Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 48(6): 1077-1080, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29924973

ABSTRACT

Four studies recently reported in the New England Journal of Medicine highlight advances in treatment with immune checkpoint blockade across the cancer care continuum. These findings demonstrate efficacy of these agents in the treatment of early and late-stage disease, as monotherapy or in combination, and in addition to-or in place of-standard front-line therapy.


Subject(s)
Antibodies, Monoclonal, Humanized , Immunotherapy , Continuity of Patient Care , Humans , Melanoma
2.
Nature ; 577(7791): 549-555, 2020 01.
Article in English | MEDLINE | ID: mdl-31942075

ABSTRACT

Treatment with immune checkpoint blockade (ICB) has revolutionized cancer therapy. Until now, predictive biomarkers1-10 and strategies to augment clinical response have largely focused on the T cell compartment. However, other immune subsets may also contribute to anti-tumour immunity11-15, although these have been less well-studied in ICB treatment16. A previously conducted neoadjuvant ICB trial in patients with melanoma showed via targeted expression profiling17 that B cell signatures were enriched in the tumours of patients who respond to treatment versus non-responding patients. To build on this, here we performed bulk RNA sequencing and found that B cell markers were the most differentially expressed genes in the tumours of responders versus non-responders. Our findings were corroborated using a computational method (MCP-counter18) to estimate the immune and stromal composition in this and two other ICB-treated cohorts (patients with melanoma and renal cell carcinoma). Histological evaluation highlighted the localization of B cells within tertiary lymphoid structures. We assessed the potential functional contributions of B cells via bulk and single-cell RNA sequencing, which demonstrate clonal expansion and unique functional states of B cells in responders. Mass cytometry showed that switched memory B cells were enriched in the tumours of responders. Together, these data provide insights into the potential role of B cells and tertiary lymphoid structures in the response to ICB treatment, with implications for the development of biomarkers and therapeutic targets.


Subject(s)
B-Lymphocytes/immunology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/immunology , Immunotherapy , Melanoma/drug therapy , Melanoma/immunology , Tertiary Lymphoid Structures/immunology , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Biomarkers, Tumor/analysis , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/surgery , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/immunology , Clone Cells/cytology , Clone Cells/immunology , Clone Cells/metabolism , Dendritic Cells, Follicular/cytology , Dendritic Cells, Follicular/immunology , Gene Expression Regulation, Neoplastic , Humans , Immunologic Memory/immunology , Mass Spectrometry , Melanoma/pathology , Melanoma/surgery , Neoplasm Metastasis/genetics , Phenotype , Prognosis , RNA-Seq , Receptors, Immunologic/immunology , Single-Cell Analysis , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transcriptome
3.
Trends Immunol ; 39(11): 900-920, 2018 11.
Article in English | MEDLINE | ID: mdl-30392721

ABSTRACT

The human microbiome is a complex aggregate of microorganisms, and their genomes exert a number of influences crucial to the metabolic, immunologic, hormonal, and homeostatic function of the host. Recent work, both in preclinical mouse models and human studies, has shed light on the impact of gut and tumor microbiota on responses to systemic anticancer therapeutics. In light of this, strategies to target the microbiome to improve therapeutic responses are underway, including efforts to target gut and intratumoral microbes. Here, we discuss mechanisms by which microbiota may impact systemic and antitumor immunity, in addition to outstanding questions in the field. A deeper understanding of these is critical as we devise putative strategies to target the microbiome.


Subject(s)
Gastrointestinal Tract/immunology , Immunotherapy/methods , Microbiota/physiology , Neoplasms/therapy , Animals , Gastrointestinal Tract/microbiology , Homeostasis , Host-Pathogen Interactions , Humans , Immunity , Mice , Neoplasms/microbiology , Tumor Microenvironment
4.
Methods Mol Biol ; 2582: 13-21, 2023.
Article in English | MEDLINE | ID: mdl-36370340

ABSTRACT

CCN4 (also known as WNT1-Inducible Signaling Pathway Protein 1 or WISP1) is a 367 amino acid, 40 kDa protein located on chromosome 8q24.1-8q24.3. Prior studies have provided support for a pro-inflammatory role for CCN4. We have shown recently that CCN4 expression is associated with advanced disease, epithelial-mesenchymal transition, and an inflamed tumor microenvironment in multiple solid tumors. We detail here the CCN4 tissue microarray immunofluorescence protocol related to these findings.


Subject(s)
CCN Intercellular Signaling Proteins , Neoplasms , Humans , CCN Intercellular Signaling Proteins/genetics , CCN Intercellular Signaling Proteins/metabolism , Tumor Microenvironment , Epithelial-Mesenchymal Transition , Fluorescent Antibody Technique
5.
Immunotherapy ; 15(12): 921-932, 2023 08.
Article in English | MEDLINE | ID: mdl-37431608

ABSTRACT

Background: There are limited clinical data comparing extended dosing (ED) versus standard dosing (SD) of pembrolizumab for metastatic non-small-cell lung cancer. Methods: This retrospective study included patients with metastatic non-small-cell lung cancer and PD-L1 tumor proportion score ≥50% treated with one or more cycles of single-agent pembrolizumab with SD or ED from January 2018 to December 2020. Results: A higher proportion of patients were alive in the ED group (vs SD) at 6 months (94 vs 51%), 12 months (94 vs 33%) and data cutoff (94 vs 26%) (p < 0.001 for all). The rate (44 vs 32%; p = 0.407) and severity of grade ≥3 immune-related adverse events were similar (50 vs 52%); however, ED patients more frequently discontinued treatment due to toxicity (45 vs 15%; p < 0.001). Conclusion: A greater proportion of ED patients were alive at data cutoff, and the rate and severity of immune-related adverse events were similar between groups.


Cancer in the lungs can be treated with drugs that use your immune system to kill cancer. This study showed that patients lived longer when the drugs were given further apart, and that the extended treatment was equally safe as the standard dosing schedule.


Subject(s)
COVID-19 , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Pandemics , Retrospective Studies , B7-H1 Antigen , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
6.
Cancer Treat Res Commun ; 34: 100678, 2023.
Article in English | MEDLINE | ID: mdl-36608489

ABSTRACT

BACKGROUND: Durvalumab following chemoradiation in unresectable stage III non-small cell lung cancer (NSCLC) has led to improved outcomes. The schedule of administration has been determined by pharmacokinetic studies. This study evaluates real-world efficacy and safety outcomes of extended dosing (ED) vs. standard dosing (SD) of durvalumab. METHODS: Stage III NSCLC patients treated at the Cancer center of Southeastern Ontario with consolidative durvalumab from March 2017-December 2020 were included. Patient characteristics and outcomes were evaluated through retrospective review. Comparisons were made using chi-square and t-tests. Kaplan-Meier curves were used to analyze overall survival (OS). RESULTS: A total of 35 patients were included; 15 (43%) switched to ED. Distant recurrence rates were higher in the ED group (53% vs. 20%, p = 0.07), with no differences in the sites of disease recurrence. A similar proportion of patients were alive in the ED vs. SD group (93% vs. 80%, p = 0.3), with no significant difference in OS. There were less grade 3 or greater immune-related adverse events in the ED group (0% vs. 20%). Treatment discontinuation occurred in 47% vs. 50% in the ED vs. SD groups, respectively, owing to toxicity in 20% of patients in the ED group vs. 40% in the SD group. CONCLUSIONS: Extended dosing has similar efficacy and toxicity to standard dosing; however, there was a higher rate of toxicity necessitating discontinuation in the SD group, which may have impacted the clinical decision-making to switch to ED. Our data is limited by a small sample size and should be further validated in larger cohorts.


Subject(s)
Antineoplastic Agents, Immunological , COVID-19 , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Pandemics , Antineoplastic Agents, Immunological/adverse effects , Neoplasm Staging , Neoplasm Recurrence, Local/drug therapy
7.
Diagnostics (Basel) ; 13(11)2023 May 27.
Article in English | MEDLINE | ID: mdl-37296725

ABSTRACT

It is believed that fetal hemoglobin (HbF) expression in adults is largely genetically regulated. The increased expression of HbF in pregnancy has been reported in a small number of articles. Different mechanisms have been proposed, but the description of HbF expression during pregnancy remains unclear. The objectives of this study were to document HbF expression during peri and postpartum periods, confirm its maternal origin, and assess clinical and biochemical parameters potentially associated with HbF modulation. In this observational prospective study, 345 pregnant women were followed. At baseline, 169 had HbF expression (≥1% of total hemoglobin) and 176 did not have HbF expression. Women were followed at the obstetric clinic during their pregnancy. Clinical and biochemical parameters were measured at each visit. Analyses were made to determine which parameters had a significant correlation to HbF expression. Results show that HbF expression of ≥1% during peri and postpartum periods in pregnant women without influencing comorbidities is at its highest peak during the first trimester. In all women, it was proven that HbF was of maternal origin. A significant positive correlation between HbF expression, ßeta-human chorionic gonadotropin (ß-HCG), and glycosylated hemoglobin (HbA1c) was present. A significant negative association between HbF expression and total hemoglobin was found. HbF expression induction during pregnancy is probably associated with an increase in ß-HCG and HbA1C, and a decrease in total hemoglobin, which could temporarily reactivate the fetal erythropoietic system.

8.
Nat Med ; 29(10): 2559-2569, 2023 10.
Article in English | MEDLINE | ID: mdl-37814061

ABSTRACT

Circulating tumor DNA (ctDNA) has shown promise in capturing primary resistance to immunotherapy. BR.36 is a multi-center, randomized, ctDNA-directed, phase 2 trial of molecular response-adaptive immuno-chemotherapy for patients with lung cancer. In the first of two independent stages, 50 patients with advanced non-small cell lung cancer received pembrolizumab as standard of care. The primary objectives of stage 1 were to ascertain ctDNA response and determine optimal timing and concordance with radiologic Response Evaluation Criteria in Solid Tumors (RECIST) response. Secondary endpoints included the evaluation of time to ctDNA response and correlation with progression-free and overall survival. Maximal mutant allele fraction clearance at the third cycle of pembrolizumab signified molecular response (mR). The trial met its primary endpoint, with a sensitivity of ctDNA response for RECIST response of 82% (90% confidence interval (CI): 52-97%) and a specificity of 75% (90% CI: 56.5-88.5%). Median time to ctDNA response was 2.1 months (90% CI: 1.5-2.6), and patients with mR attained longer progression-free survival (5.03 months versus 2.6 months) and overall survival (not reached versus 7.23 months). These findings are incorporated into the ctDNA-driven interventional molecular response-adaptive second stage of the BR.36 trial in which patients at risk of progression are randomized to treatment intensification or continuation of therapy. ClinicalTrials.gov ID: NCT04093167 .


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Antibodies, Monoclonal, Humanized , Progression-Free Survival
9.
J Thorac Oncol ; 18(6): 813-819, 2023 06.
Article in English | MEDLINE | ID: mdl-36841541

ABSTRACT

Immune checkpoint inhibitors have activity in mesothelioma. IND.227 was a phase 2 trial (120 patients planned) comparing progression-free survival of standard platinum and pemetrexed (CP) versus CP + pembrolizumab (pembro) versus pembro. Accrual to the pembro arm was discontinued on the basis of interim analysis (IA-16 wk disease control rate). CP + pembro was tolerable, with progression-free survival similar between arms and median survival and overall response rate higher than those of CP alone (19.8 mo [95% confidence interval or CI: 8.4-41.36] versus 8.9 mo [95% CI: 5.3-12.8] and 47% [95% CI: 24%-71%] versus 19% [95% CI: 5%-42%], respectively). The subsequent phase 3 trial has completed accrual; results are expected in 2023.


Subject(s)
Lung Neoplasms , Mesothelioma, Malignant , Mesothelioma , Pleural Neoplasms , Humans , Lung Neoplasms/pathology , Canada , Mesothelioma/pathology , Pemetrexed/pharmacology , Pemetrexed/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Pleural Neoplasms/pathology
10.
J Immunother Cancer ; 10(6)2022 06.
Article in English | MEDLINE | ID: mdl-35715004

ABSTRACT

With the advent of immunotherapy as one of the keystones of the treatment of our patients with cancer, a number of atypical patterns of response to these agents has been identified. These include pseudoprogression, where the tumor initially shows objective growth before decreasing in size, and hyperprogression, hypothesized to be a drug-induced acceleration of the tumor burden. Despite it being >10 years since the first immune-oncology drug was approved, neither the biology behind these paradoxical responses has been well understood, nor their incidence, identification criteria, predictive biomarkers, or clinical impact have been fully described. Immune-based Response Evaluation Criteria in Solid Tumors (iRECIST) guidelines have been published as a revision to the RECIST V.1.1 criteria for use in trials of immunotherapeutics, and the iRECIST subcommittee (of the RECIST Working Group) is working on elucidating these aspects, with data sharing a current major challenge to move forward with this unmet need in immuno-oncology.


Subject(s)
Immunotherapy , Neoplasms , Disease Progression , Humans , Neoplasms/therapy , Response Evaluation Criteria in Solid Tumors , Tumor Burden
11.
Nat Commun ; 12(1): 2606, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33972557

ABSTRACT

Understanding resistance mechanisms to targeted therapies and immune checkpoint blockade in mutant KRAS lung cancers is critical to developing novel combination therapies and improving patient survival. Here, we show that MEK inhibition enhanced PD-L1 expression while PD-L1 blockade upregulated MAPK signaling in mutant KRAS lung tumors. Combined MEK inhibition with anti-PD-L1 synergistically reduced lung tumor growth and metastasis, but tumors eventually developed resistance to sustained combinatorial therapy. Multi-platform profiling revealed that resistant lung tumors have increased infiltration of Th17 cells, which secrete IL-17 and IL-22 cytokines to promote lung cancer cell invasiveness and MEK inhibitor resistance. Antibody depletion of IL-17A in combination with MEK inhibition and PD-L1 blockade markedly reduced therapy-resistance in vivo. Clinically, increased expression of Th17-associated genes in patients treated with PD-1 blockade predicted poorer overall survival and response in melanoma and predicated poorer response to anti-PD1 in NSCLC patients. Here we show a triple combinatorial therapeutic strategy to overcome resistance to combined MEK inhibitor and PD-L1 blockade.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Th17 Cells/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/immunology , Drug Synergism , Female , Humans , Immune Checkpoint Inhibitors/immunology , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/immunology , Neoplasm Metastasis , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/metabolism , Th17 Cells/immunology , Tumor Suppressor Protein p53/metabolism
12.
Oncoimmunology ; 10(1): 1992880, 2021.
Article in English | MEDLINE | ID: mdl-34777916

ABSTRACT

Targeted and immunotherapy regimens have revolutionized the treatment of advanced melanoma patients. Despite this, only a subset of patients respond durably. Recently, combination strategies of BRAF/MEK inhibitors with immune checkpoint inhibitor monotherapy (α-CTLA-4 or α-PD-1) have increased the rate of durable responses. Based on evidence from our group and others, these therapies appear synergistic, but at the cost of significant toxicity. We know from other treatment paradigms (e.g. hematologic malignancies) that combination strategies with multi-drug regimens (>4 drugs) are associated with more durable disease control. To better understand the mechanism of these improved outcomes, and to identify and prioritize new strategies for testing, we studied several multi-drug regimens combining BRAF/MEK targeted therapy and immunotherapy combinations in a Braf-mutant murine melanoma model (BrafV600E/Pten-/- ). Short-term treatment with α-PD-1 and α-CTLA-4 monotherapies were relatively ineffective, while treatment with α-OX40 demonstrated some efficacy [17% of mice with no evidence of disease, (NED), at 60-days]. Outcomes were improved in the combined α-OX40/α-PD-1 group (42% NED). Short-term treatment with quadruplet therapy of immunotherapy doublets in combination with targeted therapy [dabrafenib and trametinib (DT)] was associated with excellent tumor control, with 100% of mice having NED after combined DT/α-CTLA-4/α-PD-1 or DT/α-OX40/α-PD-1. Notably, tumors from mice in these groups demonstrated a high proportion of effector memory T cells, and immunologic memory was maintained with tumor re-challenge. Together, these data provide important evidence regarding the potential utility of multi-drug therapy in treating advanced melanoma and suggest these models can be used to guide and prioritize combinatorial treatment strategies.


Subject(s)
Melanoma , Pharmaceutical Preparations , Animals , Humans , Immunotherapy , Melanoma/drug therapy , Melanoma/genetics , Memory T Cells , Mice , Mitogen-Activated Protein Kinase Kinases , Proto-Oncogene Proteins B-raf/genetics
13.
Nat Med ; 27(8): 1432-1441, 2021 08.
Article in English | MEDLINE | ID: mdl-34239137

ABSTRACT

Treatment with combined immune checkpoint blockade (CICB) targeting CTLA-4 and PD-1 is associated with clinical benefit across tumor types, but also a high rate of immune-related adverse events. Insights into biomarkers and mechanisms of response and toxicity to CICB are needed. To address this, we profiled the blood, tumor and gut microbiome of 77 patients with advanced melanoma treated with CICB, with a high rate of any ≥grade 3 immune-related adverse events (49%) with parallel studies in pre-clinical models. Tumor-associated immune and genomic biomarkers of response to CICB were similar to those identified for ICB monotherapy, and toxicity from CICB was associated with a more diverse peripheral T-cell repertoire. Profiling of gut microbiota demonstrated a significantly higher abundance of Bacteroides intestinalis in patients with toxicity, with upregulation of mucosal IL-1ß in patient samples of colitis and in pre-clinical models. Together, these data offer potential new therapeutic angles for targeting toxicity to CICB.


Subject(s)
CTLA-4 Antigen/immunology , Gastrointestinal Microbiome , Programmed Cell Death 1 Receptor/immunology , Animals , Cell Line, Tumor , Female , Humans , Interleukin-1beta/immunology , Melanoma , Mice , Mice, Inbred C57BL
14.
Science ; 374(6575): 1632-1640, 2021 Dec 24.
Article in English | MEDLINE | ID: mdl-34941392

ABSTRACT

Gut bacteria modulate the response to immune checkpoint blockade (ICB) treatment in cancer, but the effect of diet and supplements on this interaction is not well studied. We assessed fecal microbiota profiles, dietary habits, and commercially available probiotic supplement use in melanoma patients and performed parallel preclinical studies. Higher dietary fiber was associated with significantly improved progression-free survival in 128 patients on ICB, with the most pronounced benefit observed in patients with sufficient dietary fiber intake and no probiotic use. Findings were recapitulated in preclinical models, which demonstrated impaired treatment response to anti­programmed cell death 1 (anti­PD-1)­based therapy in mice receiving a low-fiber diet or probiotics, with a lower frequency of interferon-γ­positive cytotoxic T cells in the tumor microenvironment. Together, these data have clinical implications for patients receiving ICB for cancer.


Subject(s)
Dietary Fiber , Gastrointestinal Microbiome , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/therapy , Probiotics , Animals , Cohort Studies , Fatty Acids, Volatile/analysis , Fecal Microbiota Transplantation , Feces/chemistry , Feces/microbiology , Female , Humans , Immunotherapy , Male , Melanoma/immunology , Melanoma/microbiology , Melanoma, Experimental/immunology , Melanoma, Experimental/microbiology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Progression-Free Survival , T-Lymphocytes
15.
J Thorac Oncol ; 16(1): 127-139, 2021 01.
Article in English | MEDLINE | ID: mdl-33096269

ABSTRACT

INTRODUCTION: The combination of programmed cell death protein-1 or programmed death-ligand 1 immune checkpoint blockade and chemotherapy has revolutionized the treatment of advanced NSCLC, but the mechanisms underlying this synergy remain incompletely understood. In this study, we explored the relationships between neoadjuvant chemotherapy and the immune microenvironment (IME) of resectable NSCLC to identify novel mechanisms by which chemotherapy may enhance the effect of immune checkpoint blockade. METHODS: Genomic, transcriptomic, and immune profiling data of 511 patients treated with neoadjuvant chemotherapy followed by surgery (NCT) versus upfront surgery (US) were compared with determined differential characteristics of the IMEs derived from whole-exome sequencing (NCT = 18; US = 73), RNA microarray (NCT = 45; US = 202), flow cytometry (NCT = 17; US = 39), multiplex immunofluorescence (NCT = 10; US = 72), T-cell receptor sequencing (NCT = 16 and US = 63), and circulating cytokines (NCT = 18; US = 73). RESULTS: NCT was associated with increased infiltration of cytotoxic CD8+ T cells and CD20+ B cells. Moreover, NCT was associated with increases in CD8+CD103+ and CD4+CD103+PD-1+TIM3- tissue resident memory T cells. Gene expression profiling supported memory function of CD8+ and CD4+ T cells. However, NCT did not affect T-cell receptor clonality, richness, or tumor mutational burden. Finally, NCT was associated with decreased plasma BDNF (TrkB) at baseline and week 4 after surgery. CONCLUSIONS: Our study supports that, in the context of resectable NSCLC, neoadjuvant chemotherapy promotes antitumor immunity through T and B cell recruitment in the IME and through a phenotypic change toward cytotoxic and memory CD8+ and CD4+ memory helper T cells.


Subject(s)
Lung Neoplasms , Neoadjuvant Therapy , B-Lymphocytes , CD8-Positive T-Lymphocytes , Humans , Immunologic Memory , Lung Neoplasms/drug therapy , Tumor Microenvironment
16.
Clin Lung Cancer ; 21(4): 384-388, 2020 07.
Article in English | MEDLINE | ID: mdl-32299768

ABSTRACT

Current strategies to improve clinical outcomes in v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog-mutant non-small-cell lung cancer (NSCLC) patients include mitogen-activated protein kinase kinase 1 inhibitor and programmed death (PD) 1 (PD-1)/PD ligand 1 (PD-L1) immune checkpoint blockade combinations. Experience from treatment of melanoma suggests that anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and anti-PD-1/PD-L1 combinations improve outcomes, but similar benefits remain to be seen for treatment of NSCLC. This report describes a single center, investigator-initiated phase I/II clinical trial to compare 2 combination schedules of intermittent or continuous selumetinib (AZD6244, ARRY-142886), tremelimumab (anti-CTLA-4), and durvalumab (anti-PD-L1) treatment with historical controls in patients with previously treated, unresectable NSCLC. Forty patients will be accrued at the University of Texas M.D. Anderson Cancer Center. Primary objectives include maximum tolerated dose (dose escalation phase) and progression-free survival (dose expansion phase). Secondary objectives include response rate according to Response Evaluation Criteria In Solid Tumors version 1.1, disease control rate, overall survival, safety, and duration of response. Exploratory objectives are to assess biomarkers of response and resistance on the basis of biopsies and peripheral blood taken before and after treatment using immune profiling, transcriptome, and protein readouts.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Immunotherapy/methods , Lung Neoplasms/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Benzimidazoles/administration & dosage , Carcinoma, Non-Small-Cell Lung/pathology , Female , Follow-Up Studies , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Prognosis , Research Design , Survival Rate , Young Adult
17.
Oncoimmunology ; 8(5): e1581545, 2019.
Article in English | MEDLINE | ID: mdl-31069142

ABSTRACT

Background: WNT1-Inducible Signaling Pathway Protein 1 (WISP1) is implicated in prostate cancer growth and metastasis and the regulation of inflammation in diverse benign diseases. The objectives of this study were to assess the prognostic value of WISP1, its association to inflammation and its relevance as a biomarker for immune checkpoint blockade (ICB) response. Methods: Publicly available RNA-seq datasets were used to evaluate the prognostic value of WISP1 gene expression and its association with tumor-infiltrating lymphocytes, inflamed tumor microenvironment, and anti-PD-1 ICB response. A tissue microarray (TMA) including 285 radical prostatectomy specimens was used to confirm these associations in prostate cancer. The effect of recombinant WISP1 (rWISP1) on inflammatory cytokines was assessed in vitro. Results: High levels of WISP1 correlated with BCR-free survival in prostate adenocarcinoma and overall survival in primary melanoma, low-grade glioma, and kidney papillary cell carcinoma. Some effects could be accounted for by higher WISP1 expression in advanced disease. High WISP1 expression in prostate adenocarcinoma was correlated with CD8+ cells density. In vitro, rWISP1 increased inflammatory cytokine production. High WISP1 gene expression in RNA-seq datasets was correlated with gene signatures of multiple immune cell types as well as an inflammatory cytokine, immune checkpoint, and epithelial-mesenchymal transition (EMT) gene expression. WISP1 mRNA expression was associated with primary resistance to ICB in datasets showing EMT. Conclusions: Our results support an association between WISP1 expression and advanced disease, EMT and an inflamed tumor microenvironment in multiple solid tumors. The consequences of WISP1 expression on cancer immunotherapy remains to be addressed.

18.
Cell Res ; 29(10): 846-861, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31481761

ABSTRACT

PD-1 blockade represents a major therapeutic avenue in anticancer immunotherapy. Delineating mechanisms of secondary resistance to this strategy is increasingly important. Here, we identified the deleterious role of signaling via the type I interferon (IFN) receptor in tumor and antigen presenting cells, that induced the expression of nitric oxide synthase 2 (NOS2), associated with intratumor accumulation of regulatory T cells (Treg) and myeloid cells and acquired resistance to anti-PD-1 monoclonal antibody (mAb). Sustained IFNß transcription was observed in resistant tumors, in turn inducing PD-L1 and NOS2 expression in both tumor and dendritic cells (DC). Whereas PD-L1 was not involved in secondary resistance to anti-PD-1 mAb, pharmacological or genetic inhibition of NOS2 maintained long-term control of tumors by PD-1 blockade, through reduction of Treg and DC activation. Resistance to immunotherapies, including anti-PD-1 mAb in melanoma patients, was also correlated with the induction of a type I IFN signature. Hence, the role of type I IFN in response to PD-1 blockade should be revisited as sustained type I IFN signaling may contribute to resistance to therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Interferon Type I/metabolism , Programmed Cell Death 1 Receptor/immunology , Signal Transduction/drug effects , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/metabolism , Cell Line, Tumor , Dendritic Cells/cytology , Dendritic Cells/metabolism , Drug Resistance, Neoplasm , Humans , Kaplan-Meier Estimate , Melanoma/drug therapy , Melanoma/mortality , Melanoma/pathology , Mice , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/mortality , Neoplasms/pathology , Nitric Oxide Synthase Type II/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
19.
Clin Lung Cancer ; 18(1): 13-22, 2017 01.
Article in English | MEDLINE | ID: mdl-27461776

ABSTRACT

Lung carcinoma is the leading cause of death by cancer worldwide. When possible, surgery is the best treatment strategy for patients with non-small-cell lung cancer. However, even with curative-intent therapy, most patients will develop local or systemic recurrence and, ultimately, succumb to their disease. In recent years, evidence on the role of the antitumor activity of the immune system and the understanding of tumor immunosurveillance have resulted in the emergence of immunotherapy as a promising therapeutic approach in lung cancer. The main approaches are immune checkpoint inhibition, such as blockade of the cytotoxic T-lymphocyte antigen-4 and programmed cell death-1 receptors and the programmed cell death-1 ligand, and vaccine therapy, which elicits specific antitumor immunity against relevant tumor-associated antigens. We have reviewed recently reported results from clinical trials and the possible future role of vaccine therapy and immune checkpoint inhibition in the treatment of small cell lung cancer and non-small-cell lung cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunotherapy , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Animals , Humans , Prognosis
20.
Biomark Cancer ; 8(Suppl 2): 15-33, 2016.
Article in English | MEDLINE | ID: mdl-27168728

ABSTRACT

Prostate cancer (PC) is the second most common form of cancer in men worldwide. Biomarkers have emerged as essential tools for treatment and assessment since the variability of disease behavior, the cost and diversity of treatments, and the related impairment of quality of life have given rise to a need for a personalized approach. High-throughput technology platforms in proteomics and genomics have accelerated the development of biomarkers. Furthermore, recent successes of several new agents in PC, including immunotherapy, have stimulated the search for predictors of response and resistance and have improved the understanding of the biological mechanisms at work. This review provides an overview of currently established biomarkers in PC, as well as a selection of the most promising biomarkers within these particular fields of development.

SELECTION OF CITATIONS
SEARCH DETAIL