Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
J Cell Mol Med ; 28(5): e18065, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38116696

ABSTRACT

Colorectal cancer (CRC) is the most prevalent malignancy of the digestive system. Glucose metabolism plays a crucial role in CRC development. However, the heterogeneity of glucose metabolic patterns in CRC is not well characterized. Here, we classified CRC into specific glucose metabolic subtypes and identified the key regulators. 2228 carbohydrate metabolism-related genes were screened out from the GeneCards database, 202 of them were identified as prognosis genes in the TCGA database. Based on the expression patterns of the 202 genes, three metabolic subtypes were obtained by the non-negative matrix factorization clustering method. The C1 subtype had the worst survival outcome and was characterized with higher immune cell infiltration and more activation in extracellular matrix pathways than the other two subtypes. The C2 subtype was the most prevalent in CRC and was characterized by low immune cell infiltration. The C3 subtype had the smallest number of individuals and had a better prognosis, with higher levels of NRF2 and TP53 pathway expression. Secreted frizzled-related protein 2 (SFRP2) and thrombospondin-2 (THBS2) were confirmed as biomarkers for the C1 subtype. Their expression levels were elevated in high glucose condition, while their knockdown inhibited migration and invasion of HCT 116 cells. The analysis of therapeutic potential found that the C1 subtype was more sensitive to immune and PI3K-Akt pathway inhibitors than the other subtypes. To sum up, this study revealed a novel glucose-related CRC subtype, characterized by SFRP2 and THBS2, with poor prognosis but possible therapeutic benefits from immune and targeted therapies.


Subject(s)
Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , Glucose , Transcriptome , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Glucose/metabolism , Transcriptome/genetics , Prognosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Thrombospondins/genetics , Thrombospondins/metabolism , Cell Movement/genetics , Gene Expression Profiling , HCT116 Cells , Signal Transduction , Membrane Proteins/genetics , Membrane Proteins/metabolism
2.
Int J Mol Sci ; 25(14)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39062908

ABSTRACT

Autoimmunity refers to an organism's immune response against its own healthy cells, tissues, or components, potentially leading to irreversible damage to vital organs. Central and peripheral tolerance mechanisms play crucial roles in preventing autoimmunity by eliminating self-reactive T and B cells. The disruption of immunological tolerance, characterized by the failure of these mechanisms, results in the aberrant activation of autoreactive lymphocytes that target self-tissues, culminating in the pathogenesis of autoimmune disorders. Genetic predispositions, environmental exposures, and immunoregulatory disturbances synergistically contribute to the susceptibility and initiation of autoimmune pathologies. Within the realm of immune therapies for autoimmune diseases, cytokine therapies have emerged as a specialized strategy, targeting cytokine-mediated regulatory pathways to rectify immunological imbalances. Proinflammatory cytokines are key players in inducing and propagating autoimmune inflammation, highlighting the potential of cytokine therapies in managing autoimmune conditions. This review discusses the etiology of autoimmune diseases, current therapeutic approaches, and prospects for future drug design.


Subject(s)
Autoimmune Diseases , Autoimmunity , Cytokines , Humans , Cytokines/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/therapy , Animals , Immune Tolerance
3.
Allergy ; 77(1): 100-110, 2022 01.
Article in English | MEDLINE | ID: mdl-34245591

ABSTRACT

Coronavirus disease-19 (COVID-19) is a complex disorder caused by the pandemic diffusion of a novel coronavirus named SARS-CoV-2. Clinical manifestations vary from silent infection to severe pneumonia, disseminated thrombosis, multi-organ failure, and death. COVID-19 pathogenesis is still not fully elucidated, while increasing evidence suggests that disease phenotypes are strongly related to the virus-induced immune system's dysregulation. Indeed, when the virus-host cross talk is out of control, the occurrence of an aberrant systemic inflammatory reaction, named "cytokine storm," leads to a detrimental impairment of the adaptive immune response. Dendritic cells (DCs) are the most potent antigen-presenting cells able to support innate immune and promote adaptive responses. Besides, DCs play a key role in the anti-viral defense. The aim of this review is to focus on DC involvement in SARS-CoV-2 infection to better understand pathogenesis and clinical behavior of COVID-19 and explore potential implications for immune-based therapy strategies.


Subject(s)
COVID-19 , Vaccines , Dendritic Cells , Humans , Pandemics , SARS-CoV-2
4.
Med Princ Pract ; 30(1): 1-16, 2021.
Article in English | MEDLINE | ID: mdl-32841942

ABSTRACT

Cancer immunotherapy, which aims to control the immune system to eradicate cancer cells and prevent their spread, needs to be personalized because anticancer immune responses can be inhibited in several ways that vary from patient to patient. Cancer immunotherapy includes pharmaceuticals such as immune checkpoint inhibitors and monoclonal antibodies (MAbs) as well as cell therapy, immunogene therapy, and vaccines. Combination of programmed cell death protein 1 (PD-1)/programmed cell death protein ligand 1 (PD-L1) drugs with other immunotherapy drugs, for example, antibody-drug conjugates, as well as combination of PD-1/PD-L1 drugs with other therapies, for example, chemotherapy and radiation therapy, are being explored. Biomarkers are important for predicting the response to immunotherapy. Molecular diagnostics and sequencing are important technologies for guiding treatment in immuno-oncology. Genomic profiling of tumor mutational burden may enhance the predictive utility of PD-L1 expression and facilitate personalized combination immunotherapy. Optimization of personalized immuno-oncology requires integration of several technologies and selection of those best suited for an individual patient. Advances in immuno-oncology are also attributed to technologies for targeted delivery of anticancer therapeutics such as antigen-capturing nanoparticles for precision targeting and selective delivery. A breakthrough in cell therapy of cancer is a chimeric antigen receptors-T cell, which combines the antigen-binding site of a MAb with the signal activating machinery of a T cell, freeing antigen recognition from major histocompatibility complex restriction. Gene-editing tools such as clustered regularly interspaced short palindromic repeats have a promising application for removing alloreactivity and decreasing immunogenicity of third-party T cells. In conclusion, personalized immuno-oncology is one of the most promising approaches to management of cancer.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , Precision Medicine/methods , Adjuvants, Immunologic/pharmacology , Adoptive Transfer/methods , Biomarkers, Tumor , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Combined Modality Therapy , Genomics/methods , Granulocytes/metabolism , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive/methods , Lab-On-A-Chip Devices , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Chimeric Antigen/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , Transcriptome
5.
Cytotherapy ; 22(11): 629-641, 2020 11.
Article in English | MEDLINE | ID: mdl-32778404

ABSTRACT

BACKGROUND: In recent years, therapies with CD4+CD25highFoxP3+ regulatory T cells (Tregs) have been successfully tested in many clinical trials. The important issue regarding the use of this treatment in autoimmune conditions remains the specificity toward particular antigen, as because of epitope spread, there are usually multiple causative autoantigens to be regulated in such conditions. METHODS: Here we show a method of generation of Tregs enriched with antigen-reactive clones that potentially covers the majority of such autoantigens. In our research, Tregs were expanded with anti-CD28 and anti-CD154 antibodies and autologous monocytes and loaded with a model peptide, such as whole insulin or insulin Ɵ chain peptide 9-23. The cells were then sorted into cells recognizing the presented antigen. The reactivity was verified with functional assays in which Tregs suppressed proliferation or interferon gamma production of autologous effector T cells (polyclonal and antigen-specific) used as responders challenged with the model peptide. Finally, we analyzed clonotype distribution and TRAV gene usage in the specific Tregs. RESULTS: Altogether, the applied technique had a good yield and allowed us to obtain a Treg product enriched with a specific subset, as confirmed in the functional tests. The product consisted of many clones; nevertheless, the content of these clones was different from that found in polyclonal or unspecific Tregs. CONCLUSIONS: The presented technique might be used to generate populations of Tregs enriched with cells reactive to any given peptide, which can be used as a cellular therapy medicinal product in antigen-targeted therapies.


Subject(s)
Antibodies/metabolism , CD28 Antigens/metabolism , CD40 Ligand/metabolism , Monocytes/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Proliferation , Cells, Cultured , Clone Cells , Forkhead Transcription Factors/metabolism , Humans , Interferon-gamma/metabolism , Receptors, Antigen, T-Cell/metabolism
6.
Semin Cancer Biol ; 52(Pt 2): 158-165, 2018 10.
Article in English | MEDLINE | ID: mdl-29307568

ABSTRACT

Immuno-therapeutics aim to activate the body's own immune system against cancer and are one of the most promising cancer treatment strategies, but currently limited by a variable response rate. Biomarkers may help to distinguish those patients most likely to respond to therapy; they may also help guide clinical decision making for combination therapies, dosing schedules, and determining progression versus relapse. However, there is a need to confirm such biomarkers in preferably prospective clinical trials before they can be used in practice. Accordingly, it is essential that clinical trials for immuno-therapeutics incorporate biomarkers. Here, focusing on the specific setting of immune therapies, we discuss both the scientific and logistical hurdles to identifying potential biomarkers and testing them in clinical trials.


Subject(s)
Biomarkers, Tumor/immunology , Neoplasms/immunology , Neoplasms/pathology , Animals , Clinical Trials as Topic , Humans , Medical Oncology/methods , Prognosis
8.
Immunology ; 155(2): 273-282, 2018 10.
Article in English | MEDLINE | ID: mdl-29772075

ABSTRACT

CD6 is a type I T-cell surface receptor that modulates antigen receptor signalling. Its activity is regulated by binding of its membrane proximal domain (domain 3) to a cell surface ligand, CD166. CD6 monoclonal antibodies (mAbs) specific for the membrane distal domain (domain 1) perturb CD6 function including itolizumab (Alzumab™), which has reached the clinic for treatment of autoimmune disease. We characterized molecular and functional properties of several CD6 mAbs including itolizumab to define potential mechanisms of action. Epitope mapping using the crystal structure of CD6 to design mutants identified two distinct binding sites on different faces of domain 1, one containing residue R77, crucial for MT605 and T12.1 binding and the other, E63, which is crucial for itolizumab and MEM98. Analysis of binding kinetics revealed that itolizumab has a lower affinity compared with other CD6 domain 1 mAbs. We compared potential agonistic (triggering) and antagonistic (blocking) properties of CD6 mAbs in assays where the mechanism of action was well defined. CD6 domain 1 and 3 mAbs were equally effective in triggering interleukin-2 production by a cell line expressing a chimeric antigen receptor containing the extracellular region of CD6. CD6 domain 1 mAbs hindered binding of multivalent immobilized CD166 but were inferior compared with blocking by soluble CD166 or a CD6 domain 3 mAb. Characterization of CD6 mAbs provides an insight into how their functional effects in vivo may be interpreted and their therapeutic use optimized.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Epitopes/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Antigens, CD/chemistry , Antigens, CD/genetics , Antigens, Differentiation, T-Lymphocyte/chemistry , Antigens, Differentiation, T-Lymphocyte/genetics , Cell Line, Tumor , Humans , Mice , Models, Molecular , Mutagenesis , Mutation , Protein Conformation , Protein Domains/immunology , Protein Interaction Domains and Motifs , Rats , Signal Transduction/drug effects
9.
Curr Oncol Rep ; 19(10): 66, 2017 Aug 12.
Article in English | MEDLINE | ID: mdl-28803390

ABSTRACT

PURPOSE OF REVIEW: The knowledge base of malignant cell growth and resulting targets is rapidly increasing every day. Clonal theory is essential to understand the changes required for a cell to become malignant. These changes are then clues to therapeutic intervention strategies. Immune system optimization is a critical piece to find, recognize, and eliminate all cancer cells from the host. Only by administering (1) multiple therapies that counteract the cancer cell's mutational and externally induced survival traits and (2) by augmenting the immune system to combat immune suppression processes and by enhancing specific tumor trait recognition can cancer begin to be treated with a truly targeted focus. RECENT FINDINGS: Since the sequencing of the human genome during the 1990s, steady progress in understanding genetic alterations and gene product functions are being unraveled. In cancer, this is proceeding very fast and demonstrates that genetic mutations occur very rapidly to allow for selection of survival traits within various cancer clones. Hundreds of mutations have been identified in single individual cancers, but spread across many clones in the patient's body. Precision oncology will require accurate measurement of these cancer survival-benefiting mutations to develop strategies for effective therapy. Inhibiting these cellular mechanisms is a first step, but these malignant cells need to be eliminated by the host's mechanisms, which we are learning to direct more specifically. Cancer is one of the most complicated cellular aberrations humans have encountered. Rapidly developing significant survival traits require prompt, repeated, and total body measurements of these attributes to effectively develop multi-agent treatment of the individual's malignancy. Focused drug development to inhibit these beneficial mutations is critical to slowing cancer cell growth and, perhaps, triggering apoptosis. In many cases, activation and targeting of the immune system to kill the remaining malignant cells is essential to a cure.


Subject(s)
Clonal Evolution/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Apoptosis/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Clonal Evolution/immunology , Humans , Mutation , Neoplasms/pathology
10.
Mult Scler ; 22(13): 1732-1740, 2016 11.
Article in English | MEDLINE | ID: mdl-22653657

ABSTRACT

BACKGROUND: Few studies have analysed long-term effects of immunomodulatory disease modifying drugs (DMDs). OBJECTIVE: Assessment of the efficacy of DMDs on long-term evolution of multiple sclerosis, using a Bayesian approach to overcome methodological problems related to open-label studies. METHODS: MS patients from three different Italian multiple sclerosis centres were divided into subgroups according to the presence of treatment in their disease history before the endpoint, which was represented by secondary progression. Patients were stratified on the basis of the risk score BREMS (Bayesian risk estimate for multiple sclerosis), which is able to predict the unfavourable long-term evolution of MS at an early stage. RESULTS: We analysed data from 1178 patients with a relapsing form of multiple sclerosis at onset and at least 10 years of disease duration, treated (59%) or untreated with DMDs. The risk of secondary progression was significantly lower in patients treated with DMDs, regardless of the initial prognosis predicted by BREMS. CONCLUSIONS: DMDs significantly reduce the risk of multiple sclerosis progression both in patients with initial high-risk and patients with initial low-risk. These findings reinforce the role of DMDs in modifying the natural course of the disease, suggesting that they have a positive effect not only on the inflammatory but also on the neurodegenerative process. The study also confirms the capability of the BREMS score to predict MS evolution.


Subject(s)
Disease Progression , Immunotherapy/methods , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Outcome Assessment, Health Care , Adult , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Severity of Illness Index
11.
Cancer Treat Res ; 169: 15-22, 2016.
Article in English | MEDLINE | ID: mdl-27696255

ABSTRACT

There has been great progress in the management and patient outcome in multiple myeloma due to the use of novel agents including immunomodulatory drugs and proteasome inhibitors; nonetheless, novel agents remain an urgent need. The three promising Achilles heals or vulnerabilities to be targetted in novel therapies include: protein degradation by the ubiquitin proteasome or aggresome pathways; restoring autologous antimyeloma immunity; and targeting aberrant biology resulting from constitutive and ongoing DNA damage in tumour cells. Scientifically based therapies targeting these vulnerabilities used early in the disease course, ie smouldering multiple myeloma, have the potential to significantly alter the natural history and transform myeloma into a chronic and potentially curable disease.


Subject(s)
Immunotherapy/trends , Molecular Targeted Therapy/trends , Multiple Myeloma/therapy , Animals , Humans
12.
Pharmacol Res ; 98: 69-75, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25014184

ABSTRACT

Islet transplantation has been demonstrated to improve glycometabolic control, to reduce hypoglycemic episodes and to halt the progression of diabetic complications. However, the exhaustion of islet function and the side effects related to chronic immunosuppression limit the spread of this technique. Consequently, new immunoregulatory protocols have been developed, with the aim to avoid the use of a life-time immunosuppression. Several approaches have been tested in preclinical models, and some are now under clinical evaluation. The development of new small molecules and new monoclonal or polyclonal antibodies is continuous and raises the possibility of targeting new costimulatory pathways or depleting particular cell types. The use of stem cells and regulatory T cells is underway to take advantage of their immunological properties and to induce tolerance. Xenograft islet transplantation, although having severe problems in terms of immunological compatibility, could theoretically provide an unlimited source of donors; using pigs carrying human immune antigens has showed indeed promising results. A completely different approach, the use of encapsulated islets, has been developed; synthetic structures are used to hide islet alloantigen from the immune system, thus preserving islet endocrine function. Once one of these strategies is demonstrated safe and effective, it will be possible to establish clinical islet transplantation as a treatment for patients with type 1 diabetes long before the onset of diabetic-related complications.


Subject(s)
Immunosuppression Therapy/methods , Immunosuppressive Agents/therapeutic use , Islets of Langerhans Transplantation/immunology , Islets of Langerhans Transplantation/methods , Animals , Diabetes Mellitus, Type 1/therapy , Heterografts , Humans , Swine , Transplantation, Autologous
13.
J Anesth Analg Crit Care ; 4(1): 28, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689337

ABSTRACT

BACKGROUND: In the last decades, several adjunctive treatments have been proposed to reduce mortality in septic shock patients. Unfortunately, mortality due to sepsis and septic shock remains elevated and NO trials evaluating adjunctive therapies were able to demonstrate any clear benefit. In light of the lack of evidence and conflicting results from previous studies, in this multidisciplinary consensus, the authors considered the rational, recent investigations and potential clinical benefits of targeted adjunctive therapies. METHODS: A panel of multidisciplinary experts defined clinical phenotypes, treatments and outcomes of greater interest in the field of adjunctive therapies for sepsis and septic shock. After an extensive systematic literature review, the appropriateness of each treatment for each clinical phenotype was determined using the modified RAND/UCLA appropriateness method. RESULTS: The consensus identified two distinct clinical phenotypes: patients with overwhelming shock and patients with immune paralysis. Six different adjunctive treatments were considered the most frequently used and promising: (i) corticosteroids, (ii) blood purification, (iii) immunoglobulins, (iv) granulocyte/monocyte colony-stimulating factor and (v) specific immune therapy (i.e. interferon-gamma, IL7 and AntiPD1). Agreement was achieved in 70% of the 25 clinical questions. CONCLUSIONS: Although clinical evidence is lacking, adjunctive therapies are often employed in the treatment of sepsis. To address this gap in knowledge, a panel of national experts has provided a structured consensus on the appropriate use of these treatments in clinical practice.

14.
Vet Med (Auckl) ; 15: 15-29, 2024.
Article in English | MEDLINE | ID: mdl-38371487

ABSTRACT

Atopic dermatitis (AD) is a common inflammatory and pruritic allergic skin disease in humans and dogs worldwide. The pathogenesis of AD is multifactorial, immunologically complex, and may involve genetic factors, epidermal barrier dysfunction, microbiome changes, immune dysregulation, and allergic sensitization. Across species, prevalence of AD is on the rise. At present, there is no cure for canine AD (CAD). The treatment for CAD is multifaceted and aimed at controlling the pruritus, associated inflammation, and infections, repairing the skin barrier function, and dietary management. This review presents data on prevalence, impact, and complex immunological interactions in AD with a focus on subsequent management of the disease in the canine population. A multimodal approach for management of CAD to address varying clinical signs and responses to therapies is discussed.

15.
Clin Immunol ; 149(3): 339-44, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23537861

ABSTRACT

Class II major histocompatibility molecules (MHC) confer disease risk for multiple autoimmune disorders including type 1 diabetes. The interaction between the components of the trimolecular complex (CD4(+) T cell receptors, self-peptide, and MHC class II molecules) plays a pivotal role in autoimmune disease pathogenesis. The development of therapies targeting various components of the trimolecular complex for the prevention of type 1 diabetes is actively being pursued. This review focuses on the components of the anti-insulin trimolecular complex, registers of insulin peptide binding to 'diabetogenic' MHC class II molecules, and therapies targeting each component of the trimolecular complex.


Subject(s)
Autoantigens/immunology , Diabetes Mellitus, Type 1/drug therapy , Histocompatibility Antigens Class II/immunology , Hypoglycemic Agents/therapeutic use , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , Amino Acid Motifs , Antibodies, Monoclonal/therapeutic use , Autoantigens/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Histocompatibility Antigens Class II/metabolism , Humans , Insulin/immunology , Insulin/metabolism , Molecular Sequence Data , Peptides/metabolism , Receptors, Antigen, T-Cell/metabolism
16.
Gynecol Oncol ; 130(3): 667-73, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23800697

ABSTRACT

While therapeutic vaccines for ovarian cancer represent only a small fraction of active clinical trials, growing interest in this area and the accumulated data supporting the use of vaccines in cancer treatment portend further expansion of trials incorporating these strategies. This review explores the rationale for the use of vaccines for the treatment of ovarian cancer. It examines vaccine platforms that have been investigated and reviews the data from these studies. We also highlight recently reported phase 2 and 3 clinical trials with clinical outcomes as endpoints. Finally, we consider directions for the next generation of vaccines in light of these findings and our emerging understanding of agents that may augment vaccine responses by targeting the immunosuppressive impact of the tumor microenvironment.


Subject(s)
Cancer Vaccines/therapeutic use , Ovarian Neoplasms/drug therapy , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Female , Humans , Treatment Outcome
17.
Expert Opin Ther Targets ; 27(12): 1177-1188, 2023.
Article in English | MEDLINE | ID: mdl-37997756

ABSTRACT

INTRODUCTION: Lysine-specific histone demethylase 1A (KDM1A/LSD1) has emerged as an important therapeutic target in various cancer types. LSD1 regulates a wide range of biological processes that influence cancer development, progression, metastasis, and therapy resistance. However, recent studies have revealed novel aspects of LSD1 biology, shedding light on its involvement in immunogenicity, antitumor immunity, and DNA damage response. These emerging findings have the potential to be leveraged in the design of effective LSD1-targeted therapies. AREAS COVERED: This paper discusses the latest developments in the field of LSD1 biology, focusing on its role in regulating immunogenicity, antitumor immunity, and DNA damage response mechanisms. The newfound understanding of these mechanisms has opened possibilities for the development of novel LSD1-targeted therapies for cancer treatment. Additionally, the paper provides an overview of LSD1 inhibitor-based combination therapies for the treatment of cancer. EXPERT OPINION: Exploiting LSD1 role in antitumor immunity and DNA damage response provides cues to not only understand the LSD1-resistant mechanisms but also rationally design new combination therapies that are more efficient and less toxic than monotherapy. The exploration of LSD1 biology and the development of LSD1-targeted therapies hold great promise for the future of cancer treatment.


Subject(s)
Lysine , Neoplasms , Humans , Neoplasms/pathology , Histone Demethylases
18.
Am J Cancer Res ; 13(6): 2426-2438, 2023.
Article in English | MEDLINE | ID: mdl-37424810

ABSTRACT

Lung cancer is the most common cancer type with poor prognosis. While G protein-coupled receptor 35 (GPR35) is a potent stimulator of tumor growth, group 2 innate lymphoid cells (ILC2) have shown dual effects in tumorigenesis. Intriguingly, inflammation induced GPR35 activation leads to an upregulation in the markers associated with ILC2. Here, we reported that GPR35 knockout mice exhibited a significantly reduced tumor growth and altered immune infiltration in tumors. Furthermore, activating GPR35 in different mouse models promoted tumor development by enhancing the production of IL-5 and IL-13, thereby facilitating the formation of the ILC2-MDSC axis. Moreover, we found that GPR35 was a poor prognostic factor in patients with lung adenocarcinoma. Together, our findings suggest the potential application of targeting GPR35 in cancer immunotherapy.

19.
Brain Sci ; 13(12)2023 Dec 09.
Article in English | MEDLINE | ID: mdl-38137148

ABSTRACT

Low-Grade Gliomas (LGGs) represent a diverse group of brain tumors originating from glial cells, characterized by their unique histopathological and molecular features. This article offers a comprehensive exploration of LGGs, shedding light on their subtypes, histological and molecular aspects. By delving into the World Health Organization's grading system, 5th edition, various specificities were added due to an in-depth understanding of emerging laboratory techniques, especially genomic analysis. Moreover, treatment modalities are extensively discussed. The degree of surgical resection should always be considered according to postoperative quality of life and cognitive status. Adjuvant therapies focused on chemotherapy and radiotherapy depend on tumor grading and invasiveness. In the current literature, emerging targeted molecular therapies are well discussed due to their succinctly therapeutic effect; in our article, those therapies are summarized based on posttreatment results and possible adverse effects. This review serves as a valuable resource for clinicians, researchers, and medical professionals aiming to deepen their knowledge on LGGs and enhance patient care.

20.
Front Immunol ; 13: 1088124, 2022.
Article in English | MEDLINE | ID: mdl-36618375

ABSTRACT

Huntington's disease (HD) is a progressive neurodegenerative disease characterized by preferential loss of neurons in the striatum in patients, which leads to motor and cognitive impairments and death that often occurs 10-15 years after the onset of symptoms. The expansion of a glutamine repeat (>36 glutamines) in the N-terminal region of huntingtin (HTT) has been defined as the cause of HD, but the mechanism underlying neuronal death remains unclear. Multiple mechanisms, including inflammation, may jointly contribute to HD pathogenesis. Altered inflammation response is evident even before the onset of classical symptoms of HD. In this review, we summarize the current evidence on immune and inflammatory changes, from HD animal models to clinical phenomenon of patients with HD. The understanding of the impact of inflammation on HD would help develop novel strategies to treat HD.


Subject(s)
Huntington Disease , Neurodegenerative Diseases , Animals , Huntington Disease/therapy , Neuroinflammatory Diseases , Disease Models, Animal , Inflammation
SELECTION OF CITATIONS
SEARCH DETAIL