Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 477
Filter
1.
Cells ; 13(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38786085

ABSTRACT

Innovations in cancer immunotherapy have resulted in the development of several novel immunotherapeutic strategies that can disrupt immunosuppression. One key advancement lies in immune checkpoint inhibitors (ICIs), which have shown significant clinical efficacy and increased survival rates in patients with various therapy-resistant cancers. This immune intervention consists of monoclonal antibodies directed against inhibitory receptors (e.g., PD-1) on cytotoxic CD8 T cells or against corresponding ligands (e.g., PD-L1/PD-L2) overexpressed on cancer cells and other cells in the tumor microenvironment (TME). However, not all cancer cells respond-there are still poor clinical responses, immune-related adverse effects, adaptive resistance, and vulnerability to ICIs in a subset of patients with cancer. This challenge showcases the heterogeneity of cancer, emphasizing the existence of additional immunoregulatory mechanisms in many patients. Therefore, it is essential to investigate PD-L1's interaction with other oncogenic genes and pathways to further advance targeted therapies and address resistance mechanisms. Accordingly, our aim was to investigate the mechanisms governing PD-L1 expression in tumor cells, given its correlation with immune evasion, to uncover novel mechanisms for decreasing PD-L1 expression and restoring anti-tumor immune responses. Numerous studies have demonstrated that the upregulation of Raf Kinase Inhibitor Protein (RKIP) in many cancers contributes to the suppression of key hyperactive pathways observed in malignant cells, alongside its broadening involvement in immune responses and the modulation of the TME. We, therefore, hypothesized that the role of PD-L1 in cancer immune surveillance may be inversely correlated with the low expression level of the tumor suppressor Raf Kinase Inhibitor Protein (RKIP) expression in cancer cells. This hypothesis was investigated and we found several signaling cross-talk pathways between the regulations of both RKIP and PD-L1 expressions. These pathways and regulatory factors include the MAPK and JAK/STAT pathways, GSK3ß, cytokines IFN-γ and IL-1ß, Sox2, and transcription factors YY1 and NFκB. The pathways that upregulated PD-L1 were inhibitory for RKIP expression and vice versa. Bioinformatic analyses in various human cancers demonstrated the inverse relationship between PD-L1 and RKIP expressions and their prognostic roles. Therefore, we suspect that the direct upregulation of RKIP and/or the use of targeted RKIP inducers in combination with ICIs could result in a more targeted anti-tumor immune response-addressing the therapeutic challenges related to PD-1/PD-L1 monotherapy alone.


Subject(s)
B7-H1 Antigen , Neoplasms , Phosphatidylethanolamine Binding Protein , Humans , B7-H1 Antigen/metabolism , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/metabolism , Neoplasms/drug therapy , Phosphatidylethanolamine Binding Protein/metabolism , Phosphatidylethanolamine Binding Protein/genetics , Tumor Microenvironment/immunology , Tumor Escape/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Signal Transduction/drug effects , Immune Evasion
2.
Eur J Pharmacol ; 972: 176565, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38599309

ABSTRACT

Blockade of PD-1/PD-L1 immune checkpoint is wildly used for multiple types of cancer treatment, while the low response rate for patients is still completely unknown. As nuclear hormone receptor, PPARδ (peroxisome-proliferator-activated receptor) regulates cell proliferation, inflammation, and tumor progression, while the effect of PPARδ on tumor immune escape is still unclear. Here we found that PPARδ antagonist GSK0660 significantly reduced colon cancer cell PD-L1 protein and gene expression. Luciferase analysis showed that GSK0660 decreased PD-L1 gene transcription activity. Moreover, reduced PD-L1 expression in colon cancer cells led to increased T cell activity. Further analysis showed that GSK0660 decreased PD-L1 expression in a PPARδ dependent manner. Implanted tumor model analysis showed that GSK0660 inhibited tumor immune escape and the combined PD-1 antibody with GSK0660 effectively enhanced colorectal cancer immunotherapy. These findings suggest that GSK0660 treatment could be an effective strategy for cancer immunotherapy.


Subject(s)
B7-H1 Antigen , Immunotherapy , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , Humans , Animals , Immunotherapy/methods , Mice , Cell Line, Tumor , PPAR delta/genetics , PPAR delta/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Colonic Neoplasms/immunology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Colonic Neoplasms/genetics , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tumor Escape/drug effects , Mice, Inbred BALB C
3.
Gut ; 73(6): 985-999, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38123979

ABSTRACT

OBJECTIVE: The gain of function (GOF) CTNNB1 mutations (CTNNB1 GOF ) in hepatocellular carcinoma (HCC) cause significant immune escape and resistance to anti-PD-1. Here, we aimed to investigate the mechanism of CTNNB1 GOF HCC-mediated immune escape and raise a new therapeutic strategy to enhance anti-PD-1 efficacy in HCC. DESIGN: RNA sequencing was performed to identify the key downstream genes of CTNNB1 GOF associated with immune escape. An in vitro coculture system, murine subcutaneous or orthotopic models, spontaneously tumourigenic models in conditional gene-knock-out mice and flow cytometry were used to explore the biological function of matrix metallopeptidase 9 (MMP9) in tumour progression and immune escape. Single-cell RNA sequencing and proteomics were used to gain insight into the underlying mechanisms of MMP9. RESULTS: MMP9 was significantly upregulated in CTNNB1 GOF HCC. MMP9 suppressed infiltration and cytotoxicity of CD8+ T cells, which was critical for CTNNB1 GOF to drive the suppressive tumour immune microenvironment (TIME) and anti-PD-1 resistance. Mechanistically, CTNNB1 GOF downregulated sirtuin 2 (SIRT2), resulting in promotion of ß-catenin/lysine demethylase 4D (KDM4D) complex formation that fostered the transcriptional activation of MMP9. The secretion of MMP9 from HCC mediated slingshot protein phosphatase 1 (SSH1) shedding from CD8+ T cells, leading to the inhibition of C-X-C motif chemokine receptor 3 (CXCR3)-mediated intracellular of G protein-coupled receptors signalling. Additionally, MMP9 blockade remodelled the TIME and potentiated the sensitivity of anti-PD-1 therapy in HCC. CONCLUSIONS: CTNNB1 GOF induces a suppressive TIME by activating secretion of MMP9. Targeting MMP9 reshapes TIME and potentiates anti-PD-1 efficacy in CTNNB1 GOF HCC.


Subject(s)
CD8-Positive T-Lymphocytes , Carcinoma, Hepatocellular , Liver Neoplasms , Matrix Metalloproteinase 9 , beta Catenin , beta Catenin/metabolism , beta Catenin/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Animals , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Mice , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , CD8-Positive T-Lymphocytes/immunology , Humans , Mutation , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Tumor Escape/genetics , Tumor Escape/drug effects , Tumor Microenvironment/immunology , Cell Line, Tumor
4.
Nat Cell Biol ; 24(2): 230-241, 2022 02.
Article in English | MEDLINE | ID: mdl-35145222

ABSTRACT

Many cancers have an unusual dependence on glutamine. However, most previous studies have focused on the contribution of glutamine to metabolic building blocks and the energy supply. Here, we report that cancer cells with aberrant expression of glutamate decarboxylase 1 (GAD1) rewire glutamine metabolism for the synthesis of γ-aminobutyric acid (GABA)-a prominent neurotransmitter-in non-nervous tissues. An analysis of clinical samples reveals that increased GABA levels predict poor prognosis. Mechanistically, we identify a cancer-intrinsic pathway through which GABA activates the GABAB receptor to inhibit GSK-3ß activity, leading to enhanced ß-catenin signalling. This GABA-mediated ß-catenin activation both stimulates tumour cell proliferation and suppresses CD8+ T cell intratumoural infiltration, such that targeting GAD1 or GABABR in mouse models overcomes resistance to anti-PD-1 immune checkpoint blockade therapy. Our findings uncover a signalling role for tumour-derived GABA beyond its classic function as a neurotransmitter that can be targeted pharmacologically to reverse immunosuppression.


Subject(s)
Cell Proliferation , Neoplasms/metabolism , Tumor Escape , Tumor Microenvironment/immunology , beta Catenin/metabolism , gamma-Aminobutyric Acid/metabolism , A549 Cells , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Immune Checkpoint Inhibitors/pharmacology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Inbred C57BL , Mice, Nude , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Receptors, GABA-B/metabolism , Tumor Burden , Tumor Escape/drug effects , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , beta Catenin/genetics
5.
BMC Cancer ; 22(1): 46, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34996407

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors (ICIs) are currently one of the most promising therapy options in the field of oncology. Although the first pivotal ICI trial results were published in 2011, few biomarkers exist to predict their therapy outcome. PD-L1 expression and tumor mutational burden (TMB) were proven to be sometimes-unreliable biomarkers. We have previously suggested the analysis of processing escapes, a qualitative measurement of epitope structure alterations under immune system pressure, to provide predictive information on ICI response. Here, we sought to further validate this approach and characterize interactions with different forms of immune pressure. METHODS: We identified a cohort consisting of 48 patients with advanced non-small cell lung cancer (NSCLC) treated with nivolumab as ICI monotherapy. Tumor samples were subjected to targeted amplicon-based sequencing using a panel of 22 cancer-associated genes covering 98 mutational hotspots. Altered antigen processing was predicted by NetChop, and MHC binding verified by NetMHC. The NanoString nCounter® platform was utilized to provide gene expression data of 770 immune-related genes. Patient data from 408 patients with NSCLC were retrieved from The Cancer Genome Atlas (TCGA) as a validation cohort. RESULTS: The two immune escape mechanisms of PD-L1 expression (TPS score) (n = 18) and presence of altered antigen processing (n = 10) are mutually non-exclusive and can occur in the same patient (n = 6). Both mechanisms have exclusive influence on different genes and pathways, according to differential gene expression analysis and gene set enrichment analysis, respectively. Interestingly, gene expression patterns associated with altered processing were enriched in T cell and NK cell immune activity. Though both mechanisms influence different genes, they are similarly linked to increased immune activity. CONCLUSION: Pressure from the immune system will lay the foundations for escape mechanisms, leading to acquisition of resistance under therapy. Both PD-L1 expression and altered antigen processing are induced similarly by pronounced immunoactivity but in different context. The present data help to deepen our understanding of the underlying mechanisms behind those immune escapes.


Subject(s)
Immune Checkpoint Inhibitors , Immunotherapy , Transcriptome , Tumor Escape , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Computational Biology , Deep Learning , Female , High-Throughput Nucleotide Sequencing , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Male , Middle Aged , Nivolumab/pharmacology , Nivolumab/therapeutic use , Retrospective Studies , Transcriptome/drug effects , Transcriptome/genetics , Transcriptome/immunology , Tumor Escape/drug effects , Tumor Escape/genetics , Tumor Escape/immunology
6.
Semin Cancer Biol ; 79: 68-82, 2022 02.
Article in English | MEDLINE | ID: mdl-32201368

ABSTRACT

In the last decade, a large amount of research has focused on elucidating the mechanisms that account for homing disseminated cancer cells (DCCs) from solid tumours to distant organs, which successively progress to overt metastatic disease; this is currently incurable. A better understanding of DCC behaviour is expected to allow detectable metastasis prevention by more effectively targeting 'metastatic seeds before they sprout'. As DCC biology co-evolved with that of the primary tumour, and due to the many similarities between them, the term 'niche' has been borrowed from normal adult stem cells (ASCs) to define the site of DCC metastatic colonisation. Moreover, heterogeneity, survival, protection, stemness and plasticity as well as the prolonged G0-G1 dormant state in the metastatic niche have been the main aspects of intense investigation. Consistent with these findings, in solid cancers with minimal residual disease (MRD), it has been proposed to prolong adjuvant therapy by targeting specific molecular pathway(s) involving DCC dormancy. However, so far, few disappointing clinical data have been reported. As an alternative strategy, because immune-surveillance contributes to the steady state of the DCC population and likely to the G0-G1 state of cancer cells, we have used prolonged immune-modulatory cytostatic chemotherapy, active immune stimulation with an INF-ß/IL-2 sequence or drugs inhibiting myeloid-derived suppressor cell (MDSC)/Treg-mediated immune suppression. This strategy, mainly aimed at boosting the immune response, is based on recent findings suggesting the downregulation of immune escape mechanisms as well as other principal hallmarks during the G0-G1 state and/or in MRD. Preliminary clinical and/or laboratory data suggest the efficacy of this strategy in gastrointestinal and some endocrine-dependent cancers. Following this, we propose therapeutic schedules to prevent DCC activation and proliferation in solid cancers at a high risk of relapse or as maintenance therapy in metastatic patients after complete response (CR) to conventional treatment.


Subject(s)
Immunologic Factors/therapeutic use , Immunotherapy/methods , Neoplasm Metastasis/prevention & control , Neoplasm Recurrence, Local/prevention & control , Neoplasm, Residual/therapy , Neoplastic Cells, Circulating/pathology , Cell Proliferation/drug effects , Humans , Interleukin-2/metabolism , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Neoplasm Metastasis/drug therapy , Neoplasm Recurrence, Local/drug therapy , Neoplasm, Residual/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tumor Escape/drug effects , Tumor Escape/immunology
7.
Cancer Sci ; 113(1): 28-40, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34727389

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the commonest lethal malignancies worldwide, and often diagnosed at an advanced stage, without any curative therapy. Immune checkpoint blockers targeting the programmed death receptor 1 (PD-1) have shown impressive antitumor activity in patients with advanced-stage HCC, while the response rate is only 30%. Inducible PD-L1 overexpression may result in a lack of response to cancer immunotherapy, which is attributed to a mechanism of adaptive immune resistance. Our study investigated that the overexpression of PD-L1 promoted the invasion and migration of liver cancer cells in vitro, and the induced overexpression of PD-L1 in the tumor microenvironment could weaken the effects of anti-PD-1 immunotherapy in a BALB/c mouse model of liver cancer. CPI-203, a small-molecule bromodomain-containing protein 4 (BRD4) inhibitor, which can potently inhibit PD-L1 expression in vitro and in vivo, combined with PD-1 antibody improved the response to immunotherapy in a liver cancer model. Cell transfection and chromatin immunoprecipitation assay manifested that BRD4 plays a key role in PD-L1 expression; CPI-203 can inhibit PD-L1 expression by inhibiting the BRD4 occupation of the PD-L1 promoter region. This study indicates a potential clinical immunotherapy method to reduce the incidence of clinical resistance to immunotherapy in patients with HCC.


Subject(s)
Acetamides/administration & dosage , Azepines/administration & dosage , B7-H1 Antigen/genetics , Cell Cycle Proteins/metabolism , Immune Checkpoint Inhibitors/administration & dosage , Lung Neoplasms/drug therapy , Transcription Factors/metabolism , Up-Regulation/drug effects , Acetamides/pharmacology , Animals , Azepines/pharmacology , Cell Line, Tumor , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Promoter Regions, Genetic/drug effects , Tumor Escape/drug effects , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
8.
Article in English | MEDLINE | ID: mdl-34653581

ABSTRACT

High-density lipoproteins (HDL) are well known for their protective role against the development and progression of atherosclerosis. Atheroprotection is mainly due to the key role of HDL within the reverse cholesterol transport, and to their ability to exert a series of antioxidant and anti-inflammatory activities. Through the same mechanisms HDL could also affect cancer cell proliferation and tumor progression. Many types of cancers share common alterations of cellular metabolism, including lipid metabolism. In this context, not only fatty acids but also cholesterol and its metabolites play a key role. HDL were shown to reduce cancer cell content of cholesterol, overall rewiring cholesterol homeostasis. In addition, HDL reduce oxidative stress and the levels of pro-inflammatory molecules in cancer cells and in the tumor microenvironment (TME). Here, HDL can also help in reverting tumor immune escape and in inhibiting angiogenesis. Interestingly, HDL are good candidates for drug delivery, targeting antineoplastic agents to the tumor mass mainly through their binding to the scavenger receptor BI. Since they could affect cancer development and progression per se, HDL-based drug delivery systems may render cancer cells more sensitive to antitumor agents and reduce the development of drug resistance.


Subject(s)
Cholesterol, HDL/therapeutic use , Cholesterol/metabolism , Lipoproteins, HDL/therapeutic use , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Cholesterol/therapeutic use , Cholesterol, HDL/metabolism , Drug Delivery Systems , Drug Resistance, Neoplasm/genetics , Humans , Lipoproteins, HDL/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Oxidative Stress/drug effects , Tumor Escape/drug effects , Tumor Microenvironment/drug effects
9.
Leuk Res ; 112: 106751, 2022 01.
Article in English | MEDLINE | ID: mdl-34808592

ABSTRACT

Acute myeloblastic leukemia (AML) is one of the most common types of blood malignancies that results in an AML-associated high mortality rate each year. Several causes have been reported as prognostic factors for AML in children and adults, the most important of which are cytogenetic abnormalities and environmental risk factors. Following the discovery of numerous drugs for AML treatment, leukemic cells sought a way to escape from the cytotoxic effects of chemotherapy drugs, leading to treatment failure. Nowadays, comprehensive studies have looked at the role of extracellular vesicles (EVs) secreted by AML blasts and how the microenvironment of the tumor changes in favor of cancer progression and survival to discover the mechanisms of treatment failure to choose the well-advised treatment. Reports show that malignant cells secrete EVs that transmit messages to adjacent cells and the tumor's microenvironment. By secreting EVs, containing immune-inhibiting cytokines, AML cells inactivate the immune system against malignant cells, thus ensuring their survival. Also, increased secretion of EVs in various malignancies indicates an unfavorable prognostic factor and the possibility of drug resistance. In this study, we briefly reviewed the challenges of treating AML with a glance at the EVs' role in this process. It is hoped that with a deeper understanding of EVs, new therapies will be developed to eliminate the relapse of leukemic cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/drug effects , Extracellular Vesicles/drug effects , Leukemia, Myeloid/drug therapy , Tumor Escape/drug effects , Tumor Microenvironment/drug effects , Acute Disease , Antineoplastic Agents/immunology , Drug Resistance, Neoplasm/genetics , Extracellular Vesicles/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Leukemia, Myeloid/genetics , Treatment Failure , Tumor Escape/genetics , Tumor Microenvironment/genetics
10.
Cancer Treat Rev ; 102: 102322, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34922151

ABSTRACT

Advances in understanding the molecular mechanisms of tumor progression have achieved impressive progress in the treatment of cancer and so-called immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy. Indeed, antibody-based drugs blocking immune escape of tumor cells by modulation of T cell responses are increasingly utilized for a wide range of tumor entities. Nonetheless, response rates remain limited, and the development of secondary resistance is a common problem. In addition, by increasing the immune response a variety of severe side effects are provoked. Next to autoimmune responses, activation of the complement system and skin toxicity, an increased incidence for thrombotic complications has been observed associated with an increased mortality rate. Based on this, it can be postulated that the interplay of coagulation with inflammation in the tumor microenvironment is relevant for each step in the tumor life cycle. This review focuses on the coagulation as central player fostering mechanisms associated with tumor progression. Thus, a better understanding of the molecular pathways involved in the complex interaction of circulating tumor cells, the plasmatic coagulation and immune cells may help to improve therapeutic concepts reducing mortality and morbidity associated with cancer.


Subject(s)
Blood Coagulation/immunology , Heparin, Low-Molecular-Weight/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Inflammation/blood , Neoplasms/blood , Neoplasms/drug therapy , Anticoagulants/therapeutic use , Blood Coagulation/drug effects , Humans , Inflammation/immunology , Inflammation/pathology , Neoplasms/immunology , Thrombosis/blood , Thrombosis/drug therapy , Thrombosis/pathology , Tumor Escape/drug effects
12.
Biomolecules ; 11(12)2021 12 20.
Article in English | MEDLINE | ID: mdl-34944555

ABSTRACT

Immunotherapy has made great progress in recent years, yet the efficacy of solid tumors remains far less than expected. One of the main hurdles is to overcome the immune-suppressive tumor microenvironment (TME). Among all cells in TME, tumor-associated macrophages (TAMs) play pivotal roles because of their abundance, multifaceted interactions to adaptive and host immune systems, as well as their context-dependent plasticity. Underlying the highly plastic characteristic, lots of research interests are focused on repolarizing TAMs from M2-like pro-tumor phenotype towards M1-like antitumoral ones. Nanotechnology offers great opportunities for targeting and modulating TAM polarization to mount the therapeutic efficacy in cancer immunotherapy. Here, this mini-review highlights those emerging nano-approaches for TAM repolarization in the last three years.


Subject(s)
Nanoparticles/therapeutic use , Neoplasms/drug therapy , Cell Polarity/drug effects , Humans , Immunotherapy , Neoplasms/immunology , Tumor Escape/drug effects , Tumor Microenvironment/drug effects , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology
13.
Front Immunol ; 12: 783236, 2021.
Article in English | MEDLINE | ID: mdl-34899747

ABSTRACT

Hepatocellular carcinoma (HCC), one of the most fatal malignancies in the world, is usually diagnosed in advanced stages due to late symptom manifestation with very limited therapeutic options, which leads to ineffective intervention and dismal prognosis. For a decade, tyrosine kinase inhibitors (TKIs) have offered an overall survival (OS) benefit when used in a first-line (sorafenib and lenvatinib) and second-line setting (regorafenib and cabozantinib) in advanced HCC, while long-term response remains unsatisfactory due to the onset of primary or acquired resistance. Recently, immunotherapy has emerged as a promising therapy in the treatment of several solid tumors, such as melanoma and non-small cell lung cancer. Moreover, as the occurrence of HCC is associated with immune tolerance and immunosurveillance escape, there is a potent rationale for employing immunotherapy in HCC. However, immunotherapy monotherapy, mainly including immune checkpoint inhibitors (ICIs) that target checkpoints programmed death-1 (PD-1), programmed death-ligand 1 (PD-L1), and the cytotoxic T lymphocyte antigen-4 (CTLA-4), has a relatively low response rate. Thus, the multi-ICIs or the combination of immunotherapy with other therapies, like antiangiogenic drugs and locoregional therapies, has become a novel strategy to treat HCC. Combining different ICIs may have a synergistical effect attributed to the complementary effects of the two immune checkpoint pathways (CTLA-4 and PD-1/PD-L1 pathways). The incorporation of antiangiogenic drugs in ICIs can enhance antitumor immune responses via synergistically regulating the vasculature and the immune microenvironment of tumor. In addition, locoregional treatments can improve antitumor immunity by releasing the neoplasm antigens from killed tumor cells; in turn, this antitumor immune response can be intensified by immunotherapy. Therefore, the combination of locoregional treatments and immunotherapy may achieve greater efficacy through further synergistic effects for advanced HCC. This review aims to summarize the currently reported results and ongoing trials of the ICIs-based combination therapies for HCC to explore the rational combination strategies and further improve the survival of patients with HCC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular/therapy , Immunotherapy/methods , Liver Neoplasms/therapy , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Chemoembolization, Therapeutic/methods , Chemoradiotherapy, Adjuvant/methods , Chemotherapy, Adjuvant/methods , Clinical Trials as Topic , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Liver/immunology , Liver/pathology , Liver/surgery , Liver Neoplasms/immunology , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Progression-Free Survival , Radiofrequency Ablation/methods , Radiosurgery/methods , Tumor Escape/drug effects , Tumor Escape/immunology , Tumor Escape/radiation effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
14.
Int J Mol Sci ; 22(24)2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34948368

ABSTRACT

Triple-negative breast cancer (TNBC) is defined based on the absence of estrogen, progesterone, and human epidermal growth factor receptor 2 receptors. Currently, chemotherapy is the major therapeutic approach for TNBC patients; however, poor prognosis after a standard chemotherapy regimen is still commonplace due to drug resistance. Abnormal tumor metabolism and infiltrated immune or stromal cells in the tumor microenvironment (TME) may orchestrate mammary tumor growth and metastasis or give rise to new subsets of cancer cells resistant to drug treatment. The immunosuppressive mechanisms established in the TME make cancer cell clones invulnerable to immune recognition and killing, and turn immune cells into tumor-supporting cells, hence allowing cancer growth and dissemination. Phytochemicals with the potential to change the tumor metabolism or reprogram the TME may provide opportunities to suppress cancer metastasis and/or overcome chemoresistance. Furthermore, phytochemical intervention that reprograms the TME away from favoring immunoevasion and instead towards immunosurveillance may prevent TNBC metastasis and help improve the efficacy of combination therapies as phyto-adjuvants to combat drug-resistant TNBC. In this review, we summarize current findings on selected bioactive plant-derived natural products in preclinical mouse models and/or clinical trials with focus on their immunomodulatory mechanisms in the TME and their roles in regulating tumor metabolism for TNBC prevention or therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Phytogenic/pharmacology , Drug Development , Female , Humans , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Tumor Escape/drug effects
15.
Nat Commun ; 12(1): 6938, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34836966

ABSTRACT

Primary brain tumors, such as glioblastoma (GBM), are remarkably resistant to immunotherapy, even though pre-clinical models suggest effectiveness. To understand this better in patients, here we take advantage of our recent neoadjuvant treatment paradigm to map the infiltrating immune cell landscape of GBM and how this is altered following PD-1 checkpoint blockade using high dimensional proteomics, single cell transcriptomics, and quantitative multiplex immunofluorescence. Neoadjuvant PD-1 blockade increases T cell infiltration and the proportion of a progenitor exhausted population of T cells found within the tumor. We identify an early activated and clonally expanded CD8+ T cell cluster whose TCR overlaps with a CD8+ PBMC population. Distinct changes are also observed in conventional type 1 dendritic cells that may facilitate T cell recruitment. Macrophages and monocytes still constitute the majority of infiltrating immune cells, even after anti-PD-1 therapy. Interferon-mediated changes in the myeloid population are consistently observed following PD-1 blockade; these also mediate an increase in chemotactic factors that recruit T cells. However, sustained high expression of T-cell-suppressive checkpoints in these myeloid cells continue to prevent the optimal activation of the tumor infiltrating T cells. Therefore, future immunotherapeutic strategies may need to incorporate the targeting of these cells for clinical benefit.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/therapy , Immune Checkpoint Inhibitors/pharmacology , Neoadjuvant Therapy/methods , Neoplasm Recurrence, Local/therapy , Brain/drug effects , Brain/immunology , Brain/pathology , Brain/surgery , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Immune Checkpoint Inhibitors/therapeutic use , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Neurosurgical Procedures , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , RNA-Seq , Single-Cell Analysis , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Escape/drug effects , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology
16.
Nat Commun ; 12(1): 6667, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34795269

ABSTRACT

Inhibition of HER2 in HER2-amplified breast cancer has been remarkably successful clinically, as demonstrated by the efficacy of HER-kinase inhibitors and HER2-antibody treatments. Whilst resistance to HER2 inhibition is common in the metastatic setting, the specific programs downstream of HER2 driving resistance are not established. Through genomic profiling of 733 HER2-amplified breast cancers, we identify enrichment of somatic alterations that promote MEK/ERK signaling in metastatic tumors with shortened progression-free survival on anti-HER2 therapy. These mutations, including NF1 loss and ERBB2 activating mutations, are sufficient to mediate resistance to FDA-approved HER2 kinase inhibitors including tucatinib and neratinib. Moreover, resistant tumors lose AKT dependence while undergoing a dramatic sensitization to MEK/ERK inhibition. Mechanistically, this driver pathway switch is a result of MEK-dependent activation of CDK2 kinase. These results establish genetic activation of MAPK as a recurrent mechanism of anti-HER2 therapy resistance that may be effectively combated with MEK/ERK inhibitors.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Tumor Escape/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Drug Resistance, Neoplasm/genetics , Female , Humans , Lapatinib/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mutation , Oxazoles/pharmacology , Pyridines/pharmacology , Quinazolines/pharmacology , Quinolines/pharmacology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
17.
Nat Commun ; 12(1): 6742, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34795289

ABSTRACT

Immunotherapy has become a powerful cancer treatment, but only a small fraction of patients have achieved durable benefits due to the immune escape mechanism. In this study, epigenetic regulation is combined with gene therapy-mediated immune checkpoint blockade to relieve this immune escape mechanism. PPD (i.e., mPEG-b-PLG/PEI-RT3/DNA) is developed to mediate plasmid-encoding shPD-L1 delivery by introducing multiple interactions (i.e., electrostatic, hydrogen bonding, and hydrophobic interactions) and polyproline II (PPII)-helix conformation, which downregulates PD-L1 expression on tumour cells to relieve the immunosuppression of T cells. Zebularine (abbreviated as Zeb), a DNA methyltransferase inhibitor (DNMTi), is used for the epigenetic regulation of the tumour immune microenvironment, thus inducing DC maturation and MHC I molecule expression to enhance antigen presentation. PPD plus Zeb combination therapy initiates a systemic anti-tumour immune response and effectively prevents tumour relapse and metastasis by generating durable immune memory. This strategy provides a scheme for tumour treatment and the inhibition of relapse and metastasis.


Subject(s)
Epigenesis, Genetic/drug effects , Genetic Therapy , Immunotherapy , Neoplasms/therapy , Tumor Escape/drug effects , Animals , B7-H1 Antigen/drug effects , B7-H1 Antigen/metabolism , Cell Line, Tumor , Combined Modality Therapy , Cytidine/analogs & derivatives , Cytidine/pharmacology , DNA Methylation/drug effects , Humans , Immune Checkpoint Inhibitors , Immunity/drug effects , Methyltransferases/antagonists & inhibitors , Neoplasm Metastasis/therapy , T-Lymphocytes/immunology , Tumor Microenvironment/immunology
18.
Int J Mol Sci ; 22(22)2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34830221

ABSTRACT

Dendritic cells (DCs) are immune specialized cells playing a critical role in promoting immune response against antigens, and may represent important targets for therapeutic interventions in cancer. DCs can be stimulated ex vivo with pro-inflammatory molecules and loaded with tumor-specific antigen(s). Protocols describing the specific details of DCs vaccination manufacturing vary widely, but regardless of the employed protocol, the DCs vaccination safety and its ability to induce antitumor responses is clearly established. Many years of studies have focused on the ability of DCs to provide overall survival benefits at least for a selection of cancer patients. Lessons learned from early trials lead to the hypothesis that, to improve the efficacy of DCs-based immunotherapy, this should be combined with other treatments. Thus, the vaccine's ultimate role may lie in the combinatorial approaches of DCs-based immunotherapy with chemotherapy and radiotherapy, more than in monotherapy. In this review, we address some key questions regarding the integration of DCs vaccination with multimodality therapy approaches for cancer treatment paradigms.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive/methods , Neoplasms/drug therapy , Neoplasms/radiotherapy , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Combined Modality Therapy/methods , Humans , Neoplasms/immunology , Progression-Free Survival , Tumor Escape/drug effects , Tumor Escape/radiation effects
19.
Gastroenterology ; 161(6): 1813-1829, 2021 12.
Article in English | MEDLINE | ID: mdl-34606846

ABSTRACT

Chronic inflammation is a known risk factor for gastrointestinal cancer. The evidence that nonsteroidal anti-inflammatory drugs suppress the incidence, growth, and metastasis of gastrointestinal cancer supports the concept that a nonsteroidal anti-inflammatory drug target, cyclooxygenase, and its downstream bioactive lipid products may provide one of the links between inflammation and cancer. Preclinical studies have demonstrated that the cyclooxygenase-2-prostaglandin E2 pathway can promote gastrointestinal cancer development. Although the role of this pathway in cancer has been investigated extensively for 2 decades, only recent studies have described its effects on host defenses against transformed epithelial cells. Overcoming tumor-immune evasion remains one of the major challenges in cancer immunotherapy. This review summarizes the impacts of the cyclooxygenase-2-prostaglandin E2 pathway on gastrointestinal cancer development. Our focus was to highlight recent advances in our understanding of how this pathway induces tumor immune evasion.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Gastrointestinal Neoplasms/enzymology , Inflammation Mediators/metabolism , Tumor Escape , Tumor Microenvironment/immunology , Animals , Antineoplastic Agents/therapeutic use , Cancer-Associated Fibroblasts/enzymology , Cancer-Associated Fibroblasts/immunology , Cyclooxygenase 2 Inhibitors/therapeutic use , Epithelial Cells/enzymology , Epithelial Cells/immunology , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/immunology , Gastrointestinal Neoplasms/pathology , Humans , Immunotherapy , Lymphocytes, Tumor-Infiltrating/enzymology , Lymphocytes, Tumor-Infiltrating/immunology , Signal Transduction , Tumor Escape/drug effects , Tumor-Associated Macrophages/enzymology , Tumor-Associated Macrophages/immunology
20.
Nat Commun ; 12(1): 5733, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34593794

ABSTRACT

In addition to increasing the expression of programmed death-ligand 1 (PD-L1), tumor cells can also secrete exosomal PD-L1 to suppress T cell activity. Emerging evidence has revealed that exosomal PD-L1 resists immune checkpoint blockade, and may contribute to resistance to therapy. In this scenario, suppressing the secretion of tumor-derived exosomes may aid therapy. Here, we develop an assembly of exosome inhibitor (GW4869) and ferroptosis inducer (Fe3+) via amphiphilic hyaluronic acid. Cooperation between the two active components in the constructed nanounit induces an anti-tumor immunoresponse to B16F10 melanoma cells and stimulates cytotoxic T lymphocytes and immunological memory. The nanounit enhances the response to PD-L1 checkpoint blockade and may represent a therapeutic strategy for enhancing the response to this therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Carriers/chemistry , Exosomes/drug effects , Ferroptosis/drug effects , Melanoma, Experimental/drug therapy , Skin Neoplasms/drug therapy , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Benzylidene Compounds/pharmacology , Benzylidene Compounds/therapeutic use , Cell Line, Tumor/transplantation , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Exosomes/immunology , Exosomes/metabolism , Female , Ferroptosis/immunology , Humans , Hyaluronic Acid/chemistry , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunologic Memory , Lymphocyte Activation/drug effects , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Escape/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...