| | - Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
| |
Collapse
|
9
|
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
|
10
|
Arenas M, Fargas-Saladié M, Moreno-Solé M, Moyano-Femenia L, Jiménez-Franco A, Canela-Capdevila M, Castañé H, Martínez-Navidad C, Camps J, Joven J. Metabolomics and triple-negative breast cancer: A systematic review. Heliyon 2024; 10:e23628. [PMID: 38187259 PMCID: PMC10770474 DOI: 10.1016/j.heliyon.2023.e23628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 01/09/2024] Open
Abstract
Triple-negative breast cancer stands out as the most aggressive subtype of breast malignancy and is characterized by an unfavourable prognosis. Objective: This systematic review summarizes the insights gleaned from metabolomic analyses of individuals afflicted with this cancer variant. The overarching goal was to delineate the molecular alterations associated with triple-negative breast cancer, pinpointing potential therapeutic targets and novel biomarkers. Methods: We systematically searched for evidence using the PubMed database and followed the PRISMA and STARLITE guidelines. The search parameters were delimited to articles published within the last 13 years. Results: From an initial pool of 148 scrutinized articles, 17 studies involving 1686 participants were deemed eligible for inclusion. The current body of research shows a paucity of studies, and the available evidence presents conflicting outcomes. Notwithstanding, Pathway Enrichment Analysis identified the urea and glucose-alanine cycles as the most affected metabolic pathways, followed by arginine, proline, and aspartate metabolism. Conclusion: Future investigations need to focus on elucidating which of those metabolites and/or pathways might be reliable candidates for novel therapeutic interventions or reliable biomarkers for diagnosis and prognosis of this subtype of breast cancer.
Collapse
Affiliation(s)
- Meritxell Arenas
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut D'Investigació Sanitària Pere Virgili, Universitat Rovira I Virgili, Reus, Spain
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan de Reus, Institut D'Investigació Sanitària Pere Virgili, Universitat Rovira I Virgili, Reus, Spain
| | - Maria Fargas-Saladié
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut D'Investigació Sanitària Pere Virgili, Universitat Rovira I Virgili, Reus, Spain
| | - Marta Moreno-Solé
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut D'Investigació Sanitària Pere Virgili, Universitat Rovira I Virgili, Reus, Spain
| | - Lucía Moyano-Femenia
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut D'Investigació Sanitària Pere Virgili, Universitat Rovira I Virgili, Reus, Spain
| | - Andrea Jiménez-Franco
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan de Reus, Institut D'Investigació Sanitària Pere Virgili, Universitat Rovira I Virgili, Reus, Spain
| | - Marta Canela-Capdevila
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut D'Investigació Sanitària Pere Virgili, Universitat Rovira I Virgili, Reus, Spain
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan de Reus, Institut D'Investigació Sanitària Pere Virgili, Universitat Rovira I Virgili, Reus, Spain
| | - Helena Castañé
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan de Reus, Institut D'Investigació Sanitària Pere Virgili, Universitat Rovira I Virgili, Reus, Spain
| | - Cristian Martínez-Navidad
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan de Reus, Institut D'Investigació Sanitària Pere Virgili, Universitat Rovira I Virgili, Reus, Spain
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan de Reus, Institut D'Investigació Sanitària Pere Virgili, Universitat Rovira I Virgili, Reus, Spain
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan de Reus, Institut D'Investigació Sanitària Pere Virgili, Universitat Rovira I Virgili, Reus, Spain
| |
Collapse
|
11
|
Santos MD, Barros I, Brandão P, Lacerda L. Amino Acid Profiles in the Biological Fluids and Tumor Tissue of CRC Patients. Cancers (Basel) 2023; 16:69. [PMID: 38201497 PMCID: PMC10778074 DOI: 10.3390/cancers16010069] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Amino acids are the building blocks of proteins and essential players in pathways such as the citric acid and urea cycle, purine and pyrimidine biosynthesis, and redox cell signaling. Therefore, it is unsurprising that these molecules have a significant role in cancer metabolism and its metabolic plasticity. As one of the most prevalent malign diseases, colorectal cancer needs biomarkers for its early detection, prognostic, and prediction of response to therapy. However, the available biomarkers for this disease must be more powerful and present several drawbacks, such as high costs and complex laboratory procedures. Metabolomics has gathered substantial attention in the past two decades as a screening platform to study new metabolites, partly due to the development of techniques, such as mass spectrometry or liquid chromatography, which have become standard practice in diagnostic procedures for other diseases. Extensive metabolomic studies have been performed in colorectal cancer (CRC) patients in the past years, and several exciting results concerning amino acid metabolism have been found. This review aims to gather and present findings concerning alterations in the amino acid plasma pool of colorectal cancer patients.
Collapse
Affiliation(s)
- Marisa Domingues Santos
- Colorectal Unit, Hospital de Santo António, Centro Hospitalar Universitário de Santo António, 4050-651 Porto, Portugal;
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (I.B.); (L.L.)
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
| | - Ivo Barros
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (I.B.); (L.L.)
| | - Pedro Brandão
- Colorectal Unit, Hospital de Santo António, Centro Hospitalar Universitário de Santo António, 4050-651 Porto, Portugal;
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (I.B.); (L.L.)
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
| | - Lúcia Lacerda
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (I.B.); (L.L.)
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
- Genetic Laboratory Service, Centro de Genética Médica Jacinto de Magalhães, Centro Hospitalar Universitário de Santo António, 4050-651 Porto, Portugal
| |
Collapse
|
12
|
Alvarez-Frutos L, Barriuso D, Duran M, Infante M, Kroemer G, Palacios-Ramirez R, Senovilla L. Multiomics insights on the onset, progression, and metastatic evolution of breast cancer. Front Oncol 2023; 13:1292046. [PMID: 38169859 PMCID: PMC10758476 DOI: 10.3389/fonc.2023.1292046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
Breast cancer is the most common malignant neoplasm in women. Despite progress to date, 700,000 women worldwide died of this disease in 2020. Apparently, the prognostic markers currently used in the clinic are not sufficient to determine the most appropriate treatment. For this reason, great efforts have been made in recent years to identify new molecular biomarkers that will allow more precise and personalized therapeutic decisions in both primary and recurrent breast cancers. These molecular biomarkers include genetic and post-transcriptional alterations, changes in protein expression, as well as metabolic, immunological or microbial changes identified by multiple omics technologies (e.g., genomics, epigenomics, transcriptomics, proteomics, glycomics, metabolomics, lipidomics, immunomics and microbiomics). This review summarizes studies based on omics analysis that have identified new biomarkers for diagnosis, patient stratification, differentiation between stages of tumor development (initiation, progression, and metastasis/recurrence), and their relevance for treatment selection. Furthermore, this review highlights the importance of clinical trials based on multiomics studies and the need to advance in this direction in order to establish personalized therapies and prolong disease-free survival of these patients in the future.
Collapse
Affiliation(s)
- Lucia Alvarez-Frutos
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Daniel Barriuso
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Mercedes Duran
- Laboratory of Molecular Genetics of Hereditary Cancer, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Mar Infante
- Laboratory of Molecular Genetics of Hereditary Cancer, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, Paris, France
| | - Roberto Palacios-Ramirez
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Laura Senovilla
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| |
Collapse
|
13
|
Cai Y, Wang Z, Guo S, Lin C, Yao H, Yang Q, Wang Y, Yu X, He X, Sun W, Qiu S, Guo Y, Tang S, Xie Y, Zhang A. Detection, mechanisms, and therapeutic implications of oncometabolites. Trends Endocrinol Metab 2023; 34:849-861. [PMID: 37739878 DOI: 10.1016/j.tem.2023.08.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 08/10/2023] [Accepted: 08/28/2023] [Indexed: 09/24/2023]
Abstract
Metabolic abnormalities are a hallmark of cancer cells and are essential to tumor progression. Oncometabolites have pleiotropic effects on cancer biology and affect a plethora of processes, from oncogenesis and metabolism to therapeutic resistance. Targeting oncometabolites, therefore, could offer promising therapeutic avenues against tumor growth and resistance to treatments. Recent advances in characterizing the metabolic profiles of cancer cells are shedding light on the underlying mechanisms and associated metabolic networks. This review summarizes the diverse detection methods, molecular mechanisms, and therapeutic targets of oncometabolites, which may lead to targeting oncometabolism for cancer therapy.
Collapse
Affiliation(s)
- Ying Cai
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China; Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Zhibo Wang
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China; Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Sifan Guo
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China; Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Chunsheng Lin
- Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hong Yao
- First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Qiang Yang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yan Wang
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China
| | - Xiaodan Yu
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China
| | - Xiaowen He
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China
| | - Wanying Sun
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China
| | - Shi Qiu
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China.
| | - Yu Guo
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China.
| | - Songqi Tang
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China.
| | - Yiqiang Xie
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China.
| | - Aihua Zhang
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan General Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Medical University, Haikou 571199, China; Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| |
Collapse
|
14
|
Orsini A, Diquigiovanni C, Bonora E. Omics Technologies Improving Breast Cancer Research and Diagnostics. Int J Mol Sci 2023; 24:12690. [PMID: 37628869 PMCID: PMC10454385 DOI: 10.3390/ijms241612690] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Breast cancer (BC) has yielded approximately 2.26 million new cases and has caused nearly 685,000 deaths worldwide in the last two years, making it the most common diagnosed cancer type in the world. BC is an intricate ecosystem formed by both the tumor microenvironment and malignant cells, and its heterogeneity impacts the response to treatment. Biomedical research has entered the era of massive omics data thanks to the high-throughput sequencing revolution, quick progress and widespread adoption. These technologies-liquid biopsy, transcriptomics, epigenomics, proteomics, metabolomics, pharmaco-omics and artificial intelligence imaging-could help researchers and clinicians to better understand the formation and evolution of BC. This review focuses on the findings of recent multi-omics-based research that has been applied to BC research, with an introduction to every omics technique and their applications for the different BC phenotypes, biomarkers, target therapies, diagnosis, treatment and prognosis, to provide a comprehensive overview of the possibilities of BC research.
Collapse
Affiliation(s)
| | - Chiara Diquigiovanni
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40131 Bologna, Italy; (A.O.); (E.B.)
| | | |
Collapse
|
15
|
Tardito S, MacKay C. Rethinking our approach to cancer metabolism to deliver patient benefit. Br J Cancer 2023; 129:406-415. [PMID: 37340094 PMCID: PMC10403540 DOI: 10.1038/s41416-023-02324-9] [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: 02/28/2023] [Revised: 05/25/2023] [Accepted: 06/12/2023] [Indexed: 06/22/2023] Open
Abstract
Altered cellular metabolism is a major mechanism by which tumours support nutrient consumption associated with increased cellular proliferation. Selective dependency on specific metabolic pathways provides a therapeutic vulnerability that can be targeted in cancer therapy. Anti-metabolites have been used clinically since the 1940s and several agents targeting nucleotide metabolism are now well established as standard of care treatment in a range of indications. However, despite great progress in our understanding of the metabolic requirements of cancer and non-cancer cells within the tumour microenvironment, there has been limited clinical success for novel agents targeting pathways outside of nucleotide metabolism. We believe that there is significant therapeutic potential in targeting metabolic processes within cancer that is yet to be fully realised. However, current approaches to identify novel targets, test novel therapies and select patient populations most likely to benefit are sub-optimal. We highlight recent advances in technologies and understanding that will support the identification and validation of novel targets, re-evaluation of existing targets and design of optimal clinical positioning strategies to deliver patient benefit.
Collapse
Affiliation(s)
- Saverio Tardito
- The Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Craig MacKay
- Cancer Research Horizons, The Cancer Research UK Beatson Institute, Glasgow, UK.
| |
Collapse
|
16
|
Matou-Nasri S, Aldawood M, Alanazi F, Khan AL. Updates on Triple-Negative Breast Cancer in Type 2 Diabetes Mellitus Patients: From Risk Factors to Diagnosis, Biomarkers and Therapy. Diagnostics (Basel) 2023; 13:2390. [PMID: 37510134 PMCID: PMC10378597 DOI: 10.3390/diagnostics13142390] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is usually the most malignant and aggressive mammary epithelial tumor characterized by the lack of expression for estrogen receptors and progesterone receptors, and the absence of epidermal growth factor receptor (HER)2 amplification. Corresponding to 15-20% of all breast cancers and well-known by its poor clinical outcome, this negative receptor expression deprives TNBC from targeted therapy and makes its management therapeutically challenging. Type 2 diabetes mellitus (T2DM) is the most common ageing metabolic disorder due to insulin deficiency or resistance resulting in hyperglycemia, hyperinsulinemia, and hyperlipidemia. Due to metabolic and hormonal imbalances, there are many interplays between both chronic disorders leading to increased risk of breast cancer, especially TNBC, diagnosed in T2DM patients. The purpose of this review is to provide up-to-date information related to epidemiology and clinicopathological features, risk factors, diagnosis, biomarkers, and current therapy/clinical trials for TNBC patients with T2DM compared to non-diabetic counterparts. Thus, in-depth investigation of the diabetic complications on TNBC onset, development, and progression and the discovery of biomarkers would improve TNBC management through early diagnosis, tailoring therapy for a better outcome of T2DM patients diagnosed with TNBC.
Collapse
Affiliation(s)
- Sabine Matou-Nasri
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia
- Biosciences Department, Faculty of the School for Systems Biology, George Mason University, Manassas, VA 22030, USA
| | - Maram Aldawood
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia
- Post Graduate and Zoology Department, King Saud University, Riyadh 12372, Saudi Arabia
| | - Fatimah Alanazi
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh 11481, Saudi Arabia
- Biosciences Department, Faculty of the School for Systems Biology, George Mason University, Manassas, VA 22030, USA
| | - Abdul Latif Khan
- Tissue Biobank, KAIMRC, MNG-HA, Riyadh 11481, Saudi Arabia
- Pathology and Clinical Laboratory Medicine, King Abdulaziz Medical City (KAMC), Riyadh 11564, Saudi Arabia
| |
Collapse
|
17
|
Danzi F, Pacchiana R, Mafficini A, Scupoli MT, Scarpa A, Donadelli M, Fiore A. To metabolomics and beyond: a technological portfolio to investigate cancer metabolism. Signal Transduct Target Ther 2023; 8:137. [PMID: 36949046 PMCID: PMC10033890 DOI: 10.1038/s41392-023-01380-0] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/08/2023] [Accepted: 02/15/2023] [Indexed: 03/24/2023] Open
Abstract
Tumour cells have exquisite flexibility in reprogramming their metabolism in order to support tumour initiation, progression, metastasis and resistance to therapies. These reprogrammed activities include a complete rewiring of the bioenergetic, biosynthetic and redox status to sustain the increased energetic demand of the cells. Over the last decades, the cancer metabolism field has seen an explosion of new biochemical technologies giving more tools than ever before to navigate this complexity. Within a cell or a tissue, the metabolites constitute the direct signature of the molecular phenotype and thus their profiling has concrete clinical applications in oncology. Metabolomics and fluxomics, are key technological approaches that mainly revolutionized the field enabling researchers to have both a qualitative and mechanistic model of the biochemical activities in cancer. Furthermore, the upgrade from bulk to single-cell analysis technologies provided unprecedented opportunity to investigate cancer biology at cellular resolution allowing an in depth quantitative analysis of complex and heterogenous diseases. More recently, the advent of functional genomic screening allowed the identification of molecular pathways, cellular processes, biomarkers and novel therapeutic targets that in concert with other technologies allow patient stratification and identification of new treatment regimens. This review is intended to be a guide for researchers to cancer metabolism, highlighting current and emerging technologies, emphasizing advantages, disadvantages and applications with the potential of leading the development of innovative anti-cancer therapies.
Collapse
Affiliation(s)
- Federica Danzi
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Raffaella Pacchiana
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Andrea Mafficini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Maria T Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, Biology and Genetics Section, University of Verona, Verona, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- ARC-NET Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy.
| | - Alessandra Fiore
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| |
Collapse
|
18
|
Baranovicova E, Racay P, Zubor P, Smolar M, Kudelova E, Halasova E, Dvorska D, Dankova Z. Circulating metabolites in the early stage of breast cancer were not related to cancer stage or subtypes but associated with ki67 level. Promising statistical discrimination from controls. Mol Cell Probes 2022; 66:101862. [PMID: 36162596 DOI: 10.1016/j.mcp.2022.101862] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/30/2022] [Accepted: 09/13/2022] [Indexed: 12/30/2022]
Abstract
It was documented that the presence of malignancy in an organism causes metabolomic alterations in blood plasma which applies also to breast cancer. Breast cancer is a heterogeneous disease and there are only limited known relations of plasma metabolomic signatures with the tumour characteristics in early BC and knowing them would be of great advantage in noninvasive diagnostics. In this study, we focused on the metabolic alterations in early BC in blood plasma with the aim to identify metabolomic characteristics of BC subtypes. We used 50 early BC patients (FIGO stage I and II), where no additional metabolomic changes from metastatically changed remote organs were to be expected. We compared plasma levels of metabolites against controls and among various molecular and histological BC subtypes. BC patients showed decreased plasma levels of branched-chain amino acids BCAAs (and related keto-acids), histidine pyruvate and alanine balanced with an increased level of 3-hydroxybutyrate. The levels of circulating metabolites were not related to BC molecular subtypes (luminal A/luminal B), histological finding or grade, eventually stage, which indicate that in early BC, the BC patients share common metabolomics fingerprint in blood plasma independent of grade, stage or molecular subtype of BC. We observed statistically significant correlations between tumour proliferation marker Ki-67 level and circulating metabolites: alanine, citrate, tyrosine, glutamine, histidine and proline. This may point out the metabolites those levels could be associated with tumour growth, and conversely, the rate of tumour proliferation could be potentially estimated from plasma metabolites. When analyzing metabolomic changes in BC, we concluded that some of them could be associated with the metabolomic features of cancer cells, but the other observed alterations in blood plasma are the results of the complex mutual biochemical pathways in the comprehensive inter-organ metabolic exchange and communication. In the end, statistical discrimination against controls performed with AUC >0.91 showed the very promising potential of plasma metabolomics in the search for biomarkers for oncologic diseases.
Collapse
Affiliation(s)
- Eva Baranovicova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Mala Hora 4, 036 01, Martin, Slovakia.
| | - Peter Racay
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Mala Hora 4, 036 01, Martin, Slovakia.
| | - Pavol Zubor
- OBGY Health & Care, Ltd., 01001, Zilina, Slovak Republic; Department of Gynecologic Oncology, The Norwegian Radium Hospital, Oslo University Hospital, 0379, Oslo, Norway; Department of Obstetrics and Gynecology, The University Hospital of North Norway, 8516, Narvik, Norway; Vi Kan helse -Metro legesenter AS, 1473, Lørenskog, Norway.
| | - Marek Smolar
- Clinic of Surgery and Transplant Centre, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Kollarova 2, 036 01, Martin, Slovakia.
| | - Eva Kudelova
- Clinic of Surgery and Transplant Centre, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Kollarova 2, 036 01, Martin, Slovakia.
| | - Erika Halasova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Mala Hora 4, 036 01, Martin, Slovakia.
| | - Dana Dvorska
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Mala Hora 4, 036 01, Martin, Slovakia.
| | - Zuzana Dankova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Mala Hora 4, 036 01, Martin, Slovakia.
| |
Collapse
|
19
|
Nascentes Melo LM, Lesner NP, Sabatier M, Ubellacker JM, Tasdogan A. Emerging metabolomic tools to study cancer metastasis. Trends Cancer 2022; 8:988-1001. [PMID: 35909026 DOI: 10.1016/j.trecan.2022.07.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/24/2022] [Accepted: 07/06/2022] [Indexed: 12/24/2022]
Abstract
Metastasis is responsible for 90% of deaths in patients with cancer. Understanding the role of metabolism during metastasis has been limited by the development of robust and sensitive technologies that capture metabolic processes in metastasizing cancer cells. We discuss the current technologies available to study (i) metabolism in primary and metastatic cancer cells and (ii) metabolic interactions between cancer cells and the tumor microenvironment (TME) at different stages of the metastatic cascade. We identify advantages and disadvantages of each method and discuss how these tools and technologies will further improve our understanding of metastasis. Studies investigating the complex metabolic rewiring of different cells using state-of-the-art metabolomic technologies have the potential to reveal novel biological processes and therapeutic interventions for human cancers.
Collapse
Affiliation(s)
| | - Nicholas P Lesner
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marie Sabatier
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jessalyn M Ubellacker
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen and German Cancer Consortium, Partner Site, Essen, Germany.
| |
Collapse
|
20
|
Jin Y, Fan S, Jiang W, Zhang J, Yang L, Xiao J, An H, Ren L. Two effective models based on comprehensive lipidomics and metabolomics can distinguish BC versus HCs, and TNBC versus non-TNBC. Proteomics Clin Appl 2022; 17:e2200042. [PMID: 36443927 DOI: 10.1002/prca.202200042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/10/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Lipidomics and metabolomics are closely related to tumor phenotypes, and serum lipoprotein subclasses and small-molecule metabolites are considered as promising biomarkers for breast cancer (BC) diagnosis. This study aimed to explore potential biomarker models based on lipidomic and metabolomic analysis that could distinguish BC from healthy controls (HCs) and triple-negative BC (TNBC) from non-TNBC. METHODS Blood samples were collected from 114 patients with BC and 75 HCs. A total of 112 types of lipoprotein subclasses and 30 types of small-molecule metabolites in the serum were detected by 1 H-NMR. All lipoprotein subclasses and small-molecule metabolites were subjected to a three-step screening process in the order of significance (p < 0.05), univariate regression (p < 0.1), and lasso regression (nonzero coefficient). Discriminant models of BC versus HCs and TNBC versus non-TNBC were established using binary logistic regression. RESULTS We developed a valid discriminant model based on three-biomarker panel (formic acid, TPA2, and L6TG) that could distinguish patients with BC from HCs. The area under the receiver operating characteristic curve (AUC) was 0.999 (95% confidence interval [CI]: 0.995-1.000) and 0.990 (95% CI: 0.959-1.000) in the training and validation sets, respectively. Based on the panel (D-dimer, CA15-3, CEA, L5CH, glutamine, and ornithine), a discriminant model was established to differentiate between TNBC and non-TNBC, with AUC of 0.892 (95% CI: 0.778-0.967) and 0.905 (95% CI: 0.754-0.987) in the training and validation sets, respectively. CONCLUSION This study revealed lipidomic and metabolomic differences between BC versus HCs and TNBC versus non-TNBC. Two validated discriminatory models established against lipidomic and metabolomic differences can accurately distinguish BC from HCs and TNBC from non-TNBC. IMPACT Two validated discriminatory models can be used for early BC screening and help BC patients avoid time-consuming, expensive, and dangerous BC screening.
Collapse
Affiliation(s)
- Yu Jin
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Shuoqing Fan
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Wenna Jiang
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Jingya Zhang
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Lexin Yang
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Jiawei Xiao
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Haohua An
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Li Ren
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
21
|
Irajizad E, Wu R, Vykoukal J, Murage E, Spencer R, Dennison JB, Moulder S, Ravenberg E, Lim B, Litton J, Tripathym D, Valero V, Damodaran S, Rauch GM, Adrada B, Candelaria R, White JB, Brewster A, Arun B, Long JP, Do KA, Hanash S, Fahrmann JF. Application of Artificial Intelligence to Plasma Metabolomics Profiles to Predict Response to Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer. Front Artif Intell 2022; 5:876100. [PMID: 36034598 PMCID: PMC9403735 DOI: 10.3389/frai.2022.876100] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
There is a need to identify biomarkers predictive of response to neoadjuvant chemotherapy (NACT) in triple-negative breast cancer (TNBC). We previously obtained evidence that a polyamine signature in the blood is associated with TNBC development and progression. In this study, we evaluated whether plasma polyamines and other metabolites may identify TNBC patients who are less likely to respond to NACT. Pre-treatment plasma levels of acetylated polyamines were elevated in TNBC patients that had moderate to extensive tumor burden (RCB-II/III) following NACT compared to those that achieved a complete pathological response (pCR/RCB-0) or had minimal residual disease (RCB-I). We further applied artificial intelligence to comprehensive metabolic profiles to identify additional metabolites associated with treatment response. Using a deep learning model (DLM), a metabolite panel consisting of two polyamines as well as nine additional metabolites was developed for improved prediction of RCB-II/III. The DLM has potential clinical value for identifying TNBC patients who are unlikely to respond to NACT and who may benefit from other treatment modalities.
Collapse
Affiliation(s)
- Ehsan Irajizad
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ranran Wu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jody Vykoukal
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Eunice Murage
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rachelle Spencer
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jennifer B. Dennison
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Stacy Moulder
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Elizabeth Ravenberg
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Bora Lim
- Breast Cancer Research Program, Baylor College of Medicine, Houston, TX, United States
| | - Jennifer Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Debu Tripathym
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Senthil Damodaran
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Gaiane M. Rauch
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Beatriz Adrada
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rosalind Candelaria
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jason B. White
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Abenaa Brewster
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Banu Arun
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - James P. Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kim Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sam Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: Sam Hanash
| | - Johannes F. Fahrmann
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Johannes F. Fahrmann
| |
Collapse
|
22
|
Zhuang J, Yang X, Zheng Q, Li K, Cai L, Yu H, Lv J, Bai K, Cao Q, Li P, Yang H, Wang J, Lu Q. Metabolic Profiling of Bladder Cancer Patients' Serum Reveals Their Sensitivity to Neoadjuvant Chemotherapy. Metabolites 2022; 12:metabo12060558. [PMID: 35736490 PMCID: PMC9229374 DOI: 10.3390/metabo12060558] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/07/2022] [Accepted: 06/13/2022] [Indexed: 02/07/2023] Open
Abstract
Numerous patients with muscle-invasive bladder cancer develop low responsiveness to cisplatin. Our purpose was to explore differential metabolites derived from serum in bladder cancer patients treated with neoadjuvant chemotherapy (NAC). Data of patients diagnosed with cT2-4aNxM0 was collected. Blood samples were retained prospectively before the first chemotherapy for untargeted metabolomics by 1H-NMR and UPLC-MS. To identify characterized metabolites, multivariate statistical analyses were applied, and the intersection of the differential metabolites discovered by the two approaches was used to identify viable biomarkers. A total of 18 patients (6 NAC-sensitive patients and 12 NAC-resistant patients) were enrolled. There were 29 metabolites detected by 1H-NMR and 147 metabolites identified by UPLC-MS. Multivariate statistics demonstrated that in the sensitive group, glutamine and taurine were considerably increased compared to their levels in the resistant group, while glutamate and hypoxanthine were remarkably decreased. Pathway analysis and enrichment analysis showed significant alterations in amino acid pathways, suggesting that response to chemotherapy may be related to amino acid metabolism. In addition, hallmark analysis showed that DNA repair played a regulatory role. Overall, serum metabolic profiles of NAC sensitivity are significantly different in bladder cancer patients. Glycine, hypoxanthine, taurine and glutamine may be the potential biomarkers for clinical treatment. Amino acid metabolism has potential value in enhancing drug efficacy.
Collapse
Affiliation(s)
- Juntao Zhuang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (J.Z.); (X.Y.); (K.L.); (L.C.); (H.Y.); (J.L.); (K.B.); (Q.C.); (P.L.); (H.Y.)
| | - Xiao Yang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (J.Z.); (X.Y.); (K.L.); (L.C.); (H.Y.); (J.L.); (K.B.); (Q.C.); (P.L.); (H.Y.)
| | - Qi Zheng
- Center of Molecular Metabolism, Nanjing University of Science and Technology, Nanjing 210094, China;
| | - Kai Li
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (J.Z.); (X.Y.); (K.L.); (L.C.); (H.Y.); (J.L.); (K.B.); (Q.C.); (P.L.); (H.Y.)
| | - Lingkai Cai
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (J.Z.); (X.Y.); (K.L.); (L.C.); (H.Y.); (J.L.); (K.B.); (Q.C.); (P.L.); (H.Y.)
| | - Hao Yu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (J.Z.); (X.Y.); (K.L.); (L.C.); (H.Y.); (J.L.); (K.B.); (Q.C.); (P.L.); (H.Y.)
| | - Jiancheng Lv
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (J.Z.); (X.Y.); (K.L.); (L.C.); (H.Y.); (J.L.); (K.B.); (Q.C.); (P.L.); (H.Y.)
| | - Kexin Bai
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (J.Z.); (X.Y.); (K.L.); (L.C.); (H.Y.); (J.L.); (K.B.); (Q.C.); (P.L.); (H.Y.)
| | - Qiang Cao
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (J.Z.); (X.Y.); (K.L.); (L.C.); (H.Y.); (J.L.); (K.B.); (Q.C.); (P.L.); (H.Y.)
| | - Pengchao Li
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (J.Z.); (X.Y.); (K.L.); (L.C.); (H.Y.); (J.L.); (K.B.); (Q.C.); (P.L.); (H.Y.)
| | - Haiwei Yang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (J.Z.); (X.Y.); (K.L.); (L.C.); (H.Y.); (J.L.); (K.B.); (Q.C.); (P.L.); (H.Y.)
| | - Junsong Wang
- Center of Molecular Metabolism, Nanjing University of Science and Technology, Nanjing 210094, China;
- Correspondence: (J.W.); (Q.L.)
| | - Qiang Lu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (J.Z.); (X.Y.); (K.L.); (L.C.); (H.Y.); (J.L.); (K.B.); (Q.C.); (P.L.); (H.Y.)
- Correspondence: (J.W.); (Q.L.)
| |
Collapse
|