به جمع مشترکان مگیران بپیوندید!

تنها با پرداخت 70 هزارتومان حق اشتراک سالانه به متن مقالات دسترسی داشته باشید و 100 مقاله را بدون هزینه دیگری دریافت کنید.

برای پرداخت حق اشتراک اگر عضو هستید وارد شوید در غیر این صورت حساب کاربری جدید ایجاد کنید

عضویت
جستجوی مقالات مرتبط با کلیدواژه

tumor microenvironment

در نشریات گروه پزشکی
  • Junfeng Wang, Dalu Kong *
    Objective (s)

    The use of cryoablation for colorectal liver metastases (CLM) remains limited and controversial. This study aimed to investigate the antitumor immune response following cryoablation combined with granulocyte-macrophage colony-stimulating factor (GM-CSF) treatment in a CLM mouse model.

    Materials and Methods

    A CLM mouse model was established using BALB/c mice. The tumor-bearing mice were randomly divided into Control group, GM-CSF group, cryoablation group, and cryoablation + GM-CSF group. Tumor size, survival time, dendritic cells (DCs) count, serum cytokine levels (IL-4, IFN-γ), and the Th1/Th2 ratio (IFN-γ/IL-4) were compared among the four groups.

    Results

    The combination of cryoablation and GM-CSF demonstrated synergistic effects, resulting in the smallest tumor lesion, longest mean survival time, and highest DC count on day 21 post-treatment compared to other groups. Both cryoablation alone and combined with GM-CSF significantly increased serum IFN-γ levels and suppressed IL-4 levels on day 21 compared to pre-treatment levels (P<0.05). Notably, the combination of cryoablation and GM-CSF significantly elevated the Th1/Th2 ratio (P<0.05).

    Conclusion

    Combining cryoablation with GM-CSF treatment holds promise for CLM management. It exhibits increased DC infiltration within the tumor microenvironment, enhanced immune responses, and prolonged survival in tumor-bearing mice.

    Keywords: Colorectal Neoplasms, Cryosurgery, Cytokines, Dendritic Cells, Granulocyte-Macrophage - Colony-Stimulating Factor, Immunity, Liver Neoplasms, Tumor Microenvironment
  • Nazanin Jamshidi, Negar Jamshidi, Amir Modarresi Chahardehi, Elahe Shams, Vahid Chaleshi *

    Pancreatic ductal adenocarcinoma (PDAC) stands as the fourth leading cause of cancer-related deaths, primarily attributable to its resistance to chemotherapy, resulting in a nearly universal fatality rate. Despite the promise exhibited by numerous drugs in preclinical studies, their subsequent failure in clinical trials underscores the inherent limitations of conventional two-dimensional cell culture models commonly employed in early drug screening endeavors. The inadequacies of two-dimensional (2D) models prompted the exploration of three-dimensional (3D) culture systems, which more faithfully recapitulate the native tumor microenvironment. These 3D systems have distinct advantages over 2D models in morphology, proliferation, drug response, and protein expression. Among these 3D platforms, tumor organoids and spheroids, generated through different methodologies, have emerged as next-generation models that closely mirror aspects of pancreatic tumor biology. This comprehensive review scrutinizes pancreatic cancer spheroids' techniques, tissue sources, and applications, offering a nuanced analysis of their advantages and limitations. By comparing these distinct 3D culture systems, researchers gain valuable insights to inform the selection of optimal model designs aligned with their specific experimental objectives. The utilization of these advanced models holds significant promise for enhancing the clinical relevance of both in vitro and in vivo cancer research, thereby contributing to the development of improved therapeutics against pancreatic cancer.

    Keywords: 3D Cell Culture, Spheroid, Tumor Microenvironment, Pancreatic Cancer, In Vitro Cancer Model
  • Milad Asadi, Venus Zafari, Sanam Sadeghi-Mohammadi, Dariush Shanehbandi, Ufuk Mert, Zahra Soleimani, Ayşe Caner, Habib Zarredar *
    Introduction

    The tumor microenvironment (TME) plays a pivotal role in cancer progression, influencing tumor initiation, growth, invasion, metastasis, and response to therapies. This study explores the dynamic interactions within the TME, particularly focusing on self-organization—a process by which tumor cells and their microenvironment reciprocally shape one another, leading to cancer progression and resistance. Understanding these interactions can reveal new prognostic markers and therapeutic targets within the TME, such as extracellular matrix (ECM) components, immune cells, and cytokine signaling pathways.

    Methods

    A comprehensive search method was employed to investigate the current academic literature on TME, particularly focusing on self-organization in the context of cancer progression and resistance across the PubMed, Google Scholar, and Science Direct databases.

    Results

    Recent studies suggest that therapies that disrupt TME self-organization could improve patient outcomes by defeating drug resistance and increasing the effectiveness of conventional therapy. Additionally, this research highlights the essential of understanding the biophysical properties of the TME, like cytoskeletal alterations, in the development of more effective malignancy therapy.

    Conclusion

    This review indicated that targeting the ECM and immune cells within the TME can improve therapy effectiveness. Also, by focusing on TME self-organization, we can recognize new therapeutic plans to defeat drug resistance.

    Keywords: Tumor Microenvironment, Self-Organization, Extracellular Matrix, Metastasis, Invasion
  • Yurong Liu, Jianlan Liu, Keyu Jiang, Xiaolong Cheng, Sitong Di, Jian Tang, Binlin Luo*

    Regulatory T cells (Tregs) are central to establishing an immunosuppressive tumor microenvironment (TME), which promotes cancer progression and influences therapeutic outcomes. However, the prognostic significance of Treg-related genes (TRGs) in predicting immunotherapy response in melanoma remains insufficiently characterized. This study seeks to elucidate the role of TRGs in the antitumor immune response of melanoma. The ordinary transcriptome and single-cell RNA sequencing (scRNA-seq) data were obtained from the gene expression omnibus and the cancer genome atlas databases. A multi-tiered quality control process was applied to scRNA-seq data, followed by cell annotation, cell-cell communication, and enrichment analysis to investigate Treg function in the melanoma microenvironment. Weighted gene coexpression network analysis (WGCNA) was employed to identify modules associated with Treg infiltration. Key prognostic genes were identified using univariate Cox regression analysis and integrated into a prognostic model through least absolute shrinkage and selection operator and stepwise regression methods. The analysis revealed a Treg-related gene signature (TRGS) comprising CHD3, FOSB, SEMA4D, PSME1, FYN, PRKACB, and ARID5A. Higher TRGS-based risk scores were significantly associated with worse prognoses, immune cell infiltration, and stromal scores. TRGS was identified as an independent prognostic indicator for melanoma, offering novel insights into the role of Tregs in modulating the TME. This study highlights the potential clinical utility of TRGs in melanoma diagnostics and personalized immunotherapy, providing a robust foundation for future therapeutic strategies.

    Keywords: Immunotherapy, Melanoma, Regulatory T Cell, Single Cell Sequencing, Tumor Microenvironment
  • Wojciech Wawrety, Anna Kedziora *

    Cancers are extremely dynamic diseases that can actively cause refractorines to be gained from applied therapies, which is why they are at the forefront of deaths worldwide. In this literature review,  we covered the most recent and important discoveries regarding the influence of human microbiota, including tumor bacteriome, on the development and treatment of cancer. Advances in research on microbial communities have enabled us to discover the role of the human microbiome in the development and course of this disease, helping us understand neoplasms better and design new potential therapies. As we show through our findings, by immunomodulation and the secretion of certain chemical substances, the correct bacteriome of the intestinal tract, respiratory system, or skin can protect humans against cancer development and help during the treatment process. Bacteria also reside inside tumors, forming part of the tumor microenvironment (TME), where they interact with immunological and cancer cells in many complex ways. Some bacteria, such as Pseudomonas aeruginosa or Akkermansia muciniphila, can stimulate anticancer cell-mediated immune responses or even directly lead to cancer cell death. We also present the clinical possibilities of using some live, usually modified bacteria to develop bacteriotherapies. Modifying the gut microbiome to stimulate standard treatment is also important. Research on the microbiome and cancer remains a challenging topic in microbiology, having a great potential for advancements in cancer therapy in the future, and is continuously becoming a more and more popular field of research, as shown by our statistical analysis of PubMed data.

    Keywords: Bacteria, Immunotherapy, Microbiota, Neoplasms, Tumor Microenvironment
  • Khadijeh Ramezani Aliakbari, Seyed Amir Jalali, Maedeh Alinejad, Mahmood Jeddi Tehrani, Mahdi Shabani *
    Background

     Myeloid Derived Suppressor Cells (MDSCs) are capable of inhibiting both innate and adaptive immune responses and accumulate in the microenvironment of breast tumors. Hence, MDSC depletion by chemotherapeutic agents can improve clinical efficacy of cancer immunotherapy. The effects of 5-FU and doxorubicin agents on MDSC reduction in 4T1 breast cancer murine model were evaluated.

    Methods

     5×105 of 4T1 tumor cells were injected into mammary fat pad of BALB/c female mice. Tumor bearing mice were randomly divided into 4 groups: PBS receiving control group, doxorubicin receiving groups at doses of 2.5 and 5 mg/kg, and 5-FU receiving group at dose of 50 mg/kg. Doxorubicin and 5-FU agents were intraperitoneally administrated at three doses with 5-day intervals and five doses for three times a week, respectively. Then, on day 20 post tumor cells injection, spleens and tumors were isolated to determine frequency of CD11b+ Gr1+ MDSCs by flow cytometry analysis.

    Results

     5-FU was able to reduce significantly both splenic and interatumoral MDSCs comparing to control group (p=0.0276 and p=0.0067, respectively). Also, Doxorubicin treatment at dose of 50 mg/kg was associated to a significant reduction of splenic MDSCs in comparison to untreated group (p=0.0382). However, only 5-FU injection led to inhibit notably tumor growth in comparison to control group (p=0.0139).

    Conclusion

     Findings show that 5-FU has inhibitory effects on MDSCs and tumor growth in 4T1 tumor model. So, more investigations are needed to study combination of 5-FU with immune based approaches to enhance the efficacy of cancer therapies.

    Keywords: Breast Cancer, Fluorouracil, Immunotherapy, Mammary Neoplasm, Tumor Microenvironment, Treatment Outcome
  • Zhuanyu Guo, Xue Wang, Shuaishuai Huang, Guo Weng
    Background

    M6A RNA methylation and the tumor microenvironment (TME) have been reported to play important roles in the progression and prognosis of clear cell renal cell carcinoma (ccRCC). However, whether m6A RNA methylation regulators affect the TME in ccRCC remains unknown. Thus, we aimed to evaluate comprehensively the effect of m6A RNA methylation regulators on the TME in ccRCC.

    Methods

    Transcriptome data of ccRCC were obtained from TCGA database. Consensus clustering analysis was conducted based on the expression of m6A RNA methylation regulators. Survival differences were evaluated by Kaplan–Meier analysis between the clusters. The DESeq2 package was used to analyze the differentially expressed genes (DEGs) between the clusters. GO and KEGG pathway analyses were performed by the ClusterProfiler R package. The CIBERSORT algorithm was used to evaluate immune infiltration.

    Results

    The expression of 15 m6A regulators significantly differed between ccRCC and normal kidney tissues. Based on the expression of these 15 m6A regulators, two clusters were identified by consensus clustering, in which cluster 1 had better overall survival (OS). Overall, 4,429 DEGs were identified between the two clusters and were enriched in immune-related biological processes. Cluster 1 had lower immune and ESTIMATE scores, higher expression of HLA and lower expression of immune checkpoint molecules. Moreover, immune infiltration and expressions of Th1/IFNγ gene signature were also significantly different between the two clusters.

    Conclusion

    Our study revealed m6A regulators were important participants in the development of ccRCC, with a close relationship with the TME.

    Keywords: M6A RNA Methylation Regulator, Tumor Microenvironment, Clear Cell Renal Cell Carcinoma
  • Amin Mehra, Reza Mehrkar, Amir Fakhri, Pedram Jabbari Fard, Mohammadreza Asgharzadeh, Mohammadhossein Ghaffari Agdam, Rasoul Sharifi *
    Background

    This study investigates the relative expression of the Na+, HCO3- cotransport gene NBCn1, and caspase-3 within the tumor microenvironment of human breast cancer, considering the in vivo microenvironment.

    Method

    In this experimental study, breast cancer MDA-MB-231 cells were cultured under normoxia/hypoxia conditions for 24, 48, and 72 hours with varying glucose concentrations (5.5, 11, and 25 mM). The mRNA expression of NBCn1 and caspase-3 was evaluated using real-time polymerase chain reaction. The stability and binding pocket of NBCn1 were assessed using DispHred and the Computed Atlas of Surface Topography of proteins (CASTp) servers, respectively. The location prediction of the protein was determined using the Transmembrane Helices; Hidden Markov Model (TMHMM) server.

    Results

    Normoxia led to an increase in NBCn1 expression during all three time periods, displaying heterogeneity. The expression was particularly elevated at glucose concentrations of 25 and 5.5 mM. In hypoxic conditions, gene expression was reduced; however, an increase in glucose concentration enhanced SLC4A7 expression. Specifically, a glucose concentration of 25 mM led to decreased caspase-3 expression under hypoxic conditions. In silico studies revealed that SLC4A7 becomes disordered when the pH falls below 7, with most amino acids in the binding pocket being nonpolar.

    Conclusion

    The heightened risk of breast cancer metastasis may be linked to the upregulation of SLC4A7 and downregulation of caspase-3 expression, underscoring their fundamental roles in cancer treatment and prevention. SLC4A7 is a transmembrane protein, and its folding is pH-dependent.

    Keywords: Tumor Microenvironment, SLC4A7, Caspase-3, Breast Neoplasms, In Silico
  • Mohd Mustafa, Badar Ul Islam, Irfan Qadir Tantry, Safia Habib*
    Introduction

    The management of advanced-stage solid tumors remains a formidable challenge in oncology, often necessitating a combination of treatment modalities with limited efficacy. Nucleic acid-based drugs have emerged as promising therapeutic options, offering novel approaches to combat treatment-resistant tumors. This editorial review briefly analyzes viral and non-viral nucleic acid-based treatments for solid tumors, including their development, clinical activity, mechanisms of action, and prospects. Viral vectors, such as adenoviruses and lentiviruses, enable the delivery of therapeutic genes directly into tumor cells or the modulation of the tumor microenvironment, resulting in regulatory approvals for several gene therapies. Non-viral delivery systems, including lipid nanoparticles and polymer-based carriers, offer alternative strategies for transporting therapeutic nucleic acids into tumor cells, with precise drug release and targeting control.

    Materials and Methods

    The literature review is based on searches of the PubMed database, Web of Science, Scopus, and Google Scholar using specific keywords. The article is based on manuscripts published between 2021 and 2024.

    Results

    The mechanistic actions of nucleic acid-based drugs are diverse, encompassing direct tumor cell killing, modulation of the immune response, and inhibition of oncogenic pathways. Importantly, these therapeutic approaches demonstrate a favorable safety profile compared to traditional chemotherapy agents, minimizing off-target effects and reducing adverse events. The tumor microenvironment significantly influences the effectiveness of nucleic acid-based therapies, requiring strategies to improve drug delivery and overcome resistance mechanisms. Ongoing clinical trials, preclinical studies, and advancements in manufacturing and stability technologies are expected to drive the next generation of nucleic acid agents, ensuring widespread accessibility and clinical utility.

    Conclusion

    Nucleic acid-based drugs represent a transformative approach to cancer therapy, potentially revolutionizing treatment outcomes and ushering in an era of precision medicine in oncology.

    Keywords: Antisense Oligonucleotide, Nucleic Acid-Based Drugs, Solid Tumors, Tumor Microenvironment, Viral, Non-Viral Approach
  • Sara Shayan, Arash Arashkia, Golnaz Bahramali, Kayhan Azadmanesh *
    Background

    Colorectal Cancer (CRC) represents a significant global health challenge, and its progression, resistance to therapy, and metastasis are strongly influenced by the tumor microenvironment, including factors like hypoxia. This study explores the impact of High Mobility Group Box 1 (HMGB1) overexpression on CRC cell migration, while identifying potential genes associated with this process.

    Methods

    To explore this, we developed oncolytic virotherapy, resulting in HSVHMGB1, an oncolytic Herpes simplex virus that expresses HMGB1. HMGB1 is known its role in cancer progression, particularly in the context of cancer cell migration.

    Results

    Contrary to expectations, our scratch assays indicated that HSV-HMGB1 did not significantly induce migration in CRC cells, suggesting that HMGB1 might not directly contribute to this process. Employing microarray analysis, we investigated gene expression changes linked to CRC cell migration, leading to construction of a Protein- Protein Interaction (PPI) network. This network revealed the presence of hub proteins, including as NDRG1, LGALS1, and ANGPTL4, which are recognized for their roles in cancer cell migration. The differential expression of these genes under hypoxic conditions was further validated using quantitative RT-PCR, aligning with the findings from our microarray data.

    Conclusion

    Our findings emphasize the complex regulation of CRC cell migration, and provides valuable insights into potential molecular mechanisms and pathways. These findings have implications for further research into cancer progression and the development of therapeutic strategies.

    Keywords: Colorectal neoplasms, Galectin 1, HMGB1 protein, Oncolytic virotherapy, Simplevirus, Tumor microenvironment
  • Belal Delshad, Hamed Abadijoo, Hossein Simaee, MohammadAli Khayamian, Mohammadreza Ghaderinia, Seyed Mojtaba Yazdanparast, Jalil Beheshti, Khosro Shamsi, Maryam Avatefi Afkham, Sepideh Mansouri, MohammadEsmaeil Akbari, Mohammad Abdolahad
    Background

    Intraoperative radiation therapy (IORT), is a promising method which has been widely applied in breast cancer lumpectomy. Although its effect on destructing remaining cancer cells was approved, maintaining or draining post intraoperative radiation therapy wound fluids (PIWF) is challenging.Moreover, the roles of immune cells in interaction with PIWFs have not been studied before which is the main investigation of this paper.

    Methods

    Surgical wound fluids were collected from 24 IDC patients one day afterlumpectomy. The patients were divided into control and IORT groups. The collected wound fluids were centrifuged for 20 minutes at 2000 rpm. The concentration of tumor-associated cytokines and inflammasomes were recorded using the immunoassays.

    Results

    PIWFs stimulate the residue of cancer cells in cavity sides causing disease progression. Here we have focused on the effect of PIWFs on the proactivation or deactivation of WBCs in the tumor bed environment. By sequential imaging in time-transient intervals from the interaction between WBCs and cancer cells, PIWFs have no additive proactivating effect on immune cells.

    Conclusion

    PIWFs have significant roles in proliferation of cancer cells but did not show an observable role in pro-activating immune cells against cancer cells. The functions of immune cells did not show any independent proactivation in the presence of PIWFs with respect to their activation in the presence of blood serum. It seems that draining the PIWFs may be required. In future research, we will use tumor samples of the patients instead of cell lines to better investigate the personalized immune-tumor interactions of patients.

    Keywords: Breast cancer, tumor microenvironment, Intraoperative radiation therapy, Immune system, cytokines
  • Jiaqi Zhong, Ying Kong, Ruming Li, Minghan Feng, Liming Li, Xiao Zhu, Lianzhou Chen
    Objective

    This paper aimed to investigate the PI3K/Akt/mTOR signal-pathway regulator factor-related lncRNA signatures (PAM-SRFLncSigs), associated with regulators of the indicated signaling pathway in patients with lung adenocarcinoma (LUAD) undergoing immunotherapy.

    Materials and Methods

    In this retrospective study, we employed univariate Cox, multivariate Cox, and least absolute shrinkage and selection operator (LASSO) regression analyses to identify prognostically relevant long non-coding RNAs (lncRNAs), construct prognostic models, and perform Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Subsequently, immunoassay and chemotherapy drug screening were conducted.Finally, the prognostic model was validated using the Imvigor210 cohort, and tumor stem cells were analyzed.

    Results

    We identified seven prognosis-related lncRNAs (AC084757.3, AC010999.2, LINC02802, AC026979.2, AC024896.1, LINC00941 and LINC01312). We also developed prognostic models to predict survival in patients with LUAD. KEGG enrichment analysis confirmed association of LUAD with the PI3K/Akt/mTOR signaling pathway. In the analysis of immune function pathways, we discovered three good prognostic pathways (Cytolytic_activity, Inflammation-promoting, T_cell_co-inhibition) in LUAD. Additionally, we screened 73 oncology chemotherapy drugs using the "pRRophetic" algorithm.

    Conclusion

    Identification of seven lncRNAs linked to regulators of the PI3K/Akt/mTOR signaling pathway provided valuable insights into predicting the prognosis of LUAD, understanding the immune microenvironment and optimizing immunotherapy strategies.

    Keywords: Immunotherapy, LncRNAs, Lung Adenocarcinoma, Prognosis, Tumor Microenvironment
  • Zanyar Pirkani, Fatemeh Kamalinejad, Yasser Zare, Seyyed Behnam Abdollahi Boraei*

    This review highlights the crucial role of polylactic acid (PLA)-based scaffolds in developing novel breast cancer treatment strategies. Despite advances in early detection and therapy, breast cancer remains a complex challenge with frequent resistance and relapse. Conventional treatments, while effective, have limitations such as restricted drug distribution and radiation toxicity. PLA scaffolds offer a promising alternative due to their biocompatibility and biodegradability, making them suitable for tissue engineering applications in oncology. The article examines the design and fabrication of PLA scaffolds that are not merely passive structures but play an active role in the therapeutic process. By tailoring their mechanical properties, these scaffolds can mimic the characteristics of actual breast tissue, creating a lifelike environment for studying cancer cell behavior. Furthermore, PLA scaffolds can mimic the tumor microenvironment, offering a three-dimensional representation that allows for a more accurate examination of tumor biology and treatment response. These scaffolds also function as advanced drug delivery systems, releasing therapeutic agents at the tumor site in a controlled manner, reducing systemic side effects, and enhancing drug efficacy. This review connects fundamental research with clinical practice, highlighting the revolutionary potential of PLA-based scaffolds in breast cancer management by mimicking the tumor microenvironment, delivering drugs locally, and enabling personalized treatment strategies.

    Keywords: Breast Cancer, Polylactic Acid, Scaffold, Tumor Microenvironment
  • سعید پیرمرادی*
    مقدمه

    سرطان همچنان یک مشکل عمده و پیشرو سلامت در سراسر جهان است. عدم تشخیص زودهنگام، مقاومت شیمیایی و عود سرطان به این معنی است که تحقیقات و توسعه گسترده در این زمینه مورد نیاز است.

    روش کار

    این مطالعه مروری بر اساس پروتکل پریزما و جستجو در پایگاه های بین المللی (PubMed، Google Scholar، Science) انجام شد. با توجه به استراتژی مورد نظر تمام مقالات منتشر شده به زبان انگلیسی تا سپامبر 2022با استفاده از کلمات کلیدی «میکرومحیط»، «سرطان» و «کاتپسین D» و ترکیب این کلمات جستجو و مقالات انتخاب شده با چک لیست PRISMA مورد ارزیابی قرار گرفتند.

    یافته ها

    پیچیدگی ریزمحیط تومور در محیط زیستی، چالش های بزرگ تری را برای درمان های ایمن، انتخابی و هدفمند داشتن ایجاد می کند. استراتژی های موجود مانند شیمی درمانی، رادیوتراپی و درمان های ضد رگ زایی، بقای بیماران را بدون پیشرفت بیماری و به طور متوسط بهبود می بخشند اما با این حال همگی انها با عوارض جانبی همراه هستند. بنابراین هدف قرار دادن کاندیداهای بالقوه در ریزمحیط، مانند کاتپسین Dخارج سلولی (CathD) که نقش مهمی در ایجاد سرطان سینه و تخمدان دارد می تواند پیشرفتی در درمان سرطان به ویژه با استفاده از روش های درمانی جدید مانند ایمونوتراپی و درمان مبتنی بر فناوری نانو باشد.

    نتیجه گیری

    در نهایت در این کارمطالعاتی مروری بر روی CathD نتیجه کلی بدینگونه بود که این ترکیب میتواند به عنوان یک عامل پیش سرطانی و به طور خاص یک عامل پیش رگ زایی که به صورت عملکردی در ریزمحیط تومور عمل می کند مورد بررسی دقیق محققین قرار گیرد. بدین خاطر راه های ممکن برای هدف قرار دادن این پروتئین به عنوان یک ترکیب هدف درمانی جدید در کارهای تحقیقاتی مبتنی بر فرایندهای ازمایشگاهی و بالینی مورد بحث وبررسی قرار می گیرد.

    کلید واژگان: میکرومحیط تومور، کاتپسین D، آنژیوژنز، MAP کینازها، ایمونوتراپی
    Saeed Pirmoradi*
    Introduction

    Cancer is still a major and leading health problem worldwide. Lack of early detection, chemoresistance and cancer recurrence mean that extensive research and development is needed in this area.

    Methodology

    This review study was conducted based on the Prism protocol and searching in international databases (PubMed, Google Scholar, Science). According to the desired strategy, all articles published in English until September 2022 were evaluated using the keywords "microenvironment", "cancer" and "cathepsin D" and the combination of these search words and selected articles with the PRISMA checklist.

    Findings

    The complexity of the tumor microenvironment in the biological environment creates greater challenges for safe, selective and targeted treatments. Existing strategies such as chemotherapy, radiotherapy and anti-angiogenic treatments improve the survival of patients without disease progression and moderately, however, they are all associated with side effects. Therefore, targeting potential candidates in the microenvironment, such as extracellular cathepsin D (CathD), which plays an important role in the development of breast and ovarian cancer, can be a breakthrough in cancer treatment, especially with the use of new therapeutic methods such as immunotherapy and nanotechnology-based therapy.

    Conclusion

    Finally, in this review study on CathD, the general conclusion was that this compound can be carefully investigated by researchers as a pro-cancer agent and specifically as a pro-angiogenic agent that functions functionally in the tumor microenvironment. take Therefore, possible ways to target this protein as a new therapeutic target compound are discussed and reviewed in research works based on laboratory and clinical processes

    Keywords: Tumor Microenvironment, Cathepsin D, Angiogenesis, MAP Kinases, Immunotherapy
  • MohammadAli Nilforoushzadeh, Hanieh Azizi, Ehsan Taghiabadi *, MohammadAmir Amirkhani
  • Nazila Jalilzadeh, Neda Barzgar Barough, Mehrdad Karami, Amir Baghbanzadeh, Kobra Velaei*
    Introduction

     Understanding the key role of the tumor microenvironment in specifying molecular markers of breast cancer subtypes is of a high importance in diagnosis and treatment. Therefore, the possibility of interconversion of luminal states and their specific markers alteration under the control of tumor microenvironment (TME), particularly cancer-associated fibroblasts (CAFs) deserves to be further investigated.

    Methods

     To activate normal human fibroblasts, liquid overlay technique or nemosis was used and α-SMA protein expression, CAFs marker, in fibroblastic spheroids was measured by blotting. The luminal A, MCF-7, and luminal B, MDA-MB 361, cell lines were treated with normal and spheroidal/activated fibroblast conditioned medium for 48 hours. The morphological changes of both luminal A and B cells were evaluated by invert light microscopy and analyzed through the shape factor formula. Moreover, chemo-sensitivity, proliferation, and changes in ER-related and proliferative genes expression levels were assessed respectively via MTT assay, Ki67 expression Immunofluorescence assay, real time PCR and Annexin V-FITC techniques.

    Results

     Activated (spheroidal) fibroblasts, expressed αSMA marker two folds more than monolayer cultured fibroblasts. Our study indicated a significant increase in IC50 of both luminal A and B cell lines after being treated with conditioned medium particularly in treated group with spheroidal conditioned medium. Studying Morphological changes using shape factor formula demonstrated more aggressiveness with gaining mesenchymal features in both luminal A and B subtypes by increasing exposure time. Changes in the expression of Ki67 were observed following treatment with fibroblastic and spheroidal paracrine secretome. Driven Data from Ki67 assay supports the luminal A and B interconversion by elevated Ki67 expression in luminal A and lowered Ki67 expression in luminal B. Gene expression analysis revealed that anti-apoptotic Bcl2 gene expression in both luminal types treated with condition medium has been increased though there has seen no interchange in expression of ER-related and proliferative genes between luminal A (MCF7) and luminal B (MDA-MB361) subtypes, the results of Annexin V-FITC flow cytometry test indicated a decrease in the population of both early and late apoptotic cells in groups treated with both fibroblastic and spheroidal condition medium compared to of control group.

    Conclusion

     Under the paracrine influence of fibroblast cells, both luminal A (MCF7) and luminal B (MDA-MB) subtypes of breast cancer gained invasive, anti-apoptotic, and chemoresistance features which are mostly increased by activated(spheroidal) fibroblasts conditioned medium mimicking CAFs. There was no strong proof for interconversion of luminal A and luminal B which share more similarities among breast cancer molecular subtypes.

    Keywords: Subtype, Breast cancer, Tumor microenvironment, Cancer-associated fibroblast
  • Arooba Farman Farman*

    Non-melanoma skin cancer is a serious malignancy and white-skinned people are highly susceptible to this cancer. About 918 deaths occurred due to NMSC in the UK following the year 2018-2019. The incidence of NMSC is 18-20 times higher as compared to melanoma skin cancer. The tumor immune microenvironment of NMSC possesses a diversity of immune cells that exert pro-tumor and anti-tumor effects on the TIME. So by recognizing the tumor-promoting entities, the TIME can be remodeled. Immunotherapy provides such a treatment that activates the person’s immune system to fight against tumorigenic cells. Radiotherapy also causes the modulation of the immune system and increases the anti-tumor responses in patients. The use of immune checkpoint inhibitors after radiotherapy has produced significant survival rates in patients. Oncolytic virus therapy is a subtype of immunotherapy with a positive response in the treatment of cancer. The synthetic viral promoter is highly specific to tumor and the introduction of transgenes help them to inhibit the tumor-promoting cells and make the tumor susceptible to anti-tumor cells, thus helping the tumor to eliminate from the body. This characteristic of oncolytic virus converts the ‘‘cold TIME’’ to ‘‘hot TIME’’ which exerts a highly positive response when used ICIs. In this article, a literature review is conducted to study the role of TIME in the progression of cancer and various methods that remodel the TIME such as immunotherapy, radiotherapy, and oncolytic viruses that might help to treat NMSC.

    Keywords: Skin cancer, Tumor microenvironment, Immunotherapy, Oncolytic viruses, Radiotherapy
  • Sepehr Torabinejad, Shokouhozaman Soleymanifard *, Sima Sayyah, Fatemeh Behnam Rasouli
    Background
    Surgery and radiotherapy are two main modalities of breast cancer treatment. However, surgery affects the tumor microenvironment negatively and promotes the growth of possible malignant cells remaining in the tumor bed.
    Objective
    The present study aimed to investigate the effects of intraoperative radiotherapy (IORT) on the tumor microenvironment. Therefore, the effect of surgical wound fluid (WF), collected from operated and irradiated patients on the growth and motility of a breast cancer cell line (MCF-7) was assessed.
    Material and Methods
    In this experimental study, preoperative blood serum (PS) and secreted WF from 18 patients who underwent breast-conserving surgery (IORT-) and 19 patients who received IORT following surgery (IORT+) were collected. The samples were purified and added to MCF-7 cultures. Two groups of the cells were treated with and without fetal bovine serum (FBS) and used as positive and negative controls. Applying 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and scratch wound healing assays, the growth and motility of MCF-7 cells were measured.
    Results
    Cell growth of the cells receiving WF from IORT+ patients (WF+) was statistically higher than the corresponding values of the cells received PS or WF from IORT- patients (WF-) (P<0.01). Both WF+ and WF- decreased the cells’ migration ability compared to PS (P<0.02) and FBS (P<0.002), although WF+ caused a more significant reduction (P<0.02). 
    Conclusion
    Wound fluid extracted from breast cancer patients who underwent both surgery and IORT increased the growth of breast tumor cells, but decreased their ability to migrate.
    Keywords: Breast cancer, Intraoperative radiotherapy, Neoplasms, Radiotherapy, Adjuvant, Tumor Microenvironment
  • Mehrnoosh Pashei, Farahnaz Ghahremanfard, Ehsan Manouchehri Doulabi, Maral Hemmati, Fatemeh Pak, Parviz Kokhaei

    T cell dysregulation and shift to T helper 2 responses, boosting tumor microenvironment support, contributes to the survival of leukemic B cells in Chronic Lymphocytic Leukemia. Interleukin (IL)-25 is involved in the initiation of T helper 2 cell responses. Signal transduction of IL-25 begins with the heterodimer receptor (IL-17RA/IL-17RB). The presence of IL-25 in the tumor microenvironment may affect the supportive effects of T cells in the surrounding tumor cell environment. The purpose of this study was to evaluate the role of IL-25 in the biology of CLL. IL-17RB expression in CD3+ and CD19+ cells was assessed in isolated peripheral blood mononuclear cells (PBMCs) of nine CLL patients and nine healthy subjects by real-time polymerase chain reaction and flow cytometry. B cells were positively enriched from PBMCs using magnetic-activated cell sorting (MACS). PBMCs and purified leukemic B cells were cultured with recombinant human IL-25 (20ng/ml) for 72 hours, then the viability and apoptosis of cultured cells were measured by MTT assay and AnnexinV/7AAD. Furthermore, the levels of CD69 expression on T lymphocytes and IL-17RB in T and B cells were determined by flow cytometry. The basal level of IL-17RB expression in CLL patients was significantly higher than that in control individuals. In addition, the percentage of IL-17RB+/CD3+, IL-17RB+/CD19+ cells and CD69+/CD3+ cells increased after 72 hours of culture with IL-25 in CLL patients compared to healthy subjects. IL-25 also reduces the apoptosis rate of tumor cells. We found that IL-25 could stimulate T cells in CLL patients and lower B cell death. This suggests that IL-25 might have a role in enhancing the survival of tumor cell by expressing receptors for inflammation, such as IL-17RB, and might be involved in the development of CLL.

    Keywords: Chronic lymphocytic leukemia, Interleukin-25, Tumor microenvironment, T helper 2 cells
  • Elham Shokati, _ Elahe Safari *, Morteza Motalebnezhad

    Astragalus polysaccharide (APS) is a functional component of Astragalus membranaceus with antitumor and immunomodulatory properties. This study evaluated the effect of APS on the peripheral blood mononuclear cell (PBMC) proliferation, cytokine secretion, and regulatory T cell (Treg) induction in an in vitro coculture model of human PBMCs and A2780 human ovarian cancer cells.
    PBMC proliferation and Treg frequency were measured by flow cytometry. Cytokine levels were assessed by enzyme-linked immunosorbent assay.
    APS significantly enhanced the PBMC proliferation, reduced Treg frequency, decreased anti-inflammatory cytokines including interleukin [IL]-10, transforming growth factor beta (TGF-β), and vascular endothelial growth factor-A (VEGF-A), and increased the pro-inflammatory cytokine IL-6.
    These findings suggest that APS may be an effective immunomodulatory supplement for cancer therapy, particularly for ovarian cancer by enhancing antitumor immune responses.

    Keywords: Astragalus polysaccharides, Inflammatory cytokines, Ovarian cancer, Regulatory T cells, Tumor microenvironment
نکته
  • نتایج بر اساس تاریخ انتشار مرتب شده‌اند.
  • کلیدواژه مورد نظر شما تنها در فیلد کلیدواژگان مقالات جستجو شده‌است. به منظور حذف نتایج غیر مرتبط، جستجو تنها در مقالات مجلاتی انجام شده که با مجله ماخذ هم موضوع هستند.
  • در صورتی که می‌خواهید جستجو را در همه موضوعات و با شرایط دیگر تکرار کنید به صفحه جستجوی پیشرفته مجلات مراجعه کنید.
درخواست پشتیبانی - گزارش اشکال