Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Mol Cell ; 82(2): 420-434.e6, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34951963

ABSTRACT

Exon back-splicing-generated circular RNAs, as a group, can suppress double-stranded RNA (dsRNA)-activated protein kinase R (PKR) in cells. We have sought to synthesize immunogenicity-free, short dsRNA-containing RNA circles as PKR inhibitors. Here, we report that RNA circles synthesized by permuted self-splicing thymidylate synthase (td) introns from T4 bacteriophage or by Anabaena pre-tRNA group I intron could induce an immune response. Autocatalytic splicing introduces ∼74 nt td or ∼186 nt Anabaena extraneous fragments that can distort the folding status of original circular RNAs or form structures themselves to provoke innate immune responses. In contrast, synthesized RNA circles produced by T4 RNA ligase without extraneous fragments exhibit minimized immunogenicity. Importantly, directly ligated circular RNAs that form short dsRNA regions efficiently suppress PKR activation 103- to 106-fold higher than reported chemical compounds C16 and 2-AP, highlighting the future use of circular RNAs as potent inhibitors for diseases related to PKR overreaction.


Subject(s)
Protein Kinase Inhibitors/pharmacology , RNA, Circular/pharmacology , eIF-2 Kinase/antagonists & inhibitors , A549 Cells , Bacteriophage T4/enzymology , Bacteriophage T4/genetics , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate/drug effects , Introns , Nucleic Acid Conformation , Protein Kinase Inhibitors/immunology , RNA Ligase (ATP)/genetics , RNA Ligase (ATP)/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Circular/genetics , RNA, Circular/immunology , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , eIF-2 Kinase/metabolism
2.
Leukemia ; 35(5): 1229-1242, 2021 05.
Article in English | MEDLINE | ID: mdl-33833387

ABSTRACT

The quest for treatment-free remission (TFR) and deep molecular response (DMR) in chronic myeloid leukemia (CML) has been profoundly impacted by tyrosine kinase inhibitors (TKIs). Immunologic surveillance of residual leukemic cells is hypothesized to be one of the critical factors in successful TFR, with self-renewing leukemic stem cells implicated in relapse. Immunological characterization in CML may help to develop novel immunotherapies that specifically target residual leukemic cells upon TKI discontinuation to improve TFR rates. This review focuses on immune dysfunction in newly diagnosed CML patients, and the role that TKIs and other therapies have in restoring immune surveillance. Immune dysfunction and immunosurveillance in CML points towards several emerging areas in the key goals of DMR and TFR, including: (1) Aspects of innate immune system, in particular natural killer cells and the newly emerging target plasmacytoid dendritic cells. (2) The adaptive immune system, with promise shown in regard to leukemia-associated antigen vaccine-induced CD8 cytotoxic T-cells (CTL) responses, increased CTL expansion, and immune checkpoint inhibitors. (3) Immune suppressive myeloid-derived suppressor cells and T regulatory cells that are reduced in DMR and TFR. (4) Immunomodulator mesenchymal stromal cells that critically contribute to leukomogenesis through immunosuppressive properties and TKI- resistance. Therapeutic strategies that leverage existing immunological approaches include donor lymphocyte infusions, that continue to be used, often in combination with TKIs, in patients relapsing following allogeneic stem cell transplant. Furthermore, previous standards-of-care, including interferon-α, hold promise in attaining TFR in the post-TKI era. A deeper understanding of the immunological landscape in CML is therefore vital for both the development of novel and the repurposing of older therapies to improve TFR outcomes.


Subject(s)
Immune System/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Animals , Chronic Disease , Dendritic Cells/drug effects , Dendritic Cells/immunology , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Protein Kinase Inhibitors/immunology , Treatment Outcome
4.
Drug Resist Updat ; 51: 100702, 2020 07.
Article in English | MEDLINE | ID: mdl-32371296

ABSTRACT

The tyrosine kinase inhibitor (TKI) sorafenib continues to be the anchor drug in the treatment of advanced stage hepatocellular carcinoma (HCC). Other TKIs as well as immune checkpoint inhibitors (ICIs) have also been approved, however the response rates remain poor and heterogeneous among HCC patients, largely due to antitumor drug resistance. Studies aimed at identifying novel biomarkers and developing new strategies to improve the response to current treatment and to overcome drug resistance, are urgently needed. Germline or somatic mutations, neoantigens, and an immunotolerogenic state against constant inflammatory stimuli in the liver, are crucial for the anti-tumor response. A pharmacogenetic approach has been attempted considering germline polymorphisms in genes encoding for proteins involved in drug-targeted pathways. Single gene and comprehensive multi-gene somatic profiling approaches have been adopted in HCC to identify tumor sensitivity scores and immunogenic profiles that can be exploited for new biomarkers and innovative therapeutic approaches. However, the high genomic heterogeneity of tumors and lack of molecularly targeted agents, hamper the discovery of specific molecular markers of resistance to therapy. Adoptive cell therapy with chimeric antigen receptor redirected T (CAR-T) cells targeting specific tumor-associated antigens (TAAs) was proposed recently. The specificity of the chosen TAA, an efficient homing of CAR-T cells to the tumor site, and the ability of CAR-T cells to survive in the tumor microenvironment are central factors in the success of CAR-T therapy. The current review describes the principal systemic treatments for HCC and the molecular evidence regarding potential predictive host and somatic genetic markers, as well as the emerging strategy of liquid biopsy for disease monitoring. Novel immunotherapeutic approaches for HCC treatment, including the use of ICIs and CAR-T, as well as strategies to overcome drug resistance, are discussed.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Immunotherapy, Adoptive/methods , Liver Neoplasms/drug therapy , Receptors, Chimeric Antigen/therapeutic use , Sorafenib/therapeutic use , Angiogenesis Inhibitors/pharmacology , Antigens, Neoplasm/drug effects , Antigens, Neoplasm/immunology , Antineoplastic Agents/immunology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cytokines/immunology , Drug Resistance, Neoplasm/immunology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Neoplasm Staging , Pharmacogenetics , Protein Kinase Inhibitors/immunology , Protein Kinase Inhibitors/therapeutic use , Radiotherapy, Adjuvant/methods , Receptors, Chimeric Antigen/immunology , Sorafenib/immunology , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Vascular Endothelial Growth Factor A/immunology
5.
J Immunol ; 204(9): 2439-2446, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32198145

ABSTRACT

We reported that protein kinase C-η (PKCη) forms a novel (to our knowledge) signaling complex with the checkpoint inhibitory protein CTLA-4 in regulatory T cells (Tregs). This complex is required for the contact-dependent suppressive activity of Tregs, including suppression of antitumor immunity. However, the importance of PKCη in protective immunity mediated by T effector cells remains unclear. We used mice with germline or conditional Treg-specific deletion of Prkch, the PKCη-encoding gene, to explore CD8+ T cell-dependent antiviral immunity using the lymphocytic choriomeningitis virus Armstrong strain acute infection model as well as the in vitro activation of murine or human CD8+ T cells. Five days following infection, germline Prkch -/- mice displayed enhanced viral clearance compared with control mice. Similarly, Prkch Treg-specific conditional knockout mice also showed improved viral clearance and displayed enhanced expression of granzyme B and IFN-γ by both virus-specific and total CD8+ T cells, demonstrating that enhanced viral clearance in germline Prkch -/- mice is caused by PKCη deficiency in Tregs and the resulting functional defect of Prkch -/- Tregs. In addition, purified Prkch -/- mouse CD8+ T cells as well as PRKCH knockdown human CD8+ T cells displayed intact, or even enhanced, T cell activation in vitro as measured by proliferation and expression of granzyme B and IFN-γ. Thus, global PKCη deletion does not impair overall CD8+ T cell-mediated immunity, including antiviral immunity, implying that selective pharmacological PKCη inhibition could be safely used in vivo to inhibit undesired contact-dependent suppression by Tregs and, thus, enhance tumor-specific and, likely, virus-specific immunity.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocyte Activation , Protein Kinase C , T-Lymphocytes, Regulatory , Virus Diseases , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cell Proliferation/drug effects , CTLA-4 Antigen/immunology , Granzymes/immunology , HEK293 Cells , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Interferon-gamma/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocytic choriomeningitis virus/immunology , Mice, Knockout , Protein Kinase C/deficiency , Protein Kinase C/immunology , Protein Kinase Inhibitors/immunology , Protein Kinase Inhibitors/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Virus Diseases/immunology
6.
Eur J Med Chem ; 163: 413-427, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30530193

ABSTRACT

With the rise of immuno-oncology, small-molecule modulators targeting immune system and inflammatory processes are becoming a research hotspot. This work mainly focuses on key kinases acting as central nodes in immune signaling pathways. Although over thirty small-molecule kinase inhibitors have been approved by FDA for the treatment of various cancers, only a few are associated with immuno-oncology. With the going deep of the research work, more and more immunity protein kinase inhibitors are approved for clinical trials to treat solid tumors and hematologic malignancies by FDA, which remain good prospects. Meanwhile, in-depth understanding of biological function of immunity protein kinases in immune system is pushing the field forward. This article focuses on the development of safe and effective small-molecule immunity protein kinase inhibitors and further work needs to keep the promises of these inhibitors for patients' welfare.


Subject(s)
Immunotherapy/methods , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Humans , Immune System/drug effects , Inflammation/drug therapy , Neoplasms/pathology , Protein Kinase Inhibitors/immunology
7.
Clin Exp Metastasis ; 35(5-6): 503-520, 2018 08.
Article in English | MEDLINE | ID: mdl-30019239

ABSTRACT

Malignant melanoma is generally chemo- and radio-resistant, and patients with advanced melanoma have a poor prognosis. However, with our increased understanding of the checkpoint immune molecules and genetic alterations of melanoma cells, more effective immunotherapy, such as anti CTLA4 antibody and anti PD-1 antibodies, and targeted drug therapy, such as BRAF inhibitors and MEK inhibitors, have been developed, resulting in improved overall survival and quality of life of patients with advanced melanoma. In addition, emerging technologies to develop prognostic and predictive biomarkers for response to systemic therapy could help clinicians make more accurate assessments of the disease and formulate more effective treatment plans. In this review, current standard systemic therapy options and recently developed novel drugs for advanced melanoma are discussed.


Subject(s)
Immunotherapy/trends , Melanoma/therapy , Molecular Targeted Therapy , Protein Kinase Inhibitors/immunology , Skin Neoplasms/therapy , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Humans , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , Melanoma/immunology , Melanoma/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Treatment Outcome , Melanoma, Cutaneous Malignant
8.
Mol Cancer Res ; 16(10): 1454-1457, 2018 10.
Article in English | MEDLINE | ID: mdl-29934327

ABSTRACT

Inhibitors of the cyclin-dependent kinases 4 and 6 (CDK4/6) were originally designed to block proliferation and cell cycle progression of cancer cells in which the activity of these kinases is dysregulated. CDK4/6 inhibitors have already been FDA approved for the treatment of estrogen receptor (ER)-positive breast cancer and are being tested in numerous other cancer types. However, several recent studies have identified novel effects of CDK4/6 inhibitors on tumor growth, most notably an indirect effect resulting from the activation of immune surveillance. This Perspective discusses these recent observations, including the effects that CDK4/6 inhibitors may have on immune cells themselves. It is likely that CDK4/6 inhibitors will have a broader impact than their expected induction of cell cycle arrest in the treatment of human cancers. Mol Cancer Res; 16(10); 1454-7. ©2018 AACR.


Subject(s)
Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/immunology , Cyclin-Dependent Kinase 6/immunology , Protein Kinase Inhibitors/therapeutic use , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Breast Neoplasms/immunology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/immunology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Estrogen Receptor alpha/immunology , Female , Humans , MCF-7 Cells , Mice , Protein Kinase Inhibitors/immunology , Xenograft Model Antitumor Assays
9.
Kidney Int ; 94(2): 280-291, 2018 08.
Article in English | MEDLINE | ID: mdl-29731111

ABSTRACT

To investigate the role of protein kinase C-α (PKC-α) in glomerulonephritis, the capacity of PKC-α inhibition to reverse the course of established nephrotoxic nephritis (NTN) was evaluated. Nephritis was induced by a single injection of nephrotoxic serum and after its onset, a PKC-α inhibitor was administered either systemically or by targeted glomerular delivery. By day seven, all mice with NTN had severe nephritis, whereas mice that received PKC-α inhibitors in either form had minimal evidence of disease. To further understand the underlying mechanism, label-free shotgun proteomic analysis of the kidney cortexes were performed, using quantitative mass spectrometry. Ingenuity pathway analysis revealed 157 differentially expressed proteins and mitochondrial dysfunction as the most modulated pathway. Functional protein groups most affected by NTN were mitochondrial proteins associated with respiratory processes. These proteins were down-regulated in the mice with NTN, while their expression was restored with PKC-α inhibition. This suggests a role for proteins that regulate oxidative phosphorylation in recovery. In cultured glomerular endothelial cells, nephrotoxic serum caused a decrease in mitochondrial respiration and membrane potential, mitochondrial morphologic changes and an increase in glycolytic lactic acid production; all normalized by PKC-α inhibition. Thus, PKC-α has a critical role in NTN progression, and the results implicate mitochondrial processes through restoring oxidative phosphorylation, as an essential mechanism underlying recovery. Importantly, our study provides additional support for targeted therapy to glomeruli to reverse the course of progressive disease.


Subject(s)
Glomerulonephritis/drug therapy , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/immunology , Autoantigens/immunology , Collagen Type IV/immunology , Disease Models, Animal , Drug Delivery Systems/methods , Female , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Humans , Hybridomas , Immune Sera/administration & dosage , Immune Sera/immunology , Immunoglobulin Fragments/immunology , Immunoglobulin G/immunology , Kidney Glomerulus/drug effects , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/immunology , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Protein Kinase C-alpha/immunology , Protein Kinase C-alpha/metabolism , Protein Kinase Inhibitors/immunology , Protein Kinase Inhibitors/therapeutic use , Treatment Outcome
10.
Nat Med ; 24(2): 203-212, 2018 02.
Article in English | MEDLINE | ID: mdl-29334371

ABSTRACT

Intratumor heterogeneity is a key factor contributing to therapeutic failure and, hence, cancer lethality. Heterogeneous tumors show partial therapy responses, allowing for the emergence of drug-resistant clones that often express high levels of the receptor tyrosine kinase AXL. In melanoma, AXL-high cells are resistant to MAPK pathway inhibitors, whereas AXL-low cells are sensitive to these inhibitors, rationalizing a differential therapeutic approach. We developed an antibody-drug conjugate, AXL-107-MMAE, comprising a human AXL antibody linked to the microtubule-disrupting agent monomethyl auristatin E. We found that AXL-107-MMAE, as a single agent, displayed potent in vivo anti-tumor activity in patient-derived xenografts, including melanoma, lung, pancreas and cervical cancer. By eliminating distinct populations in heterogeneous melanoma cell pools, AXL-107-MMAE and MAPK pathway inhibitors cooperatively inhibited tumor growth. Furthermore, by inducing AXL transcription, BRAF/MEK inhibitors potentiated the efficacy of AXL-107-MMAE. These findings provide proof of concept for the premise that rationalized combinatorial targeting of distinct populations in heterogeneous tumors may improve therapeutic effect, and merit clinical validation of AXL-107-MMAE in both treatment-naive and drug-resistant cancers in mono- or combination therapy.


Subject(s)
Immunoconjugates/pharmacology , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Genetic Heterogeneity/drug effects , Humans , Immunoconjugates/immunology , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Mice , Oligopeptides/chemistry , Oligopeptides/immunology , Oligopeptides/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/immunology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/immunology , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/pharmacology , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
11.
Biol Blood Marrow Transplant ; 24(2): 267-275, 2018 02.
Article in English | MEDLINE | ID: mdl-29128554

ABSTRACT

Pathogenesis of chronic graft-versus-host disease (cGVHD) is incompletely defined, involving donor-derived CD4 and CD8-positive T lymphocytes as well as B cells. Standard treatment is lacking for steroid-dependent/refractory cases; therefore, the potential usefulness of tyrosine kinase inhibitors (TKIs) has been suggested, based on their potent antifibrotic effect. However, TKIs seem to have pleiotropic activity. We sought to evaluate the in vitro and in vivo impact of different TKIs on lymphocyte phenotype and function. Peripheral blood mononuclear cells (PBMCs) from healthy donors were cultured in the presence of increasing concentrations of nilotinib, imatinib, dasatinib, and ponatinib; in parallel, 44 PBMC samples from 15 patients with steroid-dependent/refractory cGVHD treated with nilotinib in the setting of a phase I/II trial were analyzed at baseline, after 90, and after 180 days of therapy. Flow cytometry was performed after labeling lymphocytes with a panel of monoclonal antibodies (CD3, CD4, CD16, CD56, CD25, CD19, CD45RA, FoxP3, CD127, and 7-amino actinomycin D). Cytokine production was assessed in supernatants of purified CD3+ T cells and in plasma samples from nilotinib-treated patients. Main T lymphocyte subpopulations were not significantly affected by therapeutic concentrations of TKIs in vitro, whereas proinflammatory cytokine (in particular, IL-2, IFN-γ, tumor necrosis factor-α, and IL-10) and IL-17 production showed a sharp decline. Frequency of T regulatory, B, and natural killer (NK) cells decreased progressively in presence of therapeutic concentrations of all TKIs tested in vitro, except for nilotinib, which showed little effect on these subsets. Of note, naive T regulatory cell (Treg) subset accumulated after exposure to TKIs. Results obtained in vivo on nilotinib-treated patients were largely comparable, both on lymphocyte subset kinetics and on cytokine production by CD3-positive cells. This study underlines the anti-inflammatory and immunomodulatory effects of TKIs and supports their potential usefulness as treatment for patients with steroid-dependent/refractory cGVHD. In addition, both in vitro and in vivo data point out that compared with other TKIs, nilotinib could better preserve the integrity of some important regulatory subsets, such as Treg and NK cells.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Blood Specimen Collection , Cells, Cultured , Cytokines/drug effects , Humans , Immunologic Factors/pharmacology , Killer Cells, Natural/drug effects , Protein Kinase Inhibitors/immunology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , T-Lymphocyte Subsets/drug effects , T-Lymphocytes, Regulatory/drug effects
12.
Mol Cancer Ther ; 16(11): 2572-2585, 2017 11.
Article in English | MEDLINE | ID: mdl-28830985

ABSTRACT

Approximately 10% of non-small cell lung cancer (NSCLC) patients in the United States and 40% of NSCLC patients in Asia have activating epidermal growth factor receptor (EGFR) mutations and are eligible to receive targeted anti-EGFR therapy. Despite an extension of life expectancy associated with this treatment, resistance to EGFR tyrosine kinase inhibitors and anti-EGFR antibodies is almost inevitable. To identify additional signaling routes that can be cotargeted to overcome resistance, we quantified tumor-specific molecular changes that govern resistant cancer cell growth and survival. Mass spectrometry-based quantitative proteomics was used to profile in vivo signaling changes in 41 therapy-resistant tumors from four xenograft NSCLC models. We identified unique and tumor-specific tyrosine phosphorylation rewiring in tumors resistant to treatment with the irreversible third-generation EGFR-inhibitor, osimertinib, or the novel dual-targeting EGFR/Met antibody, JNJ-61186372. Tumor-specific increases in tyrosine-phosphorylated peptides from EGFR family members, Shc1 and Gab1 or Src family kinase (SFK) substrates were observed, underscoring a differential ability of tumors to uniquely escape EGFR inhibition. Although most resistant tumors within each treatment group displayed a marked inhibition of EGFR as well as SFK signaling, the combination of EGFR inhibition (osimertinib) and SFK inhibition (saracatinib or dasatinib) led to further decrease in cell growth in vitro This result suggests that residual SFK signaling mediates therapeutic resistance and that elimination of this signal through combination therapy may delay onset of resistance. Overall, analysis of individual resistant tumors captured unique in vivo signaling rewiring that would have been masked by analysis of in vitro cell population averages. Mol Cancer Ther; 16(11); 2572-85. ©2017 AACR.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/genetics , Piperazines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-met/genetics , Acrylamides , Adaptor Proteins, Signal Transducing/genetics , Aniline Compounds , Animals , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/adverse effects , Benzodioxoles/administration & dosage , Benzodioxoles/adverse effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Humans , Mice , Mutation , Protein Kinase Inhibitors/immunology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Quinazolines/administration & dosage , Quinazolines/adverse effects , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics , Xenograft Model Antitumor Assays , src-Family Kinases/genetics
13.
Clin Cancer Res ; 23(19): 5737-5744, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28679767

ABSTRACT

Purpose: We evaluated tumor burden dynamics in patients with advanced non-small cell lung cancer (NSCLC) treated with commercial PD-1 inhibitors to identify imaging markers associated with improved overall survival (OS).Experimental Design: The study included 160 patients with advanced NSCLC treated with commercial nivolumab or pembrolizumab monotherapy as a part of clinical care. Tumor burden dynamics were studied for the association with OS.Results: Tumor burden change at best overall response (BOR) ranged from -100% to +278% (median, +3.5%). Response rate (RR) was 18% (29/160). Current and former smokers had a higher RR than never smokers (P = 0.04). Durable disease control for at least 6 months was noted in 26 patients (16%), which included 10 patients with stable disease as BOR. Using a landmark analysis, patients with <20% tumor burden increase from baseline within 8 weeks of therapy had longer OS than patients with ≥20% increase (median OS, 12.4 vs. 4.6 months, P < 0.001). Patients with <20% tumor burden increase throughout therapy had significantly reduced hazards of death (HR, 0.24; Cox P < 0.0001) after adjusting for smoking (HR, 0.86; P = 0.61) and baseline tumor burden (HR, 1.55; P = 0.062), even though some patients met criteria for RECIST progression while on therapy. One patient (0.6%) had atypical response pattern consistent with pseudoprogression.Conclusions: Objective response or durable disease control was noted in 24% of patients with advanced NSCLC treated with commercial PD-1 inhibitors. A tumor burden increase of <20% from baseline during therapy was associated with longer OS, proposing a practical marker of treatment benefit. Pseudoprogression is rare in NSCLCs treated with PD-1 inhibitors. Clin Cancer Res; 23(19); 5737-44. ©2017 AACR.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Programmed Cell Death 1 Receptor/genetics , Protein Kinase Inhibitors/administration & dosage , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Disease Progression , Disease-Free Survival , Erlotinib Hydrochloride/administration & dosage , Erlotinib Hydrochloride/adverse effects , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Nivolumab , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Protein Kinase Inhibitors/immunology , Treatment Outcome , Tumor Burden/genetics
14.
Am Soc Clin Oncol Educ Book ; 37: 661-672, 2017.
Article in English | MEDLINE | ID: mdl-28561662

ABSTRACT

There has been a therapeutic revolution in the treatment of metastatic melanoma over the past decade. Patients presenting with inoperable disease have several therapeutic options, which can include both targeted and immune therapy. Immune checkpoint inhibitors have demonstrated an improvement in overall survival and led to some durable responses. However, toxicity, especially in combination regimens, can be severe. Adverse events should be anticipated, diagnosed as early as possible, monitored, and managed. Combination BRAF and MEK inhibition has also been shown to improve overall survival in patients with V600E-mutated melanoma. Responses to therapy are often rapid, and treatment is not associated with immune-related adverse events. Current trials are under way to determine which option is optimal as frontline therapy for patients with V600E melanoma. In patients with progressive disease despite standard therapies, clinical trials are recommended. There are several promising agents in development.


Subject(s)
Immunotherapy , MAP Kinase Kinase Kinase 1/genetics , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/drug therapy , Drug-Related Side Effects and Adverse Reactions/immunology , Drug-Related Side Effects and Adverse Reactions/pathology , Humans , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase Kinase 1/immunology , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Molecular Targeted Therapy , Mutation , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/immunology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/immunology , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Treatment Outcome
15.
Clin. transl. oncol. (Print) ; 18(11): 1072-1081, nov. 2016. tab
Article in English | IBECS | ID: ibc-156872

ABSTRACT

Treatment with regorafenib has demonstrated statistically significant improvements in terms of overall survival, progression-free survival and disease control when compared with placebo in pretreated patients with metastatic colorectal cancer in two placebo-controlled, randomized, phase III trials (CORRECT and CONCUR). Similar results were observed in two open-label, single-arm studies (REBECCA and CONSIGN) performed in the real-world setting. But several authors have suggested that the benefit provided by regorafenib may not be clinically meaningful for these patients. Moreover, it has been suggested that not all subgroups of patients might benefit from regorafenib. The intention of this review is to provide an overview of the existing evidence for regorafenib in terms of efficacy, tolerability and quality of life in different subpopulations according to clinical and biological characteristics. Additionally, the magnitude of the clinical benefit provided by regorafenib to these patients has been explored and whether there are poorer outcomes in certain subpopulations (AU)


No disponible


Subject(s)
Humans , Male , Female , Colonic Neoplasms/complications , Colonic Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Neoplasm Metastasis/therapy , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/drug therapy , Protein Kinase Inhibitors/analysis , Protein Kinase Inhibitors/immunology , Quality of Life , Placebos/therapeutic use , Phenylurea Compounds/therapeutic use
17.
Oncol Rep ; 34(6): 2864-70, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26503698

ABSTRACT

EGFR/HER2 are frequently expressed in MPM tissues, however, no studies have shown the clinical benefit of using EGFR/HER2-targeting drugs in patients with malignant pleural mesothelioma (MPM). It was reported that the tyrosine kinase inhibitor (TKI) lapatinib enhanced trastuzumab-mediated antibody-dependent cellular cytotoxicity (ADCC) in HER2-positive breast cancer, suggesting that this combination is a promising strategy for MPM treatment. The aim of the present study was to explore the possibility of a TKI combined with trastuzumab to enhance ADCC in MPM cells. Five MPM cell lines were used to test the effects of TKIs targeting EGFR (gefitinib, afatinib and lapatinib) on cell proliferation and the expression of the HER family receptor. The combined effects of TKI with trastuzumab on ADCC were evaluated using the LDH release assay. Additionally, MPM cells were isolated from patients and evaluated for lapatinib-induced upregulation of HER family receptors and trastuzumab- or cetuximab­mediated ADCC. In MPM cell lines, HER2 expression was upregulated by lapatinib, downregulated by afatinib and unaffected by gefitinib. As expected, more trastuzumab bound to MPM cells pretreated with lapatinib than untreated cells, resulting in the enhancement of trastuzumab-mediated ADCC in MPM cells. In patient-derived MPM cells, both HER2 and EGFR were upregulated by lapatinib, resulting in the enhancement of both trastuzumab- and cetuximab-mediated ADCC. Of the three TKIs, only lapatinib enhanced trastuzumab-mediated ADCC via the upregulation of HER2 expression in MPM cells, suggesting that sequential combination of lapatinib and trastuzumab may be a promising strategy for MPM treatment.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Quinazolines/administration & dosage , Receptor, ErbB-2/biosynthesis , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/immunology , Antibody-Dependent Cell Cytotoxicity/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cetuximab/administration & dosage , Cetuximab/immunology , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Humans , Lapatinib , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mesothelioma/immunology , Mesothelioma/pathology , Mesothelioma, Malignant , Mice , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/immunology , Trastuzumab/administration & dosage , Trastuzumab/immunology , Xenograft Model Antitumor Assays
18.
Biol Pharm Bull ; 38(10): 1652-7, 2015.
Article in English | MEDLINE | ID: mdl-26424026

ABSTRACT

In this paper, we describe the production of the first specific antibodies against the tyrosine kinase inhibitors lapatinib and nilotinib. Anti-lapatinib antibody was obtained by immunizing rabbits with an antigen conjugated with bovine serum albumin using 3-chloro-4-((3-fluorobenzyl)oxy)aniline. Anti-nilotinib antibody was produced by immunizing mice with an antigen conjugated with bovine serum albumin using 2-(5-amino-2-methylanilino)-4-(3-pyridyl)pyrimidine. The generated antibodies were used to develop highly sensitive and specific enzyme-linked immunosorbent assays (ELISAs) for lapatinib and nilotinib in human serum. The assays were capable of detecting lapatinib and nilotinib at serum concentrations as low as 40 and 8 ng/mL, respectively. Using the two ELISAs, drugs levels were easily measured in the serum of rats after a single dose oral administration of lapatinib or nilotinib. The assays are therefore expected be valuable tools for therapeutic drug monitoring in the clinical setting and pharmacokinetic studies of lapatinib and nilotinib.


Subject(s)
Antibodies/immunology , Protein Kinase Inhibitors/immunology , Pyrimidines/immunology , Quinazolines/immunology , Animals , Drug Monitoring , Enzyme-Linked Immunosorbent Assay , Female , Horseradish Peroxidase , Humans , Lapatinib , Male , Protein Kinase Inhibitors/blood , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/blood , Quinazolines/blood , Rabbits , Rats, Wistar
19.
BMC Ophthalmol ; 15: 64, 2015 Jun 27.
Article in English | MEDLINE | ID: mdl-26115916

ABSTRACT

BACKGROUND: Previous studies demonstrate changes of autoantibody concentrations against retinal and optic nerve head antigens in the serum of glaucoma patients in comparison to healthy persons. These antibodies belong to the natural autoimmunity. Previous studies showed up regulated, but also significantly down-regulated autoantibody levels. These antibodies have the ability to influence protein profiles of neuroretinal cells and possibly hold neuroprotective potential, as we have been able to demonstrate before. Aim of this study was to analyse the serum and antibody effect of glaucoma patients on neuroretinal cells in more detail and also determine the impact of antibodies found down-regulated in glaucoma patients on the pathogenesis of the neurodegenerative disease glaucoma. METHODS: Neuroretinal cells (RGC-5) were incubated with serum either from glaucoma patients or healthy controls for 24 h. Mass spectrometric analysis was performed after cell lysis. Furthermore the neuroretinal cells were preincubated with different and concentrations of 14-3-3 antibodies (0.005, 0.1, 0.5, 1, 5 and 10 µg/ml) and then stressed with H2O2, staurosporine or glutamate. Viability tests were performed with crystal violet and ROS tests with DCFH-DA. Antibody location in the cell after antibody incubation was performed with immunocytochemical methods. Additionally mass spectrometric analysis was performed with the cells after antibody incubation. RESULTS: Protein expression analysis with Maldi-Orbitrap MS showed changes in the expression level of regulatory proteins in cells incubated with glaucoma serum, e.g. an up-regulation of 14-3-3 and a down-regulation of Calmodulin. After preincubation of the cells with anti-14-3-3 antibody and stressing the cells, we detected an increase in viability of up to 22 % and a decrease in reactive oxygen species (ROS) of up to 31 %. Proteomic 1 analysis involvement of the mitochondrial apoptosis pathway in this protective effect and immunohistochemical analysis showed an antibody uptake in the cells. CONCLUSION: We found significant effects of serum antibodies on proteins of neuroretinal cells especially of the mitochondrial apoptosis pathway. Furthermore we detected a protective potential of antibodies down-regulated in glaucoma patients. The changed autoantibodies belong to the natural autoimmunity. We conclude that changes in the natural autoimmunity of patients with glaucoma can negatively impact regulatory functions.


Subject(s)
14-3-3 Proteins/immunology , Apoptosis/physiology , Autoantibodies/physiology , Glaucoma, Open-Angle/blood , Mitochondria/metabolism , Protein Kinase Inhibitors/immunology , Retinal Ganglion Cells/cytology , Cell Line , Cell Survival , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique, Indirect , Glutamic Acid/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Oxidative Stress , Proteomics , Reactive Oxygen Species/metabolism , Retinal Ganglion Cells/drug effects , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Staurosporine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...