1
|
Liu L, He Y, Du H, Tang M, Wang T, Tan J, Zha L, Yang L, Ashrafizadeh M, Tian Y, Zhou H. Biological profile of breast cancer brain metastasis. Acta Neuropathol Commun 2025; 13:78. [PMID: 40253355 PMCID: PMC12008903 DOI: 10.1186/s40478-025-01983-4] [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: 07/29/2024] [Accepted: 03/08/2025] [Indexed: 04/21/2025] Open
Abstract
Breast cancer is one of the leading causes of death worldwide. The aggressive behaviour of breast tumor results from their metastasis. Notably, the brain tissue is one of the common regions of metastasis, thereby reducing the overall survival of patients. Moreover, the metastatic tumors demonstrate poor response or resistance to therapies. In addition, breast cancer brain metastasis provides the poor prognosis of patients. Therefore, it is of importance to understand the mechanisms in breast cancer brain metastasis. Both cell lines and animal models have been developed for the evaluation of breast cancer brain metastasis. Moreover, different tumor microenvironment components and other factors such as lymphocytes and astrocytes can affect brain metastasis. The breast cancer cells can disrupt the blood-brain barrier (BBB) during their metastasis into brain, developing blood-tumor barrier to enhance carcinogenesis. The breast cancer brain metastasis can be increased by the dysregulation of chemokines, STAT3, Wnt, Notch and PI3K/Akt. On the other hand, the effective therapeutics have been developed for the brain metastasis such as introduction of nanoparticles. Moreover, the disruption of BBB by ultrasound can increase the entrance of bioactive compounds to the brain tissue. In order to improve specificity and selectivity, the nanoparticles for the delivery of therapeutics and crossing over BBB have been developed to suppress breast cancer brain metastasis.
Collapse
Affiliation(s)
- Li Liu
- Department of Oncology, Suining Central Hospital, Suning, 629000, China
| | - Yuan He
- Department of Oncology, Yunyang County People's Hospital, Chongqing, 404500, China
| | - Hongyu Du
- Department of General Medicine, The Seventh People's Hospital of Chongqing, The Central Hospital Affiliated to Chongging University of Technology, Chongqing, 400054, China
| | - Min Tang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401120, China
| | - Tingting Wang
- Department of Gynecology and Obstetrics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jieren Tan
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, PR China
| | - Lisha Zha
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, PR China
| | - Li Yang
- Department of Nephrology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou, Guangdong Province, 510515, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250000, China.
| | - Yu Tian
- School of Public Health, Benedictine University, No.5700 College Road, Lisle, IL, 60532, USA.
- Research Center, the Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, Guangdong, China.
| | - Hui Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, No. 19 Nonglinxia Road, Guangzhou, 510080, China.
| |
Collapse
|
2
|
Mannan A, Mohan M, Singh TG. Revenge unraveling the fortress: Exploring anticancer drug resistance mechanisms in BC for enhanced therapeutic strategies. Crit Rev Oncol Hematol 2025; 210:104707. [PMID: 40122355 DOI: 10.1016/j.critrevonc.2025.104707] [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: 01/22/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025] Open
Abstract
Breast cancer (BC) is the most prevalent form of cancer in women worldwide and the main cause of cancer-related fatalities in females. BC can be classified into various types based on where cancer has begun to grow or spread, specific characteristics that influence how cancer behaves, and treatment choices. BC is multifaceted, and due to its diverse nature, the mechanisms involved are complex and have not yet been understood. Overexpression and expression of various factors involved in the functioning of mechanisms lead to abnormal changes, providing an environment supporting cancer cell growth. Understanding BC risk factors and early diagnosis through screening techniques like mammography and diagnostic techniques such as imaging and biopsies has advanced significantly. A wide range of treatment options, including surgery, radiation, chemotherapy, targeted treatments, and hormonal therapies, are now available. Daily advancements are being made in the clinical treatment of BC. Still, BC drug resistance cases remain highly prevalent and are currently one of the biggest problems faced by medical science. To increase response rates and possibly lengthen survival, there is a critical requirement for novel medicines with minimal sensitivity to overcome drug resistance. This review classifies different mechanisms that are involved in the development of BC and workable pharmacological targets and explains how they relate to the development of BC drug resistance. By concentrating on the mechanisms covered in this review, we can have a deep understanding of different mechanisms and learn innovative ways to develop novel therapeutics for the disease to combat medication resistance.
Collapse
Affiliation(s)
- Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
3
|
Yilmaz A, Ari Yuka S. The role of ceRNAs in breast cancer microenvironmental regulation and therapeutic implications. J Mol Med (Berl) 2025; 103:33-49. [PMID: 39641797 DOI: 10.1007/s00109-024-02503-y] [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: 05/06/2024] [Revised: 11/09/2024] [Accepted: 11/17/2024] [Indexed: 12/07/2024]
Abstract
The tumor microenvironment, which is the tailored physiological milieu of heterogeneous cancer cell populations surrounded by stromal and immune cells as well as extracellular matrix components, is a leading modulator of critical cancer hallmarks and one of the most significant prognostic indicators in breast cancer. In the last few decades, with the discovery of the interactions of ncRNAs with diverse cellular molecules, considerable emphasis has been devoted to understanding their direct and indirect roles in specific functions in breast cancer. Collectively, all of these have revealed that the competitive action of protein-coding RNAs and ncRNAs such as circRNAs and lncRNAs, which have a shared affinity for miRNAs, play a vital role in the molecular regulation of breast cancer. This phenomenon, termed as competing endogenous RNAs (ceRNAs), facilitates modeling the microenvironment through intercellular shuttles. Microenvironment ceRNA interactions have emerged as a frontier in the deep understanding of the complex mechanisms of breast cancer. In this review, we first discuss cellular ceRNAs in four key biological processes critical for microenvironmental regulation in breast cancer tissues: hypoxia, angiogenesis, immune regulations, and ECM remodeling. Further, we draw a complete portrait of microenvironment regulation by cell-to-cell cross-talk of shuttled ceRNAs and offer a framework of potential applications and challenges in overcoming the aggressive phenotype of the breast cancer microenvironment.
Collapse
Affiliation(s)
- Alper Yilmaz
- Department of Molecular Biology and Genetics, Yildiz Technical University, Istanbul, 34220, Turkey
| | - Selcen Ari Yuka
- Department of Genetics and Bioengineering, Alanya Alaaddin Keykubat University, Antalya, 07425, Turkey.
- Health Biotechnology Joint Research and Application Center of Excellence, Yildiz Technical University, Istanbul, 34220, Turkey.
| |
Collapse
|
4
|
Naseem N, Kushwaha P, Haider F. Leveraging nanostructured lipid carriers to enhance targeted delivery and efficacy in breast cancer therapy: a comprehensive review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:449-468. [PMID: 39196394 DOI: 10.1007/s00210-024-03408-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
Cancer, characterized by uncontrolled cell growth and proliferation, continues to be a major global health concern. Breast cancer, the most commonly diagnosed cancer among women, remains a leading cause of cancer-related deaths worldwide. Conventional treatment modalities such as surgery, radiation, and chemotherapy have made significant strides in improving patient outcomes. However, these approaches often face challenges such as limited efficacy, systemic toxicity, and multidrug resistance. Nanotechnology has emerged as a promising avenue for revolutionizing cancer therapy, offering targeted drug delivery, enhanced efficacy, and reduced side effects. Among the various nanocarrier systems, nanostructured lipid carriers (NLCs) have gained considerable attention for their unique advantages. Comprising a blend of solid and liquid lipids, NLCs offer improved drug loading capacity, enhanced stability, sustained release, and biocompatibility. This manuscript provides a comprehensive overview of the role of NLCs in breast cancer management, covering their formulation, methods of preparation, advantages, and disadvantages. Additionally, several studies are presented to illustrate the efficacy of NLCs in delivering anticancer drugs to breast tumors. These studies demonstrate the ability of NLCs to enhance drug cytotoxicity, improve tumor suppression, and minimize systemic toxicity. This manuscript aims to contribute to the existing literature by consolidating current knowledge and providing insights into the future directions of NLC-based therapeutics in breast cancer management.
Collapse
Affiliation(s)
- Nazish Naseem
- Faculty of Pharmacy, Integral University, Dasauli-Kursi Road, Lucknow, India
| | - Poonam Kushwaha
- Faculty of Pharmacy, Integral University, Dasauli-Kursi Road, Lucknow, India.
| | - Faheem Haider
- Faculty of Pharmacy, Integral University, Dasauli-Kursi Road, Lucknow, India
| |
Collapse
|
5
|
Liu X, Hyun Kim J, Li X, Liu R. Application of mesenchymal stem cells exosomes as nanovesicles delivery system in the treatment of breast cancer. Int J Pharm 2024; 666:124732. [PMID: 39304093 DOI: 10.1016/j.ijpharm.2024.124732] [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: 05/28/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
As people's living standards continue to improve and human life span expectancy increases, the incidence and mortality rates of breast cancer are continuously rising. Early detection of breast cancer and targeted therapy for different breast cancer subtypes can significantly reduce the mortality rate and alleviate the suffering of patients. Exosomes are extracellular vesicles secreted by various cells in the body. They participate in physiological and pathological responses by releasing active substances and play an important role in regulating intercellular communication. In recent years, research on exosomes has gradually expanded, and their special membrane structure and targetable characteristics are being increasingly applied in various clinical studies. Mesenchymal stem cells (MSCs)-derived exosomes play an important role in regulating the progression of breast cancer. In this review, we summarize the current treatment methods for breast cancer, the connection between MSCs, exosomes, and breast cancer, as well as the application of exosomes derived from MSCs from different sources in cancer treatment. We highlight how the rational design of modified MSCs-derived exosomes (MSCs-Exos) delivery systems can overcome the uncertainties of stem cell therapy and overcome the clinical translation challenges of nanomaterials. This work aims to promote future research on the application of MSCs-Exos in breast cancer treatment.
Collapse
Affiliation(s)
- Xiaofan Liu
- Department of Biotechnology, College of Engineering, The University of Suwon, Hwaseong 18323, Republic of Korea; Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - June Hyun Kim
- Department of Biotechnology, College of Engineering, The University of Suwon, Hwaseong 18323, Republic of Korea
| | - Xuemei Li
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China.
| | - Rui Liu
- Department of Biotechnology, College of Engineering, The University of Suwon, Hwaseong 18323, Republic of Korea.
| |
Collapse
|
6
|
Arciero I, Buonvino S, Palumbo V, Scimeca M, Melino S. A 3D-Printable Cell Array for In Vitro Breast Cancer Modeling. Int J Mol Sci 2024; 25:13068. [PMID: 39684779 DOI: 10.3390/ijms252313068] [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: 11/14/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Breast cancer is the most common cancer and the second leading cause of cancer-related death in women. In advanced stages of the disease, breast cancer can spread and metastasize to the bone, contributing to malignant progression. The roles of tissue stiffness and remodeling of the tumor microenvironment are relevant in influencing cancer progression and invasiveness, but they are still poorly understood. In this study, we aimed to investigate the effect of bone tissue stiffness on breast cancer cell behavior, using 3D cell-biomaterial systems to model the in vivo conditions. For this purpose, we developed a 3D-printable cell array, which is a tunable and reproducible platform on small scale, where each compartment could mimic the physiological cancer environment with a shape and rigidity close to bone tissue. In this system, we observed that in the highly metastatic breast cancer line MDA-MB-231, embedded in PEG-silk fibroin (PSF) hydrogel spheres in the array's cavities, increasing stiffness promotes trans-differentiation into osteoblast-like cells and the production of breast microcalcifications. Moreover, we also tested this 3D model as a platform to evaluate the cell response to the therapy, in particular, investigating the drug sensitivity of the cancer cells to chemotherapeutics, observing a decrease in drug resistance over time in the array.
Collapse
Affiliation(s)
- Ilaria Arciero
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", Via Della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Silvia Buonvino
- Departmental Faculty of Medicine, UniCamillus-Saint Camillus International University of Health Sciences, Via di Sant'Alessandro 8, 00131 Rome, Italy
| | - Valeria Palumbo
- Department of Experimental Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Manuel Scimeca
- Department of Experimental Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Sonia Melino
- Department of Experimental Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
7
|
Mussa A, Ismail NH, Hamid M, Al-Hatamleh MAI, Bragoli A, Hajissa K, Mokhtar NF, Mohamud R, Uskoković V, Hassan R. Understanding the role of TNFR2 signaling in the tumor microenvironment of breast cancer. J Exp Clin Cancer Res 2024; 43:312. [PMID: 39609700 PMCID: PMC11603874 DOI: 10.1186/s13046-024-03218-1] [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: 08/21/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed malignancy among women. It is characterized by a high level of heterogeneity that emerges from the interaction of several cellular and soluble components in the tumor microenvironment (TME), such as cytokines, tumor cells and tumor-associated immune cells. Tumor necrosis factor (TNF) receptor 2 (TNFR2) appears to play a significant role in microenvironmental regulation, tumor progression, immune evasion, drug resistance, and metastasis of many types of cancer, including BC. However, the significance of TNFR2 in BC biology is not fully understood. This review provides an overview of TNFR2 biology, detailing its activation and its interactions with important signaling pathways in the TME (e.g., NF-κB, MAPK, and PI3K/Akt pathways). We discuss potential therapeutic strategies targeting TNFR2, with the aim of enhancing the antitumor immune response to BC. This review provides insights into role of TNFR2 as a major immune checkpoint for the future treatment of patients with BC.
Collapse
Affiliation(s)
- Ali Mussa
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
- Department of Biology, Faculty of Education, Omdurman Islamic University, P.O. Box 382, Omdurman, Sudan
| | - Nor Hayati Ismail
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
| | - Mahasin Hamid
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Hunan Province, Changsha, 410013, China
- Department of Zoology, Faculty of Sciences and Information Technology, University of Nyala, Nyala, 63311, Sudan
| | - Mohammad A I Al-Hatamleh
- Division of Hematology and Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Anthony Bragoli
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Khalid Hajissa
- Department of Zoology, Faculty of Science and Technology, Omdurman Islamic University, P.O. Box 382, Omdurman, Sudan
| | - Noor Fatmawati Mokhtar
- Institute for Research in Molecular Medicine (iNFORMM), Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia.
| | - Vuk Uskoković
- TardigradeNano LLC, Irvine, CA, 92604, USA
- Division of Natural Sciences, Fullerton College, Fullerton, CA, 92832, USA
| | - Rosline Hassan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia.
| |
Collapse
|
8
|
Narvaez-Rojas AR, Linhares S, Sedighim S, Klingbeil KD, Milikowski C, Elgart G, Jaimes N, Feun L, Lutzky J, De la Cruz Ku G, Avisar E, Möller MG. Is primary breast melanoma a true pathological entity? The argument against it. Heliyon 2024; 10:e37224. [PMID: 39309840 PMCID: PMC11414497 DOI: 10.1016/j.heliyon.2024.e37224] [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: 09/25/2023] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Background Previous studies have reported cases of primary melanoma of the breast parenchyma (PMBP), but the pathogenesis of this disease remains poorly understood. We review the presentation and outcomes of reported cases and provide detailed pathological analysis of four additional cases. Furthermore, we discuss potential theories regarding the pathogenesis of this clinical presentation. Results We identified 29 published studies (n = 95 patients) and report four new cases (n = 99). Ninety-one (92 %) patients were female, with a median age of 50 years. Previous skin melanomas were reported by 56 % of patients, with the trunk being the most common location (32.7 %) followed by the upper extremities (20 %). The most common tumor location reported (n = 73) was the right (49 %) upper outer quadrant (56 %). The median time from skin melanoma diagnosis to the presence of a breast mass was 65 months (1-192). Nodal status at presentation was reported in n = 67 (68 %) patients. Of these, positive nodal metastases were seen in 40.3 %, while distant metastatic disease at presentation was reported in 30 % of patients. Surgery was performed in 66 %, being partial mastectomy (PM) the most common procedure in 82 %. Adjuvant therapy was described in 38 patients. The reported (n = 12) median survival was 11.5 (1-70) months. Conclusion Melanomas identified in the breast parenchyma are likely the result of nodal or hematogenous spread from previously known or unknown melanomas, and should not be considered as PMBP. Management should be multidisciplinary, including surgical excision aimed at obtaining negative margins with lymphadenectomy of clinically positive nodes and neoadjuvant/adjuvant immunotherapy.
Collapse
Affiliation(s)
- Alexis R. Narvaez-Rojas
- Department of Radiation Oncology, Maimonides Cancer Center, Brooklyn, NY, USA
- International Coalition on Surgical Research, Universidad Nacional Autónoma de Nicaragua, UNAN, Managua, Nicaragua
| | | | - Shaina Sedighim
- Department of Surgery, University of California, Irvine, Orange, CA, USA
| | - Kyle Daniel Klingbeil
- Department of Surgery, University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Clara Milikowski
- University of Miami Miller School of Medicine, Miami, FL, USA
- Departments of Pathology, Division of Hematology Oncology at University of Miami Miller School of Medicine, Miami, FL, USA
| | - George Elgart
- University of Miami Miller School of Medicine, Miami, FL, USA
- Departments of Dermatology, Division of Hematology Oncology at University of Miami Miller School of Medicine, Miami, FL, USA
| | - Natalia Jaimes
- University of Miami Miller School of Medicine, Miami, FL, USA
- Departments of Dermatology, Division of Hematology Oncology at University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lynn Feun
- University of Miami Miller School of Medicine, Miami, FL, USA
- Departments of Medicine, Division of Hematology Oncology at University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jose Lutzky
- University of Miami Miller School of Medicine, Miami, FL, USA
- Departments of Medicine, Division of Hematology Oncology at University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Eli Avisar
- University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, Division of Surgical Oncology at University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mecker G. Möller
- University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, Division of Surgical Oncology at University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
9
|
Song X, Hao C, Li Y, Li Y, Dong H, Wei Q, Wei M, Li H, Zhao L. Chiral inorganic nanomaterials in the tumor microenvironment: A new chapter in cancer therapy. Pharmacol Res 2024; 208:107386. [PMID: 39216840 DOI: 10.1016/j.phrs.2024.107386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/20/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Chirality plays a crucial function in the regulation of normal physiological processes and is widespread in organisms. Chirality can be imparted to nanomaterials, whether they are natural or manmade, through the process of asymmetric assembly and/or grafting of molecular chiral groups or linkers. Chiral inorganic nanomaterials possess unique physical and chemical features that set them apart from regular nanomaterials. They also have the ability to interact with cells and tissues in a specific manner, making them useful in various biomedical applications, particularly in the treatment of tumors. Despite the growing amount of research on chiral inorganic nanomaterials in the tumor microenvironment (TME) and their promising potential applications, there is a lack of literature that comprehensively summarizes the intricate interactions between chiral inorganic nanomaterials and TME. In this review, we introduce the fundamental concept, classification, synthesis methods, and physicochemical features of chiral inorganic nanomaterials. Next, we briefly outline the components of TME, such as T cells, macrophages, dendritic cells, and weak acids, and then discuss the anti-tumor effects of several chiral inorganic nanoparticles targeting these components and their potential for possible application during cancer therapy. Finally, the present challenges faced by chiral inorganic nanomaterials in cancer treatment and their future areas of investigation are disclosed.
Collapse
Affiliation(s)
- Xueyi Song
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Chenjing Hao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Yao Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Yunong Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Hongzhi Dong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Qian Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Heran Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China.
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
10
|
Shukla P, Bera AK, Ghosh A, Kiranmai G, Pati F. Assessment and process optimization of high throughput biofabrication of immunocompetent breast cancer model for drug screening applications. Biofabrication 2024; 16:035030. [PMID: 38876096 DOI: 10.1088/1758-5090/ad586b] [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: 03/02/2024] [Accepted: 06/14/2024] [Indexed: 06/16/2024]
Abstract
Recent advancements in 3D cancer modeling have significantly enhanced our ability to delve into the intricacies of carcinogenesis. Despite the pharmaceutical industry's substantial investment of both capital and time in the drug screening and development pipeline, a concerning trend persists: drug candidates screened on conventional cancer models exhibit a dismal success rate in clinical trials. One pivotal factor contributing to this discrepancy is the absence of drug testing on pathophysiologically biomimetic 3D cancer models during pre-clinical stages. Unfortunately, current manual methods of 3D cancer modeling, such as spheroids and organoids, suffer from limitations in reproducibility and scalability. In our study, we have meticulously developed 3D bioprinted breast cancer model utilizing decellularized adipose tissue-based hydrogel obtained via a detergent-free decellularization method. Our innovative printing techniques allows for rapid, high-throughput fabrication of 3D cancer models in a 96-well plate format, demonstrating unmatched scalability and reproducibility. Moreover, we have conducted extensive validation, showcasing the efficacy of our platform through drug screening assays involving two potent anti-cancer drugs, 5-Fluorouracil and PRIMA-1Met. Notably, our platform facilitates effortless imaging and gene expression analysis, streamlining the evaluation process. In a bid to enhance the relevance of our cancer model, we have introduced a heterogeneous cell population into the DAT-based bioink. Through meticulous optimization and characterization, we have successfully developed a biomimetic immunocompetent breast cancer model, complete with microenvironmental cues and diverse cell populations. This breakthrough paves the way for rapid multiplex drug screening and the development of personalized cancer models, marking a paradigm shift in cancer research and pharmaceutical development.
Collapse
Affiliation(s)
- Priyanshu Shukla
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| | - Ashis Kumar Bera
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| | - Amit Ghosh
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| | - Gaddam Kiranmai
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| | - Falguni Pati
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| |
Collapse
|
11
|
Otterlei Fjørtoft M, Huse K, Rye IH. The Tumor Immune Microenvironment in Breast Cancer Progression. Acta Oncol 2024; 63:359-367. [PMID: 38779867 PMCID: PMC11332517 DOI: 10.2340/1651-226x.2024.33008] [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: 12/01/2023] [Accepted: 02/12/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND The tumor microenvironment significantly influences breast cancer development, progression, and metastasis. Various immune cell populations, including T cells, B cells, NK cells, and myeloid cells exhibit diverse functions in different breast cancer subtypes, contributing to both anti-tumor and pro-tumor activities. PURPOSE This review provides an overview of the predominant immune cell populations in breast cancer subtypes, elucidating their suppressive and prognostic effects. We aim to outline the role of the immune microenvironment from normal breast tissue to invasive cancer and distant metastasis. METHODS A comprehensive literature review was conducted to analyze the involvement of immune cells throughout breast cancer progression. RESULTS In breast cancer, tumors exhibit increased immune cell infiltration compared to normal tissue. Variations exist across subtypes, with higher levels observed in triple-negative and HER2+ tumors are linked to better survival. In contrast, ER+ tumors display lower immune infiltration, associated with poorer outcomes. Furthermore, metastatic sites commonly exhibit a more immunosuppressive microenvironment. CONCLUSION Understanding the complex interaction between tumor and immune cells during breast cancer progression is essential for future research and the development of immune-based strategies. This comprehensive understanding may pave the way for more effective treatment approaches and improved patients outcomes.
Collapse
Affiliation(s)
- Marit Otterlei Fjørtoft
- Department of Cancer Genetics, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, Radium Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kanutte Huse
- Department of Cancer Immunology, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, Radium Hospital, Oslo, Norway
| | - Inga Hansine Rye
- Department of Cancer Genetics, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, Radium Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
12
|
Baylie T, Kasaw M, Getinet M, Getie G, Jemal M, Nigatu A, Ahmed H, Bogale M. The role of miRNAs as biomarkers in breast cancer. Front Oncol 2024; 14:1374821. [PMID: 38812786 PMCID: PMC11133523 DOI: 10.3389/fonc.2024.1374821] [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: 01/22/2024] [Accepted: 04/08/2024] [Indexed: 05/31/2024] Open
Abstract
Breast cancer (BC) is the second most common cause of deaths reported in women worldwide, and therefore there is a need to identify BC patients at an early stage as timely diagnosis would help in effective management and appropriate monitoring of patients. This will allow for proper patient monitoring and effective care. However, the absence of a particular biomarker for BC early diagnosis and surveillance makes it difficult to accomplish these objectives. miRNAs have been identified as master regulators of the molecular pathways that are emphasized in various tumors and that lead to the advancement of malignancies. Small, non-coding RNA molecules known as miRNAs target particular mRNAs to control the expression of genes. miRNAs dysregulation has been linked to the start and development of a number of human malignancies, including BC, since there is compelling evidence that miRNAs can function as tumor suppressor genes or oncogenes. The current level of knowledge on the role of miRNAs in BC diagnosis, prognosis, and treatment is presented in this review. miRNAs can regulate the tumorigenesis of BC through targeting PI3K pathway and can be used as prognostic or diagnostic biomarkers for BC therapy. Some miRNAs, like miR-9, miR-10b, and miR-17-5p, are becoming known as biomarkers of BC for diagnosis, prognosis, and therapeutic outcome prediction. Other miRNAs, like miR-30c, miR-187, and miR-339-5p, play significant roles in the regulation of hallmark functions of BC, including invasion, metastasis, proliferation, resting death, apoptosis, and genomic instability. Other miRNAs, such as miR-155 and miR-210, are circulating in bodily fluids and are therefore of interest as novel, conveniently accessible, reasonably priced, non-invasive methods for the customized care of patients with BC.
Collapse
Affiliation(s)
- Temesgen Baylie
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Mulugeta Kasaw
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, Bahir Dar University, Bahir Dar, Ethiopia
| | - Mamaru Getinet
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Gedefaw Getie
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Mohammed Jemal
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Amare Nigatu
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, Woldia University, Woldia, Ethiopia
| | - Hassen Ahmed
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, Woldia University, Woldia, Ethiopia
| | - Mihiret Bogale
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, Wollo University, Wollo, Ethiopia
| |
Collapse
|
13
|
Ferreira Almeida C, Correia-da-Silva G, Teixeira N, Amaral C. Influence of tumor microenvironment on the different breast cancer subtypes and applied therapies. Biochem Pharmacol 2024; 223:116178. [PMID: 38561089 DOI: 10.1016/j.bcp.2024.116178] [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: 12/28/2023] [Revised: 03/15/2024] [Accepted: 03/28/2024] [Indexed: 04/04/2024]
Abstract
Despite the significant improvements made in breast cancer therapy during the last decades, this disease still has increasing incidence and mortality rates. Different targets involved in general processes, like cell proliferation and survival, have become alternative therapeutic options for this disease, with some of them already used in clinic, like the CDK4/6 inhibitors for luminal A tumors treatment. Nevertheless, there is a demand for novel therapeutic strategies focused not only on tumor cells, but also on their microenvironment. Tumor microenvironment (TME) is a very complex and dynamic system that, more than surrounding and supporting tumor cells, actively participates in tumor development and progression. During the last decades, it has become clear that the cellular and acellular components of TME differ between the various breast cancer subtypes and shape the differences regarding their severity and prognosis. The pivotal role of the TME in controlling tumor growth and influencing responses to therapy represents a potential source for novel targets and therapeutic strategies. In this review, we present a description of the multiple therapeutic options used for different breast cancer subtypes, as well as the influence that the TME may exert on the development of the disease and on the response to the distinct therapies, which in some cases may explain their failure by the occurrence of relapses and resistance. Furthermore, the ongoing studies focused on the use of TME components for developing potential cancer treatments are described.
Collapse
Affiliation(s)
- Cristina Ferreira Almeida
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal.
| | - Natércia Teixeira
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Cristina Amaral
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal.
| |
Collapse
|
14
|
Sang B, Fan Y, Wang X, Dong L, Gong Y, Zou W, Zhao G, He J. The prognostic value of absolute lymphocyte count and neutrophil-to-lymphocyte ratio for patients with metastatic breast cancer: a systematic review and meta-analysis. Front Oncol 2024; 14:1360975. [PMID: 38515567 PMCID: PMC10955091 DOI: 10.3389/fonc.2024.1360975] [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: 12/24/2023] [Accepted: 02/16/2024] [Indexed: 03/23/2024] Open
Abstract
Background Neutrophil-to-lymphocyte ratio (NLR) is considered a potential prognostic marker in early breast cancer. However, the prognosis of absolute lymphocyte count (ALC) and NLR in metastatic breast cancer (MBC) has been reported in a few studies, and conclusions are still conflicting. This present manuscript aims to provide further solid evidence regarding the prognostic values of ALC and NLR in MBC patients. Method Eligible studies that reported the associations between ALC or NLR and MBC were included by searching relative electronic databases. Overall survival (OS) and progression-free survival (PFS) were used as outcome measures. The hazard ratio (HR) values and 95% confidence interval (CI) of the outcome measures were collected as effect sizes, and further analysis and discussion were conducted according to the pooled HR, subgroup analysis, publication bias, and interstudy heterogeneity. Results Twenty-nine studies comprising 3,973 patients with MBC were included. According to our findings, lower ALC was significantly associated with poorer prognosis of OS (HR = 0.57, 95% CI 0.48 to 0.68) and PFS (HR = 0.68, 95% CI 0.58 to 0.79), and greater NLR was associated with poorer OS (HR = 1.50, 95% CI 1.35 to 1.67) and PFS (HR = 1.82, 95% CI 1.42 to 2.35). Furthermore, the prognostic values of ALC and NLR in MBC were also observed in the subgroup analyses regarding cutoff values and ethnicities. Conclusion Low ALC and elevated NLR were observed to be significantly associated with adverse OS and PFS in MBC, indicating that ALC and NLR may act as potential prognostic biomarkers of MBC patients. Meanwhile, our results will also provide some novel evidence and research clues for the selection and development of clinical treatment strategies for MBC patients. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42021224114.
Collapse
Affiliation(s)
- Bulin Sang
- Clinical Pharmacology Research Center, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| | - Yuxin Fan
- Clinical Pharmacology Research Center, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| | - Xurao Wang
- College of Pharmacy, Dali University, Dali, China
| | - Lixian Dong
- Clinical Pharmacology Research Center, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| | - Yuanyuan Gong
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Wenhong Zou
- Clinical Pharmacology Research Center, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| | - Guanhua Zhao
- Clinical Pharmacology Research Center, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| | - Jianchang He
- Clinical Pharmacology Research Center, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| |
Collapse
|
15
|
Kim HS, Cho JY. Exosome proteomes reveal glycolysis-related enzyme enrichment in primary canine mammary gland tumor compared to metastases. Proteome Sci 2024; 22:4. [PMID: 38419074 PMCID: PMC10900604 DOI: 10.1186/s12953-023-00226-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/20/2023] [Indexed: 03/02/2024] Open
Abstract
OBJECTIVE Numerous evidence has highlighted the differences between primary tumors and metastases. Nonetheless, the differences in exosomal proteins derived from primary tumor and metastases remain elusive. Here, we aimed to identify differentially expressed exosomal proteins from primary canine mammary gland tumor and metastases to understand how they shape their own tumor microenvironment. METHODS We clearly distinguished primary canine mammary gland tumors (CHMp) from metastases (CHMm) and profiled the proteins within their secreted exosomes using LC-MS/MS. Moreover, the abundance of glycolysis enzymes (GPI, LDHA) in CHMp exosome was verified with Western blotting, To broaden the scope, we extended to human colorectal cancer-derived exosomes (SW480 vs. SW620) for comparison. RESULTS We identified significant differences in 87 and 65 proteins derived from CHMp and CHMm, respectively. Notably, glycolysis enzymes (GPI, LDHA, LDHB, TPI1, and ALDOA) showed specific enrichment in exosomes from the primary tumor. CONCLUSION We observed significant differences in the cellular proteome between primary tumors and metastases, and intriguingly, we identified a parallel heterogeneity the protein composition of exosomes. Specifically, we reported that glycolysis enzymes were significantly enriched in CHMp exosomes compared to CHMm exosomes. We further demonstrated that this quantitative difference in glycolysis enzymes persisted across primary and metastases, extending to human colorectal cancer-derived exosomes (SW480 vs. SW620). Our findings of the specific enrichment of glycolysis enzymes in primary tumor-derived exosomes contribute to a better understanding of tumor microenvironment modulation and heterogeneity between primary tumors and metastases.
Collapse
Affiliation(s)
- Hui-Su Kim
- Department of Biochemistry, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Gwanak-ro1, Gwanak-Gu, Seoul, 08826, Republic of Korea
- Comparative Medicine Disease Research Center (CDRC), Science Research Center (SRC), Seoul National University, Seoul, 08826, Republic of Korea
| | - Je-Yoel Cho
- Department of Biochemistry, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Gwanak-ro1, Gwanak-Gu, Seoul, 08826, Republic of Korea.
- Comparative Medicine Disease Research Center (CDRC), Science Research Center (SRC), Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
16
|
Neagu AN, Whitham D, Bruno P, Arshad A, Seymour L, Morrissiey H, Hukovic AI, Darie CC. Onco-Breastomics: An Eco-Evo-Devo Holistic Approach. Int J Mol Sci 2024; 25:1628. [PMID: 38338903 PMCID: PMC10855488 DOI: 10.3390/ijms25031628] [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: 12/20/2023] [Revised: 01/21/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Known as a diverse collection of neoplastic diseases, breast cancer (BC) can be hyperbolically characterized as a dynamic pseudo-organ, a living organism able to build a complex, open, hierarchically organized, self-sustainable, and self-renewable tumor system, a population, a species, a local community, a biocenosis, or an evolving dynamical ecosystem (i.e., immune or metabolic ecosystem) that emphasizes both developmental continuity and spatio-temporal change. Moreover, a cancer cell community, also known as an oncobiota, has been described as non-sexually reproducing species, as well as a migratory or invasive species that expresses intelligent behavior, or an endangered or parasite species that fights to survive, to optimize its features inside the host's ecosystem, or that is able to exploit or to disrupt its host circadian cycle for improving the own proliferation and spreading. BC tumorigenesis has also been compared with the early embryo and placenta development that may suggest new strategies for research and therapy. Furthermore, BC has also been characterized as an environmental disease or as an ecological disorder. Many mechanisms of cancer progression have been explained by principles of ecology, developmental biology, and evolutionary paradigms. Many authors have discussed ecological, developmental, and evolutionary strategies for more successful anti-cancer therapies, or for understanding the ecological, developmental, and evolutionary bases of BC exploitable vulnerabilities. Herein, we used the integrated framework of three well known ecological theories: the Bronfenbrenner's theory of human development, the Vannote's River Continuum Concept (RCC), and the Ecological Evolutionary Developmental Biology (Eco-Evo-Devo) theory, to explain and understand several eco-evo-devo-based principles that govern BC progression. Multi-omics fields, taken together as onco-breastomics, offer better opportunities to integrate, analyze, and interpret large amounts of complex heterogeneous data, such as various and big-omics data obtained by multiple investigative modalities, for understanding the eco-evo-devo-based principles that drive BC progression and treatment. These integrative eco-evo-devo theories can help clinicians better diagnose and treat BC, for example, by using non-invasive biomarkers in liquid-biopsies that have emerged from integrated omics-based data that accurately reflect the biomolecular landscape of the primary tumor in order to avoid mutilating preventive surgery, like bilateral mastectomy. From the perspective of preventive, personalized, and participatory medicine, these hypotheses may help patients to think about this disease as a process governed by natural rules, to understand the possible causes of the disease, and to gain control on their own health.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Carol I bvd. 20A, 700505 Iasi, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Pathea Bruno
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Aneeta Arshad
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Logan Seymour
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Hailey Morrissiey
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Angiolina I. Hukovic
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Costel C. Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| |
Collapse
|
17
|
Yang Y, Hou X, Kong S, Zha Z, Huang M, Li C, Li N, Ge F, Chen W. Intraoperative radiotherapy in breast cancer: Alterations to the tumor microenvironment and subsequent biological outcomes (Review). Mol Med Rep 2023; 28:231. [PMID: 37888611 PMCID: PMC10636769 DOI: 10.3892/mmr.2023.13118] [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: 07/13/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023] Open
Abstract
Intraoperative radiotherapy (IORT) is a precise, single high‑dose irradiation directly targeting the tumor bed during surgery. In comparison with traditional external beam RT, it minimizes damage to other normal tissues, ensures an adequate dose to the tumor bed and results in improved cosmetic outcomes and quality of life. Furthermore, IORT offers a shorter treatment duration, lower economic costs and therapeutic efficacy comparable with traditional RT. However, its relatively higher local recurrence rate limits its further clinical applications. Identifying effective radiosensitizing drugs and rational RT protocols will improve its advantages. Furthermore, IORT may not only damage DNA to directly kill breast tumor cells but also alter the tumor microenvironment (TME) to exert a sustained antitumor effect. Specific doses of IORT may exert anti‑angiogenic effects, and consequently antitumor effects, by impacting post‑radiation peripheral blood levels of vascular endothelial growth factor and delta‑like 4. IORT may also modify the postoperative wound fluid composition to continuously inhibit tumor growth, e.g. by reducing components such as microRNA (miR)‑21, miR‑221, miR‑115, oncostatin M, TNF‑β, IL‑6 and IL‑8, and by elevating levels of components such as miR‑223, to inhibit the ability of postoperative wound fluid to induce proliferation, invasion and migration of residual cancer cells. IORT can also modify cancer cell glucose metabolism to inhibit the proliferation of residual tumor cells. In addition, IORT can induce a bystander effect, eliminating the postoperative wound fluid‑induced epithelial‑mesenchymal transition and tumor stem cell phenotype. Insights gained at the molecular level may provide new directions for identifying novel therapeutic targets and approaches. A more comprehensive understanding of the effects of IORT on the breast cancer (BC) TME may further its clinical application. Hence, the present article reviews the primary effects of IORT on BC and its impact on the TME, aiming to offer fresh research perspectives for relevant professionals.
Collapse
Affiliation(s)
- Yang Yang
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Hospital, Kunming, Yunnan 650118, P.R. China
| | - Xiaochen Hou
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Hospital, Kunming, Yunnan 650118, P.R. China
| | - Shujia Kong
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Hospital, Kunming, Yunnan 650118, P.R. China
| | - Zhuocen Zha
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Hospital, Kunming, Yunnan 650118, P.R. China
| | - Mingqing Huang
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Hospital, Kunming, Yunnan 650118, P.R. China
| | - Chenxi Li
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Hospital, Kunming, Yunnan 650118, P.R. China
| | - Na Li
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Hospital, Kunming, Yunnan 650118, P.R. China
| | - Fei Ge
- Department of Breast Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Wenlin Chen
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Hospital, Kunming, Yunnan 650118, P.R. China
| |
Collapse
|
18
|
Chatterjee P, Banerjee S. Unveiling the mechanistic role of the Aryl hydrocarbon receptor in environmentally induced Breast cancer. Biochem Pharmacol 2023; 218:115866. [PMID: 37863327 DOI: 10.1016/j.bcp.2023.115866] [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: 07/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 10/22/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a crucial cytosolic evolutionary conserved ligand-activated transcription factor and a pleiotropic signal transducer. The biosensor activity of the AhR is attributed to the promiscuity of its ligand-binding domain. Evidence suggests exposure to environmental toxins such as polycyclic aromatic hydrocarbons, polychlorinated biphenyls and halogenated aromatic hydrocarbons activates the AhR signaling pathway. The constitutive activation of the receptor signaling system leads to multiple health adversities and enhances the risk of several cancers, including breast cancer (BC). This review evaluates several mechanisms that integrate the tumor-inducing property of such environmental contaminants with the AhR pathway assisting in BC tumorigenesis, progress and metastasis. Intriguingly, immune evasion is identified as a prominent hallmark in BC. Several emerging pieces of evidence have identified AhR as a potent immunosuppressive effector in several cancers. Through AhR signaling pathways, some tumors can avoid immune detection. Thus the relevance of AhR in the immunomodulation of breast tumors and its putative mode of action in the breast tumor microenvironment are discussed in this review. Additionally, the work also explores BC stemness and its associated inflammation in response to several environmental cues. The review elucidates the context-dependent ambiguous behavior of AhR either as an oncogene or a tumor suppressor with respect to its ligand. Conclusively, this holistic piece of literature attempts to potentiate AhR as a promising pharmacological target in BC and updates on the therapeutic manipulation of its various exogenous and endogenous ligands.
Collapse
Affiliation(s)
- Prarthana Chatterjee
- School of BioSciences and Technology, Vellore Institute of Technology, Vellore- 632014, Tamil Nadu, India
| | - Satarupa Banerjee
- School of BioSciences and Technology, Vellore Institute of Technology, Vellore- 632014, Tamil Nadu, India.
| |
Collapse
|
19
|
Guo S, Huang J, Li G, Chen W, Li Z, Lei J. The role of extracellular vesicles in circulating tumor cell-mediated distant metastasis. Mol Cancer 2023; 22:193. [PMID: 38037077 PMCID: PMC10688140 DOI: 10.1186/s12943-023-01909-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023] Open
Abstract
Current research has demonstrated that extracellular vesicles (EVs) and circulating tumor cells (CTCs) are very closely related in the process of distant tumor metastasis. Primary tumors are shed and released into the bloodstream to form CTCs that are referred to as seeds to colonize and grow in soil-like distant target organs, while EVs of tumor and nontumor origin act as fertilizers in the process of tumor metastasis. There is no previous text that provides a comprehensive review of the role of EVs on CTCs during tumor metastasis. In this paper, we reviewed the mechanisms of EVs on CTCs during tumor metastasis, including the ability of EVs to enhance the shedding of CTCs, protect CTCs in circulation and determine the direction of CTC metastasis, thus affecting the distant metastasis of tumors.
Collapse
Affiliation(s)
- Siyin Guo
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jing Huang
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Genpeng Li
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wenjie Chen
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhihui Li
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jianyong Lei
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
20
|
Liu Z, Lin J, Li B, Zhou Y, Li C, Cui Y, Tian F, Tang R, Wang X. Manganese-mineralized cancer cells as immunogenic cancer vaccines for tumor immunotherapy. J Mater Chem B 2023; 11:10923-10928. [PMID: 37934507 DOI: 10.1039/d3tb01538f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
The strategy of using tumor cells to construct whole-cell cancer vaccines has received widespread attention. However, the limited immunogenicity of inactivated tumor cells and the challenge of overcoming immune suppression in solid tumors have hindered the application of whole-cell-based cancer immune therapy. Inspired by the regulatory effects of MnO2 and spatiotemporal control capability of material layers in cell surface engineering, we developed a manganese (Mn)-mineralized tumor cell, B16F10@MnO2, by inactivating B16F10 melanoma cells with KMnO4 to generate manganese-mineralized tumor cells. The cell-based composite was formed by combining amorphous MnO2 with the membrane structure of cells based on the redox reaction between KMnO4 and tumor cells. The MnO2 layer induced a stronger phagocytosis of ovalbumin (OVA)-expressing tumor cells by antigen presenting cells than formaldehyde-fixed cells did, resulting in specific antigen-presentation in vitro and in vivo and subsequent immune responses. Intratumoral therapy with B16F10@MnO2 inhibited B16F10 tumor growth. Moreover, the infiltration of CD8+ T cells within B16F10 solid tumors and the proportion of central memory T cells both increased in B16F10@MnO2 treated tumor-bearing mice, indicating enhanced adaptive immunity. This study provides a convenient and effective method to improve whole-cell-based anti-tumor therapy.
Collapse
Affiliation(s)
- Zhenyu Liu
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310058, China.
| | - Jiake Lin
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310058, China.
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Benke Li
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Yuemin Zhou
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310058, China.
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Chen Li
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Yihao Cui
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Fengchao Tian
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Xiaoyu Wang
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
21
|
Jiang Z, Ju YJ, Ali A, Chung PED, Wang DY, Liu JC, Li H, Vorobieva I, Mwewa E, Ghanbari-Azarnier R, Shrestha M, Ben-David Y, Zacksenhaus E. Thinking (Metastasis) outside the (Primary Tumor) Box. Cancers (Basel) 2023; 15:5315. [PMID: 38001575 PMCID: PMC10670606 DOI: 10.3390/cancers15225315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
The metastasis of tumor cells into vital organs is a major cause of death from diverse types of malignancies [...].
Collapse
Affiliation(s)
- Zhe Jiang
- Toronto General Research Institute—University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada (Y.-J.J.); (A.A.); (D.-Y.W.); (H.L.); (E.M.); (R.G.-A.); (M.S.)
| | - Young-Jun Ju
- Toronto General Research Institute—University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada (Y.-J.J.); (A.A.); (D.-Y.W.); (H.L.); (E.M.); (R.G.-A.); (M.S.)
| | - Amjad Ali
- Toronto General Research Institute—University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada (Y.-J.J.); (A.A.); (D.-Y.W.); (H.L.); (E.M.); (R.G.-A.); (M.S.)
| | - Philip E. D. Chung
- Toronto General Research Institute—University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada (Y.-J.J.); (A.A.); (D.-Y.W.); (H.L.); (E.M.); (R.G.-A.); (M.S.)
- Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Dong-Yu Wang
- Toronto General Research Institute—University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada (Y.-J.J.); (A.A.); (D.-Y.W.); (H.L.); (E.M.); (R.G.-A.); (M.S.)
| | - Jeff C. Liu
- The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada;
| | - Huiqin Li
- Toronto General Research Institute—University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada (Y.-J.J.); (A.A.); (D.-Y.W.); (H.L.); (E.M.); (R.G.-A.); (M.S.)
| | - Ioulia Vorobieva
- Toronto General Research Institute—University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada (Y.-J.J.); (A.A.); (D.-Y.W.); (H.L.); (E.M.); (R.G.-A.); (M.S.)
- Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ethel Mwewa
- Toronto General Research Institute—University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada (Y.-J.J.); (A.A.); (D.-Y.W.); (H.L.); (E.M.); (R.G.-A.); (M.S.)
| | - Ronak Ghanbari-Azarnier
- Toronto General Research Institute—University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada (Y.-J.J.); (A.A.); (D.-Y.W.); (H.L.); (E.M.); (R.G.-A.); (M.S.)
- Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Mariusz Shrestha
- Toronto General Research Institute—University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada (Y.-J.J.); (A.A.); (D.-Y.W.); (H.L.); (E.M.); (R.G.-A.); (M.S.)
- Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China;
- The Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Eldad Zacksenhaus
- Toronto General Research Institute—University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada (Y.-J.J.); (A.A.); (D.-Y.W.); (H.L.); (E.M.); (R.G.-A.); (M.S.)
- Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| |
Collapse
|
22
|
Domingues M, Leite Pereira C, Sarmento B, Castro F. Mimicking 3D breast tumor-stromal interactions to screen novel cancer therapeutics. Eur J Pharm Sci 2023; 190:106560. [PMID: 37557927 DOI: 10.1016/j.ejps.2023.106560] [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: 04/11/2023] [Revised: 07/31/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023]
Abstract
Most of the 3D breast tumor models used in drug screening studies only comprise tumor cells, keeping out other essential cell players of the tumor microenvironment. Tumor-associated macrophages and fibroblasts are frequently correlated with tumor progression and therapy resistance, and targeting these cells at the tumor site has been appointed as a promising therapeutic strategy. However, the translation of new therapies to the clinic has been hampered by the absence of cellular models that more closely mimic the features of in vivo breast tumor microenvironment. Therefore, the development of innovative 3D models able to provide consistent and predictive responses about the in vivo efficacy of novel therapeutics is still an unmet preclinical need. Herein, we have established an in vitro 3D heterotypic spheroid model including MCF-7 breast tumor cells, human mammary fibroblasts and human macrophages. To establish this model, different cell densities have been combined and characterized through the evaluation of the spheroid size and metabolic activity, as well as histological and immunohistochemistry analysis of the 3D multicellular structures. The final optimized 3D model consisted in a multicellular spheroid seeded at the initial density of 5000 cells and cell ratio of 1:2:1 (MCF-7:monocytes:fibroblasts). Our model recapitulates several features of the breast tumor microenvironment, including the formation of a necrotic core, spatial organization, and extracellular matrix production. Further, it was validated as a platform for drug screening studies, using paclitaxel, a currently approved drug for breast cancer treatment, and Gefitinib, a chemotherapeutic approved for lung cancer and in preclinical evaluation for breast cancer. Generally, the impact on the cell viability of the 3D model was less evident than in 2D model, reinforcing the relevance of such complex 3D models in addressing novel treatment approaches. Overall, the use of a 3D heterotypic spheroid of breast cancer could be a valuable tool to predict the therapeutic effect of new treatments for breast cancer patients, by recapitulating key features of the breast cancer microenvironment.
Collapse
Affiliation(s)
- Mariana Domingues
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; FEUP - Faculdade de Engenharia da Universidade do Porto, Rua Doutor Roberto Frias, Porto 4200-465, Portugal
| | - Catarina Leite Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, Gandra 4585-116, Portugal.
| | - Flávia Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal.
| |
Collapse
|
23
|
Rosa ML, Reinert T, Pauletto MM, Sartori G, Graudenz M, Barrios CH. Implications of tumor-infiltrating lymphocytes in early-stage triple-negative breast cancer: clinical oncologist perspectives. TRANSLATIONAL BREAST CANCER RESEARCH : A JOURNAL FOCUSING ON TRANSLATIONAL RESEARCH IN BREAST CANCER 2023; 5:4. [PMID: 38751669 PMCID: PMC11093080 DOI: 10.21037/tbcr-23-43] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/09/2023] [Indexed: 05/18/2024]
Abstract
Breast cancer (BC) is the most common neoplasm in women worldwide and one of the leading causes of female death. The triple-negative subtype, characterized by the absence of hormone receptor (HR) and human epidermal growth factor receptor 2 (HER2), tends to occur in younger patients, be more aggressive and less differentiated. Furthermore, this subtype is considered the most immunogenic and associated with higher levels of tumor cell infiltration, mainly lymphocytes. Tumor-infiltrating lymphocytes (TILs) play a crucial role in the interaction of the host's immune system and cancer cells. The microenvironment is critical in tumor development and progression. Assessment of infiltrating lymphocytes can provide valuable information about the immune response and, given the lack of biomarkers to guide treatment decisions and predict outcomes in triple-negative tumors and can be considered as a potential biomarker. Some evidence suggests that higher levels of these lymphocytes are associated with better responses to systemic treatment, longer progression-free survival and overall survival (OS). However, treatment escalation or de-escalation strategies for triple-negative BC (TNBC) currently do not consider the presence or density of TILs for therapeutic decisions. TILs appear to be useful predictive and prognostic indicators. Further clinical studies are needed to confirm these relationships and integrate TILs as a biomarker consistently into clinical practice. This article summarizes key concepts relating to the role of the immune infiltrate in BC, along with the current status and future prospects regarding TILs as a predictive and prognostic biomarker.
Collapse
Affiliation(s)
- Mahira Lopes Rosa
- Postgraduate Program in Medical Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Oncoclínicas, Porto Alegre, Brazil
| | - Tomas Reinert
- Oncoclínicas, Porto Alegre, Brazil
- Grupo Brasileiro de Estudos em Câncer de Mama (GBECAM), Porto Alegre, Brazil
| | | | - Guilherme Sartori
- Postgraduate Program in Medical Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Centro de Pesquisa da Serra Gaúcha (CEPESG), Caxias do Sul, Brazil
| | - Marcia Graudenz
- Postgraduate Program in Medical Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Pathology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Carlos Henrique Barrios
- Oncoclínicas, Porto Alegre, Brazil
- Latin American Cooperative Oncology Group (LACOG), Porto Alegre, Brazil
| |
Collapse
|
24
|
Ngaha TYS, Zhilenkova AV, Essogmo FE, Uchendu IK, Abah MO, Fossa LT, Sangadzhieva ZD, D. Sanikovich V, S. Rusanov A, N. Pirogova Y, Boroda A, Rozhkov A, Kemfang Ngowa JD, N. Bagmet L, I. Sekacheva M. Angiogenesis in Lung Cancer: Understanding the Roles of Growth Factors. Cancers (Basel) 2023; 15:4648. [PMID: 37760616 PMCID: PMC10526378 DOI: 10.3390/cancers15184648] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Research has shown the role of growth factors in lung cancer angiogenesis. Angiogenesis promotes lung cancer progression by stimulating tumor growth, enhancing tumor invasion, contributing to metastasis, and modifying immune system responses within the tumor microenvironment. As a result, new treatment techniques based on the anti-angiogenic characteristics of compounds have been developed. These compounds selectively block the growth factors themselves, their receptors, or the downstream signaling pathways activated by these growth factors. The EGF and VEGF families are the primary targets in this approach, and several studies are being conducted to propose anti-angiogenic drugs that are increasingly suitable for the treatment of lung cancer, either as monotherapy or as combined therapy. The efficacy of the results are encouraging, but caution must be placed on the higher risk of toxicity, outlining the importance of personalized follow-up in the management of these patients.
Collapse
Affiliation(s)
- Tchawe Yvan Sinclair Ngaha
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
- Department of Public Health, James Lind Institute, Rue de la Cité 1, 1204 Geneva, Switzerland
| | - Angelina V. Zhilenkova
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Freddy Elad Essogmo
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Ikenna K. Uchendu
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
- Medical Laboratory Science Department, Faculty of Health Science and Technology, College of Medicine, University of Nigeria, Enugu Campus, Enugu 410001, Nigeria
| | - Moses Owoicho Abah
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Lionel Tabola Fossa
- Department of Oncology, Bafoussam Regional Hospital, Bafoussam 980, Cameroon;
| | - Zaiana D. Sangadzhieva
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Varvara D. Sanikovich
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Alexander S. Rusanov
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Yuliya N. Pirogova
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Alexander Boroda
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Alexander Rozhkov
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Jean D. Kemfang Ngowa
- Faculty of Medicine and Biomedical Sciences, University of Yaounde I, Yaounde 1364, Cameroon;
| | - Leonid N. Bagmet
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| | - Marina I. Sekacheva
- Institute for Personalized Oncology, Center for Digital Biodesign and Personalized Healthcare, First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russia; (T.Y.S.N.); (A.V.Z.); (F.E.E.); (I.K.U.); (M.O.A.); (Z.D.S.); (V.D.S.); (A.S.R.); (Y.N.P.); (A.B.); (A.R.); (L.N.B.)
| |
Collapse
|
25
|
Terceiro LEL, Ikeogu NM, Lima MF, Edechi CA, Nickel BE, Fischer G, Leygue E, McManus KJ, Myal Y. Navigating the Blood-Brain Barrier: Challenges and Therapeutic Strategies in Breast Cancer Brain Metastases. Int J Mol Sci 2023; 24:12034. [PMID: 37569410 PMCID: PMC10418424 DOI: 10.3390/ijms241512034] [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] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Breast cancer (BC) is the most common cancer in women, with metastatic BC being responsible for the highest number of deaths. A frequent site for BC metastasis is the brain. Brain metastasis derived from BC involves the cooperation of multiple genetic, epigenetic, angiogenic, and tumor-stroma interactions. Most of these interactions provide a unique opportunity for development of new therapeutic targets. Potentially targetable signaling pathways are Notch, Wnt, and the epidermal growth factor receptors signaling pathways, all of which are linked to driving BC brain metastasis (BCBM). However, a major challenge in treating brain metastasis remains the blood-brain barrier (BBB). This barrier restricts the access of unwanted molecules, cells, and targeted therapies to the brain parenchyma. Moreover, current therapies to treat brain metastases, such as stereotactic radiosurgery and whole-brain radiotherapy, have limited efficacy. Promising new drugs like phosphatase and kinase modulators, as well as BBB disruptors and immunotherapeutic strategies, have shown the potential to ease the disease in preclinical studies, but remain limited by multiple resistance mechanisms. This review summarizes some of the current understanding of the mechanisms involved in BC brain metastasis and highlights current challenges as well as opportunities in strategic designs of potentially successful future therapies.
Collapse
Affiliation(s)
- Lucas E. L. Terceiro
- Department of Pathology and Laboratory Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; (L.E.L.T.); (C.A.E.); (B.E.N.); (G.F.)
| | - Nnamdi M. Ikeogu
- Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada;
| | - Matheus F. Lima
- Department of Physiology and Pathophysiology, CancerCare Manitoba Research Institute, University of Manitoba, Winnipeg, MB R3E 0V9, Canada;
| | - Chidalu A. Edechi
- Department of Pathology and Laboratory Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; (L.E.L.T.); (C.A.E.); (B.E.N.); (G.F.)
| | - Barbara E. Nickel
- Department of Pathology and Laboratory Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; (L.E.L.T.); (C.A.E.); (B.E.N.); (G.F.)
| | - Gabor Fischer
- Department of Pathology and Laboratory Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; (L.E.L.T.); (C.A.E.); (B.E.N.); (G.F.)
| | - Etienne Leygue
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (E.L.); (K.J.M.)
| | - Kirk J. McManus
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (E.L.); (K.J.M.)
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Yvonne Myal
- Department of Pathology and Laboratory Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; (L.E.L.T.); (C.A.E.); (B.E.N.); (G.F.)
- Department of Physiology and Pathophysiology, CancerCare Manitoba Research Institute, University of Manitoba, Winnipeg, MB R3E 0V9, Canada;
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| |
Collapse
|
26
|
Loric S, Denis JA, Desbene C, Sabbah M, Conti M. Extracellular Vesicles in Breast Cancer: From Biology and Function to Clinical Diagnosis and Therapeutic Management. Int J Mol Sci 2023; 24:7208. [PMID: 37108371 PMCID: PMC10139222 DOI: 10.3390/ijms24087208] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/03/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
Breast cancer (BC) is the first worldwide most frequent cancer in both sexes and the most commonly diagnosed in females. Although BC mortality has been thoroughly declining over the past decades, there are still considerable differences between women diagnosed with early BC and when metastatic BC is diagnosed. BC treatment choice is widely dependent on precise histological and molecular characterization. However, recurrence or distant metastasis still occurs even with the most recent efficient therapies. Thus, a better understanding of the different factors underlying tumor escape is mainly mandatory. Among the leading candidates is the continuous interplay between tumor cells and their microenvironment, where extracellular vesicles play a significant role. Among extracellular vesicles, smaller ones, also called exosomes, can carry biomolecules, such as lipids, proteins, and nucleic acids, and generate signal transmission through an intercellular transfer of their content. This mechanism allows tumor cells to recruit and modify the adjacent and systemic microenvironment to support further invasion and dissemination. By reciprocity, stromal cells can also use exosomes to profoundly modify tumor cell behavior. This review intends to cover the most recent literature on the role of extracellular vesicle production in normal and cancerous breast tissues. Specific attention is paid to the use of extracellular vesicles for early BC diagnosis, follow-up, and prognosis because exosomes are actually under the spotlight of researchers as a high-potential source of liquid biopsies. Extracellular vesicles in BC treatment as new targets for therapy or efficient nanovectors to drive drug delivery are also summarized.
Collapse
Affiliation(s)
- Sylvain Loric
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | | | - Cédric Desbene
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | - Michèle Sabbah
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | - Marc Conti
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
- INTEGRACELL SAS, 91160 Longjumeau, France
| |
Collapse
|
27
|
Fröhlich E. The Variety of 3D Breast Cancer Models for the Study of Tumor Physiology and Drug Screening. Int J Mol Sci 2023; 24:ijms24087116. [PMID: 37108283 PMCID: PMC10139112 DOI: 10.3390/ijms24087116] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/01/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Breast cancer is the most common cancer in women and responsible for multiple deaths worldwide. 3D cancer models enable a better representation of tumor physiology than the conventional 2D cultures. This review summarizes the important components of physiologically relevant 3D models and describes the spectrum of 3D breast cancer models, e.g., spheroids, organoids, breast cancer on a chip and bioprinted tissues. The generation of spheroids is relatively standardized and easy to perform. Microfluidic systems allow control over the environment and the inclusion of sensors and can be combined with spheroids or bioprinted models. The strength of bioprinting relies on the spatial control of the cells and the modulation of the extracellular matrix. Except for the predominant use of breast cancer cell lines, the models differ in stromal cell composition, matrices and fluid flow. Organoids are most appropriate for personalized treatment, but all technologies can mimic most aspects of breast cancer physiology. Fetal bovine serum as a culture supplement and Matrigel as a scaffold limit the reproducibility and standardization of the listed 3D models. The integration of adipocytes is needed because they possess an important role in breast cancer.
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, 8010 Graz, Austria
- Research Center Pharmaceutical Engineering GmbH, 8010 Graz, Austria
| |
Collapse
|
28
|
Liu Y, Ge J, Chen Y, Liu T, Chen L, Liu C, Ma D, Chen Y, Cai Y, Xu Y, Shao Z, Yu K. Combined Single-Cell and Spatial Transcriptomics Reveal the Metabolic Evolvement of Breast Cancer during Early Dissemination. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205395. [PMID: 36594618 PMCID: PMC9951304 DOI: 10.1002/advs.202205395] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/10/2022] [Indexed: 06/17/2023]
Abstract
Breast cancer is now the most frequently diagnosed malignancy, and metastasis remains the leading cause of death in breast cancer. However, little is known about the dynamic changes during the evolvement of dissemination. In this study, 65 968 cells from four patients with breast cancer and paired metastatic axillary lymph nodes are profiled using single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics. A disseminated cancer cell cluster with high levels of oxidative phosphorylation (OXPHOS), including the upregulation of cytochrome C oxidase subunit 6C and dehydrogenase/reductase 2, is identified. The transition between glycolysis and OXPHOS when dissemination initiates is noticed. Furthermore, this distinct cell cluster is distributed along the tumor's leading edge. The findings here are verified in three different cohorts of breast cancer patients and an external scRNA-seq dataset, which includes eight patients with breast cancer and paired metastatic axillary lymph nodes. This work describes the dynamic metabolic evolvement of early disseminated breast cancer and reveals a switch between glycolysis and OXPHOS in breast cancer cells as the early event during lymph node metastasis.
Collapse
Affiliation(s)
- Yi‐Ming Liu
- Department of Breast SurgeryShanghai Cancer Center and Cancer InstituteFudan UniversityShanghai200032P. R. China
- Shanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Jing‐Yu Ge
- Shanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Yu‐Fei Chen
- Shanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Tong Liu
- Department of Breast SurgeryHarbin Medical University Cancer HospitalHarbinHeilongjiang150081P. R. China
| | - Lie Chen
- Shanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Cui‐Cui Liu
- Department of Breast SurgeryShanghai Cancer Center and Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Ding Ma
- Department of Breast SurgeryShanghai Cancer Center and Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Yi‐Yu Chen
- Shanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Yu‐Wen Cai
- Shanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Ying‐Ying Xu
- Department of Breast SurgeryThe First Affiliated Hospital of China Medical UniversityShenyangLiaoning110000P. R. China
| | - Zhi‐Ming Shao
- Department of Breast SurgeryShanghai Cancer Center and Cancer InstituteFudan UniversityShanghai200032P. R. China
- Key Laboratory of Breast Cancer in ShanghaiShanghai200032P. R. China
| | - Ke‐Da Yu
- Department of Breast SurgeryShanghai Cancer Center and Cancer InstituteFudan UniversityShanghai200032P. R. China
- Shanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Key Laboratory of Breast Cancer in ShanghaiShanghai200032P. R. China
| |
Collapse
|
29
|
Gong X, Chi H, Strohmer DF, Teichmann AT, Xia Z, Wang Q. Exosomes: A potential tool for immunotherapy of ovarian cancer. Front Immunol 2023; 13:1089410. [PMID: 36741380 PMCID: PMC9889675 DOI: 10.3389/fimmu.2022.1089410] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/30/2022] [Indexed: 01/19/2023] Open
Abstract
Ovarian cancer is a malignant tumor of the female reproductive system, with a very poor prognosis and high mortality rates. Chemotherapy and radiotherapy are the most common treatments for ovarian cancer, with unsatisfactory results. Exosomes are a subpopulation of extracellular vesicles, which have a diameter of approximately 30-100 nm and are secreted by many different types of cells in various body fluids. Exosomes are highly stable and are effective carriers of immunotherapeutic drugs. Recent studies have shown that exosomes are involved in various cellular responses in the tumor microenvironment, influencing the development and therapeutic efficacy of ovarian cancer, and exhibiting dual roles in inhibiting and promoting tumor development. Exosomes also contain a variety of genes related to ovarian cancer immunotherapy that could be potential biomarkers for ovarian cancer diagnosis and prognosis. Undoubtedly, exosomes have great therapeutic potential in the field of ovarian cancer immunotherapy. However, translation of this idea to the clinic has not occurred. Therefore, it is important to understand how exosomes could be used in ovarian cancer immunotherapy to regulate tumor progression. In this review, we summarize the biomarkers of exosomes in different body fluids related to immunotherapy in ovarian cancer and the potential mechanisms by which exosomes influence immunotherapeutic response. We also discuss the prospects for clinical application of exosome-based immunotherapy in ovarian cancer.
Collapse
Affiliation(s)
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Dorothee Franziska Strohmer
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Alexander Tobias Teichmann
- Sichuan Provincial Center for Gynecology and Breast Diseases (Gynecology), Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany,*Correspondence: Zhijia Xia, ; Qin Wang,
| | - Qin Wang
- Sichuan Provincial Center for Gynecology and Breast Diseases (Gynecology), Affiliated Hospital of Southwest Medical University, Luzhou, China,*Correspondence: Zhijia Xia, ; Qin Wang,
| |
Collapse
|
30
|
The Binomial "Inflammation-Epigenetics" in Breast Cancer Progression and Bone Metastasis: IL-1β Actions Are Influenced by TET Inhibitor in MCF-7 Cell Line. Int J Mol Sci 2022; 23:ijms232315422. [PMID: 36499741 PMCID: PMC9741332 DOI: 10.3390/ijms232315422] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/25/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
The existence of a tight relationship between inflammation and epigenetics that in primary breast tumor cells can lead to tumor progression and the formation of bone metastases was investigated. It was highlighted how the induction of tumor progression and bone metastasis by Interleukin-1 beta, in a non-metastatic breast cancer cell line, MCF-7, was dependent on the de-methylating actions of ten-eleven translocation proteins (TETs). In fact, the inhibition of their activity by the Bobcat339 molecule, an inhibitor of TET enzymes, determined on the one hand, the modulation of the epithelial-mesenchymal transition process, and on the other hand, the reduction in the expression of markers of bone metastasis, indicating that the epigenetic action of TETs is a prerequisite for IL-1β-dependent tumor progression and bone metastasis formation.
Collapse
|
31
|
Ennis CS, Llevenes P, Qiu Y, Dries R, Denis GV. The crosstalk within the breast tumor microenvironment in type II diabetes: Implications for cancer disparities. Front Endocrinol (Lausanne) 2022; 13:1044670. [PMID: 36531496 PMCID: PMC9751481 DOI: 10.3389/fendo.2022.1044670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/17/2022] [Indexed: 12/04/2022] Open
Abstract
Obesity-driven (type 2) diabetes (T2D), the most common metabolic disorder, both increases the incidence of all molecular subtypes of breast cancer and decreases survival in postmenopausal women. Despite this clear link, T2D and the associated dysfunction of diverse tissues is often not considered during the standard of care practices in oncology and, moreover, is treated as exclusion criteria for many emerging clinical trials. These guidelines have caused the biological mechanisms that associate T2D and breast cancer to be understudied. Recently, it has been illustrated that the breast tumor microenvironment (TME) composition and architecture, specifically the surrounding cellular and extracellular structures, dictate tumor progression and are directly relevant for clinical outcomes. In addition to the epithelial cancer cell fraction, the breast TME is predominantly made up of cancer-associated fibroblasts, adipocytes, and is often infiltrated by immune cells. During T2D, signal transduction among these cell types is aberrant, resulting in a dysfunctional breast TME that communicates with nearby cancer cells to promote oncogenic processes, cancer stem-like cell formation, pro-metastatic behavior and increase the risk of recurrence. As these cells are non-malignant, despite their signaling abnormalities, data concerning their function is never captured in DNA mutational databases, thus we have limited insight into mechanism from publicly available datasets. We suggest that abnormal adipocyte and immune cell exhaustion within the breast TME in patients with obesity and metabolic disease may elicit greater transcriptional plasticity and cellular heterogeneity within the expanding population of malignant epithelial cells, compared to the breast TME of a non-obese, metabolically normal patient. These challenges are particularly relevant to cancer disparities settings where the fraction of patients seen within the breast medical oncology practice also present with co-morbid obesity and metabolic disease. Within this review, we characterize the changes to the breast TME during T2D and raise urgent molecular, cellular and translational questions that warrant further study, considering the growing prevalence of T2D worldwide.
Collapse
Affiliation(s)
- Christina S. Ennis
- Boston University-Boston Medical Center Cancer Center, Boston University School of Medicine, Boston, MA, United States
- Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, United States
| | - Pablo Llevenes
- Boston University-Boston Medical Center Cancer Center, Boston University School of Medicine, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Yuhan Qiu
- Boston University-Boston Medical Center Cancer Center, Boston University School of Medicine, Boston, MA, United States
| | - Ruben Dries
- Boston University-Boston Medical Center Cancer Center, Boston University School of Medicine, Boston, MA, United States
- Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, United States
- Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA, United States
| | - Gerald V. Denis
- Boston University-Boston Medical Center Cancer Center, Boston University School of Medicine, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Shipley Prostate Cancer Research Professor, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
32
|
Overcoming challenges to enable targeting of metastatic breast cancer tumour microenvironment with nano-therapeutics: Current status and future perspectives. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
33
|
Bai R, Li Y, Jian L, Yang Y, Zhao L, Wei M. The hypoxia-driven crosstalk between tumor and tumor-associated macrophages: mechanisms and clinical treatment strategies. Mol Cancer 2022; 21:177. [PMID: 36071472 PMCID: PMC9454207 DOI: 10.1186/s12943-022-01645-2] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/25/2022] [Indexed: 02/08/2023] Open
Abstract
Given that hypoxia is a persistent physiological feature of many different solid tumors and a key driver for cancer malignancy, it is thought to be a major target in cancer treatment recently. Tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME), which have a large impact on tumor development and immunotherapy. TAMs massively accumulate within hypoxic tumor regions. TAMs and hypoxia represent a deadly combination because hypoxia has been suggested to induce a pro-tumorigenic macrophage phenotype. Hypoxia not only directly affects macrophage polarization, but it also has an indirect effect by altering the communication between tumor cells and macrophages. For example, hypoxia can influence the expression of chemokines and exosomes, both of which have profound impacts on the recipient cells. Recently, it has been demonstrated that the intricate interaction between cancer cells and TAMs in the hypoxic TME is relevant to poor prognosis and increased tumor malignancy. However, there are no comprehensive literature reviews on the molecular mechanisms underlying the hypoxia-mediated communication between tumor cells and TAMs. Therefore, this review has the aim to collect all recently available data on this topic and provide insights for developing novel therapeutic strategies for reducing the effects of hypoxia.
Collapse
Affiliation(s)
- Ruixue Bai
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, People's Republic of China.,Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Yunong Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, People's Republic of China
| | - Lingyan Jian
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Yuehui Yang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China. .,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, People's Republic of China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China. .,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, People's Republic of China. .,Shenyang Kangwei Medical Laboratory Analysis Co. LTD, Shenyang, 110000, People's Republic of China.
| |
Collapse
|
34
|
Analysis of Genomic Alterations Associated with Recurrence in Early Stage HER2-Positive Breast Cancer. Cancers (Basel) 2022; 14:cancers14153650. [PMID: 35954313 PMCID: PMC9367395 DOI: 10.3390/cancers14153650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/20/2022] [Accepted: 07/23/2022] [Indexed: 12/25/2022] Open
Abstract
We aimed to compare gene expression in primary tumors of patients with recurrence and nonrecurrence to gain insight into the biology of high-risk HER2-positive early breast cancer. Patients who underwent curative resection and received adjuvant trastuzumab for HER2-positive early breast cancer were evaluated. Gene expression analyses were performed using NanoString Technologies’ nCounter Breast Cancer 360 Panel. PAM50 intrinsic subtypes and Breast Cancer Signatures including tumor inflammation signature (TIS) were evaluated. Of 247 patients, 28 (11.3%) had recurrence at a median follow-up of 54.2 months. Patients with pathological stage III, tumor size > 5 cm, axillary lymph node metastases, and hormone receptor-negativity were more frequently observed in the recurrent group compared with the nonrecurrent group. In patients with recurrence, seven genes were upregulated significantly, including WNT11, HAPLN1, FGF10, BBOX1, CXADR, NDP, and EREG, and two genes were downregulated, including CXCL9 and GNLY. TIS score was significantly lower in patients with recurrence compared with controls without recurrence. These findings suggest that activation of oncogenic signaling pathways related to cell proliferation, adhesion, cancer stemness, and noninflamed tumor microenvironment are associated with the risk of recurrence in early stage, HER2-positive breast cancer.
Collapse
|
35
|
Kalinke L, Janes SM. Two phenotypes that predict prognosis in lung adenocarcinoma. Eur Respir J 2022; 60:60/1/2200569. [PMID: 35798373 DOI: 10.1183/13993003.00569-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 03/21/2022] [Indexed: 11/05/2022]
Affiliation(s)
- Lukas Kalinke
- UCL Respiratory, University College London, London, UK
| | - Sam M Janes
- UCL Respiratory, University College London, London, UK
| |
Collapse
|
36
|
Neill T, Iozzo RV. The Role of Decorin Proteoglycan in Mitophagy. Cancers (Basel) 2022; 14:804. [PMID: 35159071 PMCID: PMC8834502 DOI: 10.3390/cancers14030804] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/02/2022] [Indexed: 02/04/2023] Open
Abstract
Proteoglycans are emerging as critical regulators of intracellular catabolism. This rise in prominence has transformed our basic understanding and alerted us to the existence of non-canonical pathways, independent of nutrient deprivation, that potently control the autophagy downstream of a cell surface receptor. As a member of the small leucine-rich proteoglycan gene family, decorin has single-handedly pioneered the connection between extracellular matrix signaling and autophagy regulation. Soluble decorin evokes protracted endothelial cell autophagy via Peg3 and breast carcinoma cell mitophagy via mitostatin by interacting with VEGFR2 or the MET receptor tyrosine kinase, respectively. In this paper, we give a mechanistic perspective of the vital factors underlying the nutrient-independent, SLRP-dependent programs utilized for autophagic and/or mitophagic progression in breast cancer. Future protein therapies based on decorin (or fellow proteoglycan members) will represent a quantum leap forward in transforming autophagic progression into a powerful tool to control intracellular cell catabolism from the outside.
Collapse
Affiliation(s)
- Thomas Neill
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|