1
|
Wu H, Liu Q, Li J, Leng X, He Y, Liu Y, Zhang X, Ouyang Y, Liu Y, Liang W, Xu C. Tumor-Resident Microbiota-Based Risk Model Predicts Neoadjuvant Therapy Response of Locally Advanced Esophageal Squamous Cell Carcinoma Patients. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309742. [PMID: 39268829 DOI: 10.1002/advs.202309742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 07/11/2024] [Indexed: 09/15/2024]
Abstract
Few predictive biomarkers exist for identifying patients who may benefit from neoadjuvant therapy (NAT). The intratumoral microbial composition is comprehensively profiled to predict the efficacy and prognosis of patients with esophageal squamous cell carcinoma (ESCC) who underwent NAT and curative esophagectomy. Least Absolute Shrinkage and Selection Operator (LASSO) regression analysis is conducted to screen for the most closely related microbiota and develop a microbiota-based risk prediction (MRP) model on the genera of TM7x, Sphingobacterium, and Prevotella. The predictive accuracy and prognostic value of the MRP model across multiple centers are validated. The MRP model demonstrates good predictive accuracy for therapeutic responses in the training, validation, and independent validation sets. The MRP model also predicts disease-free survival (p = 0.00074 in the internal validation set and p = 0.0017 in the independent validation set) and overall survival (p = 0.00023 in the internal validation set and p = 0.11 in the independent validation set) of patients. The MRP-plus model basing on MRP, tumor stage, and tumor size can also predict the patients who can benefit from NAT. In conclusion, the developed MRP and MRP-plus models may function as promising biomarkers and prognostic indicators accessible at the time of diagnosis.
Collapse
Affiliation(s)
- Hong Wu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, P. R. China
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, P. R. China
- Jinfeng Laboratory, Chongqing, 400039, P. R. China
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, P. R. China
| | - Qianshi Liu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, P. R. China
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, P. R. China
- Jinfeng Laboratory, Chongqing, 400039, P. R. China
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, P. R. China
| | - Jingpei Li
- Thoracic Surgery Department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510230, P. R. China
| | - Xuefeng Leng
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, P. R. China
| | - Yazhou He
- Department of Oncology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Yiqiang Liu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, P. R. China
- Jinfeng Laboratory, Chongqing, 400039, P. R. China
| | - Xia Zhang
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, P. R. China
- Institute of Pathology and Southwest Cancer Center, Ministry of Education of China, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Chongqing, 400038, P. R. China
| | - Yujie Ouyang
- Acupuncture and Massage College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, P. R. China
| | - Yang Liu
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, P. R. China
| | - Wenhua Liang
- Thoracic Surgery Department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510230, P. R. China
| | - Chuan Xu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, P. R. China
- Jinfeng Laboratory, Chongqing, 400039, P. R. China
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, P. R. China
| |
Collapse
|
2
|
Guevara-Ramírez P, Cadena-Ullauri S, Paz-Cruz E, Ruiz-Pozo VA, Tamayo-Trujillo R, Cabrera-Andrade A, Zambrano AK. Gut Microbiota Disruption in Hematologic Cancer Therapy: Molecular Insights and Implications for Treatment Efficacy. Int J Mol Sci 2024; 25:10255. [PMID: 39408584 PMCID: PMC11476909 DOI: 10.3390/ijms251910255] [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: 07/29/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 10/20/2024] Open
Abstract
Hematologic malignancies (HMs), including leukemia, lymphoma, and multiple myeloma, involve the uncontrolled proliferation of abnormal blood cells, posing significant clinical challenges due to their heterogeneity and varied treatment responses. Despite recent advancements in therapies that have improved survival rates, particularly in chronic lymphocytic leukemia and acute lymphoblastic leukemia, treatments like chemotherapy and stem cell transplantation often disrupt gut microbiota, which can negatively impact treatment outcomes and increase infection risks. This review explores the complex, bidirectional interactions between gut microbiota and cancer treatments in patients with HMs. Gut microbiota can influence drug metabolism through mechanisms such as the production of enzymes like bacterial β-glucuronidases, which can alter drug efficacy and toxicity. Moreover, microbial metabolites like short-chain fatty acids can modulate the host immune response, enhancing treatment effectiveness. However, therapy often reduces the diversity of beneficial bacteria, such as Bifidobacterium and Faecalibacterium, while increasing pathogenic bacteria like Enterococcus and Escherichia coli. These findings highlight the critical need to preserve microbiota diversity during treatment. Future research should focus on personalized microbiome-based therapies, including probiotics, prebiotics, and fecal microbiota transplantation, to improve outcomes and quality of life for patients with hematologic malignancies.
Collapse
Affiliation(s)
- Patricia Guevara-Ramírez
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| | - Santiago Cadena-Ullauri
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| | - Elius Paz-Cruz
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| | - Viviana A. Ruiz-Pozo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| | - Rafael Tamayo-Trujillo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| | - Alejandro Cabrera-Andrade
- Escuela de Enfermería, Facultad de Ciencias de la Salud, Universidad de Las Américas, Quito 170124, Ecuador
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170124, Ecuador
| | - Ana Karina Zambrano
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| |
Collapse
|
3
|
El-Tanani M, Rabbani SA, El-Tanani Y, Matalka II. Metabolic vulnerabilities in cancer: A new therapeutic strategy. Crit Rev Oncol Hematol 2024; 201:104438. [PMID: 38977145 DOI: 10.1016/j.critrevonc.2024.104438] [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: 05/09/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024] Open
Abstract
Cancer metabolism is now a key area for therapeutic intervention, targeting unique metabolic reprogramming crucial for tumor growth and survival. This article reviews the therapeutic potential of addressing metabolic vulnerabilities through glycolysis and glutaminase inhibitors, which disrupt cancer cell metabolism. Challenges such as tumor heterogeneity and adaptive resistance are discussed, with strategies including personalized medicine and predictive biomarkers to enhance treatment efficacy. Additionally, integrating diet and lifestyle changes with metabolic targeting underscores a holistic approach to improving therapy outcomes. The article also examines the benefits of incorporating these strategies into standard care, highlighting the potential for more tailored, safer treatments. In conclusion, exploiting metabolic vulnerabilities promises a new era in oncology, positioning metabolic targeting at the forefront of personalized cancer therapy and transforming patient care.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates.
| | - Syed Arman Rabbani
- RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates.
| | - Yahia El-Tanani
- Medical School, St George's University of London, Cranmer Terrace, Tooting, London, UK
| | - Ismail I Matalka
- RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates; Department of Pathology and Microbiology, Medicine, Jordan University of Science and Technology, Irbid, Jordan.
| |
Collapse
|
4
|
Glieca S, Quarta E, Bottari B, Lal VC, Sonvico F, Buttini F. The role of airways microbiota on local and systemic diseases: a rationale for probiotics delivery to the respiratory tract. Expert Opin Drug Deliv 2024; 21:991-1005. [PMID: 39041243 DOI: 10.1080/17425247.2024.2380334] [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: 02/15/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Recent discoveries in the field of lung microbiota have enabled the investigation of new therapeutic interventions involving the use of inhaled probiotics. AREAS COVERED This review provides an overview of what is known about the correlation between airway dysbiosis and the development of local and systemic diseases, and how this knowledge can be exploited for therapeutic interventions. In particular, the review focused on attempts to formulate probiotics that can be deposited directly on the airways. EXPERT OPINION Despite considerable progress since the emergence of respiratory microbiota restoration as a new research field, numerous clinical implications and benefits remain to be determined. In the case of local diseases, once the pathophysiology is understood, manipulating the lung microbiota through probiotic administration is an approach that can be exploited. In contrast, the effect of pulmonary dysbiosis on systemic diseases remains to be clarified; however, this approach could represent a turning point in their treatment.
Collapse
Affiliation(s)
| | - Eride Quarta
- Food and Drug Department, University of Parma, Parma, Italy
| | | | | | - Fabio Sonvico
- Food and Drug Department, University of Parma, Parma, Italy
- Interdepartmental Center for Innovation in Health Products, Biopharmanet_TEC, University of Parma, Parma, Italy
| | - Francesca Buttini
- Food and Drug Department, University of Parma, Parma, Italy
- Interdepartmental Center for Innovation in Health Products, Biopharmanet_TEC, University of Parma, Parma, Italy
| |
Collapse
|
5
|
Rayan M, Sayed TS, Hussein OJ, Therachiyil L, Maayah ZH, Maccalli C, Uddin S, Prehn JHM, Korashy HM. Unlocking the secrets: exploring the influence of the aryl hydrocarbon receptor and microbiome on cancer development. Cell Mol Biol Lett 2024; 29:33. [PMID: 38448800 PMCID: PMC10918910 DOI: 10.1186/s11658-024-00538-0] [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: 09/11/2023] [Accepted: 01/17/2024] [Indexed: 03/08/2024] Open
Abstract
Gut microbiota regulates various aspects of human physiology by producing metabolites, metabolizing enzymes, and toxins. Many studies have linked microbiota with human health and altered microbiome configurations with the occurrence of several diseases, including cancer. Accumulating evidence suggests that the microbiome can influence the initiation and progression of several cancers. Moreover, some microbiotas of the gut and oral cavity have been reported to infect tumors, initiate metastasis, and promote the spread of cancer to distant organs, thereby influencing the clinical outcome of cancer patients. The gut microbiome has recently been reported to interact with environmental factors such as diet and exposure to environmental toxicants. Exposure to environmental pollutants such as polycyclic aromatic hydrocarbons (PAHs) induces a shift in the gut microbiome metabolic pathways, favoring a proinflammatory microenvironment. In addition, other studies have also correlated cancer incidence with exposure to PAHs. PAHs are known to induce organ carcinogenesis through activating a ligand-activated transcriptional factor termed the aryl hydrocarbon receptor (AhR), which metabolizes PAHs to highly reactive carcinogenic intermediates. However, the crosstalk between AhR and the microbiome in mediating carcinogenesis is poorly reviewed. This review aims to discuss the role of exposure to environmental pollutants and activation of AhR on microbiome-associated cancer progression and explore the underlying molecular mechanisms involved in cancer development.
Collapse
Affiliation(s)
- Menatallah Rayan
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Tahseen S Sayed
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
| | - Ola J Hussein
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
| | - Lubna Therachiyil
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Zaid H Maayah
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
| | | | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Hesham M Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar.
| |
Collapse
|
6
|
Hwang IC, Vasquez R, Song JH, Engstrand L, Valeriano VD, Kang DK. Alterations in the gut microbiome and its metabolites are associated with the immune response to mucosal immunization with Lactiplantibacillus plantarum-displaying recombinant SARS-CoV-2 spike epitopes in mice. Front Cell Infect Microbiol 2023; 13:1242681. [PMID: 37705931 PMCID: PMC10495993 DOI: 10.3389/fcimb.2023.1242681] [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: 06/19/2023] [Accepted: 08/11/2023] [Indexed: 09/15/2023] Open
Abstract
Lactic acid bacteria (LAB) expressing foreign antigens have great potential as mucosal vaccines. Our previous study reported that recombinant Lactiplantibacillus plantarum SK156 displaying SARS-CoV-2 spike S1 epitopes elicited humoral and cell-mediated immune responses in mice. Here, we further examined the effect of the LAB-based mucosal vaccine on gut microbiome composition and function, and gut microbiota-derived metabolites. Forty-nine (49) female BALB/c mice were orally administered L. plantarum SK156-displaying SARS-CoV-2 spike S1 epitopes thrice (at 14-day intervals). Mucosal immunization considerably altered the gut microbiome of mice by enriching the abundance of beneficial gut bacteria, such as Muribaculaceae, Mucispirillum, Ruminococcaceae, Alistipes, Roseburia, and Clostridia vadinBB60. Moreover, the predicted function of the gut microbiome showed increased metabolic pathways for amino acids, energy, carbohydrates, cofactors, and vitamins. The fecal concentration of short-chain fatty acids, especially butyrate, was also altered by mucosal immunization. Notably, alterations in gut microbiome composition, function, and butyrate levels were positively associated with the immune response to the vaccine. Our results suggest that the gut microbiome and its metabolites may have influenced the immunogenicity of the LAB-based SARS-CoV-2 vaccine.
Collapse
Affiliation(s)
- In-Chan Hwang
- Department of Animal Biotechnology, Dankook University, Cheonan, Republic of Korea
| | - Robie Vasquez
- Department of Animal Biotechnology, Dankook University, Cheonan, Republic of Korea
| | - Ji Hoon Song
- Department of Animal Biotechnology, Dankook University, Cheonan, Republic of Korea
| | - Lars Engstrand
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Valerie Diane Valeriano
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Dae-Kyung Kang
- Department of Animal Biotechnology, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
7
|
Guevara-Ramírez P, Cadena-Ullauri S, Paz-Cruz E, Tamayo-Trujillo R, Ruiz-Pozo VA, Zambrano AK. Role of the gut microbiota in hematologic cancer. Front Microbiol 2023; 14:1185787. [PMID: 37692399 PMCID: PMC10485363 DOI: 10.3389/fmicb.2023.1185787] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023] Open
Abstract
Hematologic neoplasms represent 6.5% of all cancers worldwide. They are characterized by the uncontrolled growth of hematopoietic and lymphoid cells and a decreased immune system efficacy. Pathological conditions in hematologic cancer could disrupt the balance of the gut microbiota, potentially promoting the proliferation of opportunistic pathogens. In this review, we highlight studies that analyzed and described the role of gut microbiota in different types of hematologic diseases. For instance, myeloma is often associated with Pseudomonas aeruginosa and Clostridium leptum, while in leukemias, Streptococcus is the most common genus, and Lachnospiraceae and Ruminococcaceae are less prevalent. Lymphoma exhibits a moderate reduction in microbiota diversity. Moreover, certain factors such as delivery mode, diet, and other environmental factors can alter the diversity of the microbiota, leading to dysbiosis. This dysbiosis may inhibit the immune response and increase susceptibility to cancer. A comprehensive analysis of microbiota-cancer interactions may be useful for disease management and provide valuable information on host-microbiota dynamics, as well as the possible use of microbiota as a distinguishable marker for cancer progression.
Collapse
|
8
|
Caleça T, Ribeiro P, Vitorino M, Menezes M, Sampaio-Alves M, Mendes AD, Vicente R, Negreiros I, Faria A, Costa DA. Breast Cancer Survivors and Healthy Women: Could Gut Microbiota Make a Difference?-"BiotaCancerSurvivors": A Case-Control Study. Cancers (Basel) 2023; 15:cancers15030594. [PMID: 36765550 PMCID: PMC9913170 DOI: 10.3390/cancers15030594] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
In this first analysis, samples from 23 BC survivors (group 1) and 291 healthy female controls (group 2) were characterised through the V3 and V4 regions that encode the "16S rRNA" gene of each bacteria. The samples were sequenced by next-generation sequencing (NGS), and the taxonomy was identified by resorting to Kraken2 and improved with Bracken, using a curated database called 'GutHealth_DB'. The α and β-diversity analyses were used to determine the richness and evenness of the gut microbiota. A non-parametric Mann-Whitney U test was applied to assess differential abundance between both groups. The Firmicutes/Bacteroidetes (F/B) ratio was calculated using a Kruskal-Wallis chi-squared test. The α-diversity was significantly higher in group 1 (p = 0.28 × 10-12 for the Chao index and p = 1.64 × 10-12 for the ACE index). The Shannon index, a marker of richness and evenness, was not statistically different between the two groups (p = 0.72). The microbiota composition was different between the two groups: a null hypothesis was rejected for PERMANOVA (p = 9.99 × 10-5) and Anosim (p = 0.04) and was not rejected for β-dispersion (p = 0.158), using Unifrac weighted distance. The relative abundance of 14 phyla, 29 classes, 25 orders, 64 families, 116 genera, and 74 species differed significantly between both groups. The F/B ratio was significantly lower in group 1 than in group 2, p < 0.001. Our study allowed us to observe significant taxonomic disparities in the two groups by testing the differences between BC survivors and healthy controls. Additional studies are needed to clarify the involved mechanisms and explore the relationship between microbiota and BC survivorship.
Collapse
Affiliation(s)
- Telma Caleça
- Medical Oncology Department, Hospital Professor Doutor Fernando Fonseca, 2720-276 Amadora, Portugal
- Correspondence: (T.C.); (D.A.C.)
| | - Pedro Ribeiro
- Laboratory Medicine Centre Germano de Sousa, 1600-513 Lisbon, Portugal
| | - Marina Vitorino
- Medical Oncology Department, Hospital Professor Doutor Fernando Fonseca, 2720-276 Amadora, Portugal
| | - Maria Menezes
- Medical Oncology Department, Hospital do Espírito Santo de Évora, 7000-811 Évora, Portugal
| | - Mafalda Sampaio-Alves
- PTSurg–Portuguese Surgical Research Collaborative, 1600 Lisbon, Portugal
- Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal
| | - Ana Duarte Mendes
- Medical Oncology Department, Hospital Professor Doutor Fernando Fonseca, 2720-276 Amadora, Portugal
| | - Rodrigo Vicente
- Medical Oncology Department, Hospital Professor Doutor Fernando Fonseca, 2720-276 Amadora, Portugal
| | - Ida Negreiros
- Hospital CUF, Breast Cancer Unit, 1998-018 Lisbon, Portugal
| | - Ana Faria
- Faculdade de Ciências Médicas, NOVA Medical School, 1169-056 Lisbon, Portugal
- Comprehensive Health Research Centre (CHRC), Faculdade de Ciências Médicas, NOVA Medical School, 1150-082 Lisbon, Portugal
- CINTESIS-Center for Health Technology Services Research, Faculdade de Ciências Médicas, NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Diogo Alpuim Costa
- Hospital CUF, Breast Cancer Unit, 1998-018 Lisbon, Portugal
- Faculdade de Ciências Médicas, NOVA Medical School, 1169-056 Lisbon, Portugal
- Medical Oncology Department, Hospital de Cascais Dr. José de Almeida, 2755-009 Cascais, Portugal
- Correspondence: (T.C.); (D.A.C.)
| |
Collapse
|
9
|
Molecular and Cellular Mechanisms of Propolis and Its Polyphenolic Compounds against Cancer. Int J Mol Sci 2022; 23:ijms231810479. [PMID: 36142391 PMCID: PMC9499605 DOI: 10.3390/ijms231810479] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 12/12/2022] Open
Abstract
In recent years, interest in natural products such as alternative sources of pharmaceuticals for numerous chronic diseases, including tumors, has been renewed. Propolis, a natural product collected by honeybees, and polyphenolic/flavonoid propolis-related components modulate all steps of the cancer progression process. Anticancer activity of propolis and its compounds relies on various mechanisms: cell-cycle arrest and attenuation of cancer cells proliferation, reduction in the number of cancer stem cells, induction of apoptosis, modulation of oncogene signaling pathways, inhibition of matrix metalloproteinases, prevention of metastasis, anti-angiogenesis, anti-inflammatory effects accompanied by the modulation of the tumor microenvironment (by modifying macrophage activation and polarization), epigenetic regulation, antiviral and bactericidal activities, modulation of gut microbiota, and attenuation of chemotherapy-induced deleterious side effects. Ingredients from propolis also "sensitize" cancer cells to chemotherapeutic agents, likely by blocking the activation of the transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). In this review, we summarize the current knowledge related to the the effects of flavonoids and other polyphenolic compounds from propolis on tumor growth and metastasizing ability, and discuss possible molecular and cellular mechanisms involved in the modulation of inflammatory pathways and cellular processes that affect survival, proliferation, invasion, angiogenesis, and metastasis of the tumor.
Collapse
|
10
|
Local Breast Microbiota: A "New" Player on the Block. Cancers (Basel) 2022; 14:cancers14153811. [PMID: 35954474 PMCID: PMC9367283 DOI: 10.3390/cancers14153811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Microbiota plays a fundamental role in the induction, training and function of the human immune system. The interactions between microbiota and immune cells have consequences in several settings, namely in carcinogenesis but also in anticancer activity. Immunotherapy, already widely used in the treatment of several solid cancers, modulates the action of the immune system, promoting antitumour effects. Recently, there has been a growing interest in studying the microbiota composition as a possible modulator of the tumour microenvironment and consequently of the response to certain therapies such as immunotherapy. Abstract The tumour microenvironment (TME) comprises a complex ecosystem of different cell types, including immune cells, cells of the vasculature and lymphatic system, cancer-associated fibroblasts, pericytes, and adipocytes. Cancer proliferation, invasion, metastasis, drug resistance and immune escape are all influenced by the dynamic interaction between cancer cells and TME. Microbes, such as bacteria, fungi, viruses, archaea and protists, found within tumour tissues, constitute the intratumour microbiota, which is tumour type-specific and distinct among patients with different clinical outcomes. Growing evidence reveals a significant relevance of local microbiota in the colon, liver, breast, lung, oral cavity and pancreas carcinogenesis. Moreover, there is a growing interest in the tumour immune microenvironment (TIME) pointed out in several cross-sectional studies on the correlation between microbiota and TME. It is now known that microorganisms have the capacity to change the density and function of anticancer and suppressive immune cells, enabling the promotion of an inflammatory environment. As immunotherapy (such as immune checkpoint inhibitors) is becoming a promising therapy using TIME as a therapeutic target, the analysis and comprehension of local microbiota and its modulating strategies can help improve cancer treatments.
Collapse
|