Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 227
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 182(6): 1401-1418.e18, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32810439

ABSTRACT

Blood myeloid cells are known to be dysregulated in coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2. It is unknown whether the innate myeloid response differs with disease severity and whether markers of innate immunity discriminate high-risk patients. Thus, we performed high-dimensional flow cytometry and single-cell RNA sequencing of COVID-19 patient peripheral blood cells and detected disappearance of non-classical CD14LowCD16High monocytes, accumulation of HLA-DRLow classical monocytes (Human Leukocyte Antigen - DR isotype), and release of massive amounts of calprotectin (S100A8/S100A9) in severe cases. Immature CD10LowCD101-CXCR4+/- neutrophils with an immunosuppressive profile accumulated in the blood and lungs, suggesting emergency myelopoiesis. Finally, we show that calprotectin plasma level and a routine flow cytometry assay detecting decreased frequencies of non-classical monocytes could discriminate patients who develop a severe form of COVID-19, suggesting a predictive value that deserves prospective evaluation.


Subject(s)
Coronavirus Infections , Coronavirus , Pandemics , Pneumonia, Viral , Betacoronavirus , COVID-19 , Flow Cytometry , Humans , Leukocyte L1 Antigen Complex , Monocytes , Myeloid Cells , Prospective Studies , SARS-CoV-2
3.
Curr Oncol Rep ; 25(8): 857-867, 2023 08.
Article in English | MEDLINE | ID: mdl-37129706

ABSTRACT

PURPOSE OF REVIEW: This review presents the rationale for intratumoral immunotherapy, technical considerations and safety. Clinical results from the latest trials are provided and discussed. RECENT FINDINGS: Intratumoral immunotherapy is feasible and safe in a wide range of cancer histologies and locations, including lung and liver. Studies mainly focused on multi-metastatic patients, with some positive trials such as T-VEC in melanoma, but evidence of clinical benefit is still lacking. Recent results showed improved outcomes in patients with a low tumor burden. Intratumoral immunotherapy can lower systemic toxicities and boost local and systemic immune responses. Several studies have proven the feasibility, repeatability, and safety of this approach, with some promising results in clinical trials. The clinical benefit might be improved in patients with a low tumor burden. Future clinical trials should focus on adequate timing of treatment delivery during the course of the disease, particularly in the neoadjuvant setting.


Subject(s)
Melanoma , Humans , Melanoma/pathology , Neoadjuvant Therapy , Immunotherapy/methods , Immunity
4.
Cancer Immunol Immunother ; 70(1): 221-232, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32700090

ABSTRACT

BACKGROUND: New patterns of progression under immune-oncology (IO) antibodies (mAb) have been described such as pseudoprogression. Except for melanoma, variations between studies reveal difficulties to establish their prevalence. METHODS: This retrospective study enrolled patients participating in IO phase I trials at Gustave Roussy cancer center for solid tumors excluding melanoma. Radiological assessment according to iRECIST was correlated with prospectively registered patient characteristics and outcomes. Pseudoprogression (PsPD) was defined as RECIST-defined progression followed by stabilization or decrease at the next imaging, and dissociated response (DisR) as concomitant decrease in some tumor lesions and increase in others at a same timepoint. RESULTS: Among 360 patients included, 74% received IO mAb combination: 45% with another IO mAb, 20% with targeted therapy and 10% with radiotherapy. The overall response rate was 19.7%. PsPD were observed in 10 (2.8%) patients and DisR in 12 (3.3%) patients. Atypical responses (AR), including PsPD and DisR, were not associated with any patient's baseline characteristics. Compare with typical responder patients, patients experiencing AR presented a shorter iPFS (HR 0.34; p < 0.001) and OS (HR 0.27; p = 0.026). Among the 203 patients who progressed in 12 weeks, 80 (39.4%) patients were treated beyond progression. PD was confirmed in 80% of cases, while 10% of patients presented a response. CONCLUSION: Pseudoprogression and dissociated response are uncommon patterns of progression. Their prevalence should be balanced with the rate of real progressing patients treated beyond progression. Prognosis or on-treatment biomarkers are needed to identify early patients who will benefit from immunotherapy.


Subject(s)
Antibodies/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Melanoma/drug therapy , Melanoma/immunology , Adult , Aged , Aged, 80 and over , Disease Progression , Female , Humans , Immunotherapy/methods , Male , Medical Oncology/methods , Middle Aged , Prognosis , Response Evaluation Criteria in Solid Tumors , Retrospective Studies
5.
Ann Rheum Dis ; 80(1): 36-48, 2021 01.
Article in English | MEDLINE | ID: mdl-32327425

ABSTRACT

BACKGROUND: Rheumatic and musculoskeletal immune-related adverse events (irAEs) are observed in about 10% of patients with cancer receiving checkpoint inhibitors (CPIs). Given the recent emergence of these events and the lack of guidance for rheumatologists addressing them, a European League Against Rheumatism task force was convened to harmonise expert opinion regarding their identification and management. METHODS: First, the group formulated research questions for a systematic literature review. Then, based on literature and using a consensus procedure, 4 overarching principles and 10 points to consider were developed. RESULTS: The overarching principles defined the role of rheumatologists in the management of irAEs, highlighting the shared decision-making process between patients, oncologists and rheumatologists. The points to consider inform rheumatologists on the wide spectrum of musculoskeletal irAEs, not fulfilling usual classification criteria of rheumatic diseases, and their differential diagnoses. Early referral and facilitated access to rheumatologist are recommended, to document the target organ inflammation. Regarding therapeutic, three treatment escalations were defined: (1) local/systemic glucocorticoids if symptoms are not controlled by symptomatic treatment, then tapered to the lowest efficient dose, (2) conventional synthetic disease-modifying antirheumatic drugs, in case of inadequate response to glucocorticoids or for steroid sparing and (3) biological disease-modifying antirheumatic drugs, for severe or refractory irAEs. A warning has been made on severe myositis, a life-threatening situation, requiring high dose of glucocorticoids and close monitoring. For patients with pre-existing rheumatic disease, baseline immunosuppressive regimen should be kept at the lowest efficient dose before starting immunotherapies. CONCLUSION: These statements provide guidance on diagnosis and management of rheumatic irAEs and aim to support future international collaborations.


Subject(s)
Antirheumatic Agents/therapeutic use , Glucocorticoids/therapeutic use , Immune Checkpoint Inhibitors/adverse effects , Neoplasms/drug therapy , Rheumatic Diseases/therapy , Advisory Committees , Analgesics/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arthralgia/chemically induced , Arthralgia/diagnosis , Arthralgia/immunology , Arthralgia/therapy , Arthritis, Psoriatic/chemically induced , Arthritis, Psoriatic/diagnosis , Arthritis, Psoriatic/immunology , Arthritis, Psoriatic/therapy , Arthritis, Reactive/chemically induced , Arthritis, Reactive/diagnosis , Arthritis, Reactive/immunology , Arthritis, Reactive/therapy , Autoantibodies/immunology , Decision Making, Shared , Deprescriptions , Europe , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunologic Factors/therapeutic use , Medical Oncology , Methotrexate/therapeutic use , Myalgia/chemically induced , Myalgia/diagnosis , Myalgia/immunology , Myalgia/therapy , Myocarditis/chemically induced , Myocarditis/diagnosis , Myocarditis/immunology , Myocarditis/therapy , Myositis/chemically induced , Myositis/diagnosis , Myositis/immunology , Myositis/therapy , Plasma Exchange , Polymyalgia Rheumatica/chemically induced , Polymyalgia Rheumatica/diagnosis , Polymyalgia Rheumatica/immunology , Polymyalgia Rheumatica/therapy , Rheumatic Diseases/chemically induced , Rheumatic Diseases/diagnosis , Rheumatic Diseases/immunology , Rheumatology , Severity of Illness Index , Societies, Medical , Tumor Necrosis Factor Inhibitors/therapeutic use
6.
Lancet Oncol ; 21(10): 1353-1365, 2020 10.
Article in English | MEDLINE | ID: mdl-32919526

ABSTRACT

BACKGROUND: Tumour mutational burden (TMB) has been retrospectively correlated with response to immune checkpoint blockade. We prospectively explored the association of high tissue TMB (tTMB-high) with outcomes in ten tumour-type-specific cohorts from the phase 2 KEYNOTE-158 study, which assessed the anti-PD-1 monoclonal antibody pembrolizumab in patients with selected, previously treated, advanced solid tumours. METHODS: In the multi-cohort, open-label, non-randomised, phase 2 KEYNOTE-158 study, patients were enrolled from 81 academic facilities and community-based institutions across 21 countries in Africa, the Americas, Asia, and Europe. Eligible patients were aged 18 years or older, had a histologically or cytologically confirmed advanced (ie, unresectable or metastatic, or both) incurable solid tumour (eligible tumour types were anal, biliary, cervical, endometrial, mesothelioma, neuroendocrine, salivary, small-cell lung, thyroid, and vulvar), progression on or intolerance to one or more lines of standard therapy, had measurable disease per Response Evaluation Criteria in Solid Tumors (RECIST; version 1.1) assessed by independent central radiological review, Eastern Cooperative Oncology Group performance status of 0 or 1, life expectancy of at least 3 months, adequate organ function, and a tumour sample for biomarker analysis. Participants were given pembrolizumab 200 mg intravenously every 3 weeks for up to 35 cycles. Tissue TMB (tTMB) was assessed in formalin-fixed paraffin-embedded tumour samples using the FoundationOne CDx assay (Foundation Medicine, Cambridge, MA, USA). The prespecified definition of tTMB-high status was at least 10 mutations per megabase. The primary endpoint was the proportion of patients with an objective response (complete or partial response) as per Response Evaluation Criteria in Solid Tumours (version 1.1) by independent central review. This prespecified analysis assessed the association between antitumour activity and tTMB in treated patients with evaluable tTMB data. Efficacy was assessed in all participants who received at least one dose of pembrolizumab, had evaluable tTMB data, and were enrolled at least 26 weeks before data cutoff (June 27, 2019), and safety was assessed in all participants who received at least one dose of pembrolizumab and had tTMB-high status. KEYNOTE-158 is registered at ClinicalTrials.gov, NCT02628067, and is ongoing. FINDINGS: Between Jan 15, 2016, and June 25, 2019, 1073 patients were enrolled. 1066 participants were treated as of data cutoff (June 27, 2019), of whom 805 (76%) were evaluable for TMB, and 105 (13%) of 805 had tTMB-high status and were assessed for safety. 1050 (98%) of 1066 patients enrolled by at least 26 weeks before data cutoff, of whom 790 (75%) were evaluable for TMB and included in efficacy analyses. 102 (13%) of these 790 patients had tTMB-high status (≥10 mutations per megabase), and 688 (87%) patients had non-tTMB-high status (<10 mutations per megabase). Median study follow-up was 37·1 months (IQR 35·0-38·3). Objective responses were observed in 30 (29%; 95% CI 21-39) of 102 patients in the tTMB-high group and 43 (6%; 5-8) of 688 in the non-tTMB-high group. 11 (10%) of 105 patients had treatment-related serious adverse events. 16 (15%) participants had a grade 3-5 treatment-related adverse event, of which colitis was the only such adverse event that occurred in more than one patient (n=2). One patient had fatal pneumonia that was assessed by the investigator to be treatment related. INTERPRETATION: tTMB-high status identifies a subgroup of patients who could have a robust tumour response to pembrolizumab monotherapy. tTMB could be a novel and useful predictive biomarker for response to pembrolizumab monotherapy in patients with previously treated recurrent or metastatic advanced solid tumours. FUNDING: Merck Sharp & Dohme Corp, a subsidiary of Merck & Co, Inc.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Biomarkers, Tumor/genetics , Neoplasms/therapy , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Biomarkers, Tumor/metabolism , Drug-Related Side Effects and Adverse Reactions , Female , Humans , Immunotherapy , Male , Middle Aged , Mutation , Neoplasms/genetics , Neoplasms/pathology , Prospective Studies , Response Evaluation Criteria in Solid Tumors , Survival Analysis
7.
Oncologist ; 25(5): 369-374, 2020 05.
Article in English | MEDLINE | ID: mdl-32091646

ABSTRACT

A breakthrough in oncology over the last 5 years, immunotherapy has proved its salutary effects in a wide range of solid tumors. The targeting of the programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) pathway can restore a competent antitumor T-cell response by addressing key tumor immune evasion mechanisms. This novel mechanism of action is associated with new patterns of responses that were not observed with conventional treatments such as chemotherapy or targeted therapies. Thus, hyperprogressive disease (HPD), an unexpected acceleration of cancer evolution after starting immunotherapy, has been reported by several groups with a PD-1/PD-L1 blockade. This tumor flare-up phenomenon is associated with a poorer outcome and is suspected to be an immune-related adverse event. Despite been highly debated, the issue of HPD is currently a real challenge for oncologists' practice in terms of patients' information, diagnosis, and management. Herein, we describe the case of a 57-year-old man diagnosed with metastatic urothelial carcinoma who developed a rapid tumor growth after an anti-PD-L1+ IO combination. This case illustrates how current practice should evolve to address the HPD reality in the anticheckpoint era. KEY POINTS: Hyperprogressive disease (HPD) is an unexpected acceleration of cancer growth after starting immunotherapy that is associated with a poor outcome. Definition of HPD is based on comparing kinetics of tumor growth before and after starting immunotherapy. No predictive biomarker has been homogenously identified in the reported studies. Suspected pathophysiology includes expansion of programmed cell death protein 1 (PD-1) + regulatory T cells, exhaustion of compensatory T cells, modulation of pro-tumorigenic immune cell subsets, activation of aberrant inflammation, or activation of oncogenic signaling. HPD is one of the most controversial immune-related adverse events, as the liability of immunotherapy in this tumor deleterious flare-up phenomenon has not been proved yet. The reported incidence of HPD in retrospective studies varies across different solid tumor types from 6% to 29%. This phenomenon has been mainly suspected in non-small cell lung cancer (NSCLC), head and neck squamous cell carcinoma (HNSCC), and in urothelial carcinomas, where several randomized phase III trials have shown early crossing over of survival curves. In the context of anti-PD-1/programmed death-ligand 1 therapy, in particular for NSCLC, HNSCC, or urothelial carcinoma, the authors recommend performing an early computed tomography (CT) assessment at week 3-4. In the case of an early progression, tumor molecular characterization by tumor biopsy or circulating tumor DNA could be urged. Immunotherapy discontinuation should be discussed. Performing a confirmatory CT scan 4 weeks later to exclude pseudoprogression should not be the rule. Early switch to cytotoxic therapy may counteract the deleterious flare-up. Patients should be informed of the risk of developing HPD. Health authorities and trial sponsors could monitor and report the rates of tumor flares in trials in order to help oncologists to properly inform their patients about the expected rates of HPD.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , B7-H1 Antigen , Humans , Immunotherapy , Male , Middle Aged , Programmed Cell Death 1 Receptor , Retrospective Studies
8.
Eur J Nucl Med Mol Imaging ; 47(10): 2301-2312, 2020 09.
Article in English | MEDLINE | ID: mdl-32206839

ABSTRACT

PURPOSE: To compare the prognostic value of imaging biomarkers derived from a quantitative analysis of baseline 18F-FDG-PET/CT in patients with mucosal melanoma (Muc-M) or cutaneous melanoma (Cut-M) treated with immune checkpoint inhibitors (ICIs). METHODS: In this retrospective monocentric study, we included 56 patients with non-resectable Muc-M (n = 24) or Cut-M (n = 32) who underwent baseline 18F-FDG-PET/CT before treatment with ICIs between 2011 and 2017. Parameters were extracted from (i) tumoral tissues: SUVmax, SUVmean, TMTV (total metabolic tumor volume), and TLG (total lesion glycolysis) and (ii) lymphoid tissues: BLR (bone marrow-to-liver SUVmax ratio) and SLR (spleen-to-liver SUVmax ratio). Association with survival and response was evaluated using Cox prediction models, Student's t tests, and Spearman's correlation respectively. p < 0.05 was considered significant. RESULTS: Majority of ICIs were anti-PD1 (92.9%, n = 52/56). All 18F-FDG-PET/CT were positive. Overall (Muc-M to Cut-M), ORR was 33%:42%, DCR was 56%:69%, median follow-up was 25.0:28.9 months, median PFS was 4.7:10.7 months, and median OS was 23.9:28.3 months. In Muc-M, increased tumor SUVmax was associated with shorter OS while it was not correlated with PFS, ORR, or DCR. In Cut-M, increased TMTV and increased BLR were independently associated with shorter OS, shorter PFS, and lower response (ORR, DCR). CONCLUSION: While all Muc-M and Cut-M were FDG avid, prognostic imaging biomarkers differed. For Muc-M patients treated with ICI, the only prognostic imaging biomarker was a high baseline maximal glycolytic activity (SUVmax), whereas for Cut-M patients, baseline metabolic tumor burden or bone marrow metabolism was negatively correlated to ICI response duration.


Subject(s)
Melanoma , Skin Neoplasms , CTLA-4 Antigen , Fluorodeoxyglucose F18 , Humans , Immune Checkpoint Inhibitors , Melanoma/diagnostic imaging , Melanoma/drug therapy , Positron Emission Tomography Computed Tomography , Prognosis , Programmed Cell Death 1 Receptor , Retrospective Studies , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/drug therapy , Tumor Burden
9.
Br J Cancer ; 120(1): 3-5, 2019 01.
Article in English | MEDLINE | ID: mdl-30413824

ABSTRACT

Clinical trials have now identified over 30 cancer histotypes with sensitivity to anti-PD-(L)1 therapies. It is the first time in oncology that a class of drugs has demonstrated such a wide spectrum of activity in monotherapy. This subgroup of cancers ('PD-Lomas') is driving the clinical research strategies for the next generation of combination immunotherapy.


Subject(s)
B7-H1 Antigen/immunology , Immunotherapy , Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Clinical Trials as Topic , Humans , Neoplasms/classification , Neoplasms/genetics , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Signal Transduction
10.
Eur J Nucl Med Mol Imaging ; 46(11): 2298-2310, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31346755

ABSTRACT

PURPOSE: An imaging-based stratification tool is needed to identify melanoma patients who will benefit from anti Programmed Death-1 antibody (anti-PD1). We aimed at identifying biomarkers for survival and response evaluated in lymphoid tissue metabolism in spleen and bone marrow before initiation of therapy. METHODS: This retrospective study included 55 patients from two institutions who underwent 18F-FDG PET/CT before anti-PD1. Parameters extracted were SUVmax, SUVmean, HISUV (SUV-based Heterogeneity Index), TMTV (total metabolic tumor volume), TLG (total lesion glycolysis), BLR (Bone marrow-to-Liver SUVmax ratio), and SLR (Spleen-to-Liver SUVmax ratio). Each parameter was dichotomized using the median as a threshold. Association with survival, best overall response (BOR), and transcriptomic analyses (NanoString assay) were evaluated using Cox prediction models, Wilcoxon tests, and Spearman's correlation, respectively. RESULTS: At 20.7 months median follow-up, 33 patients had responded, and 29 patients died. Median PFS and OS were 11.4 (95%CI 2.7-20.2) and 28.5 (95%CI 13.4-43.8) months. TMTV (>25cm3), SLR (>0.77), and BLR (>0.79) correlated with shorter survival. High TMTV (>25 cm3), SLR (>0.77), and BLR (>0.79) correlated with shorter survival, with TMTV (HR PFS 2.2, p = 0.02, and HR OS 2.5, p = 0.02) and BLR (HR OS 2.3, p = 0.04) remaining significant in a multivariable analysis. Low TMTV and TLG correlated with BOR (p = 0.03). Increased glucose metabolism in bone marrow (BLR) was associated with transcriptomic profiles including regulatory T cell markers (p < 0.05). CONCLUSION: Low tumor burden correlates with survival and objective response while hematopoietic tissue metabolism correlates inversely with survival. These biomarkers should be further evaluated for potential clinical application.


Subject(s)
Biomarkers, Tumor/metabolism , Immunotherapy , Melanoma/immunology , Melanoma/therapy , Positron-Emission Tomography , Programmed Cell Death 1 Receptor/immunology , Adult , Aged , Aged, 80 and over , Biopsy , Fluorodeoxyglucose F18 , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Middle Aged , Neoplasm Metastasis , Prognosis , Retrospective Studies , Transcriptome , Treatment Outcome , Young Adult
11.
Lancet Oncol ; 19(9): 1180-1191, 2018 09.
Article in English | MEDLINE | ID: mdl-30120041

ABSTRACT

BACKGROUND: Because responses of patients with cancer to immunotherapy can vary in success, innovative predictors of response to treatment are urgently needed to improve treatment outcomes. We aimed to develop and independently validate a radiomics-based biomarker of tumour-infiltrating CD8 cells in patients included in phase 1 trials of anti-programmed cell death protein (PD)-1 or anti-programmed cell death ligand 1 (PD-L1) monotherapy. We also aimed to evaluate the association between the biomarker, and tumour immune phenotype and clinical outcomes of these patients. METHODS: In this retrospective multicohort study, we used four independent cohorts of patients with advanced solid tumours to develop and validate a radiomic signature predictive of immunotherapy response by combining contrast-enhanced CT images and RNA-seq genomic data from tumour biopsies to assess CD8 cell tumour infiltration. To develop the radiomic signature of CD8 cells, we used the CT images and RNA sequencing data of 135 patients with advanced solid malignant tumours who had been enrolled into the MOSCATO trial between May 1, 2012, and March 31, 2016, in France (training set). The genomic data, which are based on the CD8B gene, were used to estimate the abundance of CD8 cells in the samples and data were then aligned with the images to generate the radiomic signatures. The concordance of the radiomic signature (primary endpoint) was validated in a Cancer Genome Atlas [TGCA] database dataset including 119 patients who had available baseline preoperative imaging data and corresponding transcriptomic data on June 30, 2017. From 84 input variables used for the machine-learning method (78 radiomic features, five location variables, and one technical variable), a radiomics-based predictor of the CD8 cell expression signature was built by use of machine learning (elastic-net regularised regression method). Two other independent cohorts of patients with advanced solid tumours were used to evaluate this predictor. The immune phenotype internal cohort (n=100), were randomly selected from the Gustave Roussy Cancer Campus database of patient medical records based on previously described, extreme tumour-immune phenotypes: immune-inflamed (with dense CD8 cell infiltration) or immune-desert (with low CD8 cell infiltration), irrespective of treatment delivered; these data were used to analyse the correlation of the immune phenotype with this biomarker. Finally, the immunotherapy-treated dataset (n=137) of patients recruited from Dec 1, 2011, to Jan 31, 2014, at the Gustave Roussy Cancer Campus, who had been treated with anti-PD-1 and anti-PD-L1 monotherapy in phase 1 trials, was used to assess the predictive value of this biomarker in terms of clinical outcome. FINDINGS: We developed a radiomic signature for CD8 cells that included eight variables, which was validated with the gene expression signature of CD8 cells in the TCGA dataset (area under the curve [AUC]=0·67; 95% CI 0·57-0·77; p=0·0019). In the cohort with assumed immune phenotypes, the signature was also able to discriminate inflamed tumours from immune-desert tumours (0·76; 0·66-0·86; p<0·0001). In patients treated with anti-PD-1 and PD-L1, a high baseline radiomic score (relative to the median) was associated with a higher proportion of patients who achieved an objective response at 3 months (vs those with progressive disease or stable disease; p=0·049) and a higher proportion of patients who had an objective response (vs those with progressive disease or stable disease; p=0·025) or stable disease (vs those with progressive disease; p=0·013) at 6 months. A high baseline radiomic score was also associated with improved overall survival in univariate (median overall survival 24·3 months in the high radiomic score group, 95% CI 18·63-42·1; vs 11·5 months in the low radiomic score group, 7·98-15·6; hazard ratio 0·58, 95% CI 0·39-0·87; p=0·0081) and multivariate analyses (0·52, 0·35-0·79; p=0·0022). INTERPRETATION: The radiomic signature of CD8 cells was validated in three independent cohorts. This imaging predictor provided a promising way to predict the immune phenotype of tumours and to infer clinical outcomes for patients with cancer who had been treated with anti-PD-1 and PD-L1. Our imaging biomarker could be useful in estimating CD8 cell count and predicting clinical outcomes of patients treated with immunotherapy, when validated by further prospective randomised trials. FUNDING: Fondation pour la Recherche Médicale, and SIRIC-SOCRATE 2.0, French Society of Radiation Oncology.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Molecular Imaging/methods , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tomography, X-Ray Computed , Adult , Aged , Antineoplastic Agents, Immunological/adverse effects , B7-H1 Antigen/immunology , Biomarkers, Tumor/genetics , CD8-Positive T-Lymphocytes/immunology , Female , Gene Expression Profiling , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Neoplasms/genetics , Neoplasms/immunology , Phenotype , Predictive Value of Tests , Programmed Cell Death 1 Receptor/immunology , RNA, Neoplasm/genetics , Reproducibility of Results , Retrospective Studies , Sequence Analysis, RNA , Time Factors , Transcriptome , Treatment Outcome
12.
J Hepatol ; 68(6): 1181-1190, 2018 06.
Article in English | MEDLINE | ID: mdl-29427729

ABSTRACT

BACKGROUND & AIMS: Immunotherapy for metastatic cancer can be complicated by the onset of hepatic immune-related adverse events (IRAEs). This study compared hepatic IRAEs associated with anti-programmed cell death protein 1 (PD-1)/PD ligand 1 (PD-L1) and anti-cytotoxic T lymphocyte antigen 4 (CTLA-4) monoclonal antibodies (mAbs). METHODS: Among 536 patients treated with anti-PD-1/PD-L1 or CTLA-4 immunotherapies, 19 (3.5%) were referred to the liver unit for grade ≥3 hepatitis. Of these patients, nine had received anti-PD-1/PD-L1 and seven had received anti-CTLA-4 mAbs, in monotherapy or in combination with anti-PD-1. Liver investigations were undertaken in these 16 patients, including viral assays, autoimmune tests and liver biopsy, histological review, and immunostaining of liver specimens. RESULTS: In the 16 patients included in this study, median age was 63 (range 33-84) years, and nine (56%) were female. Time between therapy initiation and hepatitis was five (range, 1-49) weeks and median number of immunotherapy injections was two (range, 1-36). No patients developed hepatic failure. Histology related to anti-CTLA-4 mAbs demonstrated granulomatous hepatitis including fibrin ring granulomas and central vein endotheliitis. Histology related to anti-PD-1/PD-L1 mAbs was characterised by lobular hepatitis. The management of hepatic IRAEs was tailored according to the severity of both the biology and histology of liver injury: six patients improved spontaneously; seven received oral corticosteroids at 0.5-1 mg/kg/day; two were maintained on 0.2 mg/kg/day corticosteroids; and one patient required pulses and 2.5 mg/kg/day of corticosteroids, and the addition of a second immunosuppressive drug. In three patients, immunotherapy was reintroduced without recurrence of liver dysfunction. CONCLUSIONS: Acute hepatitis resulting from immunotherapy for metastatic cancer is rare (3.5%) and, in most cases, not severe. Histological assessment can distinguish between anti-PD-1/PD-L1 and anti-CTLA-4 mAb toxicity. The severity of liver injury is helpful for tailoring patient management, which does not require systematic corticosteroid administration. LAY SUMMARY: Immunotherapy for metastatic cancer can be complicated by immune-related adverse events in the liver. In patients receiving immunotherapy for metastatic cancer who develop immune-mediated hepatitis, liver biopsy is helpful for the diagnosis and evaluation of the severity of liver injury. This study demonstrates the need for patient-oriented management, which could eventually avoid unnecessary systemic corticosteroid treatment.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , B7-H1 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Immunotherapy/adverse effects , Liver/injuries , Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , B7-H1 Antigen/immunology , CTLA-4 Antigen/immunology , Female , Hepatitis/etiology , Hepatitis/immunology , Hepatitis/pathology , Humans , Ipilimumab/adverse effects , Liver/immunology , Liver/pathology , Male , Middle Aged , Neoplasms/immunology , Nivolumab/adverse effects
13.
Ann Rheum Dis ; 76(10): 1747-1750, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28600350

ABSTRACT

OBJECTIVES: Immune checkpoint inhibitors (ICIs) targeting cytotoxic T-lymphocyte-associated protein 4 and programmed cell death protein 1 (PD-1) have demonstrated improved survival for multiple cancers. However, these new drug classes have led to increased immune-related adverse events (IrAE). Rheumatic IrAEs have not been well described in clinical trials. We report here cases of rheumatoid arthritis (RA) and polymyalgia rheumatica (PMR) occurring after ICI treatment. METHODS: This was a retrospective study of patients receiving an ICI in whom symptoms of arthritis or arthralgia developed and revealed a diagnosis of RA or PMR. RESULTS: In 10 patients who received ICI therapy (all anti-PD-1 or anti-PDL1 antibodies), RA or PMR developed at a median of 1 month (1 to 9) after exposure. No patient had pre-existing rheumatic or autoimmune disease. RA developed in six patients; all six were positive for anti-cyclic citrullinated peptide (anti-CCP) antibodies and four for rheumatoid factor. Anti-CCP antibodies were detected in two out of three patients tested before immunotherapy. Disease-modifying antirheumatic drugs were needed for three patients; the three others received corticosteroids or non-steroid anti-inflammatory drugs. PMR was diagnosed in four patients, all responded to corticosteroids. Despite these IrAEs, immunotherapy was pursued for all but one patient until cancer progression. CONCLUSIONS: This is the first description of RA occurring after ICI therapy for cancer. PMR can also occur after ICI, particularly after anti-PD-1 therapy. All cases responded to corticosteroids or with immunosuppressive therapy. Collaboration between rheumatologists and oncologists is crucial and could lead to better recognition and care of these patients.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , Arthritis, Rheumatoid/chemically induced , Polymyalgia Rheumatica/chemically induced , Aged , Aged, 80 and over , Antibodies/blood , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/drug therapy , B7-H1 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Female , Humans , Ipilimumab , Male , Middle Aged , Nivolumab , Peptides, Cyclic/immunology , Polymyalgia Rheumatica/blood , Polymyalgia Rheumatica/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Retrospective Studies
14.
Blood ; 123(5): 678-86, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24326534

ABSTRACT

Natural killer (NK) cells mediate antilymphoma activity by spontaneous cytotoxicity and antibody-dependent cell-mediated cytotoxicity (ADCC) when triggered by rituximab, an anti-CD20 monoclonal antibody (mAb) used to treat patients with B-cell lymphomas. The balance of inhibitory and activating signals determines the magnitude of the efficacy of NK cells by spontaneous cytotoxicity. Here, using a killer-cell immunoglobulin-like receptor (KIR) transgenic murine model, we show that blockade of the interface of inhibitory KIRs with major histocompatibility complex (MHC) class I antigens on lymphoma cells by anti-KIR antibodies prevents a tolerogenic interaction and augments NK-cell spontaneous cytotoxicity. In combination with anti-CD20 mAbs, anti-KIR treatment induces enhanced NK-cell-mediated, rituximab-dependent cytotoxicity against lymphoma in vitro and in vivo in KIR transgenic and syngeneic murine lymphoma models. These results support a therapeutic strategy of combination rituximab and KIR blockade through lirilumab, illustrating the potential efficacy of combining a tumor-targeting therapy with an NK-cell agonist, thus stimulating the postrituximab antilymphoma immune response.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antigens, CD20/immunology , Killer Cells, Natural/immunology , Lymphoma/therapy , Receptors, KIR/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Antibody-Dependent Cell Cytotoxicity , Cell Line , Female , Histocompatibility Antigens Class I/immunology , Humans , Lymphoma/immunology , Male , Mice , Rituximab
15.
Ann Pathol ; 36(1): 94-102, 2016 Jan.
Article in French | MEDLINE | ID: mdl-26778219

ABSTRACT

Therapies targeting immune checkpoints, in particular programmed death 1 (PD-1) and its ligand programmed death ligand 1 (PD-L1), are major new strategies for the treatment of several malignancies including mestatatic non-small cell lung cancer (NSCLC). The identification of predictive biomarkers of response is required, considering efficacy, cost and potential adverse events. Expression of PD-L1 by immunohistochemistry has been associated with higher response rate and overall survival in several clinical trials evaluating anti-PD-1 and anti-PD-L1 monoclonal antibodies. Thus, PD-L1 immunohistochemical companion assays could be required for treatment with some of these therapies in NSCLC. However, heterogeneity in methodologies of PD-L1 assays in terms of primary antibodies and scoring algorithms, and tumor heterogenity for PD-L1 expression are important issues to be considered. More studies are required to compare the different assays, ensure their harmonization and standardization and identify the optimal conditions for testing. PD-L1 expression is likely an imperfect predictive biomarker for patient selection and association with other markers of the tumor immune microenvironment will be probably necessary in the future.


Subject(s)
B7-H1 Antigen/analysis , Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/chemistry , Lung Neoplasms/chemistry , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Clone Cells , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Immunohistochemistry/methods , Immunotherapy , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Molecular Targeted Therapy
17.
Strahlenther Onkol ; 191(7): 604-12, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25896312

ABSTRACT

INTRODUCTION: Neuroblastoma (NB) is the most frequent indication for extracranial pediatric radiotherapy. As long-term survival of high-risk localized NB has greatly improved, we reviewed treatment-related late toxicities in pediatric patients who received postoperative radiotherapy (RT) for localized NB within two French prospective clinical trials: NB90 and NB94. PATIENTS AND METHODS: From 1990-2000, 610 children were enrolled. Among these, 35 were treated with induction chemotherapy, surgery, and RT. The recommended RT dose was 24 Gy at ≤ 2 years, 34 Gy at > 2 years, ± a 5 Gy boost in both age groups. RESULTS: The 22 patients still alive after 5 years were analyzed. The median follow-up time was 14 years (range 5-21 years). Late effects after therapy occurred in 73 % of patients (16/22), within the RT field for 50 % (11/22). The most frequent in-field effects were musculoskeletal abnormalities (n = 7) that occurred only with doses > 31 Gy/1.5 Gy fraction (p = 0.037). Other effects were endocrine in 3 patients and second malignancies in 2 patients. Four patients presented with multiple in-field late effects only with doses > 31 Gy. CONCLUSION: After a median follow-up of 14 years, late effects with multimodality treatment were frequent. The most frequent effects were musculoskeletal abnormalities and the threshold for their occurrence was 31 Gy.


Subject(s)
Neuroblastoma/radiotherapy , Radiation Injuries/etiology , Radiotherapy, Adjuvant , Adolescent , Child , Child, Preschool , Dose Fractionation, Radiation , Female , France , Gene Amplification , Humans , Infant , Male , N-Myc Proto-Oncogene Protein , Neoplasm, Residual/mortality , Neoplasm, Residual/radiotherapy , Neoplasms, Radiation-Induced/etiology , Neuroblastoma/genetics , Neuroblastoma/mortality , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Prospective Studies , Radiotherapy Dosage , Survival Analysis
18.
Mutagenesis ; 30(2): 205-11, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25688113

ABSTRACT

The clinical benefits obtained with rituximab in the treatment of CD20(+) B-cell malignancies and of imatinib in the treatment of Phi(+) leukaemias have opened a new era in oncology, transforming the concepts of tumour-targeted therapies and personalised medicine into reality. Since then, many tumour-targeted monoclonal antibodies and tyrosine kinase inhibitors have been approved for the treatment of cancers. Compared to conventional chemotherapies, these new drugs have more specificity against cancer cells and less systemic toxicities. However, like conventional chemotherapies, they often provide limited therapeutic benefits with short-lasting tumour responses as the vast majority of cancers become resistant to these drugs over time. Therefore, tumour-targeted therapies are an incremental innovation as compared to historical chemotherapies. Recently, a paradigm shift has been brought to the clinic with drugs targeting immune cells rather than cancer cells with the aim of stimulating the anti-tumour immune response of patients against their own cancer. Immunomodulatory drugs such as anti-CTLA4 and anti-PD-1 have generated long-lasting tumour responses when used as single agent in patients with refractory/relapsing cancers such as metastatic melanomas, renal cell carcinoma or non-small-cell lung carcinoma. These new immune-targeted therapies are therefore a disruptive innovation in cancer treatment: they demonstrate that long-lasting clinical benefits could be obtained by targeting molecules involved in the immune tolerance of cancer cells rather than by targeting oncogenic drivers or antigens expressed by cancer cells.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Immunologic Factors/therapeutic use , Neoplasms/drug therapy , Antibodies, Monoclonal/immunology , Antineoplastic Agents/immunology , Humans , Immune System/drug effects , Neoplasms/immunology
19.
Pediatr Blood Cancer ; 62(8): 1317-25, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25808079

ABSTRACT

Monoclonal antibodies (mAbs) have inaugurated the concepts of tumor-targeted therapy and personalized medicine. A new family of mAbs is currently emerging in the clinic, which target immune cells rather than cancer cells. These immune-targeted therapies have recently demonstrated long-term tumor responses in adults with refractory/relapsing metastatic solid tumors. Pediatric cancers are different from their adult counterparts in terms of histological features and immune infiltrates. However, the same immune checkpoint targets can be expressed within the microenvironment of pediatric tumors. The benefits of immune checkpoint blockade in pediatric cancers are currently under evaluation in early phase clinical trials.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Immunotherapy, Active/methods , Neoplasms/drug therapy , Adult , Child , Humans , Neoplasms/immunology , Tumor Escape/immunology
SELECTION OF CITATIONS
SEARCH DETAIL