Gastrointestinal stromal tumors (GIST) are among the most common soft tissue tumors in the digestive system, typically arising from specialized cells known as interstitial cells of Cajal. These tumors are characterized by their formation due to genetic mutations, most of which are associated with the KIT and PDGFRA genes. There are various therapeutic strategies available for treating GIST, but drug resistance poses a major challenge. In this article, we address the role of immune cells infiltrating the tumors, specifically macrophages, their interaction with tumor cells, and how this understanding can aid in the development of new therapeutic strategies. We will also present how these immune interactions influence disease progression and recurrence risks. Join us in exploring this complex relationship between immune cells and tumors and the potential benefits that may arise from it.
Understanding Gastrointestinal Stromal Tumors
Gastrointestinal stromal tumor, commonly abbreviated as GIST, is a prevalent form of mesenchymal tumors in the digestive tract. This type of tumor develops from specialized cells known as interstitial cells of Cajal found in the intestinal wall. The treatment approach for this tumor largely depends on its biological characteristics, including genetic mutation. Two of the most commonly identified mutations associated with GIST are the mutation in the KIT gene (75-80%) and mutations in PDGFRA (5-10%). GIST is problematic due to its high rate of recurrence after treatment, necessitating modern therapeutic strategies.
Risk stratification based on tumor size, location, and cellular mitotic count is considered an important tool for predicting the likelihood of disease recurrence. The drug imatinib, a tyrosine kinase inhibitor, is typically used as the first-line treatment for patients with advanced or recurrent tumors. However, the problem of drug resistance appears to be increasing, highlighting the need for new treatment strategies for GIST. The importance of comprehensive studies on the relationship between infiltrating immune cells and tumor progression has also been emphasized.
Immune Response and Infiltrating Cell Relationship
The immune response to gastrointestinal tumors involves the unique experience of infiltrating immune cells, particularly macrophages and T-cells. Recent studies reveal that a substantial presence of these cells can play a crucial role in tumor development and treatment outcomes. For instance, results from single-cell genomic research have shown that various immune cells influence tumor development by creating complex interactions within the tumor-adjacent microenvironment.
Among the key findings was the focus on the MIF/CXCR4 axis, which represents the most common interaction between immune cells and tumor cells. As the risks of disease recurrence increased, levels of immune markers such as CD68, CD206, MIF, and CXCR4 rose. Experimental data demonstrated that cancer cells, such as GIST882 cells, could modulate macrophage responses by secreting MIF, leading to increased phenotypic changes in macrophages towards an M2-like pattern. This opens the door for exploring new immunotherapeutic strategies to enhance therapeutic efficacy.
Modern Techniques Used in Research
The scientific techniques employed to study GIST and the interactions between immune cells vary. One of the prominent methods is single-cell RNA sequencing, which allows for detailed information gathering about different cell types within the tumor microenvironment. This technique has been utilized in the described study, where samples from patients with GIST were examined. The processing of capturing enzymes, genomic molecule isolation, and obtaining accurate results were part of the experimental protocol.
Furthermore, tissue arrays and immunological techniques have been used to identify and classify cells within the tumor microenvironment. This type of analysis reflects how immune networks are formed and the location of active cells, such as T-cell macrophages, within the tumor tissue. Experts consider this an important indicator for understanding how the surrounding environment influences tumor behavior.
Implications
Clinical Outcomes and Future Research
Understanding the role of infiltrating immune cells in GIST is a pivotal step for the development of new therapeutic strategies. Studies show that immune cells are not just visible as defenses against tumors but also as factors influencing disease pathways and progression. Researchers now have the opportunity to understand how to enhance immune formation and reduce tumor response by targeting complex pathways.
These new research directions require more clinical studies to verify the effectiveness of immunotherapies and consider combination strategies, such as integrating immune inhibition with traditional therapies for better outcomes. Scientists emphasize the need to focus on providing personalized treatments that take into account the complex interactions between tumor cells and the surrounding immunity. This deep understanding is likely to contribute to significant advancements in the management and treatment of gastrointestinal sarcoma.
Analysis of Migrating Cells in Soft Tissue Cancer
The findings from single-cell analysis using RNA sequencing highlighted the cellular biodiversity within different tumors. By analyzing samples from two tumors, CD8+ T cells, macrophages, and cancer cells were among the prominent cells observed. In the G01 sample, cancer cells constituted 55.1% of the total cell count, while in the G02 sample, this percentage dropped to 27.5%. These results suggest that the variation in cell composition may play a crucial role in determining tumor function and response to treatment.
The study also showed that the number of infiltrating immune cells in the samples was significant, with a high concentration of CD8+ cells in G02, indicating a stronger immune response in low-risk tumors. On the other hand, G01 results showed a marked increase in the number of macrophages, which may indicate a pro-inflammatory environment promoting tumor growth. Overall, these differences highlight the importance of understanding the tumor microenvironment and how it influences disease progression.
Cell Interactions in Cancer Tissues
Cell interaction analysis using CellChat was employed to determine how cells communicate within cancer tissues. Seven cell types were identified, and the inputs and outputs for each cell type were analyzed through ligand-receptor partnership models. Tumor cells were the most interactive with macrophages, supporting the hypothesis that this interaction may contribute to sustained growth and cancer progression.
It was identified that the MIF/CXCR4 signaling axis represents one of the most important communication pathways between cancer and macrophage cells. This partnership was studied in both G01 and G02, where it was found that these signals play a crucial role in controlling the immune body’s response to the tumor. Enhancing cell interactions could facilitate an anti-tumor immune response if the balance is correct, but in situations where macrophages dominate, it can lead to negative outcomes that may include control of tumor growth.
Role of Immune Cells in Tumor Progression and Cancer Characteristics
The immunological characterization of tumors is a key approach to understanding how immune cells can impact tumor development. In the study, immunohistochemistry was used to identify levels of CD8+ and CD68+ cells. High levels of CD8+ cells were found in low-risk tumors, while macrophage counts significantly increased with higher risk levels.
The findings suggest that increasing macrophage presence may reflect a replacement mechanism among immune cells, where high-risk tumors show an interest in evading immune response. This is in line with previous studies indicating that macrophages associated with certain tumors may contribute to suppressing effective immune mechanisms and enhancing the tumor growth environment.
Impact
MIF/CXCR4 axis in immune cell differentiation
Highlighting the MIF/CXCR4 signaling pathway in cellular changes raises further questions about how these signals operate. In laboratory experiments, GIST-882 cells demonstrated the ability to secrete MIF, and showed an increase in differentiation components towards M2 cells. It was noted that this effect could be diminished with the use of MIF inhibitors, suggesting that balancing the immune environment using these signals may be key to effective therapy.
There has been inquiry into how this understanding can be utilized to improve therapeutic strategies for GIST patients, who face challenges in their treatment, including drug resistance. The use of MIF inhibitors can be viewed as a means to enhance immune response balance and identify the precise characteristics of each type of cancer, which may ultimately lead to increased efficacy of current therapies.
Summary and Future Analysis
The results indicate the importance of detailing and delineating the individual role of each type of cell in tumors, as risk classifications and the distribution of macrophages and related immune cells strongly overlap with disease progression. This is not only in terms of cell numbers but also in how these cells can positively or negatively influence immune responses. A more complex understanding of these dynamics could help in developing new therapeutic strategies based on modulating the tumor microenvironment and tailoring treatments more accurately. This research represents an important step towards understanding the interaction between immunotherapy and the tumor microenvironment, representing a significant direction for future research in this field.
Tumor Progression and Cellular Interactions
Tumors are complex diseases that depend on the interactions of the tumor environment with immune cells and neighboring cells. Tumor progression is highlighted in the context of two types of immune response: the inflammatory M1 type and the tumor-promoting M2 type. Studies indicate that the M1 state contributes to an effective immune response against cancer cells, while the M2 state promotes tumor progression. However, the environmental roots of these two states and understanding the supportive mechanisms for progression remain subjects of intensive research.
Research has shown that PD-1 expression is present in M2 macrophages associated with tumors, suggesting that these macrophages may contribute to increased tumor progression. By analyzing CD206 expression, the highest level of expression was found in high-risk tumors. However, the reason for the increased M2 macrophages in high-risk cases remains unclear, warranting further research to understand the relationship between macrophage gene expression and tumor progression.
Modern methods such as CellChat analysis have been utilized to understand the cellular communications between tumor cells and macrophages. The results indicated that the MIF/CXCR4 axis is a primary mode of communication, where high levels of MIF are associated with tumor development in various cancer types. It was discovered that MIF can act on cells in an autocrine or paracrine manner, leading to changes in physiological functions.
MIF/CXCR4 axis and the progression of cancer types
The MIF/CXCR4 axis is considered a critical pathway in tumor development, associated with various negative effects in tumors. Studies have shown that high expression levels of both MIF and CXCR4 are linked to an increased risk of tumor recurrence. It not only contributes to drug resistance but also plays a role in the metastatic invasion of tumors.
Through laboratory experiments, it was found that GIST882 cells can secrete MIF, enhancing M2 macrophage activity. Results indicated that high expression levels of both components are negative prognostic indicators, highlighting the importance of this axis in cancer progression. Although the precise mechanism of the MIF/CXCR4 axis remains not fully understood, the results suggest a need for further research to understand how these components influence tumor development.
From
During clinical studies, it was confirmed that elevated levels of MIF indicate a deterioration in the disease condition. As a result, targeting the MIF/CXCR4 axis is being considered as a new therapeutic strategy. This strategy may provide new hope in improving current treatments by offering a deeper understanding of how cancer cells interact with their surrounding immune environment.
Macrophages and the Immune Environment in GIST Tumors
Macrophages are key immune cells that play an important role in shaping the tumor microenvironment. M2 macrophages are the most prevalent in GIST tumors, as they can modify the immune response in favor of tumor progression. Research has shown that these macrophages contribute to creating an immunosuppressive environment, facilitating tumor cell escape and progression.
Studies in this field indicate that an increased number of M2 macrophages in the tumor is directly associated with increased disease severity and recurrence rates. By understanding how M2 macrophages are recruited, ways can be explored to modify this response to make it more beneficial in combating cancer. This knowledge may be crucial for developing new therapeutic strategies targeting the balance of immune response in tumors.
Understanding the dynamics of macrophages and their interaction with tumor cells provides new insights into the cellular mechanisms that support tumor growth and immune responses. This understanding may contribute to providing advanced therapeutic strategies that target the immune microenvironment of the tumor, potentially leading to improved treatment outcomes for patients with complex tumors such as GIST.
Gastrointestinal Stromal Tumor: Understanding Genetic and Pathological Factors
Gastrointestinal stromal tumor, also known as GIST, is a type of tumor that arises in the fibrous tissues of the digestive system. This type of cancer is characterized by the presence of genetic mutations, most notably mutations in the KIT and PDGFRA genes. These mutations are major factors contributing to tumor development and increasing its aggressiveness. In several cases, KIT mutations exhibit increased reactivity, facilitating the growth and spread of cancer cells. By the time diagnosis of GIST occurs, it is often associated with high recurrence rates, making it an important subject for research and development of treatment strategies.
Although targeted therapy using tyrosine kinase inhibitors like imatinib is the primary treatment, drug resistance poses a significant challenge. Statistics show that up to 50% of patients develop resistance to treatment over time. Hence, there is an increasing need to develop new therapies that take into account the tumor’s microstructure and its interactions with immune cells.
The Interaction Between Tumor Cells and the Immune System
In recent years, there has been an increase in studies examining how tumor cells interact with immune cells, particularly those associated with tumors like macrophages. Immune checkpoints play a vital role in how tumors respond to treatments, and predictive markers such as PD-1/PD-L1 and the diversity of T cells are key factors in this. Research has shown a significant presence of infiltrating immune cells in the tumor microenvironment of GIST, suggesting that immune responses may be impaired in many patients. While cells like CD8+ T can activate the immune response, they can become exhausted as the disease progresses, leading to an increase in macrophages that may contribute to tumor progression.
Moreover, macrophages are believed to participate in regulating the environments around tumors, and studies have shown that the M2 macrophage polarization pattern contributes to tumor progression and adaptation to the responsive immune environment. Therefore, understanding the role of macrophages and their strategies in immune evasion may be key to developing more effective treatments for GIST.
Strategies
The New Treatment: Combining Antibodies and Immunotherapy
In parallel with research concerning targeted therapy, unmodified antibodies, such as those targeting the immune checkpoints PD-1 and CTLA-4, have begun to gain attention as potential treatments in many tumors. The use of these therapies has led to improved survival rates in some cases, but not all patients respond to them. Factors such as the tumor’s mutational burden and deviations in the immune system may help improve treatment options for the patient.
It’s worth noting that combining immunotherapies with targeted therapy may represent a step towards improving outcomes in patients with GIST. This requires expanding the knowledge base regarding patient responses to treatment, how the immune system interacts with tumors, and how therapies can be effectively integrated.
Future Research and Treatment Administration
The rapid changes in research aspects concerning GIST treatment and genetics indicate the beginning of a new chapter in clinical medicine. Research focuses on new strategies, such as utilizing single-cell RNA sequencing technology to determine how immune cells interact with tumor cells. This helps in understanding the intricate dynamics of their relationship and offers new opportunities for developing treatments.
The shift towards personalizing treatment based on the genetic composition of the patient and identifying individual patterns of response to treatment marks a turning point in how GIST and immunotherapy are managed. The focus on improving health outcomes for patients shows positive steps towards achieving better results. Therefore, investment in research and developing new strategies will help improve healing rates and reduce recurrence rates.
Re-excision of Benign Intestinal Tumors and the Importance of Immunological Test Results
Case G02 underwent endoscopic tumor removal, where immunohistochemical test results showed that CD117, DOG-1, and CD34 were positive. These antigens are recognized as key indicators of the presence of gastrointestinal tumors known as gastrointestinal stromal tumors (GIST). On the other hand, case G01 presented a tumor with a maximum diameter of 10 cm, resulting in a high mitotic rate of 12 per 50 fields of view, indicating the severity of the case. We can conclude based on the information that tumor size and nature of the mitotic index help physicians classify tumors and understand potential risks for patients. In comparison, the tumor size in case G02 was small, measuring only 2 cm, with a relatively low mitotic rate, classified as very low risk, opening the door for more hopeful and effective therapeutic strategies.
Laboratory Methods in Analyzing GIST Tumors
Tumor tissues were obtained from resection samples and cut into small pieces. These pieces were digested using collagenase IV, an enzyme used in tissue processing to release the cells. Subsequently, a cell filter was used to sort the cells, and their viability was measured using the Countess II Automated Cell Counter. These steps are essential for forming genetic libraries from individual cells, as each cell processed individually provides an accurate picture of cellular diversity.
The complete process involves using the 10X Chromium Single Cell Platform, which facilitates cell sequencing and provides accurate genetic information. Here are important stages in this process: generating gel beads in emulsion, designing barcodes, and cleaning GEM-RT. The sequencing library is then prepared, and its concentration is measured using Qubit before being run on the Illumina NovaSeq 6000 for gene sequencing analysis.
Data quality analysis was strictly applied, excluding any data that did not meet certain criteria such as RNA counts lower than 500 or greater than 98% of the sample cells. This meticulous selection ensures the provision of reliable data on gene expression, making the results practical and applicable in clinical studies. The resulting data reflect the cellular composition in tumor tissues and provide comprehensive insights into patient characteristics and the impact of tumors on the immune system.
Analysis
Cell Expression Using Miniature Tissues
A miniature tissue array of 5-micron thickness was created from wax-embedded tumor tissues collected over nine years. The inclusion criteria involved patients diagnosed solely with GIST tumors, which makes it a tightly studied group. Patients who were treated with targeted therapies or those whose tumors did not occur in the stomach or intestine were excluded. Ultimately, the study included 80 patients, and various immune analyses were performed on them, allowing researchers to understand how different cell types interact in GIST tumors.
Techniques such as immunohistochemistry and fluorescence immunology were used, relying on different antibodies to detect the expression of various antigens. Software like ImageJ was utilized to scan the stained slide and count positive cells to reveal the results. These results enhance the understanding of the role of different immune cells in tumor development and how proteins such as CD8, CD68, and CD206 can contribute to the identification of different tumor types and their stages of progression.
Data analysis showed a clear variation in cell expression among different risk groups, indicating that immune cell interactions play a vital role in tumor response and the body’s response to treatment. Understanding these dynamics is crucial for developing better treatment strategies.
Cellular Networks and Cell Communication in the Tumor Environment
Cell inflammation and relationships among them are fundamental topics in the context of tumors. Seven cell types were identified through the analysis, and the use of the CellChat tool contributed to providing quantitative analyses of cell interactions in depth. The results showed that interactions between tumor cells and macrophages were the most common, indicating the importance of these relationships in the tumor environment. The cellular networks among different cells contribute to enhancing the microbial environment surrounding the tumor and influence tumor development and growth.
The analysis showed that the MIF/CXCR4 axis was the most frequently observed between tumor cells and macrophages in both G01 and G02 samples. This highlights an important model of cell interaction, as immune cells enhance responses to GIST tumors, emphasizing the importance of collaboration between immune cells and cancer cells. As the patient’s body responds, enhancing the understanding of these relationships opens new avenues for cancer treatment by targeting these axes.
Additionally, these complex networks of communication between cells represent an opportunity to explore potential therapies aimed at disrupting or enhancing these interactions, which could help improve tumor management and future treatments.
Gene Expression and Its Impact on Tumor Progression
The expression of genes and receptors among tumor cells plays a pivotal role in tumor growth and spread. The MIF/CXCR4 signaling axis was chosen as a primary subject for further investigation in the study, encompassing interactions between tumor cells and immune cells. The data showed that tumors with higher levels of this signaling were associated with an increased risk of tumor progression.
Through immunoassays, the expression of CD8, CD68, and CD206 was analyzed in different risk groups. The results showed that CD8+ T cells were most abundant in the low-risk group, while CD68+ macrophages were most common in the high-risk group. This indicates that the surrounding immune environment can vary based on the distinct characteristics of the tumor, highlighting the necessity of understanding these dynamics to enhance treatment strategies.
This information is highly valuable for developing immunotherapies, where targeted drugs can be tailored to manage the interaction of immune cells with tumors based on unique differences between these two cell types. Focusing on the balance between activating immunity and inhibiting cancer growth may be key to developing more effective treatments.
Expression
About MIF and CXCR4 in Tumor Growth
The expression levels of MIF and CXCR4 are important factors in the study of tumor growth, with results showing that the expression of both proteins is significantly associated with disease progression. In the case of GIST, the highest expression levels were identified in high-risk categories of tumors, indicating that increased expression of these proteins correlates with a deterioration in health status and exacerbation of the disease. Quantitative analyses revealed statistically significant differences among different groups, where the expression levels of MIF and CXCR4 reflected the progression of cases from low to high risk. These findings are particularly significant as they provide evidence of the clear role these proteins play in the diagnosis and assessment of tumor severity. This phenomenon supports the notion that high expression of CXCR4 and MIF may indicate increased cancer activity and thus could be considered a biomarker in treatment decision-making.
The Role of Immune Cells in Gastrointestinal Tumors
Immune cells, such as CD8+ T cells and CD68+ macrophages, are an important part of the tumor-associated microenvironment, playing a crucial role in monitoring and developing tumors. Studies have shown that an increase in the number of macrophages correlates with cancer progression due to a shift in immune response that leads to immune evasion against tumors. As the tumor advances, we observe that the relative quantity of CD8+ cells begins to decline while the number of macrophages increases, which may suggest a strategy of the tumor to evade immune response. The relationship between the status of M1 and M2 macrophages has also been discussed, as the balance of these states can significantly influence tumor behavior. For example, the M2 state may indicate the presence of a favorable environment for tumor progression and disease exacerbation, highlighting the importance of understanding how the immune system responds in accelerating tumor growth.
Mechanism of Action of the MIF/CXCR4 Axis and Its Impact on Immune Response
The mechanism of action of the MIF/CXCR4 axis is a significant topic in the search for new approaches to cancer treatment. This mechanism shows how tumor cells can influence immune cells, particularly macrophages. Current data indicate that MIF can modify macrophage responses, facilitating tumor advancement by promoting the M2 state that causes immune suppression. An ELISA assay was used to measure MIF secretion from tumor cell lines, providing evidence that GIST882 cells secrete MIF at concentrations that increase over time. This secretion helped in creating a suppressive immune environment around the tumor. Furthermore, the application of inhibitors such as ISO-1 and WZ811 demonstrated how the intervention of these chemicals affects M2 macrophage levels, reflecting the vital role of the MIF/CXCR4 axis in immune response alterations. These discoveries are an important step toward understanding how this axis can be targeted for potential immunotherapy in tumors.
Future Directions in Cancer Treatment by Targeting Immune Axes
Findings related to the MIF/CXCR4 axis open new horizons in the treatment of GIST tumors. With the increasing focus on immunotherapy, targeting this axis represents a promising strategy to enhance immune responses. By enhancing our understanding of the relationship between tumor cells and immunity, we can develop new drugs targeting these axes to inhibit cancer growth mechanisms. It is also essential to conduct in-depth studies on drugs that enhance the efficacy of immune cells or those that reduce the impact of the tumor environment, leading to improved clinical outcomes. Current data already show how targeted drug therapies may face resistance risks, necessitating further research to explore the possibilities of combining targeted therapies with immunotherapies. Future directions inspire hope for designing integrative treatment protocols focused on immune and molecular effects to improve outcomes for GIST patients.
Tumors
Sarcoma and Clinical Patterns
Sarcoma tumors are a group of tumors that affect fibrous tissues, such as muscles, bones, and fat. These tumors differ from cancer-like tumors in their causes and clinical patterns. For example, gastrointestinal sarcomas, such as gastrointestinal stromal tumors, are considered a specific type of tumor that occurs in the digestive system and poses unique treatment challenges. The early symptoms of these tumors are often vague and resemble other gastrointestinal diseases, leading to delays in diagnosis.
Differentiating the clinical patterns of sarcoma tumors contributes to a better understanding of how to treat patients. In this context, there is a focus on the use of targeted therapies, such as “Imatinib,” which is widely used for treating gastrointestinal stromal tumors. Studies have shown that Imatinib can help reduce tumor size and improve patients’ quality of life. However, examining the progression of these tumors and evaluating their genetic characteristics is essential for understanding treatment response.
Early Diagnosis and Treatment of Sarcoma Tumors
Early diagnosis of sarcoma tumors is vital in improving health outcomes for patients. Diagnosis requires the use of advanced imaging techniques, such as X-rays, MRI, and CT scans, which provide a detailed understanding of the tumor’s location and size. For example, X-ray examinations can help identify abnormal masses in the patient’s bones, while MRI contributes to assessing the surrounding soft tissue.
In addition to imaging techniques, cellular and biological examinations play a crucial role in diagnosing these tumors. Analyzing tissue samples is linked to examining molecular symptoms, such as mutations in the “KIT” and “PDGFRA” genes, as the presence of these mutations can provide important indicators of tumor type and its response to treatment. For example, the presence of mutations in the “KIT” gene is an indicator of the effectiveness of Imatinib as a first-line treatment, helping to guide appropriate treatment pathways.
Research and Recent Developments in Immunotherapy for Sarcoma Tumors
Research on immunotherapy for sarcoma tumors has gained increasing attention in recent years. One new aspect is targeting immune checkpoints, such as “PD-1” and “PD-L1,” which play a significant role in inhibiting the immune response against cancer cells. Various studies show that using inhibitors of these checkpoints may help restore inactive T-cell activity and enhance immune defense against tumors.
Immunotherapy is particularly effective in advanced sarcoma cases, where it can lead to long-lasting responses in patients who do not respond to conventional treatments. For example, recent studies have shown that combining “PD-1” inhibitors with treatments like “Imatinib” may significantly improve patient outcomes, highlighting the importance of combination therapy strategy.
Future Challenges in Sarcoma Tumor Management
Despite significant advances in understanding and treating sarcoma tumors, there are still challenges remaining. Among these challenges is the development of drug resistance, where the tumor may lose its responsiveness to treatment over time. Ongoing research aims to understand the molecular mechanisms behind this resistance, contributing to the development of new strategies to prevent negative drug turnover. Additionally, a better understanding of the role that the tumor microenvironment plays, including immune cells, is essential for achieving effective targeted therapies.
Sustainable management of sarcoma tumors also requires collaboration among various medical specialties, including oncologists, immunotherapy specialists, and radiologists, to create comprehensive treatment protocols suitable for different cases. Research indicates that personalized treatment plans that consider the genetic characteristics of the patient can contribute to improving the overall success rate of treatment.
Link
Source: https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2024.1431535/full
Artificial intelligence was used ezycontent
“`css
}@media screen and (max-width: 480px) {
.lwrp.link-whisper-related-posts{
}
.lwrp .lwrp-title{
}.lwrp .lwrp-description{
}
.lwrp .lwrp-list-multi-container{
flex-direction: column;
}
.lwrp .lwrp-list-multi-container ul.lwrp-list{
margin-top: 0px;
margin-bottom: 0px;
padding-top: 0px;
padding-bottom: 0px;
}
.lwrp .lwrp-list-double,
.lwrp .lwrp-list-triple{
width: 100%;
}
.lwrp .lwrp-list-row-container{
justify-content: initial;
flex-direction: column;
}
.lwrp .lwrp-list-row-container .lwrp-list-item{
width: 100%;
}
.lwrp .lwrp-list-item:not(.lwrp-no-posts-message-item){
“`
“`html
}
.lwrp .lwrp-list-item .lwrp-list-link .lwrp-list-link-title-text,
.lwrp .lwrp-list-item .lwrp-list-no-posts-message{
};
}
“`
Leave a Reply