Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 208
Filtrar
1.
Int Immunopharmacol ; 136: 112273, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-38810311

RESUMO

Cholangiocarcinoma (CCA) presents a significant clinical challenge which is often identified in advanced stages, therby restricting the effectiveness of surgical interventions for most patients. The high incidence of cancer recurrence and resistance to chemotherapy further contribute to a bleak prognosis and low survival rates. To address this pressing need for effective therapeutic strategies, our study focuses on the development of an innovative cellular immunotherapy, specifically utilizing chimeric antigen receptor (CAR)-engineered natural killer (NK) cells designed to target the cMET receptor tyrosine kinase. In this investigation, we initiated the screening of a phage library displaying human single-chain variable fragment (ScFv) to identify novel ScFv molecules with specificity for cMET. Remarkably, ScFv11, ScFv72, and ScFv114 demonstrated exceptional binding affinity, confirmed by molecular docking analysis. These selected ScFvs, in addition to the well-established anti-cMET ScFvA, were integrated into a CAR cassette harboring CD28 transmembrane region-41BB-CD3ζ domains. The resulting anti-cMET CAR constructs were transduced into NK-92 cells, generating potent anti-cMET CAR-NK-92 cells. To assess the specificity and efficacy of these engineered cells, we employed KKU213A cells with high cMET expression and KKU055 cells with low cMET levels. Notably, co-culture of anti-cMET CAR-NK-92 cells with KKU213A cells resulted in significantly increased cell death, whereas no such effect was observed with KKU055 cells. In summary, our study identified cMET as a promising therapeutic target for CCA. The NK-92 cells, armed with the anti-cMET CAR molecule, have shown strong ability to kill cancer cells specifically, indicating their potential as a promising treatment for CCA in the future.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Células Matadoras Naturais , Proteínas Proto-Oncogênicas c-met , Receptores de Antígenos Quiméricos , Anticorpos de Cadeia Única , Humanos , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/uso terapêutico , Anticorpos de Cadeia Única/imunologia , Colangiocarcinoma/terapia , Colangiocarcinoma/imunologia , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Células Matadoras Naturais/imunologia , Linhagem Celular Tumoral , Neoplasias dos Ductos Biliares/terapia , Neoplasias dos Ductos Biliares/imunologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Proteínas Proto-Oncogênicas c-met/imunologia , Imunoterapia Adotiva/métodos , Imunoterapia/métodos , Medicina de Precisão
2.
Am J Physiol Cell Physiol ; 327(1): C11-C33, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38708523

RESUMO

In contrast to other types of cancers, there is no available efficient pharmacological treatment to improve the outcomes of patients suffering from major primary liver cancers, i.e., hepatocellular carcinoma and cholangiocarcinoma. This dismal situation is partly due to the existence in these tumors of many different and synergistic mechanisms of resistance, accounting for the lack of response of these patients, not only to classical chemotherapy but also to more modern pharmacological agents based on the inhibition of tyrosine kinase receptors (TKIs) and the stimulation of the immune response against the tumor using immune checkpoint inhibitors (ICIs). This review summarizes the efforts to develop strategies to overcome this severe limitation, including searching for novel drugs derived from synthetic, semisynthetic, or natural products with vectorial properties against therapeutic targets to increase drug uptake or reduce drug export from cancer cells. Besides, immunotherapy is a promising line of research that is already starting to be implemented in clinical practice. Although less successful than in other cancers, the foreseen future for this strategy in treating liver cancers is considerable. Similarly, the pharmacological inhibition of epigenetic targets is highly promising. Many novel "epidrugs," able to act on "writer," "reader," and "eraser" epigenetic players, are currently being evaluated in preclinical and clinical studies. Finally, gene therapy is a broad field of research in the fight against liver cancer chemoresistance, based on the impressive advances recently achieved in gene manipulation. In sum, although the present is still dismal, there is reason for hope in the non-too-distant future.


Assuntos
Neoplasias Hepáticas , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/genética , Animais , Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/genética , Imunoterapia/métodos , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Inibidores de Proteínas Quinases/uso terapêutico , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/imunologia , Colangiocarcinoma/patologia , Epigênese Genética/efeitos dos fármacos
3.
Front Immunol ; 15: 1376590, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38799431

RESUMO

Background: Research of immunotherapy for cholangiocarcinoma has yielded some results, but more clinical data are needed to prove its efficacy and safety. Moreover, there is a need to identify accessible indexes for selecting patients who may benefit from such treatments. Methods: The medical records of 66 cholangiocarcinoma patients who underwent immunotherapy were retrospectively collected. The effectiveness of immunotherapy was assessed by tumor response, progression-free survival (PFS), and overall survival (OS), while safety was evaluated by adverse events during treatment. Univariate and multivariate Cox regression analyses were performed to identify prognostic risk factors for PFS and OS, and Kaplan-Meier curves of potential prognostic factors were drawn. Results: Overall, in this study, immunotherapy achieved an objective response rate of 24.2% and a disease control rate of 89.4% for the included patients. The median PFS was 445 days, and the median OS was 772.5 days. Of the 66 patients, 65 experienced adverse events during treatment, but none had severe consequences. Multivariate Cox analysis indicated that tumor number is a prognostic risk factor for disease progression following immunotherapy in cholangiocarcinoma patients, while tumor differentiation and the fibrosis-4 (FIB-4) index are independent risk factors for OS. Conclusion: In general, immunotherapy for cholangiocarcinoma is safe, with adverse events remaining within manageable limits, and it can effectively control disease progression in most patients. The FIB-4 index may reflect the potential benefit of immunotherapy for patients with cholangiocarcinoma.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Imunoterapia , Humanos , Colangiocarcinoma/terapia , Colangiocarcinoma/imunologia , Colangiocarcinoma/mortalidade , Masculino , Feminino , Pessoa de Meia-Idade , Neoplasias dos Ductos Biliares/terapia , Neoplasias dos Ductos Biliares/mortalidade , Neoplasias dos Ductos Biliares/imunologia , Idoso , Prognóstico , Imunoterapia/efeitos adversos , Imunoterapia/métodos , Estudos Retrospectivos , Adulto , Fibrose , Idoso de 80 Anos ou mais , Fatores de Risco
4.
Biomed Pharmacother ; 175: 116718, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38744221

RESUMO

Advanced cholangiocarcinoma (CCA) presents a clinical challenge due to limited treatment options, necessitating exploration of innovative therapeutic approaches. Bispecific T cell engager (BTE)-armed T cell therapy shows promise in hematological and solid malignancies, offering potential advantages in safety over continuous BTE infusion. In this context, we developed a novel BTE, targeting CD3 on T cells and integrin αvß6, an antigen elevated in various epithelial malignancies, on cancer cells. The novel BTE was generated by fusing an integrin αvß6-binding peptide (A20) to an anti-CD3 (OKT3) single-chain variable fragment (scFv) through a G4S peptide linker (A20/αCD3 BTE). T cells were then armed with A20/αCD3 BTE (A20/αCD3-armed T cells) and assessed for antitumor activity. Our results highlight the specific binding of A20/αCD3 BTE to CD3 on T cells and integrin αvß6 on target cells, effectively redirecting T cells towards these targets. After co-culture, A20/αCD3-armed T cells exhibited significantly heightened cytotoxicity against integrin αvß6-expressing target cells compared to unarmed T cells in both KKU-213A cells and A375.ß6 cells. Moreover, in a five-day co-culture, A20/αCD3-armed T cells demonstrated superior cytotoxicity against KKU-213A spheroids compared to unarmed T cells. Importantly, A20/αCD3-armed T cells exhibited an increased proportion of the effector memory T cell (Tem) subset, upregulation of T cell activation markers, enhanced T cell proliferation, and increased cytolytic molecule/cytokine production, when compared to unarmed T cells in an integrin αvß6-dependent manner. These findings support the potential of A20/αCD3-armed T cells as a novel therapeutic approach for integrin αvß6-expressing cancers.


Assuntos
Antígenos de Neoplasias , Neoplasias dos Ductos Biliares , Colangiocarcinoma , Integrinas , Linfócitos T , Humanos , Colangiocarcinoma/imunologia , Colangiocarcinoma/terapia , Colangiocarcinoma/patologia , Antígenos de Neoplasias/imunologia , Linfócitos T/imunologia , Integrinas/metabolismo , Linhagem Celular Tumoral , Neoplasias dos Ductos Biliares/imunologia , Neoplasias dos Ductos Biliares/patologia , Neoplasias dos Ductos Biliares/terapia , Complexo CD3/imunologia , Anticorpos de Cadeia Única/farmacologia , Técnicas de Cocultura , Anticorpos Biespecíficos/farmacologia
5.
Cancer Lett ; 594: 216992, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38797231

RESUMO

Intrahepatic cholangiocarcinoma (iCCA) is an aggressive cancer with an extremely poor prognosis, and new treatment options are needed. Recently, immunotherapy has emerged as an efficient treatment against malignant tumors, but less effective in iCCA. Activation of stimulator of interferon genes (STING) signaling could reignite immunologically inert tumors, but the expression and role of STING in iCCA remains to be determined. Here, we show STING is expressed in iCCA, and patients with high expression of STING in early-stage iCCA have a longer overall survival than those have low expression. Increased immune cell infiltration in early-stage iCCA corresponds to elevated STING expression. In mice iCCA models, treatment with the STING agonist MSA-2 show stage-specific inhibitory effects on tumors, with beneficial effects in early-stage tumors but not with advanced-stage cancer. This discrepancy was associated with greater programmed cell death ligand 1 (PD-L1) expression in advanced-stage tumors. Combination therapy targeting PD-L1 and MSA-2 strikingly reduced tumor burden in such tumors compared to either monotherapy. Cumulatively, these data demonstrate that STING agonism monotherapy improves the immune landscape of the tumor microenvironment in early-stage iCCA, while combination therapy ameliorates advanced-stage iCCA.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Proteínas de Membrana , Colangiocarcinoma/imunologia , Colangiocarcinoma/genética , Colangiocarcinoma/patologia , Colangiocarcinoma/tratamento farmacológico , Animais , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/agonistas , Humanos , Camundongos , Neoplasias dos Ductos Biliares/patologia , Neoplasias dos Ductos Biliares/imunologia , Neoplasias dos Ductos Biliares/tratamento farmacológico , Neoplasias dos Ductos Biliares/genética , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Estadiamento de Neoplasias , Linhagem Celular Tumoral , Microambiente Tumoral/imunologia , Masculino , Feminino , Transdução de Sinais
6.
Biomed Pharmacother ; 175: 116659, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38692063

RESUMO

Cholangiocarcinoma (CCA), a rare yet notably aggressive cancer, has experienced a surge in incidence in recent years. Presently, surgical resection remains the most effective curative strategy for CCA. Nevertheless, a majority of patients with CCA are ineligible for surgical removal at the time of diagnosis. For advanced stages of CCA, the combination of gemcitabine and cisplatin is established as the standard chemotherapy regimen. Despite this, treatment efficacy is often hindered by the development of resistance. In recent times, immune checkpoint inhibitors, particularly those that block programmed death 1 and its ligand (PD1/PD-L1), have emerged as promising strategies against a variety of cancers and are being increasingly integrated into the therapeutic landscape of CCA. A growing body of research supports that the use of PD1/PD-L1 monoclonal antibodies in conjunction with chemotherapy may significantly improve patient outcomes. This article seeks to meticulously review the latest studies on PD1/PD-L1 involvement in CCA, delving into their expression profiles, prognostic significance, contribution to oncogenic processes, and their potential clinical utility.


Assuntos
Antígeno B7-H1 , Neoplasias dos Ductos Biliares , Colangiocarcinoma , Inibidores de Checkpoint Imunológico , Imunoterapia , Receptor de Morte Celular Programada 1 , Colangiocarcinoma/terapia , Colangiocarcinoma/imunologia , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/patologia , Humanos , Antígeno B7-H1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Neoplasias dos Ductos Biliares/terapia , Neoplasias dos Ductos Biliares/imunologia , Neoplasias dos Ductos Biliares/tratamento farmacológico , Neoplasias dos Ductos Biliares/patologia , Imunoterapia/métodos , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/farmacologia , Animais
7.
J Hepatol ; 80(6): 892-903, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38458319

RESUMO

BACKGROUND & AIMS: Cholangiocarcinoma (CCA) is a poorly immunogenic malignancy associated with limited survival. Syngeneic immunocompetent mouse models of CCA are an essential tool to elucidate the tumor immune microenvironment (TIME), understand mechanisms of tumor immune evasion, and test novel immunotherapeutic strategies. The scope of this study was to develop and characterize immunocompetent CCA models with distinct genetic drivers, and correlate tumor genomics, immunobiology, and therapeutic response. METHODS: A multifaceted approach including scRNA-seq, CITE-seq, whole exome and bulk RNA sequencing was employed. FDA-approved PD-1/PD-L1 antibodies were tested in humanized PD-1/PD-L1 mice (HuPD-H1). RESULTS: A genetic mouse model of intrahepatic CCA (iCCA) driven by intrabiliary transduction of Fbxw7ΔF/Akt that mimics human iCCA was generated. From the Fbxw7ΔF/Akt tumors, a murine cell line (FAC) and syngeneic model with genetic and phenotypic characteristics of human iCCA were developed. Established SB1 (YAPS127A/Akt) and KPPC (KrasG12Dp53L/L) models were compared to the FAC model. Although the models had transcriptomic similarities, they had substantial differences as well. Mutation patterns of FAC, SB1, and KPPC cells matched different mutational signatures in Western and Japanese CCA patient cohorts. KPPC tumors had a high tumor mutation burden. FAC tumors had a T cell-infiltrated TIME, while SB1 tumors had a preponderance of suppressive myeloid cells. FAC, SB1, and KPPC tumors matched different immune signatures in human iCCA cohorts. Moreover, FAC, SB1, and KPPC tumor-bearing HuPD-H1 mice displayed differential responses to nivolumab or durvalumab. CONCLUSIONS: Syngeneic iCCA models display a correlation between tumor genotype and TIME phenotype, with differential responses to FDA-approved immunotherapies. This study underscores the importance of leveraging multiple preclinical models to understand responses to immunotherapy in different genetic subsets of human CCA. IMPACT AND IMPLICATIONS: Understanding the relationship between tumor genotype and the phenotype of the immune microenvironment is an unmet need in cholangiocarcinoma (CCA). Herein, we use syngeneic murine models of intrahepatic CCA with different genetic drivers to demonstrate a correlation between tumor genotype and immune microenvironment phenotype in murine models, which is associated with differential responses to FDA-approved immunotherapies. This information will help guide other preclinical studies. Additionally, it emphasizes that immune checkpoint inhibition in patients with CCA is not a "one-size-fits-all" approach. Our observations suggest that, as for targeted therapies, patients should be stratified and selected for treatment according to their tumor genetics.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Modelos Animais de Doenças , Microambiente Tumoral , Animais , Colangiocarcinoma/imunologia , Colangiocarcinoma/genética , Camundongos , Microambiente Tumoral/imunologia , Humanos , Neoplasias dos Ductos Biliares/imunologia , Neoplasias dos Ductos Biliares/genética , Proteína 7 com Repetições F-Box-WD/genética , Linhagem Celular Tumoral
8.
Neoplasia ; 43: 100920, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37515847

RESUMO

BACKGROUND: Most patients with resected bile tract cancers (BTCs) survive for less than 5 years; however, some achieve better prognosis. The tumor microbiome can improve survival by regulating the tumor immune microenvironment. However, whether the tumor microbiome promotes immune cell infiltration in BTCs is unknown. This study aimed to determine the association between CD8+ T lymphocyte infiltration and the tumor microbiome in patients with resected BTCs. METHODS: Archived formalin-fixed paraffin-embedded tumor specimens were collected from patients with resected BTCs and analyzed using 16S rRNA gene sequencing to identify that prognosis-related and significantly differentially enriched taxa. Gene ontology (GO) analysis of the differentially enriched taxa was used to assess how CD8+ T lymphocyte infiltration is affected by the tumor microbiome of BTCs. RESULTS: We enrolled 32 patients with resected BTCs. The high CD8+ lymphocyte-infiltration (CD8hi) group had four significantly enriched taxa, and in the low CD8+ lymphocyte-infiltration (CD8low) group comprised one significantly enriched taxon. Patients with higher Clostridia abundance (enriched in the CD8hi group) experienced longer overall survival than those with lower abundance. The enrichment of Clostridia in the CD8hi group corresponded with lower CCL2 expression and downregulation of phosphatidylinositol 3-kinase activity, which might decrease myeloid-derived suppressor cell recruitment to the tumor milieu, thus increasing CD8+ lymphocyte infiltration in BTCs. CONCLUSIONS: The tumor microbiome is related to CD8+ T lymphocyte infiltration in patients with resected BTCs. The relationship between tumor Clostridia and high infiltration of CD8+ T lymphocytes might reflect decreased recruitment of myeloid-derived suppressor cells via the PI3K-CCL2-CCR2 axis.


Assuntos
Neoplasias dos Ductos Biliares , Linfócitos T CD8-Positivos , Colangiocarcinoma , Clostridium , Linfócitos do Interstício Tumoral , Microbiota , Humanos , Linfócitos T CD8-Positivos/imunologia , Quimiocina CCL2/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Células Supressoras Mieloides/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Prognóstico , Receptores CCR2/metabolismo , RNA Ribossômico 16S , Microambiente Tumoral/genética , Colangiocarcinoma/imunologia , Colangiocarcinoma/microbiologia , Neoplasias dos Ductos Biliares/imunologia , Neoplasias dos Ductos Biliares/microbiologia , Clostridium/imunologia
9.
Front Immunol ; 13: 845193, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154166

RESUMO

N6-methyladenosine (m6A) has been reported as an important mechanism of post-transcriptional regulation. Programmed death ligand 1 (PD-L1) is a primary immune inhibitory molecule expressed on tumor cells that promotes immune evasion. In addition, seven in absentia homolog 2 (Siah2), a RING E3 ubiquitin ligase, has been involved in tumorigenesis and cancer progression. However, the role of m6A-METTL14-Siah2-PD-L1 axis in immunotherapy remains to be elucidated. In this study, we showed that METTL14, a component of the m6A methyltransferase complex, induced Siah2 expression in cholangiocarcinoma (CCA). METTL14 was shown to enrich m6A modifications in the 3'UTR region of the Siah2 mRNA, thereby promoting its degradation in an YTHDF2-dependent manner. Furthermore, co-immunoprecipitation experiments demonstrated that Siah2 interacted with PD-L1 by promoting its K63-linked ubiquitination. We also observed that in vitro and in vivo Siah2 knockdown inhibited T cells expansion and cytotoxicity by sustaining tumor cell PD-L1 expression. The METTL14-Siah2-PD-L1-regulating axis was further confirmed in human CCA specimens. Analysis of specimens from patients receiving anti-PD1 immunotherapy suggested that tumors with low Siah2 levels were more sensitive to anti-PD1 immunotherapy. Taken together, our results evidenced a new regulatory mechanism of Siah2 by METTL14-induced mRNA epigenetic modification and the potential role of Siah2 in cancer immunotherapy.


Assuntos
Antígeno B7-H1/imunologia , Colangiocarcinoma/imunologia , Proteínas Nucleares/imunologia , Linfócitos T/imunologia , Ubiquitina-Proteína Ligases/imunologia , Adenosina/análogos & derivados , Adenosina/imunologia , Linhagem Celular , Colangiocarcinoma/terapia , Humanos , Imunoterapia , Metiltransferases/imunologia , RNA Mensageiro/imunologia
10.
Br J Cancer ; 126(2): 219-227, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34616011

RESUMO

BACKGROUND: Caveolin-1 (CAV1) in cancer-associated fibroblasts (CAFs) has pro- or anti-tumourigenic effect depending on the cancer type. However, its effect in intrahepatic carcinoma (ICC) remains unknown. Therefore, this study aimed to investigate the relationship between CAV1 in CAFs and tumour-infiltrating lymphocyte (TIL) numbers or PD-L1 levels in ICC patients. METHODS: Consecutive ICC patients (n = 158) were enrolled in this study. The levels of CAV1 in CAFs, CD8 + TILs, Foxp3+ TILs and PD-L1 in cancer cells were analysed using immunohistochemistry. Their association with the clinicopathological factors and prognosis were evaluated. The correlation between these factors was evaluated. RESULTS: CAV1 upregulation in CAFs was associated with a poor overall survival (OS) (P < 0.001) and recurrence-free survival (P = 0.008). Clinicopathological factors were associated with high CA19-9 levels (P < 0.001), advanced tumour stage (P = 0.046) and lymph node metastasis (P = 0.004). CAV1 level was positively correlated with Foxp3+ TIL numbers (P = 0.01). There were no significant correlations between CAV1 levels and CD8 + TIL numbers (P = 0.80) and PD-L1 levels (P = 0.97). An increased CD8 + TIL number and decreased Foxp3+ TIL number were associated with an increased OS. In multivariate analysis, positive CAV1 expression in CAFs (P = 0.013) and decreased CD8 + TIL numbers (P = 0.021) were independent poor prognostic factors. CONCLUSION: Cellular senescence, represented by CAV1 levels, may be a marker of CAFs and a prognostic indicator of ICC through Foxp3+ TIL regulation. CAV1 expression in CAFs can be a therapeutic target for ICC.


Assuntos
Antígeno B7-H1/metabolismo , Fibroblastos Associados a Câncer/patologia , Caveolina 1/metabolismo , Senescência Celular , Colangiocarcinoma/patologia , Fatores de Transcrição Forkhead/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Idoso , Antígeno B7-H1/imunologia , Neoplasias dos Ductos Biliares/imunologia , Neoplasias dos Ductos Biliares/metabolismo , Neoplasias dos Ductos Biliares/patologia , Linfócitos T CD8-Positivos/imunologia , Fibroblastos Associados a Câncer/metabolismo , Colangiocarcinoma/imunologia , Colangiocarcinoma/metabolismo , Feminino , Fatores de Transcrição Forkhead/imunologia , Humanos , Masculino , Prognóstico , Taxa de Sobrevida
11.
Hepatol Commun ; 6(1): 178-193, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34558800

RESUMO

Liver cancers consist primarily of hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). Immune checkpoint inhibitors have emerged as promising therapeutic agents against liver cancers. Programmed cell death protein 1 (PD-1) is an immunoinhibitory receptor present on T cells that interacts with its ligand programmed death-ligand 1 (PD-L1) found on cancer cells. Blocking PD-1/PD-L1 binding improves T-cell survival, proliferation and cytotoxicity, which enhances their antitumor activity. Better understanding of the molecular mechanisms governing PD-1/PD-L1 response is essential to the development of predictive markers and therapeutic combinations that could improve the efficiency of anti-PD-1/PD-L1 treatment. Chemokine-like factor (CKLF)-like MARVEL transmembrane domain-containing 6 (CMTM6) has been recently identified as a major regulator of PD-L1. Another member in the CMTM family, CKLF-like MARVEL transmembrane domain-containing 4 (CMTM4), has been shown to compensate for the effects of CMTM6 when CMTM6 is lost. Interestingly, we found that CMTM4 is the major regulator of PD-L1 in the context of liver cancer. Up-regulated CMTM4 in patients with HCC and ICC is associated with poor patient survival, potentially due to its function in stabilizing PD-L1 expression, hence facilitating escape from T cell-mediated cytotoxicity. We confirmed the role of CMTM4 as a positive regulator of PD-L1 in multiple HCC and ICC cell lines and demonstrated that CMTM4 stabilizes PD-L1 through posttranslational mechanisms. In vivo, suppression of Cmtm4 inhibited HCC growth and increased CD8+ T-cell infiltration in immunocompetent mice. Furthermore, we found that depletion of CMTM4 sensitized HCC tumor to anti-PD-L1 treatment compared with control. This suggests that CMTM4 expression level could be a predictive marker for patient response to anti-PD-L1 treatment, and CMTM4 depletion can potentially be used to enhance the clinical benefits of anti-PD-L1 immunotherapy in patients with liver cancer.


Assuntos
Carcinoma Hepatocelular/imunologia , Colangiocarcinoma/imunologia , Neoplasias Hepáticas/imunologia , Proteínas com Domínio MARVEL/genética , Receptor de Morte Celular Programada 1/imunologia , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Células Cultivadas , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/metabolismo , Hepatócitos , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Proteínas com Domínio MARVEL/antagonistas & inibidores , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Regulação para Cima
12.
Br J Cancer ; 126(1): 10-23, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34400801

RESUMO

Liver cancer is one of the most prevalent cancers, and the third most common cause of cancer-related mortality worldwide. The therapeutic options for the main types of primary liver cancer-hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA)-are very limited. HCC and CCA are immunogenic cancers, but effective immune-mediated tumour control is prevented by their immunosuppressive tumour microenvironment. Despite the critical involvement of key co-inhibitory immune checkpoint interactions in immunosuppression in liver cancer, only a minority of patients with HCC respond to monotherapy using approved checkpoint inhibitor antibodies. To develop effective (combinatorial) therapeutic immune checkpoint strategies for liver cancer, in-depth knowledge of the different mechanisms that contribute to intratumoral immunosuppression is needed. Here, we review the co-inhibitory pathways that are known to suppress intratumoral T cells in HCC and CCA. We provide a detailed description of insights from preclinical studies in cellular crosstalk within the tumour microenvironment that results in interactions between co-inhibitory receptors on different T-cell subsets and their ligands on other cell types, including tumour cells. We suggest alternative immune checkpoints as promising targets, and draw attention to the possibility of combined targeting of co-inhibitory and co-stimulatory pathways to abrogate immunosuppression.


Assuntos
Colangiocarcinoma/imunologia , Proteínas de Checkpoint Imunológico/imunologia , Terapia de Imunossupressão/métodos , Imunoterapia/métodos , Neoplasias Hepáticas/patologia , Microambiente Tumoral , Neoplasias dos Ductos Biliares/tratamento farmacológico , Neoplasias dos Ductos Biliares/imunologia , Neoplasias dos Ductos Biliares/patologia , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/patologia , Ensaios Clínicos como Assunto , Humanos , Proteínas de Checkpoint Imunológico/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/imunologia
13.
Hepatology ; 75(2): 297-308, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34510503

RESUMO

BACKGROUND AND AIMS: Cholangiocarcinoma (CCA) is a deadly and highly therapy-refractory cancer of the bile ducts, with early results from immune checkpoint blockade trials showing limited responses. Whereas recent molecular assessments have made bulk characterizations of immune profiles and their genomic correlates, spatial assessments may reveal actionable insights. APPROACH AND RESULTS: Here, we have integrated immune checkpoint-directed immunohistochemistry with next-generation sequencing of resected intrahepatic CCA samples from 96 patients. We found that both T-cell and immune checkpoint markers are enriched at the tumor margins compared to the tumor center. Using two approaches, we identify high programmed cell death protein 1 or lymphocyte-activation gene 3 and low CD3/CD4/inducible T-cell costimulator specifically in the tumor center as associated with poor survival. Moreover, loss-of-function BRCA1-associated protein-1 mutations are associated with and cause elevated expression of the immunosuppressive checkpoint marker, B7 homolog 4. CONCLUSIONS: This study provides a foundation on which to rationally improve and tailor immunotherapy approaches for this difficult-to-treat disease.


Assuntos
Antígenos CD/metabolismo , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/metabolismo , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos CD/genética , Antígenos B7/genética , Neoplasias dos Ductos Biliares/imunologia , Ductos Biliares Intra-Hepáticos , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linfócitos T CD4-Positivos , Linhagem Celular Tumoral , Colangiocarcinoma/imunologia , Feminino , Expressão Gênica , Genes Supressores de Tumor , Genômica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imuno-Histoquímica , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Mutação com Perda de Função , Masculino , Pessoa de Meia-Idade , Oncogenes/genética , Receptor de Morte Celular Programada 1/genética , Taxa de Sobrevida , Proteínas Supressoras de Tumor/genética , Ubiquitina Tiolesterase/genética , Inibidor 1 da Ativação de Células T com Domínio V-Set/genética , Adulto Jovem , Proteína do Gene 3 de Ativação de Linfócitos
14.
Clin Cancer Res ; 28(3): 540-551, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34785581

RESUMO

PURPOSE: Combined hepatocellular-cholangiocarcinoma (cHCC-CCA) is a rare malignancy associated with an overall poor prognosis. We aimed to investigate the immune profile of cHCC-CCA and determine its impact on disease outcome. EXPERIMENTAL DESIGN: We performed a multicenter study of 96 patients with cHCC-CCA. Gene expression profile was analyzed using nCounter PanCancer IO 360 Panel. Densities of main immune cells subsets were quantified from digital slides of IHC stainings. Genetic alterations were investigated using targeted next-generation sequencing. RESULTS: Two main immune subtypes of cHCC-CCA were identified by clustering analysis: an "immune-high" (IH) subtype (57% of the cases) and an "immune-low" (IL) subtype (43% of the cases). Tumors classified as IH showed overexpression of genes related to immune cells recruitment, adaptive and innate immunity, antigen presentation, cytotoxicity, immune suppression, and inflammation (P < 0.0001). IH cHCC-CCAs also displayed activation of gene signatures recently shown to be associated with response to immunotherapy in patients with HCC. Quantification of immunostainings confirmed that IH tumors were also characterized by higher densities of immune cells. Immune subtypes were not associated with any genetic alterations. Finally, multivariate analysis showed that the IH subtype was an independent predictor of improved overall survival. CONCLUSIONS: We have identified a subgroup of cHCC-CCA that displays features of an ongoing intratumor immune response, along with an activation of gene signatures predictive of response to immunotherapy in HCC. This tumor subclass is associated with an improved clinical outcome. These findings suggest that a subset of patients with cHCC-CCA may benefit from immunomodulating therapeutic approaches.


Assuntos
Neoplasias dos Ductos Biliares/imunologia , Neoplasias dos Ductos Biliares/terapia , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/terapia , Colangiocarcinoma/imunologia , Colangiocarcinoma/terapia , Imunoterapia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/terapia , Neoplasias Primárias Múltiplas/imunologia , Neoplasias Primárias Múltiplas/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias dos Ductos Biliares/diagnóstico , Neoplasias dos Ductos Biliares/genética , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/genética , Colangiocarcinoma/diagnóstico , Colangiocarcinoma/genética , Feminino , Previsões , Humanos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética , Masculino , Pessoa de Meia-Idade , Neoplasias Primárias Múltiplas/diagnóstico , Neoplasias Primárias Múltiplas/genética , Resultado do Tratamento
15.
Cells ; 10(11)2021 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-34831096

RESUMO

Cholangiocarcinomas (CHOLs), hepatobiliary malignancies, are characterized by high genetic heterogeneity, a rich tumor microenvironment, therapeutic resistance, difficulty diagnosing, and poor prognoses. Current knowledge of genetic alterations and known molecular markers for CHOL is insufficient, necessitating the need for further evaluation of the genome and RNA expression data in order to identify potential therapeutic targets, clarify the roles of these targets in the tumor microenvironment, and explore novel therapeutic drugs against the identified targets. Consequently, in our attempt to explore novel genetic markers associated with the carcinogenesis of CHOL, five genes (SNX15, ATP2A1, PDCD10, BET1, and HMGA2), collectively termed CHOL-hub genes, were identified via integration of differentially expressed genes (DEGs) from relatively large numbers of samples from CHOL GEO datasets. We further explored the biological functions of the CHOL-hub genes and found significant enrichment in several biological process and pathways associated with stem cell angiogenesis, cell proliferation, and cancer development, while the interaction network revealed high genetic interactions with a number of onco-functional genes. In addition, we established associations between the CHOL-hub genes and tumor progression, metastasis, tumor immune and immunosuppressive cell infiltration, dysfunctional T-cell phenotypes, poor prognoses, and therapeutic resistance in CHOL. Thus, we proposed that targeting CHOL-hub genes could be an ideal therapeutic approach for treating CHOLs, and we explored the potential of HLC-018, a novel benzamide-linked small molecule, using molecular docking of ligand-receptor interactions. To our delight, HLC-018 was well accommodated with high binding affinities to binding pockets of CHOL-hub genes; more importantly, we found specific interactions of HLC-018 with the conserved sequence of the AT-hook DNA-binding motif of HMGA2. Altogether, our study provides insights into the immune-oncogenic phenotypes of CHOL and provides valuable information for our ongoing experimental validation.


Assuntos
Benzamidas/farmacologia , Colangiocarcinoma/genética , Colangiocarcinoma/imunologia , Oncogenes , Transcriptoma/genética , Motivos AT-Hook , Benzamidas/química , Benzamidas/uso terapêutico , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/patologia , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Progressão da Doença , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes , Proteína HMGA2/química , Humanos , Imunofenotipagem , Ligantes , Simulação de Acoplamento Molecular , Metástase Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Prognóstico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Risco , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Resultado do Tratamento
16.
J Exp Med ; 218(10)2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34495298

RESUMO

Cholangiocarcinoma (CCA) results from the malignant transformation of cholangiocytes. Primary sclerosing cholangitis (PSC) and primary biliary cholangitis (PBC) are chronic diseases in which cholangiocytes are primarily damaged. Although PSC is an inflammatory condition predisposing to CCA, CCA is almost never found in the autoimmune context of PBC. Here, we hypothesized that PBC might favor CCA immunosurveillance. In preclinical murine models of cholangitis challenged with syngeneic CCA, PBC (but not PSC) reduced the frequency of CCA development and delayed tumor growth kinetics. This PBC-related effect appeared specific to CCA as it was not observed against other cancers, including hepatocellular carcinoma. The protective effect of PBC was relying on type 1 and type 2 T cell responses and, to a lesser extent, on B cells. Single-cell TCR/RNA sequencing revealed the existence of TCR clonotypes shared between the liver and CCA tumor of a PBC host. Altogether, these results evidence a mechanistic overlapping between autoimmunity and cancer immunosurveillance in the biliary tract.


Assuntos
Autoimunidade , Neoplasias dos Ductos Biliares/imunologia , Colangiocarcinoma/imunologia , Colangite/imunologia , Animais , Neoplasias dos Ductos Biliares/patologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Colangiocarcinoma/patologia , Colangite/patologia , Citocinas/metabolismo , Feminino , Fatores de Transcrição Forkhead/metabolismo , Fígado/imunologia , Fígado/patologia , Camundongos Endogâmicos C57BL , Monitorização Imunológica , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia
17.
Front Immunol ; 12: 705378, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34526987

RESUMO

Intrahepatic cholangiocarcinoma (ICC) is highly invasive and carries high mortality due to limited therapeutic strategies. In other solid tumors, immune checkpoint inhibitors (ICIs) target cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and programmed death 1 (PD1), and the PD1 ligand PD-L1 has revolutionized treatment and improved outcomes. However, the relationship and clinical significance of CTLA-4 and PD-L1 expression in ICC remains to be addressed. Deciphering CTLA-4 and PD-L1 interactions in ICC enable targeted therapy for this disease. In this study, immunohistochemistry (IHC) was used to detect and quantify CTLA-4, forkhead box protein P3 (FOXP3), and PD-L1 in samples from 290 patients with ICC. The prognostic capabilities of CTLA-4, FOXP3, and PD-L1 expression in ICC were investigated with the Kaplan-Meier method. Independent risk factors related to ICC survival and recurrence were assessed by the Cox proportional hazards models. Here, we identified that CTLA-4+ lymphocyte density was elevated in ICC tumors compared with peritumoral hepatic tissues (P <.001), and patients with a high density of CTLA-4+ tumor-infiltrating lymphocytes (TILsCTLA-4 High) showed a reduced overall survival (OS) rate and increased cumulative recurrence rate compared with patients with TILsCTLA-4 Low (P <.001 and P = .024, respectively). Similarly, patients with high FOXP3+ TILs (TILsFOXP3 High) had poorer prognoses than patients with low FOXP3+ TILs (P = .021, P = .034, respectively), and the density of CTLA-4+ TILs was positively correlated with FOXP3+ TILs (Pearson r = .31, P <.001). Furthermore, patients with high PD-L1 expression in tumors (TumorPD-L1 High) and/or TILsCTLA-4 High presented worse OS and a higher recurrence rate than patients with TILsCTLA-4 LowTumorPD-L1 Low. Moreover, multiple tumors, lymph node metastasis, and high TumorPD-L1/TILsCTLA-4 were independent risk factors of cumulative recurrence and OS for patients after ICC tumor resection. Furthermore, among ICC patients, those with hepatolithiasis had a higher expression of CTLA-4 and worse OS compared with patients with HBV infection or undefined risk factors (P = .018). In conclusion, CTLA-4 is increased in TILs in ICC and has an expression profile distinct from PD1/PD-L1. TumorPD-L1/TILsCTLA-4 is a predictive factor of OS and ICC recurrence, suggesting that combined therapy targeting PD1/PD-L1 and CTLA-4 may be useful in treating patients with ICC.


Assuntos
Antígeno B7-H1/fisiologia , Neoplasias dos Ductos Biliares/imunologia , Antígeno CTLA-4/fisiologia , Colangiocarcinoma/imunologia , Linfócitos do Interstício Tumoral/imunologia , Proteínas de Neoplasias/fisiologia , Receptor de Morte Celular Programada 1/fisiologia , Idoso , Antígeno B7-H1/biossíntese , Antígeno B7-H1/genética , Neoplasias dos Ductos Biliares/mortalidade , Neoplasias dos Ductos Biliares/patologia , Antígeno CTLA-4/biossíntese , Antígeno CTLA-4/genética , Colangiocarcinoma/mortalidade , Colangiocarcinoma/patologia , Feminino , Fatores de Transcrição Forkhead/análise , Perfilação da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Litíase/etiologia , Hepatopatias/etiologia , Linfócitos do Interstício Tumoral/química , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Receptor de Morte Celular Programada 1/biossíntese , Receptor de Morte Celular Programada 1/genética , Modelos de Riscos Proporcionais , Microambiente Tumoral , Regulação para Cima
18.
Int Immunopharmacol ; 99: 108039, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34426102

RESUMO

Cholangiocarcinoma (CHOL), the second most common malignant liver tumor, is clinically heterogeneous. In this study, we used gene expression profiles of CHOL obtained from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases to identify novel mutation signatures in CHOL. Hepcidin antimicrobial peptide (HAMP) was identified as a novel diagnostic biomarker for CHOL using the intersection of mutation analysis and receiver operating characteristic (ROC) analysis. We then explored the expression signatures of HAMP in CHOL. HAMP-related differentially expressed genes (DEGs) were selected for the identification of hub genes related to HAMP and for prognostic prediction model analysis. Gene set enrichment analysis (GSEA) showed that the HAMP-related DEGs were mainly enriched for signaling pathways related to cholangiocarcinoma development. Through immunohistochemistry validation, clinical cohorts analysis, and TCGA analysis, we investigated the association between HAMP and clinical parameters and found that decreased HAMP expression was correlated with advanced pathological grade and poor prognosis. Besides, we estimated the immune infiltration level in CHOL and its relationship with HAMP expression. The proportion of tumor-infiltrating cells revealed that gamma delta T cells and monocytes were positively correlated with HAMP expression. Besides, HAMP was also correlated with chemokine, CCL16. This evidence suggested that HAMP might contribute to immune activation in the CHOL microenvironment. Therefore, HAMP may play a synergistic role with these immune cells and chemokines to inhibit CHOL development. HAMP serves as a valuable biomarker in CHOL and is closely correlated with its progression.


Assuntos
Neoplasias dos Ductos Biliares/genética , Colangiocarcinoma/genética , Hepcidinas/genética , Mutação , Neoplasias dos Ductos Biliares/imunologia , Neoplasias dos Ductos Biliares/metabolismo , Neoplasias dos Ductos Biliares/patologia , Biomarcadores Tumorais/genética , Quimiocinas CC/metabolismo , Colangiocarcinoma/imunologia , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patologia , Biologia Computacional , Correlação de Dados , Mineração de Dados , Bases de Dados Genéticas , Regulação Neoplásica da Expressão Gênica , Hepcidinas/imunologia , Hepcidinas/metabolismo , Humanos , Estimativa de Kaplan-Meier , Neutrófilos/imunologia , Prognóstico , Mapas de Interação de Proteínas , Transcriptoma , Microambiente Tumoral/imunologia
19.
J Immunother ; 44(7): 254-263, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34191790

RESUMO

Cholangiocarcinoma (CC) is an uncommon malignancy with increasing incidence and dismal prognosis. We conducted a comprehensive analysis of the CC tumor immune microenvironment (TIME) based on tumor location to identify therapeutic targets. We hypothesized that the TIME of CC would vary by primary tumor location and that high tumor infiltration by CD8+ T cells and low infiltration by M2 macrophages would be associated with improved survival. A retrospective analysis was conducted of 99 CC tumor samples surgically resected between 2000 and 2014. Tissue microarrays were constructed from each tumor and stained by immunohistochemistry for 24 markers of immune cells, immune activation or inhibition, programmed cell death-ligand 1, and mesothelin. Most tumors were amply infiltrated with by CD4+, CD8+, and FoxP3+ T cells, as well as by myeloid cells. Mesothelin expression ≥1+ by immunohistochemistry was found in 68% of tumors. We identified higher densities of M1 macrophages in primary distal extrahepatic CC, as well as metastatic lesions. Mesothelin expression was also significantly higher in distal extrahepatic CC. There was no association with survival of infiltration by CD4+, CD8+, or FoxP3+ T cells, mesothelin expression, or programmed cell death-ligand 1 percentage expression, however, high CD14+ myeloid cells and high CD163+ M2 macrophages were associated with worse survival. In conclusion, the CC TIME is a heterogenous milieu highly infiltrated by innate and adaptive immune cells, which differs based on primary tumor location and between primary tumors and metastatic lesions. The correlation of intratumoral M2 macrophages and myeloid cells with a worse prognosis may suggest promising immunotherapeutic targets in CC.


Assuntos
Neoplasias dos Ductos Biliares/imunologia , Colangiocarcinoma/imunologia , Células Mieloides/imunologia , Idoso , Antígeno B7-H1/imunologia , Neoplasias dos Ductos Biliares/mortalidade , Neoplasias dos Ductos Biliares/patologia , Colangiocarcinoma/mortalidade , Colangiocarcinoma/patologia , Feminino , Humanos , Masculino , Mesotelina/imunologia , Pessoa de Meia-Idade , Prognóstico , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...