Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
BMC Cancer ; 22(1): 293, 2022 Mar 19.
Article in English | MEDLINE | ID: mdl-35305593

ABSTRACT

BACKGROUND: This study aimed to evaluate the efficiency and prognostic factors of lenvatinib plus programmed death 1 (PD-1) blockades in patients with advanced hepatocellular carcinoma (HCC), especially for those with tumor occupation ≥50% volume of liver (TO ≥50%) or invasion in Vp4, who were excluded from the trial KEYNOTE-524. METHODS: We reviewed the clinical data of patients with unresectable HCC who received lenvatinib plus PD-1 blockades. The Kaplan-Meier method was performed to compare the progression-free survival (PFS) and the overall survival (OS). Cox proportional hazards model was adopted to identify independent prognostic factors. RESULTS: The median PFS and OS of the enrolled 84 HCC patients (31 patients with TO ≥50% and 30 patients with Vp4 invasion) were 6.6 and 11.4 months respectively. TO ≥50% had significantly negative impact on the objective response rates (ORR) (p = 0.015). HCC patients with TO ≥50% had significantly worse PFS and OS than those with TO < 50% (both p value < 0.001). Conversely, invasion in Vp4 did not significantly affect the ORR, PFS or OS for HCC patients receiving lenvatinib plus PD-1 blockades (p = 0.419, 0.528 and 0.855). After multivariate analyses, TO ≥50% was the independent predictor for PFS and OS (both p value < 0.001). No significant correlation was found between any kind of AEs and TO ≥50% or invasion in Vp4. CONCLUSION: Lenvatinib plus PD-1 blockades can provide survival benefits for HCC patients with invasion in Vp4 and the indications of lenvatinib plus pembrolizumab may be further expanded. Locoregional treatments should be considered for patients with TO ≥50% during systemic therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Phenylurea Compounds/administration & dosage , Programmed Cell Death 1 Receptor/administration & dosage , Quinolines/administration & dosage , Adult , Aged , Carcinoma, Hepatocellular/pathology , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Liver Neoplasms/pathology , Male , Middle Aged , Progression-Free Survival , Retrospective Studies
2.
Zhongguo Fei Ai Za Zhi ; 24(4): 217-235, 2021 Apr 20.
Article in Chinese | MEDLINE | ID: mdl-33896153

ABSTRACT

Non-small cell lung cancer (NSCLC) is the most common pathological type of lung cancer. The systemic antitumor therapy of advanced NSCLC has undergone renovations of chemotherapy, targeted therapy and immunotherapy, which results in greatly improved survival for patients with advanced NSCLC. Immune checkpoint inhibitors (ICIs), especially targeting programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1), has changed the treatment paradigm of NSCLC. ICIs have become the standard treatment for advanced NSCLC without epidermal growth factor receptor(EGFR) mutation or anaplastic lymphomakinase(ALK) translocation in the first- or second-line setting, and for locally advanced NSCLC following concurrent radiotherapy and chemotherapy. ICIs are also promising in adjuvant/neoadjuvant therapy. More and more ICIs have been approved domestically for the treatment of NSCLC. Led by the NSCLC expert committee of Chinese Society of Clinical Oncology (CSCO), this consensus was developed and updated based on thoroughly reviewing domestic and foreign literatures, clinical trial data, systematic reviews, experts' discussion and the consensus(2019 version). This consensus will aid domestic clinicians in the treatment of NSCLC with ICIs.
.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Immune Checkpoint Inhibitors/administration & dosage , Lung Neoplasms/drug therapy , B7-H1 Antigen/administration & dosage , China , Consensus , Humans , Immunotherapy , Programmed Cell Death 1 Receptor/administration & dosage
3.
Neurosurgery ; 88(4): 855-863, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33370819

ABSTRACT

BACKGROUND: Cerebral vasospasm is a major source of morbidity and mortality following aneurysm rupture and has limited treatment options. OBJECTIVE: To evaluate the role of programmed death-1 (PD-1) in cerebral vasospasm. METHODS: Endovascular internal carotid artery perforation (ICAp) was used to induce cerebral vasospasm in mice. To evaluate the therapeutic potential of targeting PD-1, programmed death ligand-1 (PD-L1) was administered 1 h after ICAp and vasospasm was measured histologically at the level of the ICA bifurcation bilaterally. PD-1 expressing immune cell populations were evaluated by flow cytometry. To correlate these findings to patients and evaluate the potential of PD-1 as a biomarker, monocytes were isolated from the peripheral blood and analyzed by flow cytometry in a cohort of patients with ruptured cerebral aneurysms. The daily frequency of PD-1+ monocytes in the peripheral blood was correlated to transcranial Doppler velocities as well as clinical and radiographic vasospasm. RESULTS: We found that PD-L1 administration prevented cerebral vasospasm by inhibiting ingress of activated Ly6c+ and CCR2+ monocytes into the brain. Human correlative studies confirmed the presence of PD-1+ monocytes in the peripheral blood of patients with ruptured aneurysms and the frequency of these cells corresponded with cerebral blood flow velocities and clinical vasospasm. CONCLUSION: Our results identify PD-1+ monocytes as mediators of cerebral vasospasm and support PD-1 agonism as a novel therapeutic strategy.


Subject(s)
Monocytes/metabolism , Programmed Cell Death 1 Receptor/administration & dosage , Subarachnoid Hemorrhage/blood , Subarachnoid Hemorrhage/drug therapy , Vasospasm, Intracranial/blood , Vasospasm, Intracranial/prevention & control , Animals , Brain/blood supply , Brain/diagnostic imaging , Brain/drug effects , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Cohort Studies , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Subarachnoid Hemorrhage/diagnostic imaging , Ultrasonography, Doppler, Transcranial/methods , Vasospasm, Intracranial/diagnostic imaging
4.
Cancer Treat Rev ; 85: 101979, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32078962

ABSTRACT

PURPOSE: The combination of an anti-programmed death 1 (PD-1) or anti-programmed death ligand 1 (PD-L1) monoclonal antibody with platinum-based chemotherapy can improve outcomes for patients with advanced non-small-cell lung cancer (NSCLC) or small-cell lung cancer (SCLC) compared with chemotherapy alone. For patients receiving these new treatment regimens, it is important that toxicities be managed effectively. A particular challenge can be determining the etiology of an event, especially when there are overlapping symptoms that can be attributed to either immunotherapy or to platinum-based chemotherapy. Here, we evaluate adverse events (AEs) reported in clinical trials of combination therapy with an anti-PD-1 or anti-PD-L1 (anti-PD-[L]1) immunotherapy and chemotherapy to provide information on toxicity management. METHODS: We performed a systematic review of the literature focused on randomized controlled trials of anti-PD-(L)1 therapy combined with platinum-based chemotherapy for advanced/metastatic NSCLC and SCLC. RESULTS: Eleven reports from 9 randomized studies evaluating pembrolizumab, nivolumab, and atezolizumab combined with platinum-based chemotherapy in patients with advanced lung cancer were identified. Immune-mediated AEs and infusion reactions occurred more commonly in patients who received anti-PD-(L)1 immunotherapy with platinum-based chemotherapy compared with chemotherapy alone; however, there was no evidence of unexpected or unanticipated toxicity with these combinations. CONCLUSION: Combinations of anti-PD-(L)1 immunotherapy with platinum-based chemotherapy regimens improve outcomes for patients with NSCLC and SCLC, and toxicity is generally manageable. Strategies for appropriate workup of AEs to allow clinicians to make informed decisions regarding causality and treatment modifications when appropriate are an important element of management of patients receiving an anti-PD-(L)1 agent combined with platinum-based chemotherapy.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug-Related Side Effects and Adverse Reactions/etiology , Lung Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Antibodies, Monoclonal, Humanized/therapeutic use , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Disease-Free Survival , Drug-Related Side Effects and Adverse Reactions/drug therapy , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Neoplasm Invasiveness/pathology , Neoplasm Staging , Nivolumab/therapeutic use , Prognosis , Programmed Cell Death 1 Receptor/administration & dosage , Randomized Controlled Trials as Topic , Risk Assessment , Survival Analysis , Treatment Outcome
5.
Biomolecules ; 9(11)2019 10 25.
Article in English | MEDLINE | ID: mdl-31731492

ABSTRACT

Programmed cell death protein 1 (PD-1) is a biomarker on the surface of cells with a role in promoting self-tolerance by suppressing the inflammatory activity of T cells. In this work, one peptide of PD-1 was used as the template for molecular imprinting to form magnetic peptide-imprinted poly(ethylene-co-vinyl alcohol) composite nanoparticles (MPIP NPs). The nanoparticles were characterized by dynamic light scattering (DLS), high-performance liquid chromatography (HPLC), Brunauer-Emmett-Teller (BET) analysis, and superconducting quantum interference device (SQUID) analysis. Natural killer 92 (NK-92) cells were added to these composite nanoparticles and then incubated with human hepatoma (HepG2) cells. The viability and the apoptosis pathway of HepG2 were then studied using cell counting kit-8 (CCK8) and quantitative real-time polymerase chain reaction (qRT-PCR), respectively. These nanoparticles were found to significantly enhance the activity of natural killer cells toward HepG2 cells by increasing the expression of nuclear factor kappa B (NF-κB), caspase 8, and especially caspase 3.


Subject(s)
Carcinoma, Hepatocellular/therapy , Iron Compounds/administration & dosage , Killer Cells, Natural/immunology , Liver Neoplasms/therapy , Nanocomposites/administration & dosage , Peptides/administration & dosage , Polyvinyls/administration & dosage , Programmed Cell Death 1 Receptor/administration & dosage , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Cell Line , Cell Survival/drug effects , Gene Expression/drug effects , Humans , Immunotherapy , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Magnetic Phenomena , Molecular Imprinting
8.
Lancet Oncol ; 18(12): e731-e741, 2017 12.
Article in English | MEDLINE | ID: mdl-29208439

ABSTRACT

Use of immune checkpoint inhibitors targeting the programmed cell death protein-1/programmed cell death-ligand 1 and cytotoxic T lymphocyte-associated protein-4 axes has yielded impressive results in some clinical trials. However, only a subset of patients initially respond to these inhibitors, and increasing clinical evidence indicates that a substantial proportion of initial responders ultimately relapse with lethal, drug-resistant disease months or years later. Studies that have used massively parallel sequencing have shed light on the rich functional landscape of mutations that endow tumour cells with the ability to evade T-cell-mediated immunosurveillance. Cancer genomes bear signatures of clonal evolution and selection, particularly implicating acquired defects in interferon receptor signalling and antigen presentation. In this Review, we discuss the biological processes that operate in the formation of so-called immunoresistant niches, and describe the latest progress in the development of combination strategies to reinstate immunosurveillance in immune-refractory tumours.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Cell Cycle Checkpoints/immunology , Immunotherapy/methods , Neoplasms/drug therapy , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/administration & dosage , Cell Cycle Checkpoints/drug effects , Female , Humans , Male , Molecular Targeted Therapy , Neoplasms/genetics , Prognosis , Programmed Cell Death 1 Receptor/administration & dosage , Risk Assessment , Treatment Outcome
9.
Head Neck ; 39(6): 1086-1094, 2017 06.
Article in English | MEDLINE | ID: mdl-28323387

ABSTRACT

BACKGROUND: Cyclic dinucleotides (CDNs) are bacterial intracellular messengers that have demonstrated antitumor activity in melanoma and breast tumors, although their role in immunotherapy of head and neck squamous cell cancers (HNSCCs) has not been well investigated. METHODS: We measured primary tumor growth rates, mechanism of antitumor activity, and efficacy of programmed death-L1 blockade combinatorial therapy in SCCFVII tumor-bearing C3H/HeOUJ mice undergoing intratumoral injections with RR-cyclic-di-guanine (synthetic CDG), CDG (natural cyclic-di-guanine), R848 (TLR 7/8 agonist), or phosphate buffered saline (PBS, control). RESULTS: Intratumoral CDN treatment groups showed decreased tumor size and enhanced splenocyte Th1 response when compared to the PBS treatment control group (p < .05). The RR-CDG tumor microenvironment showed upregulated interferon (IFN)-γ+CD8+ and programmed death-L1. Combining programmed death-L1 blocking antibody with RR-CDG induced regression of established tumors. CONCLUSION: This study demonstrates the antitumor effects of CDNs in a HNSCC cell line. These preclinical data strongly support the future clinical development of intratumoral CDN in patients with HNSCC. © 2017 Wiley Periodicals, Inc. Head Neck 39: 1086-1094, 2017.


Subject(s)
Antibodies, Neoplasm/immunology , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Immunotherapy/methods , Membrane Proteins/drug effects , Programmed Cell Death 1 Receptor/administration & dosage , Animals , Antibodies, Neoplasm/drug effects , Carcinoma, Squamous Cell/pathology , Cell Survival/drug effects , Disease Models, Animal , Female , Head and Neck Neoplasms/pathology , Injections, Intralesional , Membrane Proteins/immunology , Mice , Mice, Inbred C3H , Programmed Cell Death 1 Receptor/immunology , Random Allocation , Sensitivity and Specificity , Squamous Cell Carcinoma of Head and Neck
10.
J Immunother Cancer ; 4: 60, 2016.
Article in English | MEDLINE | ID: mdl-27777770

ABSTRACT

BACKGROUND: Anti-PD-1 therapy is increasingly used in various advanced malignancies. Patients with baseline organ dysfunction are largely excluded from clinical trials. Therefore it is unclear whether anti-PD-1 therapy is safe or effective in this setting. Further, these patients are often not candidates for other anti-cancer therapies, highlighting their need for active treatment options. METHODS: We performed a retrospective analysis of patients from multiple centers with advanced solid tumors and baseline organ dysfunction who received anti-PD-1 therapy. Organ dysfunction was defined as cardiac (left ventricular ejection fraction ≤45 %), renal (creatinine ≥2 mg/dL or GFR ≤30 ml/min) or hepatic dysfunction (evidence of cirrhosis on imaging or AST, ALT or bilirubin ≥3x ULN). We assessed change in organ dysfunction, immune related adverse events (irAEs), response rate, progression free survival (PFS) and overall survival (OS). RESULTS: We identified 27 patients eligible for inclusion with the following diseases: renal cell carcinoma (n = 8), melanoma (10), non-small cell lung cancer (3), small cell lung cancer (2) and urothelial bladder cancer (4). Baseline organ dysfunction included renal dysfunction (n = 17), hepatic dysfunction (7), cardiac dysfunction (11), including >1 organ dysfunction (8). Worsening organ dysfunction requiring hospitalization or dose delays occurred in 8 patients (30 %) although in most cases this was thought not-drug related and resolved with supportive care. Grade 3 irAEs occurred in 2 pts (7 %; hepatitis and colitis). Thirteen of 27 patients had ongoing treatment benefit (objective response or stable disease) at data collection (48 %). Eleven patients had primary progressive disease (41 %), 11 had stable disease (41 %), 4 had partial responses (15 %), and one had a complete response (4 %). Overall, median PFS was 168 days. Median OS was not reached. CONCLUSIONS: In our experience, anti-PD-1 agents in this group of patients with cardiac, hepatic or renal dysfunction were associated with tolerable irAEs and infrequent manageable worsening of organ dysfunction. Further, objective responses and prolonged PFS were observed in a number of patients. Thus, patients with baseline organ dysfunction may be considered for anti-PD-1 therapy with appropriate clinical monitoring.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Heart/drug effects , Kidney/drug effects , Liver/drug effects , Molecular Targeted Therapy/adverse effects , Programmed Cell Death 1 Receptor/administration & dosage , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/therapeutic use , Cardiotoxicity , Female , Heart/physiopathology , Heart Function Tests , Humans , Kidney/physiopathology , Kidney Function Tests , Liver/physiopathology , Liver Function Tests , Male , Middle Aged , Neoplasm Staging , Neoplasms/complications , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/mortality , Positron Emission Tomography Computed Tomography , Retrospective Studies , Treatment Outcome
11.
EBioMedicine ; 6: 50-58, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27211548

ABSTRACT

Tumor indoleamine 2,3-dioxygenase (IDO) promotes immunosuppression by direct action on effector T cells and Tregs and through recruitment, expansion and activation of myeloid-derived suppressor cells (MDSCs). Targeting of MDSCs is clinically being explored as a therapeutic strategy, though optimal targeting strategies and biomarkers predictive of response are presently unknown. Maturation and tumor recruitment of MDSCs are dependent on signaling through the receptor tyrosine kinase CSF-1R on myeloid cells. Here, we show that MDSCs are the critical cell population in IDO-expressing B16 tumors in mediating accelerated tumor outgrowth and resistance to immunotherapy. Using a clinically relevant drug, we show that inhibition of CSF-1R signaling can functionally block tumor-infiltrating MDSCs and enhance anti-tumor T cell responses. Furthermore, inhibition of CSF-1R sensitizes IDO-expressing tumors to immunotherapy with T cell checkpoint blockade, and combination of CSF-1R blockade with IDO inhibitors potently elicits tumor regression. These findings provide evidence for a critical and functional role for MDSCs on the in vivo outcome of IDO-expressing tumors.


Subject(s)
Colonic Neoplasms/drug therapy , Immunotherapy/methods , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Melanoma, Experimental/drug therapy , Myeloid-Derived Suppressor Cells/drug effects , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , CTLA-4 Antigen/administration & dosage , Cell Line, Tumor , Colonic Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Melanoma, Experimental/metabolism , Mice , Molecular Targeted Therapy , Programmed Cell Death 1 Receptor/administration & dosage , Xenograft Model Antitumor Assays
12.
Biomed Pharmacother ; 76: 57-64, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26653551

ABSTRACT

As a key factor for tumor occurrence and development, tumor cells escape immune surveillance and inhibit the body immune killer effect through negative signaling pathways. In this research, we designed and expressed the fusion protein CRT-sPD1 to block PD1/PDL1 negative signal pathway, indirectly bind CRT to the tumor cell surface and to increase the cell immunogenicity activity. Results from western blotting, flow cytometry (FCM) and ELISA showed that the cell lines that stably express CRT, PD1 and CRT-sPD1 protein were obtained and the transfected cellular supernatant contained PD1 and CRT-sPD1 could bind to PDL1 on the surface of EL4 cells. Vitro experiments indicated the secreted mCRT-sPD1 protein could bind to PDL1 and enhance lymphocyte proliferation and CTL activity. We also found that fusion protein CRT-sPD1 could activate and induce the immune system to kill the tumor cells, specifically inhibit the tumor growth and prolong the survival period in mouse tumor model. And all these suggested that CRT-sPD1 could be used as drug development and utilization of cancer immunotherapy.


Subject(s)
Calreticulin/administration & dosage , Melanoma, Experimental/therapy , Programmed Cell Death 1 Receptor/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Animals , Blotting, Western , Calreticulin/immunology , Cell Line, Tumor , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunotherapy/methods , Lymphocytes/metabolism , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Programmed Cell Death 1 Receptor/immunology , Recombinant Fusion Proteins/immunology , Survival Rate
13.
Hum Vaccin Immunother ; 10(3): 724-33, 2014.
Article in English | MEDLINE | ID: mdl-24326266

ABSTRACT

The development of an effective T cell based HIV vaccine would need to elicit cell mediated immune responses with superior magnitude, breadth, and quality. Since blocking the interactions between inhibitory receptors with their associated ligands using soluble PD-1 (sPD-1) and soluble Tim-3 (sTim-3) have been shown to reverse T cell exhaustion and enhance cell mediated immune responses, we tested if co-administration of sPD-1 and sTim-3 with an adenovirus vectored SIV vaccine (rAd5-SIV) can enhance cell mediated immune responses. The frequency of SIV antigen specific IFN-γ spot-forming cells and the secretion of IFN-γ and TNF-α by splenocytes from rAd5-SIV immunized mice was significantly increased when stimulated ex vivo with SIV peptides in the presence of sPD-1 or sTim-3 or both sPD-1 and sTim-3. The magnitude of cell mediated immune responses elicited by rAd5-SIV was enhanced by co-administration of sPD-1 and sTim-3. Co-administration of both sPD-1 and sTim-3 induced higher frequency of SIV antigen specific IFN-γ(+) spot-forming cells to poorly immunogenic Vif and Tat. The percentage of cell mediated responses for each SIV antigen became more balanced, with reduction to Gag but induction to non-structural proteins. Furthermore, co-injection of rAd5-sPD1 and rAd5-sTim3 with rAd5-SIV in mice enhanced T cell proliferation capability and generated more antigen specific IFN-γ(+) CD4(+) and CD8(+) T cells. Our study provided a new approach to enhance vaccine induced cell mediated immune responses, which may be applicable to improve the efficacy of vaccines against SIV/HIV.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Leukocytes, Mononuclear/immunology , Programmed Cell Death 1 Receptor/administration & dosage , Receptors, Virus/administration & dosage , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/immunology , Adenoviridae/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Drug Carriers , Enzyme-Linked Immunospot Assay , Female , Genetic Vectors , Hepatitis A Virus Cellular Receptor 2 , Interferon-gamma/metabolism , Mice, Inbred C57BL , SAIDS Vaccines/genetics , Tumor Necrosis Factor-alpha/metabolism
14.
PLoS One ; 8(12): e84927, 2013.
Article in English | MEDLINE | ID: mdl-24367702

ABSTRACT

There is an urgent need for improved therapy for advanced ovarian carcinoma, which may be met by administering immune-modulatory monoclonal antibodies (mAbs) to generate a tumor-destructive immune response. Using the ID8 mouse ovarian cancer model, we investigated the therapeutic efficacy of various mAb combinations in mice with intraperitoneal (i.p.) tumor established by transplanting 3 × 10(6) ID8 cells 10 days previously. While most of the tested mAbs were ineffective when given individually or together, the data confirm our previous finding that 2 i.p. injections of a combination of anti-CD137 with anti-PD-1 mAbs doubles overall survival. Mice treated with this mAb combination have a significantly increased frequency and total number of CD8(+) T cells both in the peritoneal lavage and spleens, and these cells are functional as demonstrated by antigen-specific cytolytic activity and IFN-γ production. While administration of anti-CD137 mAb as a single agent similarly increases CD8(+) T cells, these have no functional activity, which may be attributed to up-regulation of co-inhibitory PD-1 and TIM-3 molecules induced by CD137. Addition of the anti-cancer drug cisplatin to the 2 mAb combination increased overall survival >90 days (and was probably curative) by a mechanism which included a systemic CD8(+) T cell response with tumor specificity and immunological memory. Strikingly, combined treatment of cisplatin and CD137/PD-1 mAb also gave rise to the long-term survival of mice with established TC1 lung tumors. A similar combination of the 2 mAbs and cisplatin should be considered for clinical 'translation'.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Antineoplastic Combined Chemotherapy Protocols/immunology , Cisplatin/pharmacology , Ovarian Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Analysis of Variance , Animals , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Drug Synergism , Enzyme-Linked Immunosorbent Assay , Female , Injections, Intraperitoneal , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/administration & dosage , Programmed Cell Death 1 Receptor/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/administration & dosage , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...