1
|
Pherez-Farah A, Boncompagni G, Chudnovskiy A, Pasqual G. The Bidirectional Interplay between T Cell-Based Immunotherapies and the Tumor Microenvironment. Cancer Immunol Res 2025; 13:463-475. [PMID: 39786986 PMCID: PMC7617322 DOI: 10.1158/2326-6066.cir-24-0857] [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: 09/02/2024] [Revised: 11/06/2024] [Accepted: 01/07/2025] [Indexed: 01/12/2025]
Abstract
T cell-based therapies, including tumor-infiltrating lymphocyte therapy, T-cell receptor-engineered T cells, and chimeric antigen receptor T cells, are powerful therapeutic approaches for cancer treatment. Whereas these therapies are primarily known for their direct cytotoxic effects on cancer cells, accumulating evidence indicates that they also influence the tumor microenvironment (TME) by altering the cytokine milieu and recruiting additional effector populations to help orchestrate the antitumor immune response. Conversely, the TME itself can modulate the behavior of these therapies within the host by either supporting or inhibiting their activity. In this review, we provide an overview of clinical and preclinical data on the bidirectional influences between T-cell therapies and the TME. Unraveling the interactions between T cell-based therapies and the TME is critical for a better understanding of their mechanisms of action, resistance, and toxicity, with the goal of optimizing efficacy and safety.
Collapse
Affiliation(s)
- Alfredo Pherez-Farah
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Gioia Boncompagni
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | | | - Giulia Pasqual
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Veneto Institute of Oncology IOV IRCCS, Padua, Italy
| |
Collapse
|
2
|
Hirao H, Honda M, Tomita M, Li L, Adawy A, Xue W, Hibi T. Intravital Imaging of Immune Responses in the Cancer Microenvironment. Cancer Med 2025; 14:e70899. [PMID: 40257446 PMCID: PMC12010765 DOI: 10.1002/cam4.70899] [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: 02/08/2025] [Revised: 03/18/2025] [Accepted: 04/09/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND To date, many types of immune cells have been identified, but their precise role in cancer immunity remains unclear. Understanding the immune responses involved in cancer and the cancer microenvironment is becoming increasingly important for elucidating disease mechanisms. In recent years, the application of intravital imaging in cancer research has provided new insights into the mechanisms of cancer-specific immune events, including innate and adaptive immunity. RESULTS In this review, we focus on the emerging role of intravital imaging in cancer research and describe how cancer and immune cells can be observed using intravital imaging in vivo. We also discuss new insights gained by this state-of-the-art technique. CONCLUSIONS Intravital imaging is a relatively new field of research that offers significant advantages, including the ability to directly capture cell-cell interactions, pathophysiology, and immune cell dynamics in the cancer microenvironment in vivo.
Collapse
Affiliation(s)
- Hiroki Hirao
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Masaki Honda
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Masahiro Tomita
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Lianbo Li
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Ahmad Adawy
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Weijie Xue
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Taizo Hibi
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| |
Collapse
|
3
|
de Moraes FCA, Souza MEC, Sano VKT, Moraes RA, Melo AC. Association of tumor-infiltrating lymphocytes with clinical outcomes in patients with triple-negative breast cancer receiving neoadjuvant chemotherapy: a systematic review and meta-analysis. Clin Transl Oncol 2025; 27:974-987. [PMID: 39154313 DOI: 10.1007/s12094-024-03661-8] [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: 06/29/2024] [Accepted: 07/30/2024] [Indexed: 08/19/2024]
Abstract
OBJECTIVE Triple-negative breast cancer (TNBC) presents a clinical challenge as an aggressive tumor, correlated with unfavorable prognosis. Tumor-infiltrating lymphocytes (TILs) have garnered interest as a potential prognostic biomarker. However, the disparity in outcomes between varying TILs rates remains inadequately explored. METHODS PubMed, Scopus, Web of Science, and Cochrane databases were searched for studies about the prognostic value of TILs in patients with TNBC receiving neoadjuvant chemotherapy. The hazard ratios (HRs) or odds ratios (ORs) were computed for binary endpoints, with 95% confidence intervals (CIs). RESULTS Twenty-nine studies were included, involving a population of six thousand one hundred sixty-one (80.41%) with TNBC. The cut-off TILs value ranged from 10 to 60%, with 50% being the most related value. Compared with the low-TIL expression group, the disease-free survival (DFS) (HR 0.71; 95% CI 0.61-0.82; p < 0.00001) and overall survival (OS) (HR 0.76; 95% CI 0.63-0.90; p = 0.002) rates showed significant improvement with higher TIL infiltrations. In the subgroup analyses of the lymphocyte subtypes CD4 + and CD8 + , there was statistical significance favoring higher TILs rates in both subtypes, each associated with improved DFS (HR 0.48; 95% CI 0.33-0.71; p = 0.0002) and OS (HR 0.53; 95% CI 0.36-0.78; p = 0.001), regardless of which cell subtype was predominantly infiltrated. The complete pathological response analysis showed better rates for the higher TIL group than the control for both the TIL (OR 1.29; 95% CI 1.13-1.48; p = 0.0003) and Ki-67 (OR 2.74; 95% CI 2.01-3.73; p < 0.00001) analyses. CONCLUSION Higher expressions of TILs in patients with TNBC were associated with improved significantly DFS, OS, and pCR outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Ana C Melo
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
4
|
Gakinya S, Nzioka AK, Mugo AG, Onyuma T, Ogutu J. Autophagy-related protein LC3β and its association with clinical-pathological characteristics, mismatch repair proteins and survival in colorectal carcinoma. Front Med (Lausanne) 2025; 12:1512127. [PMID: 40018347 PMCID: PMC11865083 DOI: 10.3389/fmed.2025.1512127] [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: 10/16/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
Introduction Autophagy is a metabolic process that serves to maintain cellular homeostasis as well as enable the cell to adapt to metabolic stress. In malignant cells, autophagy has been associated with drug resistance, metastasis and poor outcome. Colorectal carcinoma is a leading cause of cancer morbidity and mortality worldwide. The management and outcome are dependent on the tumor clinical and pathological characteristics. Autophagy is a potential therapeutic target as well as prognostic biomarker given its role in cancer pathogenesis. This study aimed at evaluating the autophagy status of colorectal carcinomas for tumors diagnosed at the Aga Khan University Hospital, Nairobi and establish its association with clinical-pathological characteristics including age, tumor location, tumor grade, tumor pathological stage, tumor nodal stage, tumor budding, tumor-infiltrating lymphocytes (TILs), Mismatch repair protein status (MMR), HER2 status and patient survival. Methods The study assessed the autophagy status of 114 colorectal carcinoma cases using immunohistochemistry for autophagy related protein LC3β. The clinical-pathological characteristics were determined by examining the medical records and evaluation of hematoxylin and eosin-stained slides. HER2 and MMR status were evaluated using immunohistochemistry. The treatment outcome was determined from the patient's records by checking for date of last visit or death. Results and discussion The mean age of patients in our study was 58years. There were more males 61.8% (n = 70) than females 38.6% (n = 44). Most of the patients had high pathological tumor stage of pT3 and pT4. Majority of the tumors showed intermediate tumor budding and weak tumor-infiltrating lymphocytes. The mismatch repair deficiency and HER2 overexpression were found in 14.9% (n = 17) and 2.6% (n = 3) of the cases respectively. LC3β was overexpressed in 36% (n = 41) of the cases and was significantly more common in females (p = 0.013). The LC3β status showed no significant association with age, tumor location, tumor grade, tumor stage, nodal stage, tumor budding, tumor-infiltrating lymphocytes, MMR status, HER2 status or patient survival. Future prospective studies are recommended to further explore the utility of autophagy as a prognostic and predictive biomarker.
Collapse
Affiliation(s)
- Samuel Gakinya
- Department of Pathology, Aga Khan University, Nairobi, Kenya
| | - Ancent K. Nzioka
- Department of Pathology, Kenyatta University Teaching, Referral and Research Hospital, Nairobi, Kenya
| | - Alex G. Mugo
- Department of Pathology, Aga Khan University, Nairobi, Kenya
| | - Timothy Onyuma
- Department of Pathology, Kenyatta National Hospital, Nairobi, Kenya
| | - James Ogutu
- Department of Microbiology, Kenyatta University, Nairobi, Kenya
| |
Collapse
|
5
|
Yang M, Lin W, Huang J, Mannucci A, Luo H. Novel immunotherapeutic approaches in gastric cancer. PRECISION CLINICAL MEDICINE 2024; 7:pbae020. [PMID: 39397869 PMCID: PMC11467695 DOI: 10.1093/pcmedi/pbae020] [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: 06/25/2024] [Revised: 09/08/2024] [Accepted: 09/08/2024] [Indexed: 10/15/2024] Open
Abstract
Gastric cancer is a malignant tumor that ranks third in cancer-related deaths worldwide. Early-stage gastric cancer can often be effectively managed through surgical resection. However, the majority of cases are diagnosed in advanced stages, where outcomes with conventional radiotherapy and chemotherapy remain unsatisfactory. Immunotherapy offers a novel approach to treating molecularly heterogeneous gastric cancer by modifying the immunosuppressive tumor microenvironment. Immune checkpoint inhibitors and adoptive cell therapy are regarded as promising modalities in cancer immunotherapy. Food and Drug Administration-approved programmed death-receptor inhibitors, such as pembrolizumab, in combination with chemotherapy, have significantly extended overall survival in gastric cancer patients and is recommended as a first-line treatment. Despite challenges in solid tumor applications, adoptive cell therapy has demonstrated efficacy against various targets in gastric cancer treatment. Among these approaches, chimeric antigen receptor-T cell therapy research is the most widely explored and chimeric antigen receptor-T cell therapy targeting claudin18.2 has shown acceptable safety and robust anti-tumor capabilities. However, these advancements primarily remain in preclinical stages and further investigation should be made to promote their clinical application. This review summarizes the latest research on immune checkpoint inhibitors and adoptive cell therapy and their limitations, as well as the role of nanoparticles in enhancing immunotherapy.
Collapse
Affiliation(s)
- Meng Yang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Wuhao Lin
- Department of Molecular Diagnostics, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jiaqian Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Alessandro Mannucci
- Gastroenterology and Gastrointestinal Emndoscopy Unit, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan 20132, Italy
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope; Monrovia, CA 91016, USA
| | - Huiyan Luo
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| |
Collapse
|
6
|
Islam S, Islam MM, Akhand MRN, Park BY, Akanda MR. Recent advancements in cGAS-STING activation, tumor immune evasion, and therapeutic implications. Med Oncol 2024; 41:291. [PMID: 39419913 DOI: 10.1007/s12032-024-02539-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
The cGAS-STING signaling pathway is indeed a pivotal component of the immune system and serve as a crucial link between innate and adaptive immune responses. STING is involved in the cellular response to pathogen invasion and DNA damage, and which has important consequences for host defense mechanisms and cancer regulation. Ongoing research aiming to modulate the cGAS-STING pathway for improved clinical outcomes in cancer and autoimmune diseases is underway. Indeed, the interaction between the cGAS-STING pathway and immune evasion mechanisms is a complex and critical aspect of cancer biology. Pathogens and various host factors can exploit this pathway to reduce the effectiveness of cancer therapies, particularly immunotherapies. Thus, immunotherapies or combination therapies may assist in overcoming the immune suppression and improving clinical outcomes. This review explores recent advancements in understanding the cGAS-STING signaling pathway, with particular emphasis on its activation mechanisms and role in tumor immune evasion. The dual role of the pathway in boosting immune responses while simultaneously enabling tumors to evade the immune system makes it a crucial target for innovative cancer treatment approaches.Please confirm if the author names are presented accurately and in the correct sequence (given name, middle name/initial, family name). Author 2 Given name: [Md Mazedul] Last name [Islam], Author 3 Given name: [Mst Rubaiat Nazneen] Last name [Akhand] and Author 5 Given name: [Md Rashedunnabi] Last name [Akanda]. Also, kindly confirm the details in the metadata are correct.AQ1: Here Author 4 given name: [Byung-Yong] Last name [Park] is missing. Metadata are correct.
Collapse
Affiliation(s)
- Saiful Islam
- Department of Physiology, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Md Mazedul Islam
- Faculty of Veterinary, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | | | - Byung-Yong Park
- Institute of Animal Transplantation, College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, South Korea
| | - Md Rashedunnabi Akanda
- Department of Pharmacology and Toxicology, Sylhet Agricultural University, Sylhet, 3100, Bangladesh.
| |
Collapse
|
7
|
Ibrahim E, Diab E, Hayek R, Hoyek K, Kourie H. Triple-Negative Breast Cancer: Tumor Immunogenicity and Beyond. Int J Breast Cancer 2024; 2024:2097920. [PMID: 39399414 PMCID: PMC11469932 DOI: 10.1155/2024/2097920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/19/2024] [Indexed: 10/15/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a breast malignancy with a poor prognosis and limited therapeutic options. Many studies show that TNBC exhibits heterogeneity across clinical, histopathological, and molecular levels. In this review, we discuss the immunogenic features of TNBC with a focus on immunotherapy and the current standard of care in the neoadjuvant, adjuvant, and metastatic setting. In addition, we address the ongoing research on immunotherapy, antibody-drug conjugates (ADCs), poly ADP-ribose polymerase (PARP) inhibitors, and future challenges in the treatment of this entity.
Collapse
Affiliation(s)
- Elio Ibrahim
- Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Ernest Diab
- Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
- Oncology Department, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Rony Hayek
- Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Karim Hoyek
- Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Hampig Kourie
- Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
- Oncology Department, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| |
Collapse
|
8
|
Pateriya A, Nema R, Mishra AK, Kumar A, Shrivastava A. Decreased Expression of CD247 and CD4 Immune Marker Predicts Poor Prognosis in Triple Negative Breast Cancer. Asian Pac J Cancer Prev 2024; 25:3187-3197. [PMID: 39342598 DOI: 10.31557/apjcp.2024.25.9.3187] [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: 04/26/2024] [Indexed: 10/01/2024] Open
Abstract
OBJECTIVE Triple negative breast cancer (TNBC) is an aggressive from of breast cancer and is associated with poor prognosis. Tumor microenvironment of breast cancer consists of a wide range of cell types, including tumor-infiltrating lymphocytes (TILs). Accumulating evidence indicate that TILs play a crucial role in cancer progression and resistance to standard chemotherapy. METHOD We used online computational tools to evaluate the prognostic significance of CD247 and CD4 in TNBC. RESULTS TNBC patients with lower expression of CD247 and CD4 have much shorter relapse- free survival and overall survival than the patients with higher expression of these genes. CD247 and CD4 expression show a strong positive correlation with tumor-infiltrating dendritic cells, B-cells, CD4+, CD8+, and neutrophils. CONCLUSION We've concluded that low levels of CD247 and CD4 may stop immune cells from entering the area around the tumor, which stops cancer cells from being killed and gives the patient a bad outlook. These findings suggest that CD247 and CD4 may be useful biomarkers or as a target to understand the progression of TNBC. Our findings also suggest that CD247 and CD4 targeted therapeutics should be explored in detail, and could be a potentially used as atreatment strategy for TNBC.
Collapse
Affiliation(s)
- Ankit Pateriya
- Centre for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, India
| | - Rajeev Nema
- Department of Biosciences Manipal University Jaipur, Dehmi Kalan, Jaipur-Ajmer Expressway, Jaipur, Rajasthan, India
| | - Anand Kumar Mishra
- Department of Endocrine Surgery, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal, India
| | - Ashutosh Shrivastava
- Centre for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, India
| |
Collapse
|
9
|
Huang AL, He YZ, Yang Y, Pang M, Zheng GP, Wang HL. Exploring the potential of the TCR repertoire as a tumor biomarker (Review). Oncol Lett 2024; 28:413. [PMID: 38988449 PMCID: PMC11234811 DOI: 10.3892/ol.2024.14546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/14/2024] [Indexed: 07/12/2024] Open
Abstract
T cells play an important role in adaptive immunity. Mature T cells specifically recognize antigens on major histocompatibility complex molecules through T-cell receptors (TCRs). As the TCR repertoire is highly diverse, its analysis is vital in the assessment of T cells. Advances in sequencing technology have provided convenient methods for further investigation of the TCR repertoire. In the present review, the TCR structure and the mechanisms by which TCRs function in tumor recognition are described. In addition, the potential value of the TCR repertoire in tumor diagnosis is reviewed. Furthermore, the role of the TCR repertoire in tumor immunotherapy is introduced, and the relationships between the TCR repertoire and the effects of different tumor immunotherapies are discussed. Based on the reviewed literature, it may be concluded that the TCR repertoire has the potential to serve as a biomarker for tumor prognosis. However, a wider range of cancer types and more diverse subjects require evaluation in future research to establish the TCR repertoire as a biomarker of tumor immunity.
Collapse
Affiliation(s)
- An-Li Huang
- Institute of Cancer Biology, Basic Medical Sciences Center, School of Basic Medicine, Shanxi Medical University, Jinzhong, Shanxi 030600, P.R. China
- The First Clinical Medical College, Shanxi Medical University, Jinzhong, Shanxi 030600, P.R. China
| | - Yan-Zhao He
- Institute of Cancer Biology, Basic Medical Sciences Center, School of Basic Medicine, Shanxi Medical University, Jinzhong, Shanxi 030600, P.R. China
| | - Yong Yang
- Institute of Cancer Biology, Basic Medical Sciences Center, School of Basic Medicine, Shanxi Medical University, Jinzhong, Shanxi 030600, P.R. China
| | - Min Pang
- NHC Key Laboratory of Pneumoconiosis, Shanxi Province Key Laboratory of Respiratory Disease, Department of Pulmonary and Critical Care Medicine, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Guo-Ping Zheng
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales 2145, Australia
| | - Hai-Long Wang
- Institute of Cancer Biology, Basic Medical Sciences Center, School of Basic Medicine, Shanxi Medical University, Jinzhong, Shanxi 030600, P.R. China
| |
Collapse
|
10
|
Kan H, Chen Y. Revealing endogenous conditions for Peto's paradox via an ordinary differential equation model. J Math Biol 2024; 89:27. [PMID: 38970664 PMCID: PMC11227477 DOI: 10.1007/s00285-024-02123-7] [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: 07/27/2023] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/08/2024]
Abstract
Cancer, a disease intimately linked to cellular mutations, is commonly believed to exhibit a positive association with the cell count and lifespan of a species. Despite this assumption, the observed uniformity in cancer rates across species, referred to as the Peto's paradox, presents a conundrum. Recognizing that tumour progression is not solely dependent on cancer cells but involves intricate interactions among various cell types, this study employed a Lotka-Volterra (LV) ordinary differential equation model to analyze the evolution of cancerous cells and the cancer incidence in an immune environment. As a result, this study uncovered the sufficient conditions underlying the absence of correlation in Peto's paradox and provide insights into the reasons for the equitable distribution of cancer incidence across diverse species by applying nondimensionalization and drawing an analogy between the characteristic time interval for the variation of cell populations in the ODE model and that of cell cycles of a species.
Collapse
Affiliation(s)
- Haichun Kan
- SCS Laboratory, Department of Human and Engineered Environmental Studies, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Yu Chen
- SCS Laboratory, Department of Human and Engineered Environmental Studies, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan.
| |
Collapse
|
11
|
Cazzaniga M, Cardinali M, Di Pierro F, Zonzini GB, Palazzi CM, Gregoretti A, Zerbinati N, Guasti L, Matera MR, Cavecchia I, Bertuccioli A. The Role of Short-Chain Fatty Acids, Particularly Butyrate, in Oncological Immunotherapy with Checkpoint Inhibitors: The Effectiveness of Complementary Treatment with Clostridium butyricum 588. Microorganisms 2024; 12:1235. [PMID: 38930617 PMCID: PMC11206605 DOI: 10.3390/microorganisms12061235] [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: 04/30/2024] [Revised: 06/06/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
The discovery of immune checkpoints (CTLA-4, PD-1, and PD-L1) and their impact on the prognosis of oncological diseases have paved the way for the development of revolutionary oncological treatments. These treatments do not combat tumors with drugs "against" cancer cells but rather support and enhance the ability of the immune system to respond directly to tumor growth by attacking the cancer cells with lymphocytes. It has now been widely demonstrated that the presence of an adequate immune response, essentially represented by the number of TILs (tumor-infiltrating lymphocytes) present in the tumor mass decisively influences the response to treatments and the prognosis of the disease. Therefore, immunotherapy is based on and cannot be carried out without the ability to increase the presence of lymphocytic cells at the tumor site, thereby limiting and nullifying certain tumor evasion mechanisms, particularly those expressed by the activity (under positive physiological conditions) of checkpoints that restrain the response against transformed cells. Immunotherapy has been in the experimental phase for decades, and its excellent results have made it a cornerstone of treatments for many oncological pathologies, especially when combined with chemotherapy and radiotherapy. Despite these successes, a significant number of patients (approximately 50%) do not respond to treatment or develop resistance early on. The microbiota, its composition, and our ability to modulate it can have a positive impact on oncological treatments, reducing side effects and increasing sensitivity and effectiveness. Numerous studies published in high-ranking journals confirm that a certain microbial balance, particularly the presence of bacteria capable of producing short-chain fatty acids (SCFAs), especially butyrate, is essential not only for reducing the side effects of chemoradiotherapy treatments but also for a better response to immune treatments and, therefore, a better prognosis. This opens up the possibility that favorable modulation of the microbiota could become an essential complementary treatment to standard oncological therapies. This brief review aims to highlight the key aspects of using precision probiotics, such as Clostridium butyricum, that produce butyrate to improve the response to immune checkpoint treatments and, thus, the prognosis of oncological diseases.
Collapse
Affiliation(s)
- Massimiliano Cazzaniga
- Scientific & Research Department, Velleja Research, 20125 Milan, Italy; (M.C.); (F.D.P.)
- Microbiota International Clinical Society, 10123 Torino, Italy; (A.G.); (M.R.M.); (I.C.); (A.B.)
| | - Marco Cardinali
- Department of Internal Medicine, Infermi Hospital, AUSL Romagna, 47921 Rimini, Italy;
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61122 Urbino, Italy;
| | - Francesco Di Pierro
- Scientific & Research Department, Velleja Research, 20125 Milan, Italy; (M.C.); (F.D.P.)
- Microbiota International Clinical Society, 10123 Torino, Italy; (A.G.); (M.R.M.); (I.C.); (A.B.)
- Department of Medicine and Surgery, University of Insurbia, 21100 Varese, Italy; (N.Z.); (L.G.)
| | - Giordano Bruno Zonzini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61122 Urbino, Italy;
| | - Chiara Maria Palazzi
- Microbiota International Clinical Society, 10123 Torino, Italy; (A.G.); (M.R.M.); (I.C.); (A.B.)
| | - Aurora Gregoretti
- Microbiota International Clinical Society, 10123 Torino, Italy; (A.G.); (M.R.M.); (I.C.); (A.B.)
| | - Nicola Zerbinati
- Department of Medicine and Surgery, University of Insurbia, 21100 Varese, Italy; (N.Z.); (L.G.)
| | - Luigina Guasti
- Department of Medicine and Surgery, University of Insurbia, 21100 Varese, Italy; (N.Z.); (L.G.)
| | - Maria Rosaria Matera
- Microbiota International Clinical Society, 10123 Torino, Italy; (A.G.); (M.R.M.); (I.C.); (A.B.)
| | - Ilaria Cavecchia
- Microbiota International Clinical Society, 10123 Torino, Italy; (A.G.); (M.R.M.); (I.C.); (A.B.)
| | - Alexander Bertuccioli
- Microbiota International Clinical Society, 10123 Torino, Italy; (A.G.); (M.R.M.); (I.C.); (A.B.)
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61122 Urbino, Italy;
| |
Collapse
|
12
|
Kapoor M, Sehrawat A, Karthik J, Sundriyal D. Tumor infiltrating lymphocytes in gastric cancer: Unraveling complex interactions for precision medicine. World J Clin Oncol 2024; 15:478-481. [PMID: 38689625 PMCID: PMC11056866 DOI: 10.5306/wjco.v15.i4.478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/07/2024] [Accepted: 03/19/2024] [Indexed: 04/22/2024] Open
Abstract
This editorial will focus on tumor immunity and the factors that alter the tumor immune micro-environment. The role of tumor infiltrating lymphocytes (TILs) will also be discussed in detail, including the types, mechanism of action, and role. Gastric cancer (GC) often presents in the advanced stage and has various factors predicting the outcomes. The interplay of these factors and their correlation with the TILs is discussed. A literature review revealed high intra-tumoral TILs associated with higher grade, HER2-, and Helicobacter pylori negativity. Moreover, stromal (ST) TILs correlated with lower grade and lesser recurrence risk in GC. High TILs in ST and invasive border also correlated with mismatch repair deficiency status. Further characterization of the CD3+, CD8+, and other cells is also warranted. In the future, this complex correlation of cancer cells with the immune system can be explored for therapeutic avenues.
Collapse
Affiliation(s)
- Mayank Kapoor
- Department of Medical Oncology Haematology, All India Institute of Medical Sciences Rishikesh, Rishikesh 249203, India
| | - Amit Sehrawat
- Department of Medical Oncology Haematology, All India Institute of Medical Sciences Rishikesh, Rishikesh 249203, India
| | - Jayalingappa Karthik
- Department of Medical Oncology Haematology, All India Institute of Medical Sciences Rishikesh, Rishikesh 249203, India
| | - Deepak Sundriyal
- Department of Medical Oncology Haematology, All India Institute of Medical Sciences Rishikesh, Rishikesh 249203, India
| |
Collapse
|
13
|
Yang X, Wu C. Systemic immune inflammation index and gastric cancer prognosis: A systematic review and meta‑analysis. Exp Ther Med 2024; 27:122. [PMID: 38410191 PMCID: PMC10895464 DOI: 10.3892/etm.2024.12410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/01/2023] [Indexed: 02/28/2024] Open
Abstract
The present study aimed to pool the available data on the associations between the systemic immune inflammation index (SII) and overall survival (OS) or recurrence-free survival (RFS) in patients with gastric cancer (GC). A systematic search was conducted in the PubMed, EMBASE and Scopus databases for observational studies, and a random effects model was used to conduct the statistical analysis. Pooled effect sizes were reported as hazard ratios (HRs) with corresponding 95% confidence intervals (CI). Data from 30 studies (24 conducted in China) with follow-ups ranging between 15.5 and 65.6 months were analyzed. Patients with GC and high SII levels had poor OS (HR, 1.53; 95% CI, 1.34-1.75) and recurrence free survival (HR, 1.41; 95% CI, 1.17-1.70). These increased risks were present irrespective of the treatment strategy (surgical or non-surgical management), the sample size (<500 and ≥500) and the cut-off used to define high and low SII (<600 and ≥600 x109 cells/l). The results of this meta-analysis suggest that high pretreatment SII levels were associated with poor OS and RFS in patients with GC.
Collapse
Affiliation(s)
- Xiaomao Yang
- Department of Gastrointestinal Hernia, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, Zhejiang 313000, P.R. China
| | - Chen Wu
- Department of Gastrointestinal Hernia, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, Zhejiang 313000, P.R. China
| |
Collapse
|
14
|
Sun L, Zhou H, Wu C, Peng Y. Molecular markers that predict response to combined radiotherapy and immunotherapy in patients with lung adenocarcinoma: a bioinformatics analysis. Transl Cancer Res 2023; 12:2646-2659. [PMID: 37969379 PMCID: PMC10643968 DOI: 10.21037/tcr-23-968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/19/2023] [Indexed: 11/17/2023]
Abstract
Background Immunotherapy has had a high success rate in treating lung adenocarcinoma (LUAD) for several decades. However, many patients do not benefit from immunotherapy alone. Recent studies revealed that a combination of immunotherapy and radiotherapy (RT) stimulates a good systemic immune response to LUAD. However, clinical and experimental evidence suggest that RT may give rise to primary immunodeficiency, facilitating tumor immunity escape. Little is known about the molecular mechanisms whereby RT and stereotactic body radiotherapy (SBRT) influence tumor immunogenicity and the effectiveness of immunotherapy in patients with LUAD. Methods We investigated molecular markers that predict response to combination of immunotherapy and SBRT in the treatment of LUAD using bioinformatics. Results SBRT significantly upregulated the expression of PTPRC, LILRB2, TLR8, CCR5, and PLEK and significantly downregulated the expression of CXCL13, CD19, and LTA. Among these genes, the expression of PTPRC, TLR8, and CCR5 was associated with responsiveness to immunotherapy after SBRT. However, only TLR8 and CCR5 expression were associated with an improved prognosis. Further analysis revealed that TLR8 and CCR5 expression increased responsiveness to immunotherapy by promoting M0 macrophage and memory B cell infiltration of LUAD tissues. Conclusions In patients with LUAD, TLR8 and CCR5 expression are potential markers of a favorable response to combined immunotherapy and RT.
Collapse
Affiliation(s)
- Lu Sun
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haiting Zhou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Wu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Yi Peng
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Zhang Y, He S, Yu L, Shi C, Zhang Y, Tang S. Prognostic significance of HLA-G in patients with colorectal cancer: a meta-analysis and bioinformatics analysis. BMC Cancer 2023; 23:1024. [PMID: 37875821 PMCID: PMC10594707 DOI: 10.1186/s12885-023-11522-w] [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: 06/14/2023] [Accepted: 10/13/2023] [Indexed: 10/26/2023] Open
Abstract
PURPOSE Human leukocyte antigen-G (HLA-G) has been reported to be aberrantly expressed in colorectal cancer (CRC); however, its prognostic value remains controversial. Hence, our meta-analysis aims to assess the prognostic value of HLA-G in CRC patients based on published literature and The Cancer Genome Atlas (TCGA) datasets. METHODS A systematic search was conducted on relevant studies retrieved from four electronic databases including PubMed, Embase, Web of Science and Cochrane Library. Hazard ratios (HRs) with 95% confidence intervals (CIs) were recorded to be applied as effective values. Fixed-effects models or random-effects models were applied on the basis of the value of heterogeneity (I 2). Publication bias was analyzed by Begg's and Egger's tests. In addition, the results were validated by using TCGA datasets. RESULTS Thirteen studies comprising 3896 patients were incorporated into this meta-analysis. The pooled results showed that HLA-G expression was significantly associated with poor overall survival (OS) in both the univariate analysis (HR = 1.44, 95% CI: 1.14-1.83, P = 0.002) and the multivariate analysis (HR = 1.55, 95% CI: 1.23-1.95, P < 0.001). Nevertheless, the expression of HLA-G is not related to age, sex, tumor type, tumor differentiation, TNM stage, or distant metastasis but lymph node metastasis. Notably, the prognosis of colorectal cancer was not consistent with the analysis result from TCGA data. CONCLUSION HLA-G expression was significantly related to poor OS in CRC according to the results of our meta-analysis. However, we found that the prognostic significance was inconsistent with our results according to the TCGA data in CRC. Hence, more research is still needed to further illustrate the prognostic role of HLA-G in CRC.
Collapse
Affiliation(s)
- Yingying Zhang
- Department of clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China.
| | - Siying He
- Department of clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Lisha Yu
- Department of clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Chao Shi
- Department of clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Yanyue Zhang
- Department of clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Shiyue Tang
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
16
|
Zeiny SMH, Ali SHM. Immunohistochemical study of the expressed cluster differentiation markers proteins type 20 and 56 in breast tissues from a group of Iraqi patients with breast cancers. Asian Pac J Cancer Prev 2023; 24:3621-3628. [PMID: 37898871 PMCID: PMC10770690 DOI: 10.31557/apjcp.2023.24.10.3621] [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: 07/12/2023] [Accepted: 10/24/2023] [Indexed: 10/30/2023] Open
Abstract
BACKGROUND Tumor-infiltrating lymphocytes (TIL) are important immunological components in response to cancers. Patients with higher numbers of TIL in breast cancerous tissues, comprising T- cytotoxic and T - helper cells along with B- and rare natural killer (NK) cells, have more favorable clinical outcomes. OBJECTIVE To analyze the rate of the expressed surface biomarker proteins of CD20-B cells and CD56- NK cells on the infiltrative lymphocytic subpopulations in a group of breast tumorous tissues (invasive and benign) from female patients in Iraq and explore the relations to the grade of the invasive breast cancerous tissues. PATIENTS AND METHODS One hundred and 75 archived breast tissues were enrolled in this retrospective research: 100 archived breast from female patients with invasive breast cancers (BC) [20 well differentiated BC tissues; 48 moderately differentiated BC and 32 poorly differentiated BC tissues]; 50 tissue biopsies from female patients with benign breast tumors and 25 apparently normal individuals with healthy breast tissues (included as the control group for this study). Immunohistochemistry was achieved for the detection of the expressed surface biomarker proteins related to B cell CD20 and NK cell CD56 present on the infiltrative lymphocytic subpopulations in breast tissues by using specific primary antibodies for these proteins via utilizing an immune-enzymatic antigen detection system. RESULTS The detection of IHC reactions for the expressed B cell CD20 - cell surface ( CD) biomarker proteins were observed in 53 out of 100 (53.0%) BC tissues, and in 24 out of 50 (48.0%) benign breast tumorous tissues, while CD20- positive cell surface markers was detected in apparently healthy breast tissues of the control group in a percentage of 32.0% (8 out of 25 tissues). Statistical significant differences (P<0.05) between both groups of malignant and benign breast tumors and the control group were found. However, between breast malignant and benign tumor groups, no significant difference was found ( p >0.05). Detection of CD56- IHC reactions revealed in 14% (14 out of 100 BC tissues), in 16% (8 out of 50 benign breast tissues) and none of control breast tissues revealed CD56- IHC reactions. Among all the enrolled groups, no significant differences (P>0.05) were detected. CONCLUSIONS The observed significant rates that showed highly significant differences between both studied groups of breast malignant and benign tumor in comparison to the control group indicate that the CD20- positive infiltrative B cell- lymphocytic subpopulations might contributed in the defense against these subsets of benign and malignant breast tumors. However, the observed rates of NK cell CD56 present on the lymphocytic subpopulations infiltrating the examined malignant and benign breast tumorous tissues seeming to play irrelevant roles in the defense against these studied breast tumor groups.
Collapse
Affiliation(s)
- Sarmad M H Zeiny
- Department of Microbiology, College of Medicine, University of Baghdad, Iraq.
| | - Saad Hasan Mohammed Ali
- Clinical Communicable Diseases Research unit, College of Medicine, University of Baghdad, Iraq.
| |
Collapse
|
17
|
Yaping W, Zhe W, Zhuling C, Ruolei L, Pengyu F, Lili G, Cheng J, Bo Z, Liuyin L, Guangdong H, Yaoling W, Niuniu H, Rui L. The soldiers needed to be awakened: Tumor-infiltrating immune cells. Front Genet 2022; 13:988703. [PMID: 36246629 PMCID: PMC9558824 DOI: 10.3389/fgene.2022.988703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022] Open
Abstract
In the tumor microenvironment, tumor-infiltrating immune cells (TIICs) are a key component. Different types of TIICs play distinct roles. CD8+ T cells and natural killer (NK) cells could secrete soluble factors to hinder tumor cell growth, whereas regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) release inhibitory factors to promote tumor growth and progression. In the meantime, a growing body of evidence illustrates that the balance between pro- and anti-tumor responses of TIICs is associated with the prognosis in the tumor microenvironment. Therefore, in order to boost anti-tumor response and improve the clinical outcome of tumor patients, a variety of anti-tumor strategies for targeting TIICs based on their respective functions have been developed and obtained good treatment benefits, including mainly immune checkpoint blockade (ICB), adoptive cell therapies (ACT), chimeric antigen receptor (CAR) T cells, and various monoclonal antibodies. In recent years, the tumor-specific features of immune cells are further investigated by various methods, such as using single-cell RNA sequencing (scRNA-seq), and the results indicate that these cells have diverse phenotypes in different types of tumors and emerge inconsistent therapeutic responses. Hence, we concluded the recent advances in tumor-infiltrating immune cells, including functions, prognostic values, and various immunotherapy strategies for each immune cell in different tumors.
Collapse
Affiliation(s)
- Wang Yaping
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Wang Zhe
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Chu Zhuling
- Department of General Surgery, Eastern Theater Air Force Hospital of PLA, Nanjing, China
| | - Li Ruolei
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Fan Pengyu
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Guo Lili
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Ji Cheng
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhang Bo
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Liu Liuyin
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Hou Guangdong
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Wang Yaoling
- Department of Geriatrics, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hou Niuniu
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Department of General Surgery, Eastern Theater Air Force Hospital of PLA, Nanjing, China
- *Correspondence: Hou Niuniu, ; Ling Rui,
| | - Ling Rui
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Hou Niuniu, ; Ling Rui,
| |
Collapse
|