Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.771
Filter
1.
Nat Commun ; 15(1): 5932, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39013886

ABSTRACT

PD-1/PD-L1 blockade has so far shown limited survival benefit for high-grade ovarian carcinomas. By using paired samples from the NeoPembrOv randomized phase II trial (NCT03275506), for which primary outcomes are published, and by combining RNA-seq and multiplexed immunofluorescence staining, we explore the impact of NeoAdjuvant ChemoTherapy (NACT) ± Pembrolizumab (P) on the tumor environment, and identify parameters that correlated with response to immunotherapy as a pre-planned exploratory analysis. Indeed, i) combination therapy results in a significant increase in intraepithelial CD8+PD-1+ T cells, ii) combining endothelial and monocyte gene signatures with the CD8B/FOXP3 expression ratio is predictive of response to NACT + P with an area under the curve of 0.93 (95% CI 0.85-1.00) and iii) high CD8B/FOXP3 and high CD8B/ENTPD1 ratios are significantly associated with positive response to NACT + P, while KDR and VEGFR2 expression are associated with resistance. These results indicate that targeting regulatory T cells and endothelial cells, especially VEGFR2+ endothelial cells, could overcome immune resistance of ovarian cancers.


Subject(s)
Antibodies, Monoclonal, Humanized , Neoadjuvant Therapy , Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/immunology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Neoadjuvant Therapy/methods , Antibodies, Monoclonal, Humanized/therapeutic use , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Neoplasm Grading , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/antagonists & inhibitors , Immunotherapy/methods
2.
ACS Appl Mater Interfaces ; 16(28): 36131-36141, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38979627

ABSTRACT

Cancer immunotherapy is developing as the mainstream strategy for treatment of cancer. However, the interaction between the programmed cell death protein-1 (PD-1) and the programmed death ligand 1 (PD-L1) restricts T cell proliferation, resulting in the immune escape of tumor cells. Recently, immune checkpoint inhibitor therapy has achieved clinical success in tumor treatment through blocking the PD-1/PD-L1 checkpoint pathway. However, the presence of M2 tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) will inhibit antitumor immune responses and facilitate tumor growth, which can weaken the effectiveness of immune checkpoint inhibitor therapy. The repolarization of M2 TAMs into M1 TAMs can induce the immune response to secrete proinflammatory factors and active T cells to attack tumor cells. Herein, hollow iron oxide (Fe3O4) nanoparticles (NPs) were prepared for reprogramming M2 TAMs into M1 TAMs. BMS-202, a small-molecule PD-1/PD-L1 inhibitor that has a lower price, higher stability, lower immunogenicity, and higher tumor penetration ability compared with antibodies, was loaded together with pH-sensitive NaHCO3 inside hollow Fe3O4 NPs, followed by wrapping with macrophage membranes. The formed biomimetic FBN@M could produce gaseous carbon dioxide (CO2) from NaHCO3 in response to the acidic TME, breaking up the macrophage membranes to release BMS-202. A series of in vitro and in vivo assessments revealed that FBN@M could reprogram M2 TAMs into M1 TAMs and block the PD-1/PD-L1 pathway, which eventually induced T cell activation and the secretion of TNF-α and IFN-γ to kill the tumor cells. FBN@M has shown a significant immunotherapeutic efficacy for tumor treatment.


Subject(s)
Immune Checkpoint Inhibitors , Immunotherapy , Animals , Mice , Immune Checkpoint Inhibitors/chemistry , Immune Checkpoint Inhibitors/pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/therapy , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology , Tumor Microenvironment/drug effects , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Line, Tumor , Magnetic Iron Oxide Nanoparticles/chemistry , Female , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism
3.
Nat Commun ; 15(1): 5939, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39009599

ABSTRACT

The precise regulation of protein function is essential in biological systems and a key goal in chemical biology and protein engineering. Here, we describe a straightforward method to engineer functional control into the isopeptide bond-forming SpyTag/SpyCatcher protein ligation system. First, we perform a cysteine scan of the structured region of SpyCatcher. Except for two known reactive and catalytic residues, none of these mutations abolish reactivity. In a second screening step, we modify the cysteines with disulfide bond-forming small molecules. Here we identify 8 positions at which modifications strongly inhibit reactivity. This inhibition can be reversed by reducing agents. We call such a reversibly inhibitable SpyCatcher "SpyLock". Using "BiLockCatcher", a genetic fusion of wild-type SpyCatcher and SpyLock, and SpyTagged antibody fragments, we generate bispecific antibodies in a single, scalable format, facilitating the screening of a large number of antibody combinations. We demonstrate this approach by screening anti-PD-1/anti-PD-L1 bispecific antibodies using a cellular reporter assay.


Subject(s)
Antibodies, Bispecific , Cysteine , Protein Engineering , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/immunology , Antibodies, Bispecific/chemistry , Humans , Protein Engineering/methods , Cysteine/chemistry , Cysteine/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , HEK293 Cells , Disulfides/chemistry , Animals
4.
Sci Rep ; 14(1): 16257, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39009619

ABSTRACT

In order to comprehend the dynamics of disease propagation within a society, mathematical formulations are essential. The purpose of this work is to investigate the diagnosis and treatment of lung cancer in persons with weakened immune systems by introducing cytokines ( I L 2 & I L 12 ) and anti-PD-L1 inhibitors. To find the stable position of a recently built system TCD I L 2 I L 12 Z, a qualitative and quantitative analysis are taken under sensitive parameters. Reliable bounded findings are ensured by examining the generated system's boundedness, positivity, uniqueness, and local stability analysis, which are the crucial characteristics of epidemic models. The positive solutions with linear growth are shown to be verified by the global derivative, and the rate of impact across every sub-compartment is determined using Lipschitz criteria. Using Lyapunov functions with first derivative, the system's global stability is examined in order to evaluate the combined effects of cytokines and anti-PD-L1 inhibitors on people with weakened immune systems. Reliability is achieved by employing the Mittag-Leffler kernel in conjunction with a fractal-fractional operator because FFO provide continuous monitoring of lung cancer in multidimensional way. The symptomatic and asymptomatic effects of lung cancer sickness are investigated using simulations in order to validate the relationship between anti-PD-L1 inhibitors, cytokines, and the immune system. Also, identify the actual state of lung cancer control with early diagnosis and therapy by introducing cytokines and anti-PD-L1 inhibitors, which aid in the patients' production of anti-cancer cells. Investigating the transmission of illness and creating control methods based on our validated results will both benefit from this kind of research.


Subject(s)
B7-H1 Antigen , CD8-Positive T-Lymphocytes , Lung Neoplasms , Humans , CD8-Positive T-Lymphocytes/immunology , Lung Neoplasms/immunology , Lung Neoplasms/drug therapy , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Cytokines/metabolism , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Computer Simulation
5.
Protein Sci ; 33(8): e5106, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39012010

ABSTRACT

Miniproteins constitute an excellent basis for the development of structurally demanding functional molecules. The engrailed homeodomain, a three-helix-containing miniprotein, was applied as a scaffold for constructing programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) interaction inhibitors. PD-L1 binders were initially designed using the computer-aided approach and subsequently optimized iteratively. The conformational stability was assessed for each obtained miniprotein using circular dichroism spectroscopy, indicating that numerous mutations could be introduced. The formation of a sizable hydrophobic surface at the inhibitor that fits the molecular target imposed the necessity for the incorporation of additional charged amino acid residues to retain its appropriate solubility. Finally, the miniprotein effectively binding to PD-L1 (KD = 51.4 nM) that inhibits PD-1/PD-L1 interaction in cell-based studies with EC50 = 3.9 µM, was discovered.


Subject(s)
B7-H1 Antigen , Programmed Cell Death 1 Receptor , Protein Engineering , Programmed Cell Death 1 Receptor/chemistry , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , B7-H1 Antigen/chemistry , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , Humans , Protein Binding , Models, Molecular , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics
6.
Cancer Immunol Immunother ; 73(9): 178, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38954031

ABSTRACT

Intracranial tumors present a significant therapeutic challenge due to their physiological location. Immunotherapy presents an attractive method for targeting these intracranial tumors due to relatively low toxicity and tumor specificity. Here we show that SCIB1, a TRP-2 and gp100 directed ImmunoBody® DNA vaccine, generates a strong TRP-2 specific immune response, as demonstrated by the high number of TRP2-specific IFNγ spots produced and the detection of a significant number of pentamer positive T cells in the spleen of vaccinated mice. Furthermore, vaccine-induced T cells were able to recognize and kill B16HHDII/DR1 cells after a short in vitro culture. Having found that glioblastoma multiforme (GBM) expresses significant levels of PD-L1 and IDO1, with PD-L1 correlating with poorer survival in patients with the mesenchymal subtype of GBM, we decided to combine SCIB1 ImmunoBody® with PD-1 immune checkpoint blockade to treat mice harboring intracranial tumors expressing TRP-2 and gp100. Time-to-death was significantly prolonged, and this correlated with increased CD4+ and CD8+ T cell infiltration in the tissue microenvironment (TME). However, in addition to PD-L1 and IDO, the GBM TME was found to contain a significant number of immunoregulatory T (Treg) cell-associated transcripts, and the presence of such cells is likely to significantly affect clinical outcome unless also tackled.


Subject(s)
Brain Neoplasms , Cancer Vaccines , Immune Checkpoint Inhibitors , Programmed Cell Death 1 Receptor , Vaccines, DNA , Animals , Female , Humans , Mice , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Glioblastoma/immunology , Glioblastoma/therapy , Glioblastoma/drug therapy , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Intramolecular Oxidoreductases , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use , Male , Child , Middle Aged
7.
Cancer Immunol Immunother ; 73(9): 175, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38953994

ABSTRACT

Tumor immunotherapies targeting PD-(L)1 exhibit anti-tumor efficacy in only 10-30% of patients with various cancers. Literature has demonstrated that a "hot tumor" which contains high T lymphocytes in the tumor microenvironment exhibits a better response to immunotherapies than a "cold tumor." This study aimed to investigate whether tumor-intrinsic IFNα and CXCL10 determine the recruitment and activation of CD8+ T cells to become "hot tumor." In this study, we found that CXCL10 overexpressed in a variety of tumors including lung, colon, and liver tumors with a correlation with PD-L1. High PD-L1 and CXCL10 are associated with better survival rates in tumor patients receiving immunotherapies. IFNs-downstream transcriptional factor IRF-1 and STAT1 were correlated with PD-L1 and CXCL10 expression. We demonstrated that IRF-1 and STAT1 were both bound with the promoters of PD-L1 and CXCL10, sharing the same signaling pathway and determining IFNs-mediated PD-L1 and CXCL10 expression. In addition, IFNα significantly increased activation marker IFNγ in PBMCs, promoting M1 type monocyte differentiation, CD4+ T, and CD8+ T cell activation. Particularly, we found that CD8+ T lymphocytes abundantly expressed CXCR3, a receptor of CXCL10, by flow cytometry, indicating that tumor-intrinsic CXCL10 potentially recruited CD8+ T in tumor microenvironment. To demonstrate the hypothesis, immunotherapy-sensitive CT26 and immunotherapy-resistant LL/2 were used and we found that CT26 cells exhibited higher IFNα, IFNγ, CXCL10, and PD-L1 levels compared to LL/2, leading to higher IFNγ expression in mouse splenocytes. Moreover, we found that CD8+ T cells were recruited by CXCL10 in vitro, whereas SCH546738, an inhibitor of CXCR3, inhibited T cell migration and splenocytes-mediated anti-tumor effect. We then confirmed that CT26-derived tumor was sensitive to αPD-L1 immunotherapy and LL/2-tumor was resistant, whereas αPD-L1 significantly increased T lymphocyte activation marker CD107a in CT26-derived BALB/c mice. In conclusion, this study revealed that CXCL10 expression is correlated with PD-L1 in tumors, sharing the same signaling pathway and associating with better immunotherapeutic efficacy. Further evidence in the syngeneic tumor models demonstrated that immunotherapy-sensitive CT26 intrinsically exhibited higher IFNα and CXCL10 compared to immunotherapy-resistant LL/2 to recruit and activate CD8+ T cells in the tumor microenvironment, exhibiting "hot tumor" characteristic of sensitizing αPD-L1 immunotherapies.


Subject(s)
Chemokine CXCL10 , Immunotherapy , Interferon-alpha , Tumor Microenvironment , Chemokine CXCL10/metabolism , Chemokine CXCL10/immunology , Tumor Microenvironment/immunology , Animals , Mice , Humans , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Lymphocyte Activation/immunology , Cell Line, Tumor , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Female , STAT1 Transcription Factor/metabolism
8.
Eur J Med Chem ; 275: 116622, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38959727

ABSTRACT

Blockade of the programmed cell death-1 (PD-1)/programmed cell death ligand 1 (PD-L1) pathway is an attractive strategy for immunotherapy, but the clinical application of small molecule PD-1/PD-L1 inhibitors remains unclear. In this work, based on BMS-202 and our previous work YLW-106, a series of compounds with benzo[d]isothiazol structure as scaffold were designed and synthesized. Their inhibitory activity against PD-1/PD-L1 interaction was evaluated by a homogeneous time-resolved fluorescence (HTRF) assay. Among them, LLW-018 (27c) exhibited the most potent inhibitory activity with an IC50 value of 2.61 nM. The cellular level assays demonstrated that LLW-018 exhibited low cytotoxicity against Jurkat T and MDA-MB-231. Further cell-based PD-1/PD-L1 blockade bioassays based on PD-1 NFAT-Luc Jurkat cells and PD-L1 TCR Activator CHO cells indicated that LLW-018 could interrupt PD-1/PD-L1 interaction with an IC50 value of 0.88 µM. Multi-computational methods, including molecular docking, molecular dynamics, MM/GBSA, MM/PBSA, Metadynamics, and QM/MM MD were utilized on PD-L1 dimer complexes, which revealed the binding modes and dissociation process of LLW-018 and C2-symmetric small molecule inhibitor LCH1307. These results suggested that LLW-018 exhibited promising potency as a PD-1/PD-L1 inhibitor for further investigation.


Subject(s)
B7-H1 Antigen , Drug Design , Programmed Cell Death 1 Receptor , Humans , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Structure-Activity Relationship , Molecular Structure , Dose-Response Relationship, Drug , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/chemical synthesis , Jurkat Cells , Molecular Docking Simulation , Thiazoles/pharmacology , Thiazoles/chemistry , Thiazoles/chemical synthesis , Animals , Benzothiazoles/pharmacology , Benzothiazoles/chemistry , Benzothiazoles/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry
9.
J Immunother Cancer ; 12(7)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38991728

ABSTRACT

BACKGROUND: Metabolomics has the characteristics of terminal effects and reflects the physiological state of biological diseases more directly. Several current biomarkers of multiple omics were revealed to be associated with immune-related adverse events (irAEs) occurrence. However, there is a lack of reliable metabolic biomarkers to predict irAEs. This study aims to explore the potential metabolic biomarkers to predict risk of irAEs and to investigate the association of plasma metabolites level with survival in patients with lung cancer receiving PD-1/PD-L1 inhibitor treatment. METHODS: The study collected 170 plasmas of 85 patients with lung cancer who received immune checkpoint inhibitors (ICIs) treatment. 58 plasma samples of 29 patients with irAEs were collected before ICIs treatment and at the onset of irAEs. 112 plasma samples of 56 patients who did not develop irAEs were collected before ICIs treatment and plasma matched by treatment cycles to onset of irAEs patients. Untargeted metabolomics analysis was used to identify the differential metabolites before initiating ICIs treatment and during the process that development of irAEs. Kaplan-Meier curves analysis was used to detect the associations of plasma metabolites level with survival of patients with lung cancer. RESULTS: A total of 24 differential metabolites were identified to predict the occurrence of irAEs. Baseline acylcarnitines and steroids levels are significantly higher in patients with irAEs, and the model of eight acylcarnitine and six steroid metabolites baseline level predicts irAEs occurrence with area under the curve of 0.91. Patients with lower concentration of baseline decenoylcarnitine(AcCa(10:1) 2, decenoylcarnitine(AcCa(10:1) 3 and hexanoylcarnitine(AcCa(6:0) in plasma would have better overall survival (OS). Moreover, 52 differential metabolites were identified related to irAEs during ICIs treatment, dehydroepiandrosterone sulfate, corticoserone, cortisol, thyroxine and sphinganine 1-phaosphate were significantly decreased in irAEs group while oxoglutaric acid and taurocholic acid were significantly increased in irAEs group. CONCLUSIONS: High levels of acylcarnitines and steroid hormone metabolites might be risk factor to development of irAEs, and levels of decenoylcarnitine (AcCa(10:1) 2, decenoylcarnitine (AcCa(10:1) 3 and hexanoylcarnitine (AcCa(6:0) could be used to predict OS for patients with lung cancer received ICIs treatment.


Subject(s)
Immune Checkpoint Inhibitors , Lung Neoplasms , Metabolomics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/blood , Male , Female , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/adverse effects , Metabolomics/methods , Aged , Middle Aged , B7-H1 Antigen/blood , B7-H1 Antigen/antagonists & inhibitors , Aged, 80 and over , Programmed Cell Death 1 Receptor/antagonists & inhibitors
10.
Theranostics ; 14(10): 3777-3792, 2024.
Article in English | MEDLINE | ID: mdl-38994018

ABSTRACT

Rationale: Growing evidence has demonstrated that miRNA-21 (miR-21) upregulation is closely associated with tumor pathogenesis. However, the mechanisms by which miR-21 inhibition modulates the immunosuppressive tumor microenvironment (TME) and improves tumor sensitivity to immune checkpoint blockade therapies remain largely unexplored. In this study, we demonstrate the precise delivery of anti-miR-21 using a PD-L1-targeting peptide conjugate (P21) to the PD-L1high TME. Methods: Investigating miR-21 inhibition mechanisms involved conducting quantitative real-time PCR, western blot, flow cytometry, and confocal microscopy analyses. The antitumor efficacy and immune profile of P21 monotherapy, or combined with anti-PD-L1 immune checkpoint inhibitors, were assessed in mouse models bearing CT26.CL25 tumors and 4T1 breast cancer. Results Inhibition of oncogenic miR-21 in cancer cells by P21 efficiently activates tumor suppressor genes, inducing autophagy and endoplasmic reticulum stress. Subsequent cell-death-associated immune activation (immunogenic cell death) is initiated via the release of damage-associated molecular patterns. The in vivo results also illustrated that the immunogenic cell death triggered by P21 could effectively sensitize the immunosuppressive TME. That is, P21 enhances CD8+ T cell infiltration in tumor tissues by conferring immunogenicity to dying cancer cells and promoting dendritic cell maturation. Meanwhile, combining P21 with an anti-PD-L1 immune checkpoint inhibitor elicits a highly potent antitumor effect in a CT26.CL25 tumor-bearing mouse model and 4T1 metastatic tumor model. Conclusions: Collectively, we have clarified a miR-21-related immunogenic cell death mechanism through the precise delivery of anti-miR-21 to the PD-L1high TME. These findings highlight the potential of miR-21 as a target for immunotherapeutic interventions.


Subject(s)
B7-H1 Antigen , Immunogenic Cell Death , Immunotherapy , MicroRNAs , Tumor Microenvironment , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Mice , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Immunogenic Cell Death/drug effects , Cell Line, Tumor , Immunotherapy/methods , Female , Mice, Inbred BALB C , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Autophagy/drug effects , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/genetics
12.
Magy Onkol ; 68(2): 126-135, 2024 Jul 16.
Article in Hungarian | MEDLINE | ID: mdl-39013086

ABSTRACT

Tumor hypoxia plays an important role in controlling tumor progression through signaling pathways related to the transcription factor HIF-1. In addition to enhancing migration, promoting angiogenesis and regulating metabolism, the hypoxic environment also affects immune function. In this hypoxic microenvironment an immunosuppressive milieu is established, where HIF-1 upregulates the expression of PD-L1, a key regulator of the immune response. We have found that elevated expression of PD-L1 correlates with increased HIF-1 levels in cancer cell lines and clinical samples. Thus, the co-inhibition of HIF-1 and PD-1/PD-L1 offers promising therapeutic possibilities. In this review we have examined the limitations of HIF-1 and PD-1/PD-L1 inhibition as monotherapy, explored their combined benefits and evaluated the feasibility of targeting PD-L1 with HIF-1 inhibitors.


Subject(s)
B7-H1 Antigen , Neoplasms , Programmed Cell Death 1 Receptor , Tumor Hypoxia , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/pathology , Tumor Hypoxia/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Signal Transduction/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
14.
PLoS One ; 19(7): e0307204, 2024.
Article in English | MEDLINE | ID: mdl-39008481

ABSTRACT

Malignant pleural mesothelioma (MPM) is an aggressive cancer with a very poor prognosis. Recently, immune checkpoint inhibition (ICI) has taken center stage in the currently ongoing revolution that is changing standard-of-care treatment for several malignancies, including MPM. As multiple arguments and accumulating lines of evidence are in support of the existence of a therapeutic synergism between chemotherapy and immunotherapy, as well as between different classes of immunotherapeutics, we designed a multicenter, single-arm, phase I/II trial in which both programmed-death-ligand 1 (PD-L1) inhibition and dendritic cell (DC) vaccination are integrated in the first-line conventional platinum/pemetrexed-based treatment scheme for epithelioid MPM patients (Immuno-MESODEC, ClinicalTrials.gov identifier NCT05765084). Fifteen treatment-naïve patients with unresectable epithelioid subtype MPM will be treated with four 3-weekly (±3 days) chemo-immunotherapy cycles. Standard-of-care chemotherapy consisting of cisplatinum (75mg/m2) and pemetrexed (500mg/m2) will be supplemented with the anti-PD-L1 antibody atezolizumab (1200 mg) and autologous Wilms' tumor 1 mRNA-electroporated dendritic cell (WT1/DC) vaccination (8-10 x 106 cells/vaccination). Additional atezolizumab (1680 mg) doses and/or WT1/DC vaccinations (8-10 x 106 cells/vaccination) can be administered optionally following completion of the chemo-immunotherapy scheme. Follow-up of patients will last for up to 90 days after final atezolizumab administration and/or WT1/DC vaccination or 24 months after diagnosis, whichever occurs later. The trial's primary endpoints are safety and feasibility, secondary endpoints are clinical efficacy and immunogenicity. This phase I/II trial will evaluate whether addition of atezolizumab and WT1/DC vaccination to frontline standard-of-care chemotherapy for the treatment of epithelioid MPM is feasible and safe. If so, this novel combination strategy should be further investigated as a promising advanced treatment option for this hard-to-treat cancer.


Subject(s)
Antibodies, Monoclonal, Humanized , B7-H1 Antigen , Cancer Vaccines , Dendritic Cells , Mesothelioma, Malignant , Humans , Mesothelioma, Malignant/drug therapy , Mesothelioma, Malignant/immunology , Dendritic Cells/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Cancer Vaccines/therapeutic use , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Male , Female , WT1 Proteins/immunology , Pleural Neoplasms/immunology , Pleural Neoplasms/drug therapy , Pleural Neoplasms/therapy , Immunotherapy/methods , Middle Aged , Adult , Immune Checkpoint Inhibitors/therapeutic use , Aged , Vaccination , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Pemetrexed/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Mesothelioma/drug therapy , Mesothelioma/immunology , Mesothelioma/therapy , Cisplatin/therapeutic use , Cisplatin/pharmacology
15.
Cancer Immunol Immunother ; 73(9): 162, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38953977

ABSTRACT

PURPOSE: To compare the risk of immune-associated pneumonitis between PD-1 and PD-L1 inhibitors, the meta-analysis was designed. METHOD: The difference in risk of immune-associated pneumonitis between PD-1 and PD-L1 inhibitors was assessed by two different meta-analysis methods, the Mirror-pairing and the PRISMA guidelines. RESULTS: A total of eighty-eight reports were used for meta-analysis, while thirty-two studies were used for the Mirror-pairing. Both PD-1 and PD-L1 inhibitors (used alone or combined with chemotherapy) increased the risk of developing immune-related pneumonitis (P < 0.00001; P < 0.00001). Based on indirect analyses results (subgroup analyses), the risk of PD-L1-induced pneumonitis was weaker than that of PD-1 inhibitors when the control group was chemotherapy (OR = 3.33 vs. 5.43) or placebo (OR = 2.53 vs. 3.19), while no obvious significant differences were found (P = 0.17; P = 0.53). For the Mirror-pairing-based meta-analysis, the risk of PD-1-induced pneumonitis was significantly higher than that of PD-L1 inhibitors (OR = 1.46, 95%CI [1.08, 1.98], I2 = 0%, Z = 2.47 (P = 0.01)). However, this difference was not significant, when they were combined with chemotherapy (OR = 1.05, 95%CI [0.68, 1.60], I2 = 38%, Z = 0.21 (P = 0.84)). CONCLUSION: Both PD-1 and PD-L1 inhibitors increased the risk of immune-related pneumonitis, while the risk of PD-1-induced pneumonitis was significantly higher than that of PD-L1 inhibitors.


Subject(s)
B7-H1 Antigen , Immune Checkpoint Inhibitors , Pneumonia , Programmed Cell Death 1 Receptor , Humans , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Immune Checkpoint Inhibitors/adverse effects , Neoplasms/drug therapy , Neoplasms/immunology , Pneumonia/immunology , Pneumonia/etiology , Programmed Cell Death 1 Receptor/antagonists & inhibitors
16.
BMC Cancer ; 24(1): 851, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026211

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors, such as anti-programmed cell death-1 (PD-1) and PD-1 ligand-1 (PD-L1) antibodies, have achieved breakthrough results in improving long-term survival rates in lung cancer. Although high levels of PD-L1 expression and tumor mutational burden have emerged as pivotal biomarkers, not all patients derive lasting benefits, and resistance to immune checkpoint blockade remains a prevalent issue. Comprehending the immunological intricacies of lung cancer is crucial for uncovering the mechanisms that govern responses and resistance to immunomodulatory treatments. This study aimed to explore the potential of peripheral immune markers in predicting treatment efficiency among lung cancer patients undergoing PD-1/PD-L1 checkpoint inhibitors. METHODS: This study enrolled 71 lung cancer patients undergoing PD-1/PD-L1 inhibitor therapy and 20 healthy controls. Immune cell subsets (CD4 + T cells, CD8 + T cells, B cells, NK cells, and NKT cells), phenotypic analysis of T cells and B cells, and PMA/Ionomycin-stimulated lymphocyte function assay were conducted. RESULTS: Lung cancer patients exhibited significant alterations in immune cell subsets, notably an increased percentage of Treg cells. Post-treatment, there were substantial increases in absolute numbers of CD3 + T cells, CD8 + T cells, and NKT cells, along with heightened HLA-DR expression on CD3 + T and CD8 + T cells. Comparison between complete remission and non-complete remission (NCR) groups showed higher Treg cell percentages and HLA-DR + CD4 + T cells in the NCR group. CONCLUSION: The study findings suggest potential predictive roles for immune cell subsets and phenotypes, particularly Treg cells, HLA-DR + CD4 + T cells, and naïve CD4 + T cells, in evaluating short-term PD-1/PD-L1 therapy efficacy for lung cancer patients. These insights offer valuable prospects for personalized treatment strategies and underscore the importance of immune profiling in lung cancer immunotherapy.


Subject(s)
B7-H1 Antigen , Immune Checkpoint Inhibitors , Lung Neoplasms , Programmed Cell Death 1 Receptor , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Male , Female , Middle Aged , Immune Checkpoint Inhibitors/therapeutic use , Aged , B7-H1 Antigen/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Biomarkers, Tumor , Adult
17.
Cancer Med ; 13(14): e70007, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39030820

ABSTRACT

BACKGROUND: Despite promising outcomes of first-line immunotherapy with or without chemotherapy in advanced non-small cell lung cancer (NSCLC), limited accessibility due to reimbursement was remain the problem in low to middle income countries. This study aimed to evaluate real-world effectiveness of immunotherapy in patients with advanced NSCLC in Northern Thailand. METHOD: A retrospective, single-centered cohort, was conducted. Patients with advanced NSCLC who underwent PD-L1 testing (excluding EGFR and ALK mutations) and were treated with immunotherapy or without chemotherapy or chemotherapy alone were included. The primary end point was progression-free survival (PFS). The secondary endpoints were overall survival (OS), objective response rate (ORR), and adverse events. RESULTS: A total of 123 patients, of which 21 patients received immunotherapy-based regimen and 102 patients received chemotherapy alone. The median PFS was 11.9 months in immunotherapy-based group compared to 5.93 months in the chemotherapy group, with a. hazard ratio (HR) of 0.4 (95% confidence interval [CI], 0.23 to 0.68; p = 0.001). Similarly, the median OS was 26.68 months in the immunotherapy-based group and 11.21 months in the chemotherapy group, with HR of 0.42 (95% CI 0.22-0.8; p = 0.009). ORRs were significantly higher in the immunotherapy-based group, with 65% of patients showing a response compared to 32% in the chemotherapy group (p = 0.006). CONCLUSION: The result of this real-world study in patients with advanced stage NSCLC indicate that first-line immunotherapy-based regimen was associated with significantly greater PFS, OS, and ORR with a safety profile consistent with pivotal studies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Non-Small-Cell Lung , Immune Checkpoint Inhibitors , Lung Neoplasms , Tertiary Care Centers , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Female , Male , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/therapy , Lung Neoplasms/pathology , Middle Aged , Retrospective Studies , Aged , Immune Checkpoint Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Treatment Outcome , B7-H1 Antigen/antagonists & inhibitors , Progression-Free Survival , Immunotherapy/methods , Adult , Thailand , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Aged, 80 and over
18.
Arch Dermatol Res ; 316(7): 478, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39023568

ABSTRACT

The efficacy of ritlecitinib, an oral JAK3/TEC family kinase inhibitor, on active and stable lesions was evaluated in patients with active non-segmental vitiligo in a phase 2b trial (NCT03715829). Patients were randomized to placebo or daily ritlecitinib 50 mg (with or without 4-week 100-mg or 200-mg loading dose), 30 mg, or 10 mg for 24 weeks. Active lesions showed greater baseline expression of inflammatory/immune markers IFNG and CCL5, levels of CD103, and T-cell infiltrates than stable lesions. Patients with more active than stable vitiligo lesions showed higher baseline serum levels of CXCL9 and PD-L1, while patients with more stable than active lesions showed higher baseline serum levels of HO-1. At Week 24, ritlecitinib 50 mg significantly stabilized mean percent change from baseline in depigmentation extent in both active lesions and stable lesions vs. placebo-response, with stable lesions showing greater repigmentation. After 24 weeks of treatment, ritlecitinib 50 mg increased expression of melanocyte markers in stable lesions, while Th1/Th2-related and co-stimulatory molecules decreased significantly in both stable and active lesions. Serum from patients with more active than stable lesions showed decreased levels of ICOS and NK cell activation markers. These data, confirmed at transcription/protein levels, indicate that stable lesion repigmentation occurs early with ritlecitinib, while active lesions require stabilization of inflammation first. ClinicalTrials.gov: NCT03715829.


Subject(s)
Janus Kinase 3 , Protein Kinase Inhibitors , Vitiligo , Humans , Vitiligo/drug therapy , Vitiligo/diagnosis , Vitiligo/immunology , Male , Female , Adult , Janus Kinase 3/antagonists & inhibitors , Middle Aged , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/administration & dosage , Treatment Outcome , Chemokine CXCL9/blood , Chemokine CCL5/blood , Young Adult , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , B7-H1 Antigen/blood , Melanocytes/drug effects , Double-Blind Method , Skin Pigmentation/drug effects , Administration, Oral , Interferon-gamma
19.
Mol Cancer ; 23(1): 146, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39014460

ABSTRACT

The advent of PD1/PD-L1 inhibitors has significantly transformed the therapeutic landscape for clear cell renal cell carcinoma (ccRCC). This review provides an in-depth analysis of the biological functions and regulatory mechanisms of PD1 and PD-L1 in ccRCC, emphasizing their role in tumor immune evasion. We comprehensively evaluate the clinical efficacy and safety profiles of PD1/PD-L1 inhibitors, such as Nivolumab and Pembrolizumab, through a critical examination of recent clinical trial data. Furthermore, we discuss the challenges posed by resistance mechanisms to these therapies and potential strategies to overcome them. We also explores the synergistic potential of combination therapies, integrating PD1/PD-L1 inhibitors with other immunotherapies, targeted therapies, and conventional modalities such as chemotherapy and radiotherapy. In addition, we examine emerging predictive biomarkers for response to PD1/PD-L1 blockade and biomarkers indicative of resistance, providing a foundation for personalized therapeutic approaches. Finally, we outline future research directions, highlighting the need for novel therapeutic strategies, deeper mechanistic insights, and the development of individualized treatment regimens. Our work summarizes the latest knowledge and progress in this field, aiming to provide a valuable reference for improving clinical efficacy and guiding future research on the application of PD1/PD-L1 inhibitors in ccRCC.


Subject(s)
B7-H1 Antigen , Carcinoma, Renal Cell , Immune Checkpoint Inhibitors , Kidney Neoplasms , Programmed Cell Death 1 Receptor , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/therapy , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Biomarkers, Tumor , Treatment Outcome , Animals , Drug Resistance, Neoplasm , Molecular Targeted Therapy , Immunotherapy/methods
20.
Cell Rep Med ; 5(7): 101621, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38906149

ABSTRACT

Immune checkpoint inhibitors (ICIs) activate anti-cancer immunity by blocking T cell checkpoint molecules such as programmed death 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4). Although ICIs induce some durable responses in various cancer patients, they also have disadvantages, including low response rates, the potential for severe side effects, and high treatment costs. Therefore, selection of patients who can benefit from ICI treatment is critical, and identification of biomarkers is essential to improve the efficiency of ICIs. In this review, we provide updated information on established predictive biomarkers (tumor programmed death-ligand 1 [PD-L1] expression, DNA mismatch repair deficiency, microsatellite instability high, and tumor mutational burden) and potential biomarkers currently under investigation such as tumor-infiltrated and peripheral lymphocytes, gut microbiome, and signaling pathways related to DNA damage and antigen presentation. In particular, this review aims to summarize the current knowledge of biomarkers, discuss issues, and further explore future biomarkers.


Subject(s)
Biomarkers, Tumor , Immune Checkpoint Inhibitors , Neoplasms , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Microsatellite Instability , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , Animals , Gastrointestinal Microbiome/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...