Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Am J Hematol ; 99(2): E32-E36, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37994196

ABSTRACT

The safety and efficacy of sabatolimab, a novel immunotherapy targeting T-cell immunoglobulin domain and mucin domain-3 (TIM-3), was assessed in combination with hypomethylating agents (HMAs) in patients with HMA-naive revised International Prognostic System Score (IPSS-R) high- or very high-risk myelodysplastic syndromes (HR/vHR-MDS) or chronic myelomonocytic leukemia (CMML). Sabatolimab + HMA had a safety profile similar to that reported for HMA alone and demonstrated durable clinical responses in patients with HR/vHR-MDS. These results support the ongoing evaluation of sabatolimab-based combination therapy in MDS, CMML, and acute myeloid leukemia.


Subject(s)
Antibodies, Monoclonal , Leukemia, Myeloid, Acute , Leukemia, Myelomonocytic, Chronic , Myelodysplastic Syndromes , Humans , Azacitidine/therapeutic use , Decitabine/therapeutic use , Antimetabolites, Antineoplastic/therapeutic use , Myelodysplastic Syndromes/drug therapy , Hepatitis A Virus Cellular Receptor 2/therapeutic use , Leukemia, Myelomonocytic, Chronic/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Antibodies/therapeutic use , Treatment Outcome
2.
Med Oncol ; 41(1): 40, 2023 Dec 29.
Article in English | MEDLINE | ID: mdl-38158454

ABSTRACT

Efforts in cancer immunotherapy aim to counteract evasion mechanisms and stimulate the immune system to recognise and attack cancer cells effectively. Combination therapies that target multiple aspects of immune evasion are being investigated to enhance the overall efficacy of cancer immunotherapy. PD-1 (Programmed Cell Death Protein 1), CTLA-4 (Cytotoxic T-Lymphocyte Antigen 4), LAG-3 (Lymphocyte-Activation Gene 3), and TIM-3 (T Cell Immunoglobulin and Mucin Domain-Containing Protein3) are all immune checkpoint receptors that play crucial roles in regulating the immune response and maintaining self-tolerance often exploited by cancer cells to evade immune surveillance. Antibodies targeted against immune checkpoint inhibitors such as anti-PD-1 antibodies (e.g., pembrolizumab, nivolumab), anti-CTLA-4 antibodies (e.g., Ipilimumab), and experimental drugs targeting LAG-3 and TIM-3, aim to block these interactions and unleash the immune system's ability to recognise and destroy cancer cells. The US FDA has approved different categories of immune checkpoint inhibitors that have been utilised successfully in some patients with metastatic melanoma, renal cell carcinoma, head and neck cancers, and non-small lung cancer. Although several immune checkpoint inhibitor antibodies have been developed, they exhibited immune-related adverse effects, resulting in hypophysitis, diabetes, and neurological issues. These adverse effects of antibodies can be reduced by developing aptamer against the target. Aptamers offer several advantages over traditional antibodies, such as improved specificity, reduced immunogenicity, and flexible design for reduced adverse effects that specifically target and block protein-protein or receptor-ligand interactions involved in immune checkpoint pathways. The current study aims to review the function of particular immune checkpoint inhibitors along with developed aptamer-mediated antitumor cytotoxicity in cancer treatment.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Immune Checkpoint Inhibitors , Immunotherapy , Kidney Neoplasms , Melanoma , Humans , CTLA-4 Antigen , Drug-Related Side Effects and Adverse Reactions/etiology , Hepatitis A Virus Cellular Receptor 2/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Kidney Neoplasms/drug therapy , Melanoma/drug therapy
3.
Thorac Cancer ; 14(36): 3556-3560, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37926435

ABSTRACT

Lung spindle cell carcinoma is an aggressive subtype of pleomorphic lung cancer resistant to cytotoxic chemotherapy. Programmed cell death-1 (PD-1) inhibitors have been reported to have clinical effects in patients with spindle cell carcinoma; however, the resistance mechanism to PD-1 inhibitors is yet to be fully elucidated. Herein, we report the case of an 88-year-old man with G-CSF-producing spindle cell carcinoma who acquired resistance to PD-1/PD-ligand 1 (L1) inhibitor in an early setting after a remarkable response. A histopathological review of the resistant specimen revealed a low count of CD8+ T cells and a predominant presence of M2 and TIM-3+ macrophages, indicating the presence of an immunosuppressive microenvironment. Our findings suggest a novel resistance mechanism to PD-1/PD-L1 inhibitors in G-CSF-producing spindle cell carcinoma.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma , Lung Neoplasms , Male , Humans , Aged, 80 and over , Immune Checkpoint Inhibitors/therapeutic use , Granulocyte Colony-Stimulating Factor/therapeutic use , Hepatitis A Virus Cellular Receptor 2/therapeutic use , CD8-Positive T-Lymphocytes/metabolism , Programmed Cell Death 1 Receptor/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung/pathology , B7-H1 Antigen/metabolism , Tumor Microenvironment
4.
Theranostics ; 13(12): 4166-4181, 2023.
Article in English | MEDLINE | ID: mdl-37554274

ABSTRACT

Rationale: Although neoantigen-based cancer vaccines have shown promise in various solid tumors, limited immune responses and clinical outcomes have been reported in patients with advanced disease. Cytosolic transport of neoantigen and adjuvant is required for the activation of intracellular Toll-like receptors (TLRs) and cross-presentation to prime neoantigen-specific CD8+T cells but remains a significant challenge. Methods: In this study, we aimed to develop a virus-like silicon vaccine (V-scVLPs) with a unique spike topological structure, capable of efficiently co-delivering a hepatocellular carcinoma (HCC)-specific neoantigen and a TLR9 agonist to dendritic cells (DCs) to induce a robust CD8+T cell response to prevent orthotopic tumor growth. We evaluated the antitumor efficacy of V-scVLPs by examining tumor growth and survival time in animal models, as well as analyzing tumor-infiltrating CD8+T cells and cytokine responses in the tumor microenvironment (TME). To evaluate the synergistic efficacy of V-scVLPs in combination with α-TIM-3 in HCC, we used an orthotopic HCC mouse model, a lung metastasis model, and a tumor rechallenge model after hepatectomy. Results: We found that V-scVLPs can efficiently co-deliver the hepatocellular carcinoma (HCC)-specific neoantigen and the TLR9 agonist to DCs via caveolin-mediated endocytosis. This advanced delivery strategy results in efficient lymph node draining of V-scVLPs to activate lymphoid DC maturation for promoting robust CD8+T cells and central memory T cells responses, which effectively prevents orthotopic HCC tumor growth. However, in the established orthotopic liver tumor models, the inhibitory receptor of TIM-3 was significantly upregulated in tumor-infiltrating CD8+T cells after immunization with V-scVLPs. Blocking the TIM-3 signaling further restored the antitumor activity of V-scVLPs-induced CD8+T cells, reduced the proportion of regulatory T cells, and increased the levels of cytokines to alter the tumor microenvironment to efficiently suppress established orthotopic HCC tumor growth, and inhibit lung metastasis as well as recurrence after hepatectomy. Conclusion: Overall, the developed novel spike nanoparticles with efficient neoantigen and adjuvant intracellular delivery capability holds great promise for future clinical translation to improve HCC immunotherapy.


Subject(s)
Cancer Vaccines , Carcinoma, Hepatocellular , Liver Neoplasms , Lung Neoplasms , Mice , Animals , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Hepatitis A Virus Cellular Receptor 2/therapeutic use , Toll-Like Receptor 9 , Cytokines/metabolism , CD8-Positive T-Lymphocytes , Cancer Vaccines/therapeutic use , Caveolin 1/therapeutic use , Adjuvants, Immunologic/therapeutic use , Lung Neoplasms/drug therapy , Tumor Microenvironment
5.
Int J Biol Markers ; 38(3-4): 203-213, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37518940

ABSTRACT

BACKGROUND: Epithelial ovarian cancer (EOC) is the leading cause of death associated with gynecologic tumors. EOC is asymptomatic in early stages, so most patients are not diagnosed until late stages, highlighting the need to develop new diagnostic biomarkers. Mediators of the tumoral microenvironment may influence EOC progression and resistance to treatment. AIM: To analyze immune checkpoints to evaluate them as theranostic biomarkers for EOC. PATIENTS AND METHODS: Serum levels of 16 immune checkpoints were determined in EOC patients and healthy controls using the MILLIPLEX MAP® Human Immuno-Oncology Checkpoint Protein Magnetic Bead Panel. RESULTS: Seven receptors: BTLA, CD40, CD80/B7-1, GITRL, LAG-3, TIM-3, TLR-2 are differentially expressed between EOC and healthy controls. Serum levels of immune checkpoints in EOC patients are positively significantly correlated with levels of their ligands, with a higher significant correlation between CD80 and CTLA4 than between CD28 and CD80. Four receptors, CD40, HVEM, PD-1, and PD-L1, are positively associated with the development of resistance to Taxol-platinum-based chemotherapy. All of them have an acceptable area under the curve (>0.7). CONCLUSION: This study has yielded a first panel of seven immune checkpoints (BTLA, CD40, CD80/B7-1, GITRL, LAG-3, TIM-3, TLR-2) associated with a higher risk of EOC and a second panel of four immune checkpoints (CD40, HVEM, PD-1, PD-L1) that may help physicians to identify EOC patients who are at high risk of developing resistance to EOC chemotherapy.


Subject(s)
B7-H1 Antigen , Ovarian Neoplasms , Humans , Female , Carcinoma, Ovarian Epithelial , B7-H1 Antigen/metabolism , Hepatitis A Virus Cellular Receptor 2/therapeutic use , Programmed Cell Death 1 Receptor/metabolism , Toll-Like Receptor 2 , Biomarkers , Ovarian Neoplasms/pathology , Biomarkers, Tumor , Tumor Microenvironment
6.
Orthop Surg ; 15(3): 829-838, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36519392

ABSTRACT

OBJECTIVES: The fact that studies on anti-programmed cell death 1 (PD-1) or its relevant ligand 1 (PD-L1) have yielded such few responses greatly decreases the confidence in immunotherapy with checkpoint inhibitors for advanced osteosarcoma. We intended to characterize the expression of various checkpoint molecules with immunohistochemistry in osteosarcoma specimens and analyzed the relationship of the expression of these checkpoint molecules with patients' clinical courses. METHODS: This study was a retrospective non-intervention study from August 1st 2017 to March 1st 2020. Immunohistochemistry for B7-H3 (CD276, Cluster of Differentiation 276), CD47 (Cluster of Differentiation 47), PD-L1 (programmed cell death ligand 1), TIM3 (mucin-domain containing-3), TGF-ß (TransformingGrowth Factor ß), CXCR 4 (Chemokine Receptor 4), CD27 (Cluster of Differentiation 27), IDO1 (Indoleamine 2,3-dioxygenase 1), KIRs (Killer cell Immunoglobulin-like Receptors), and SDF-1 (Stromal cell-Derived Factor-1) was performed on 35 resected osteosarcoma specimens. Patients progressed upon first-line chemotherapy with evaluable lesions were qualified for this study, and their specimens previously stored in the pathological department repository would be retrieved for analysis. Associations between the immunohischemistry markers and clinicopathological variables and survival were evaluated by the χ2 displayed by cross-table, Cox proportional hazards regression model, and Kaplan-Meier plots. RESULTS: The positive rates of B7-H3, CD47, PD-L1, TIM3, and TGF-ß expression in this sample of 35 heavily treated osteosarcomas were 29% (10/35), 15% (5/35), 9% (3/35), 6% (2/35), and 6% (2/35), respectively, and diverse staining intensities were observed. Among these advanced patients, 15/35 (43%) had positive checkpoint expression, of which 33% (5/15) showed evidence of the co-expression of more than one checkpoint molecule. We did not find any obvious correlation with clinicopathological characteristics and the positive expression of these molecules. CONCLUSIONS: The present study highlights that only a small subset of progressive osteosarcomas, which had been heavily-treated, expressed tumor immune-associated checkpoint molecules, of which B7-H3 was the most positively expressed checkpoint and might be a promising target for further osteosarcoma investigation.


Subject(s)
Bone Neoplasms , Lung Neoplasms , Osteosarcoma , Humans , Lung Neoplasms/pathology , B7-H1 Antigen/analysis , B7-H1 Antigen/metabolism , CD47 Antigen/therapeutic use , Retrospective Studies , Clinical Relevance , Hepatitis A Virus Cellular Receptor 2/therapeutic use , Osteosarcoma/pathology , Bone Neoplasms/pathology , Immunotherapy , B7 Antigens
7.
Aliment Pharmacol Ther ; 57(6): 689-708, 2023 03.
Article in English | MEDLINE | ID: mdl-36411952

ABSTRACT

BACKGROUND: Hepatitis B surface antigen (HBsAg) seroconversion is sometimes observed in hepatitis B reactivation (rHBV), probably due to immune resetting and differentiation. AIMS: To investigate sequential immune differentiation and abrogation of tolerance in patients with rHBV who achieved HBsAg seroconversion. METHODS: We included 19 patients with chronic hepatitis B (CHBV; HBV DNA log103-8 ), 67 with rHBV (raised ALT [>5XULN], HBV DNAlog104-8 ) and 10 healthy controls. Immune differentiation, tolerance and functional status of CD4, CD8, T regulatory cells (Tregs), B cells and follicular T helper (Tfh) cells were assessed at baseline and 24 weeks. RESULTS: At 24 weeks, 81% rHBV (n = 67) lost HBV DNA and HBeAg (41%), and 12 (19%) lost HBsAg and made anti-HBs titers >10 IU/ml. rHBV patients had higher Th1/17, TEM , Tfh, Tfh1/17, plasma and ATM B cells, and lower Tregs, Th2, Th17 and TEMRA expression. rHBV showed lower PD1, TIM3, LAG3, SLAM and TOX compared to CHBV. There was a significant increase in CD8, CD8EM, Tfh, Tfh1/17 and plasma B cells in seroconverters than non-seroconverters. At 24 weeks, we also observed increased plasma B cell frequency in seroconverters. While non-seroconverters showed higher expression of PD1, TIM3, LAG3, SLAM and TOX on CD4/CD8 T cells, blockade of PD1, TIM3, LAG3 and CTLA4 significantly enhanced IFN-γ, TNF-α, IL-4 and IL-21 expression on CD4/CD8 and Tfh cells in non-seroconverters. CONCLUSIONS: Non-seroconverters have increased inhibitory markers on CD4/CD8 T cells. There is a critical play of CD8, Tfh and B cells and subsets in seroclearance, along with checkpoint molecules as a potential therapy for non-seroconverters in HBV infection.


Subject(s)
Hepatitis B, Chronic , Hepatitis B , Humans , Hepatitis B Surface Antigens , Hepatitis B virus/genetics , DNA, Viral , Seroconversion , Hepatitis A Virus Cellular Receptor 2/therapeutic use , Hepatitis B/drug therapy , Hepatitis B, Chronic/drug therapy , Hepatitis B e Antigens , Antiviral Agents/therapeutic use
8.
BMC Infect Dis ; 22(1): 842, 2022 Nov 11.
Article in English | MEDLINE | ID: mdl-36368952

ABSTRACT

PURPOSE: This study was designed to analyze the liver tissue changes among the CHB patients who received treatment for at least 6 months and follow-up for at least 1 year, together with the correlation between the different disease condition and serum markers. METHODS: One-hundred and eighty-five CHB patients underwent antiviral therapy for at least 6 months were enrolled. In the 12-month follow-up, ultrasonography-guided biopsy was performed. The patients were grouped based on the serum markers and pathological changes in liver tissues. Then we determined the serum markers, virological tests and Tim-3 expression among these groups. RESULTS: Antiviral therapy significantly reduced liver inflammation indicators and serum Tim-3 level. However, the fibrosis process of liver tissue was not changed, and there are still disputes on the serum marker and hepatic lesion outcomes. Under normal liver function or negative hepatitis B e antigen (HBeAg) of CHB patients, there might be consensus between Tim-3 change and liver pathological outcome. According to the liver tissue inflammation and fibrosis conditions, Tim-3 was positively correlated with liver function indices. Besides, it was also related to fibrosis stage and inflammation grade. CONCLUSION: There were inconsistent changes between serum markers and liver tissue conditions after anti-viral therapy. Tim-3 expression was more suitable to indicate the changes of liver inflammatory and fibrosis response to some extent than ALT and AST. It may serve as a certain indicator to predict the CHB prognosis, which could be used as one of the monitoring indicators in liver pathological changes of chronic HBV infection, especially in monitoring liver tissue inflammation.


Subject(s)
Hepatitis B virus , Hepatitis B, Chronic , Humans , Hepatitis B virus/genetics , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Hepatitis A Virus Cellular Receptor 2/therapeutic use , Hepatitis B e Antigens , Liver/pathology , Prognosis , Inflammation/drug therapy , Fibrosis , Biomarkers/metabolism , Antiviral Agents/therapeutic use , DNA, Viral , Alanine Transaminase
9.
Psychosom Med ; 84(8): 976-983, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36162059

ABSTRACT

OBJECTIVE: We examined individual differences in CD4/CD8 T-cell ratio trajectories and associated risk profiles from acute HIV infection (AHI) through 144 weeks of antiretroviral therapy (ART) using a data-driven approach. METHODS: A total of 483 AHI participants began ART during Fiebig I-V and completed follow-up evaluations for 144 weeks. CD4+, CD8+, and CD4/CD8 T-cell ratio trajectories were defined followed by analyses to identify associated risk variables. RESULTS: Participants had a median viral load (VL) of 5.88 copies/ml and CD4/CD8 T-cell ratio of 0.71 at enrollment. After 144 weeks of ART, the median CD4/CD8 T-cell ratio was 1.3. Longitudinal models revealed five CD4/CD8 T-cell ratio subgroups: group 1 (3%) exhibited a ratio >1.0 at all visits; groups 2 (18%) and 3 (29%) exhibited inversion at enrollment, with normalization 4 and 12 weeks after ART, respectively; and groups 4 (31%) and 5 (18%) experienced CD4/CD8 T-cell ratio inversion due to slow CD4+ T-cell recovery (group 4) or high CD8+ T-cell count (group 5). Persistent inversion corresponded to ART onset after Fiebig II, higher VL, soluble CD27 and TIM-3, and lower eosinophil count. Individuals with slow CD4+ T-cell recovery exhibited higher VL, lower white blood cell count, lower basophil percent, and treatment with standard ART, as well as worse mental health and cognition, compared with individuals with high CD8+ T-cell count. CONCLUSIONS: Early HIV disease dynamics predict unfavorable CD4/CD8 T-cell ratio outcomes after ART. CD4+ and CD8+ T-cell trajectories contribute to inversion risk and correspond to specific viral, immune, and psychological profiles during AHI. Adjunctive strategies to achieve immune normalization merit consideration.


Subject(s)
Anti-HIV Agents , HIV Infections , Anti-HIV Agents/therapeutic use , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes , HIV Infections/drug therapy , Hepatitis A Virus Cellular Receptor 2/therapeutic use , Humans , Individuality , Viral Load
10.
Clin Breast Cancer ; 22(8): e901-e915, 2022 12.
Article in English | MEDLINE | ID: mdl-36089459

ABSTRACT

INTRODUCTION: Immunotherapy has been determined as an important choice in breast carcinomas, especially in tumors with markedly inflammatory response. About this promising subject, tumor-infiltrating lymphocytes (TIL) and the expression of immune control point receptors on TIL have gained importance. MATERIALS AND METHODS: In this study, stromal TIL and tertiary lymphoid structures (TLS) were determined in tumor tissues of 312 invasive and 68 in situ breast cancer patients. Expression rates of PD-1, LAG-3, and TIM-3 on intratumoral and stromal TIL were immunohistochemically evaluated. RESULTS: In invasive breast carcinomas, stromal TIL was found to be significantly associated with lymph node metastasis, HR and HER2 expression, and basal-like phenotype, as the presence of TLS with neoadjuvant therapy, recurrence, death, and expression of HR and HER2. PD-1, LAG-3, and TIM-3 expressions were found to be associated with HR and HER2 status, stromal TIL rates, and TLS. In multivariate analysis, high stromal TIL and PD-1 expression in intratumoral TIL were found to be independent prognostic factors in terms of overall survival and disease-free survival. CONCLUSION: Evaluation of TIL and immune control point receptor expressions in breast cancer is particularly important in terms of planning the therapeutic approaches based on immunotherapy protocols.


Subject(s)
Breast Neoplasms , Carcinoma, Intraductal, Noninfiltrating , Tertiary Lymphoid Structures , Humans , Female , Lymphocytes, Tumor-Infiltrating , Tertiary Lymphoid Structures/pathology , Programmed Cell Death 1 Receptor , Hepatitis A Virus Cellular Receptor 2/therapeutic use , Prognosis , Breast Neoplasms/drug therapy , Carcinoma, Intraductal, Noninfiltrating/pathology
11.
Med Sci Monit ; 28: e935326, 2022 Jul 21.
Article in English | MEDLINE | ID: mdl-35859350

ABSTRACT

BACKGROUND Hepatocellular carcinoma (HCC) is currently a leading cause of cancer-related death, and its prognostic evaluation remains a challenge. We aimed to explore the predictive value of T cell immunoglobulin and mucin 3 (Tim-3) in HCC patients undergoing transcatheter arterial chemoembolization (TACE). MATERIAL AND METHODS For the present study, 167 HCC patients who had undergone TACE were retrospectively recruited and randomly stratified into 2 subgroups at a ratio of about 2: 1 (training: 109; validation: 58) using EXCEL software. Pre-TACE serum was collected from all of these patients, and in a second visit a month after the initial treatment, an additional serum sample was collected from 45 patients in the training set. Tim-3 concentrations were determined by enzyme-linked immunosorbent assay. Kaplan-Meier survival curves analysis, log-rank tests, and Cox proportional hazard regression model were applied to evaluate prognostic significance. RESULTS Patients whose cancer progressed after TACE had significantly higher serum Tim-3 in both the training and validation sets (both P<0.05). Kaplan-Meier analysis revealed that patients with elevated serum Tim-3 had significantly shorter time-to-progression and overall survival in both the training and validation sets (both P<0.05). Multivariate Cox regression analysis confirmed that elevated pre-TACE Tim-3 served as a novel, independent indicator for predicting poor prognosis in HCC [progression, hazard ratio (HR) 2.197; 95% confidential interval (CI) 1.408-3.373; P<0.001; death, HR 2.570; 95% CI 1.508-4.378; P=0.001], which was further verified in the validation set (progression P=0.005; death P=0.043). Interestingly, serum Tim-3 retained its prognostic significance in the a-fetoprotein-negative subgroup. Notably, patients with high Tim-3 levels after TACE encountered dismal outcomes. CONCLUSIONS Serum Tim-3 might be a satisfactory prognostic indicator for HCC patients undergoing TACE.


Subject(s)
Carcinoma, Hepatocellular , Chemoembolization, Therapeutic , Liver Neoplasms , Carcinoma, Hepatocellular/pathology , Chemoembolization, Therapeutic/methods , Hepatitis A Virus Cellular Receptor 2/therapeutic use , Humans , Immunoglobulins , Liver Neoplasms/pathology , Mucin-3 , Prognosis , Retrospective Studies , T-Lymphocytes/pathology , Treatment Outcome
12.
Leuk Lymphoma ; 63(1): 152-161, 2022 01.
Article in English | MEDLINE | ID: mdl-34405769

ABSTRACT

Acute myeloid leukemia (AML) with t(8;21) is a heterogeneous disease and needs to be stratified. Both, cancer cells and immune cells participate in tumor initiation, growth and progression and might affect clinical outcomes. TIM-3 (T cell immunoglobulin and mucin domain-containing protein 3), an immune checkpoint molecule, is expressed not only on immune cells but also on leukemic stem cells (LSCs) in AML. This prompted us to investigate the prognostic significance of TIM-3 in t(8;21) AML. A total of 47 t(8;21) AML patients were tested for TIM-3 expression by multi-parameter flow cytometry at diagnosis. 35 of these, who received chemotherapy alone or along with allogeneic hematopoietic stem cell transplantation were followed up. The expression pattern of TIM-3 on T-cells and NK (natural killer) cells as a whole (T + NK) and LSCs were evaluated independently. High percentage of T + NK - TIM-3+ and CD34+CD38-TIM-3+ cells were significantly associated with a high 2-year cumulative incidence of relapse (CIR) (p = 0.028, 0.016). Further, concurrent high frequencies of T + NK-TIM-3+ and CD34+CD38-TIM-3+ cells at diagnosis were significantly associated with a high 2-year CIR (p < 0.0001) and this together with c-KIT D816 mutation were the independent adverse prognostic factors for relapse (hazard ratio (HR)=2.5, [95% confidence interval (CI), 1.1-6.0], p = 0.04; HR = 46.5, [95% CI, 2.7-811.5], p = 0.009). In conclusion, the expression pattern of TIM-3 on both T and NK cells and LSCs at diagnosis had prognostic significance in t (8;21) AML.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Leukemia, Myeloid, Acute , Antigens, CD34/metabolism , Flow Cytometry , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Hepatitis A Virus Cellular Receptor 2/therapeutic use , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Neoplastic Stem Cells/metabolism , Prognosis
13.
Clin Cancer Res ; 27(23): 6393-6404, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34465599

ABSTRACT

PURPOSE: Immune checkpoint inhibitors show high response rates and durable clinical benefit in microsatellite instability-high/mismatch repair-deficient (MSI-H/dMMR) tumors. However, 50%-60% do not respond to single-agent anti-programmed death-1/programmed death ligand 1 (PD-1/PD-L1) antibodies, and approximately 50% of responders relapse within 6-12 months. This phase Ib trial evaluated safety and antitumor activity of anti-PD-L1 antibody LY3300054 monotherapy or in combination with anti-TIM-3 antibody LY3321367 in patients with MSI-H/dMMR advanced solid tumors. PATIENTS AND METHODS: Eligible patients ≥18 years without prior anti-PD-1/PD-L1 therapy received LY3300054 monotherapy (N = 40) or combination (N = 20); patients with PD-1/PD-L1 inhibitor-resistant/refractory tumors received the combination (N = 22). LY3300054 (700 mg) and anti-TIM-3 antibody (cycles 1-2: 1,200 mg, cycle 3 onward: 600 mg) were administered intravenously every 2 weeks. Primary endpoints were safety and tolerability. RESULTS: Eighty-two patients were enrolled. Most had colorectal (n = 39, 47.6%) or endometrial (n = 14, 17.1%) tumors. More than 70% of patients in the PD-1/PD-L1 inhibitor-resistant/refractory combination cohort had received ≥3 treatment lines. Treatment-related adverse events (TRAE) occurred in 22 patients (55.0%) receiving monotherapy, 13 (65.0%) in the PD-1/PD-L1 inhibitor-naïve combination cohort, and 6 (27.3%) in the PD-1/PD-L1 inhibitor-resistant/refractory combination cohort. A total of 2 patients (5.0%) receiving monotherapy and 3 (7.1%) receiving the combination experienced grade ≥3 TRAEs. Objective responses occurred in 13 patients (32.5%) with monotherapy, 9 (45.0%) in the PD-1/PD-L1 inhibitor-naïve combination cohort, and 1 patient (4.5%) in the PD-1/PD-L1 inhibitor-resistant/refractory combination cohort. CONCLUSIONS: LY3300054 monotherapy and combined LY3300054/anti-TIM-3 had manageable safety profiles. Both regimens showed promising clinical activity against PD-1/PD-L1 inhibitor-naïve MSI-H/dMMR tumors. The combination had limited clinical benefit in patients with PD-1/PD-L1 inhibitor-resistant/refractory MSI-H/dMMR tumors.


Subject(s)
B7-H1 Antigen , Neoplasms , Antineoplastic Combined Chemotherapy Protocols/adverse effects , B7-H1 Antigen/adverse effects , DNA Mismatch Repair , Hepatitis A Virus Cellular Receptor 2/therapeutic use , Humans , Microsatellite Instability , Neoplasms/drug therapy , Neoplasms/genetics
14.
Expert Rev Anticancer Ther ; 21(5): 523-534, 2021 05.
Article in English | MEDLINE | ID: mdl-33334180

ABSTRACT

INTRODUCTION: Dysfunction of the immune system is a hallmark of cancer. Through increased understanding of the complex interactions between immunity and cancer, immunotherapy has emerged as a treatment modality for different types of cancer. Promising activity with immunotherapy has been reported in numerous malignancies, but challenges such as limited response rates and treatment resistance remain. Furthermore, outcomes with this therapeutic approach in hematologic malignancies are even more limited than in solid tumors. T-cell immunoglobulin domain and mucin domain 3 (TIM-3) has emerged as a potential immune checkpoint target in both solid tumors and hematologic malignancies. TIM-3 has been shown to promote immune tolerance, and overexpression of TIM-3 is associated with more aggressive or advanced disease and poor prognosis. AREAS COVERED: This review examines what is currently known regarding the biology of TIM-3 and clinical implications of targeting TIM-3 in cancer. Particular focus is given to myeloid malignancies. EXPERT OPINION: The targeting of TIM-3 is a promising therapeutic approach in cancers, including hematologic cancers such as myeloid malignancies which have not benefited much from current immunotherapeutic treatment approaches. We anticipate that with further clinical evaluation, TIM-3 blockade will emerge as an important treatment strategy in myeloid malignancies.


Subject(s)
Hematologic Neoplasms , Neoplasms , Hematologic Neoplasms/therapy , Hepatitis A Virus Cellular Receptor 2/metabolism , Hepatitis A Virus Cellular Receptor 2/therapeutic use , Humans , Immunotherapy , Neoplasms/pathology
15.
Med Sci Monit ; 23: 3593-3602, 2017 Jul 24.
Article in English | MEDLINE | ID: mdl-28736431

ABSTRACT

BACKGROUND Glioblastoma multiforme (GBM) evades immune surveillance by inducing immunosuppression via receptor-ligand interactions between immune checkpoint molecules. T cell immunoglobulin and mucin domain 3 (Tim-3) is a key checkpoint receptor responsible for exhaustion and dysfunction of T cells and plays a critical role in immunosuppression. Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) has been recently identified as a heterophilic ligand for Tim-3. MATERIAL AND METHODS We established an intracranial GBM model using C57BL/6 mice and GL261 cells, and treated the mice with single or combined monoclonal antibodies (mAbs) against Tim-3/CEACAM1. The CD4+, CD8+, and regulatory T cells in brain-infiltrating lymphocytes were analyzed using flow cytometry, and the effector function of T cells was assessed using ELISA. We performed a rechallenge by subcutaneous injection of GL261 cells in the "cured" (>90 days post-orthotopic tumor implantation) and naïve mice. RESULTS The mean survival time in the control, anti-Tim-3, anti-CEACAM1, and combined treatment groups was 29.8, 43.4, 42.3, and 86.0 days, respectively, with 80% of the mice in the combined group becoming long-term survivors showing immune memory against glioma cells. Infiltrating CD4+ and CD8+ T cells increased and immunosuppressive Tregs decreased with the combined therapy, which resulted in a markedly elevated ratio of CD4+ and CD8+ cells to Tregs. Additionally, plasma IFN-γ and TGF-ß levels were upregulated and downregulated, respectively. CONCLUSIONS Our data indicate that combined blockade of Tim-3 and CEACAM1 generates robust therapeutic efficacy in mice with intracranial tumors, and provides a promising option for GBM immunotherapy.


Subject(s)
Carcinoembryonic Antigen/therapeutic use , Glioma/pathology , Hepatitis A Virus Cellular Receptor 2/physiology , Hepatitis A Virus Cellular Receptor 2/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Antilymphocyte Serum/therapeutic use , Brain Neoplasms/metabolism , CD8-Positive T-Lymphocytes/immunology , Carcinoembryonic Antigen/metabolism , Carcinoembryonic Antigen/physiology , Disease Models, Animal , Glioblastoma , Glioma/drug therapy , Hepatitis A Virus Cellular Receptor 2/metabolism , Immune Tolerance/immunology , Immunotherapy , Mice , Mice, Inbred C57BL , Receptors, Virus/metabolism , Transforming Growth Factor beta/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...