Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Publication year range
1.
Int J Mol Sci ; 24(14)2023 Jul 24.
Article in English | MEDLINE | ID: mdl-37511609

ABSTRACT

Neoadjuvant immunotherapy has emerged as a promising approach in the treatment of various malignancies, with preclinical studies showing improved immune responses in the preoperative setting. FDA-approved neoadjuvant-immunotherapy-based approaches include triple-negative breast cancer and early non-small cell lung cancer on the basis of improvement in pathological response and event free survival. Nevertheless, current trials have only shown benefits in a fraction of patients. It is therefore crucial to identify predictive biomarkers to improve patient selection for such approaches. This review aims to provide an overview of potential biomarkers of neoadjuvant immunotherapy in early triple-negative breast cancer, bladder cancer, melanoma, non-small cell lung cancer, colorectal cancer and gastric cancer. By the extrapolation of the metastatic setting, we explore known predictive biomarkers, i.e., PD-L1, mismatch repair deficiency and tumour mutational burden, as well as potential early-disease-specific biomarkers. We also discuss the challenges of identifying reliable biomarkers and the need for standardized protocols and guidelines for their validation and clinical implementation.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Triple Negative Breast Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Neoadjuvant Therapy/methods , Standard of Care , Biomarkers, Tumor , Immunotherapy/methods , B7-H1 Antigen
2.
Nervenarzt ; 94(12): 1123-1128, 2023 Dec.
Article in German | MEDLINE | ID: mdl-37594495

ABSTRACT

Magnetic resonance imaging (MRI) is of exceptional importance in the diagnostics and monitoring of multiple sclerosis (MS); however, a close interdisciplinary cooperation between neurologists in private practice, (neuro)radiological practices, hospitals or specialized MS centers is only rarely established. In particular, there is a lack of standardized MRI protocols for image acquisition as well as established quality parameters, which guarantee the comparability of MRI records; however, this is a fundamental prerequisite for an effective application of MRI in the treatment of MS patients, e.g., for making the diagnosis or treatment monitoring. To address these challenges a group of neurologists and (neuro)radiologists developed a consensus proposal for standardization of image acquisition, interpretation and transmission of results and for improvement in interdisciplinary cooperation. This pilot project in the metropolitan area of Essen used a modified Delphi process and was based on the most up to date scientific knowledge. The recommendation takes the medical, economic, temporal and practical aspects of MRI in MS into consideration. The model of interdisciplinary cooperation between radiologists and neurologists with the aim of a regional standardization of MRI could serve as an example for other regions of Germany in order to optimize MRI for MS.


Subject(s)
Multiple Sclerosis , Humans , Multiple Sclerosis/diagnosis , Consensus , Pilot Projects , Magnetic Resonance Imaging/methods , Neurologists
3.
Rev Med Suisse ; 16(716): 2264-2270, 2020 Nov 25.
Article in French | MEDLINE | ID: mdl-33237644

ABSTRACT

Immune checkpoint inhibitors have radically changed oncology by significantly improving prognosis and survival of many patients, even at an advanced or metastatic stage. Some patients undergoing immunotherapy develop adverse immune-related events, presenting a toxicity spectrum that can affect any organ, separately or simultaneously, with different intensities depending on the treatment used and patient characteristics. We hereby suggest a diagnostic and therapeutic approach that any internist, general practitioner or emergency doctor should have facing digestive, cardiac and pulmonary toxicities.


Les inhibiteurs de points de contrôle immunitaire (IPCI) ont radicalement changé la prise en charge oncologique en améliorant significativement le pronostic ainsi que la survie de nombreux patients, même à un stade avancé ou métastatique. Une partie des patients traités peuvent développer des effets indésirables immunomédiés avec un spectre de toxicités pouvant atteindre tous les organes, de façon isolée ou simultanée, avec une sévérité et une chronologie variables en fonction du traitement utilisé et des caractéristiques de chaque patient. Nous proposons ici la conduite à tenir du médecin interniste, généraliste ou urgentiste devant les toxicités digestives, cardiaques et pulmonaires.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/diagnosis , Drug-Related Side Effects and Adverse Reactions/prevention & control , General Practitioners , Immunotherapy/adverse effects , Neoplasms/drug therapy , Neoplasms/immunology , Humans , Immunologic Factors , Neoplasms/pathology , Prognosis
4.
Rev Med Suisse ; 16(696): 1165-1168, 2020 Jun 03.
Article in French | MEDLINE | ID: mdl-32496706

ABSTRACT

Immune checkpoint inhibitors (ICI) have revolutionized the field of oncology, by reshaping the prognosis of many cancers and are progressively becoming the standard of care. One of the costs of these advances is the emergence of a new spectrum of immune-related adverse events (irAEs), of which cardiovascular irAEs are particularly feared. ICI-induced myocarditis is often a diagnostic challenge because of the vast heterogeneity of clinical presentations, and it is associated with a high mortality rate of around 50%. The present article summarizes the cardiac manifestations, the diagnostic strategy and the therapeutic management of patients with ICI-induced myocarditis used in the treatment of cancer.


Les inhibiteurs de points de contrôle immunitaire (ICI), ou immune checkpoint inhibitors (ICI), ont révolutionné la prise en charge de nombreux cancers en améliorant significativement la survie des patients et en devenant progressivement la norme de soins. Cette efficacité a néanmoins pour prix un taux élevé d'effets indésirables immunomédiés avec un large spectre d'organes touchés. Les toxicités cardiaques, dominées par la myocardite induite par les ICI, sont particulièrement redoutées du fait des difficultés diagnostiques et du risque d'évolution rapidement défavorable associée à une mortalité élevée, de l'ordre de 50 %. Le présent article s'intéresse aux manifestations cardiaques, à la stratégie diagnostique ainsi qu'à la prise en charge des patients présentant une myocardite induite par les ICI utilisés dans le traitement du cancer.


Subject(s)
Cardiotoxicity/etiology , Immunotherapy/adverse effects , Myocarditis/chemically induced , Neoplasms/drug therapy , Humans , Neoplasms/immunology , Neoplasms/pathology
5.
Lancet Oncol ; 20(1): e54-e64, 2019 01.
Article in English | MEDLINE | ID: mdl-30614479

ABSTRACT

Immune checkpoint inhibitors are reshaping the prognosis of many cancer and are progressively becoming the standard of care in the treatment of many tumour types. Immunotherapy is bringing new hope to patients, but also a whole new spectrum of toxicities for healthcare practitioners to manage. Oncologists and specialists involved in the pluridisciplinary management of patients with cancer are increasingly confronted with the therapeutic challenge of treating patients with severe and refractory immune-related adverse events. In this Personal View, we summarise the therapeutic strategies that have been used to manage such toxicities resulting from immune checkpoint inhibitor treatment. On the basis of current knowledge about their pathogenesis, we discuss the use of new biological and non-biological immunosuppressive drugs to treat severe and steroid refractory immune-related adverse events. Depending on the immune infiltrate type that is predominant, we propose a treatment algorithm for personalised management that goes beyond typical corticosteroid use. We propose a so-called shut-off strategy that aims at inhibiting key inflammatory components involved in the pathophysiological processes of immune-related adverse events, and limits potential adverse effects of drug immunosuppression on tumour response. This approach develops on current guidelines and challenges the step-by-step increase approach to drug immunosuppression.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , Drug-Related Side Effects and Adverse Reactions/drug therapy , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Immunotherapy/adverse effects , Algorithms , Drug-Related Side Effects and Adverse Reactions/immunology , Drug-Related Side Effects and Adverse Reactions/pathology , Drug-Related Side Effects and Adverse Reactions/physiopathology , Humans , Neoplasms/therapy , Precision Medicine
6.
Rev Med Suisse ; 15(N° 632-633): 17-19, 2019 Jan 09.
Article in French | MEDLINE | ID: mdl-30629360

ABSTRACT

The immune checkpoint inhibitors (ICI) have radically changed the treatment and the prognostic of certain types of cancer. This reprogramming of the immune system can sometimes lead to immuno-related adverse events (irAEs) involving, depending on the immunotherapy used, multiple organ (s) with different severity, frequency and chronology. In theory all organs can be affected, often in a reversible and rapid manner if an immunosuppressive therapy is initiated in time. In contrast, endocrinopathies and some neuropathies are of long lasting and sometimes of irreversible nature. The management of high-grade autoimmune toxicities requires, first, the suspension of treatment and second, the introduction of steroids as first-line or, in case of failure, of a second immunosuppressive agent.


Les inhibiteurs des points de contrôle immunitaire ont changé radicalement la prise en charge de certains types de cancer. Cette reprogrammation du système immunitaire peut entraîner parfois des événements indésirables (EI) immunomédiés impliquant en fonction de l'immunothérapie utilisée de multiple(s) organe(s) avec différentes sévérité, fréquence et chronologie. En théorie, tous les organes peuvent être touchés, souvent d'une manière réversible et rapide si le traitement immunosuppresseur est initié à temps. En revanche, les endocrinopathies et certaines neuropathies sont de nature durable et parfois irréversible. La gestion des toxicités autoimmunes de haut grade nécessite d'une part la suspension du traitement et d'autre part, l'introduction en première ligne d'une corticothérapie ou en cas d'échec d'un 2e immunosuppresseur.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Immunotherapy , Neoplasms , Humans , Immunologic Factors , Immunosuppressive Agents , Immunotherapy/adverse effects , Neoplasms/therapy
8.
Cancer Immunol Immunother ; 66(7): 833-840, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28409192

ABSTRACT

The abscopal effect, which is the spontaneous regression of tumors or metastases outside the radiation field, occurs rarely in cancer patients. Interestingly, radiotherapy (RT) triggers an immunogenic cell death (ICD) that is able to generate tumor-specific cytotoxic CD8+ T cells that are efficient in killing cancer cells. The key question is: why is this "abscopal effect" so uncommon in cancer patients treated with RT? Most probably, the main reason may be related to the highly immunosuppressive tumor microenvironment of well-established tumors that constantly antagonizes the anti-tumor immune responses triggered by RT. In this case, additional or combinatorial immunotherapy is needed to attenuate these immunosuppressive networks and, therefore, substantially increases the efficacy of RT. Here, we describe a potentially promising synergistic radio-immunotherapy "in situ tumor vaccination" protocol by antagonizing the tumor-immunosuppressive microenvironment with a combinatorial approach using local RT and IL-12-based TH1 response augmentation.


Subject(s)
Immune Tolerance , Immunosuppression Therapy/methods , Interleukin-12/therapeutic use , Neoplasms/therapy , Tumor Microenvironment , Animals , Combined Modality Therapy , Disease Models, Animal , Female , Humans , Immune Tolerance/radiation effects , Immunity, Cellular , Mice , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/radiotherapy , Th1 Cells/immunology , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
9.
Nat Med ; 13(1): 54-61, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17187072

ABSTRACT

Anthracyclin-treated tumor cells are particularly effective in eliciting an anticancer immune response, whereas other DNA-damaging agents such as etoposide and mitomycin C do not induce immunogenic cell death. Here we show that anthracyclins induce the rapid, preapoptotic translocation of calreticulin (CRT) to the cell surface. Blockade or knockdown of CRT suppressed the phagocytosis of anthracyclin-treated tumor cells by dendritic cells and abolished their immunogenicity in mice. The anthracyclin-induced CRT translocation was mimicked by inhibition of the protein phosphatase 1/GADD34 complex. Administration of recombinant CRT or inhibitors of protein phosphatase 1/GADD34 restored the immunogenicity of cell death elicited by etoposide and mitomycin C, and enhanced their antitumor effects in vivo. These data identify CRT as a key feature determining anticancer immune responses and delineate a possible strategy for immunogenic chemotherapy.


Subject(s)
Apoptosis/immunology , Calreticulin/immunology , Colonic Neoplasms/metabolism , Animals , Anthracyclines/pharmacology , Anthracyclines/therapeutic use , Antigens, Differentiation/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Calreticulin/genetics , Calreticulin/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/immunology , Cell Membrane/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Dendritic Cells/immunology , Electrophoresis, Gel, Two-Dimensional , Etoposide/pharmacology , Etoposide/therapeutic use , Immunoblotting , Mice , Mice, Inbred BALB C , Mice, Nude , Mitomycin/pharmacology , Mitomycin/therapeutic use , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Phagocytosis/immunology , Protein Phosphatase 1 , Protein Transport/drug effects , RNA Interference , Recombinant Proteins/pharmacology
10.
Nat Med ; 13(9): 1050-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17704786

ABSTRACT

Conventional cancer treatments rely on radiotherapy and chemotherapy. Such treatments supposedly mediate their effects via the direct elimination of tumor cells. Here we show that the success of some protocols for anticancer therapy depends on innate and adaptive antitumor immune responses. We describe in both mice and humans a previously unrecognized pathway for the activation of tumor antigen-specific T-cell immunity that involves secretion of the high-mobility-group box 1 (HMGB1) alarmin protein by dying tumor cells and the action of HMGB1 on Toll-like receptor 4 (TLR4) expressed by dendritic cells (DCs). During chemotherapy or radiotherapy, DCs require signaling through TLR4 and its adaptor MyD88 for efficient processing and cross-presentation of antigen from dying tumor cells. Patients with breast cancer who carry a TLR4 loss-of-function allele relapse more quickly after radiotherapy and chemotherapy than those carrying the normal TLR4 allele. These results delineate a clinically relevant immunoadjuvant pathway triggered by tumor cell death.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/radiotherapy , Toll-Like Receptor 4/immunology , Animals , Bone Neoplasms/drug therapy , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/radiotherapy , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Organoplatinum Compounds/therapeutic use , Osteosarcoma/drug therapy , Pyridines/therapeutic use
11.
J Neurol ; 271(4): 1947-1958, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38141128

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors (ICIs) are reshaping the prognosis of many cancers, but often cause immune-related adverse events (irAEs). Among neurological irAEs, myositis is the most frequently reported. Our aim is to describe clinical and non-clinical characteristics, treatment and outcome of all irMyositis (skeletal limb-girdle and/or ocular myositis) and irMyocarditis cases in our reference center. METHODS: We retrospectively enrolled all irMyositis/irMyocarditis patients seen between 2018 and 2022. We reviewed demographics, clinical characteristics, biological, neurophysiological, imaging workup, treatment and outcome. RESULTS: We included 14 consecutive patients. The most frequent treatments were pembrolizumab (35%) or ipilimumab-nivolumab combination (35%). Limb-girdle, ocular (non-fluctuating palpebral ptosis and/or diplopia with or without ophthalmoparesis) and cardiac phenotypes were equally distributed, overlapping in 40% of cases. Ocular involvement was frequently misdiagnosed; review of brain MRIs disclosed initially missed signs of skeletal myositis in one patient and ocular myositis in 3. Seven patients had other co-existing irAEs. When performed, myography showed a myogenic pattern. CK was elevated in 8/15 patients, troponin-T in 12/12 and troponin-I in 7/9 tested patients. ICI were discontinued in all cases, with further immunosuppressive treatment in nine patients. In most cases, neurological and cardiological outcome was good at last follow-up. CONCLUSION: Myositis is a potentially severe irAE. Despite its heterogeneous presentation, some highly suggestive clinical symptoms, such as ocular involvement, or radiological signs should raise physicians' attention to avoid misdiagnosis. We thus recommend a multidisciplinary assessment (including complete neuromuscular evaluation) even in case of isolated myocarditis. Our series underlines the importance of an early diagnosis, since suspension of ICI and adequate treatment are usually associated with good functional outcome.


Subject(s)
Antineoplastic Agents, Immunological , Myocarditis , Myositis , Humans , Immune Checkpoint Inhibitors , Myocarditis/chemically induced , Myocarditis/complications , Myocarditis/drug therapy , Antineoplastic Agents, Immunological/adverse effects , Retrospective Studies , Myositis/diagnosis
12.
Nat Commun ; 15(1): 3664, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38693123

ABSTRACT

The application of mammalian target of rapamycin inhibition (mTORi) as primary prophylactic therapy to optimize T cell effector function while preserving allograft tolerance remains challenging. Here, we present a comprehensive two-step therapeutic approach in a male patient with metastatic cutaneous squamous cell carcinoma and heart transplantation followed with concomitant longitudinal analysis of systemic immunologic changes. In the first step, calcineurin inhibitor/ mycophenolic acid is replaced by the mTORi everolimus to achieve an improved effector T cell status with increased cytotoxic activity (perforin, granzyme), enhanced proliferation (Ki67) and upregulated activation markers (CD38, CD69). In the second step, talimogene laherparepvec (T-VEC) injection further enhances effector function by switching CD4 and CD8 cells from central memory to effector memory profiles, enhancing Th1 responses, and boosting cytotoxic and proliferative activities. In addition, cytokine release (IL-6, IL-18, sCD25, CCL-2, CCL-4) is enhanced and the frequency of circulating regulatory T cells is increased. Notably, no histologic signs of allograft rejection are observed in consecutive end-myocardial biopsies. These findings provide valuable insights into the dynamics of T cell activation and differentiation and suggest that timely initiation of mTORi-based primary prophylaxis may provide a dual benefit of revitalizing T cell function while maintaining allograft tolerance.


Subject(s)
Carcinoma, Squamous Cell , Graft Rejection , Heart Transplantation , Herpesvirus 1, Human , MTOR Inhibitors , Heart Transplantation/adverse effects , Humans , Male , Graft Rejection/prevention & control , Graft Rejection/immunology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/drug therapy , MTOR Inhibitors/pharmacology , MTOR Inhibitors/therapeutic use , Biological Products/pharmacology , Biological Products/therapeutic use , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Skin Neoplasms/drug therapy , Middle Aged , Everolimus/pharmacology , Everolimus/therapeutic use , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors
13.
medRxiv ; 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38883792

ABSTRACT

Background: Immune-checkpoint inhibitors (ICI) are associated with life-threatening myocarditis but milder presentations are increasingly recognized. The same autoimmune process that causes ICI-myocarditis can manifest concurrent generalized myositis, myasthenia-like syndrome, and respiratory muscle failure. Prognostic factors for this "cardiomyotoxicity" are lacking. Methods: A multicenter registry collected data retrospectively from 17 countries between 2014-2023. A multivariable cox regression model (hazard-ratio(HR), [95%confidence-interval]) was used to determine risk factors for the primary composite outcome: severe arrhythmia, heart failure, respiratory muscle failure, and/or cardiomyotoxicity-related death. Covariates included demographics, comorbidities, cardio-muscular symptoms, diagnostics, and treatments. Time-dependent covariates were used and missing data were imputed. A point-based prognostic risk score was derived and externally validated. Results: In 748 patients (67% male, age 23-94), 30-days incidence of the primary composite outcome, cardiomyotoxic death, and overall death were 33%, 13%, and 17% respectively. By multivariable analysis, the primary composite outcome was associated with active thymoma (HR=3.60[1.93-6.72]), presence of cardio-muscular symptoms (HR=2.60 [1.58-4.28]), low QRS-voltage on presenting electrocardiogram (HR for ≤0.5mV versus >1mV=2.08[1.31-3.30]), left ventricular ejection fraction (LVEF) <50% (HR=1.78[1.22-2.60]), and incremental troponin elevation (HR=1.86 [1.44-2.39], 2.99[1.91-4.65], 4.80[2.54-9.08], for 20, 200 and 2000-fold above upper reference limit, respectively). A prognostic risk score developed using these parameters showed good performance; 30-days primary outcome incidence increased gradually from 3.9%(risk-score=0) to 81.3%(risk-score≥4). This risk-score was externally validated in two independent French and US cohorts. This risk score was used prospectively in the external French cohort to identify low risk patients who were managed with no immunosuppression resulting in no cardio-myotoxic events. Conclusions: ICI-myocarditis can manifest with high morbidity and mortality. Myocarditis severity is associated with magnitude of troponin, thymoma, low-QRS voltage, depressed LVEF, and cardio-muscular symptoms. A risk-score incorporating these features performed well. Trial registration number: NCT04294771 and NCT05454527.

14.
Front Immunol ; 14: 1213375, 2023.
Article in English | MEDLINE | ID: mdl-37622123

ABSTRACT

Therapeutic monoclonal antibodies (mAb) targeting the immune checkpoint inhibitor programmed cell death protein 1 (PD-1) have achieved considerable clinical success in anti-cancer therapy through relieving T cell exhaustion. Blockade of PD-1 interaction with its ligands PD-L1 and PD-L2 is an important determinant in promoting the functional recovery of exhausted T cells. Here, we show that anti-PD-1 mAbs act through an alternative mechanism leading to the downregulation of PD-1 surface expression on memory CD4+ and CD8+ T cells. PD-1 receptor downregulation is a distinct process from receptor endocytosis and occurs in a CD14+ monocyte dependent manner with the CD64/Fcγ receptor I acting as the primary factor for this T cell extrinsic process. Importantly, downregulation of surface PD-1 strongly enhances antigen-specific functional recovery of exhausted PD-1+CD8+ T cells. Our study demonstrates a novel mechanism for reducing cell surface levels of PD-1 and limiting the inhibitory targeting by PD-L1/2 and thereby enhancing the efficacy of anti-PD-1 Ab in restoring T cell functionality.


Subject(s)
CD8-Positive T-Lymphocytes , Receptors, IgG , B7-H1 Antigen , Cell Membrane , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use
15.
Cancer Treat Rev ; 120: 102614, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37603905

ABSTRACT

Immune-checkpoint inhibitors have revolutionized cancer therapy, yet many patients either do not derive any benefit from treatment or develop a resistance to checkpoint inhibitors. Intrinsic resistance can result from neoantigen depletion, defective antigen presentation, PD-L1 downregulation, immune-checkpoint ligand upregulation, immunosuppression, and tumor cell phenotypic changes. On the other hand, extrinsic resistance involves acquired upregulation of inhibitory immune-checkpoints, leading to T-cell exhaustion. Current data suggest that PD-1, CTLA-4, and LAG-3 upregulation limits the efficacy of single-agent immune-checkpoint inhibitors. Ongoing clinical trials are investigating novel immune-checkpoint targets to avoid or overcome resistance. This review provides an in-depth analysis of the evolving landscape of potentially targetable immune-checkpoints in cancer. We highlight their biology, emphasizing the current understanding of resistance mechanisms and focusing on promising strategies that are under investigation. We also summarize current results and ongoing clinical trials in this crucial field that could once again revolutionize outcomes for cancer patients.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Humans , Immune Checkpoint Inhibitors/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology
16.
J Immunother Cancer ; 11(6)2023 06.
Article in English | MEDLINE | ID: mdl-37380369

ABSTRACT

Cytokine storm induced by anti-human epidermal growth factor receptor-2 (HER2) therapies has not been reported. We report a patient with breast cancer treated with trastuzumab/pertuzumab who developed severe biventricular dysfunction and cardiogenic shock (CS) 6 months after starting double anti-HER2 therapy. The CS was accompanied by severe systemic inflammation, and cardiac MRI (cMRI) showed structural changes typical of myocardial inflammation. The immuno-inflammatory profile showed significantly increased levels of activation of the complement system, proinflammatory cytokines (IL-1ß, IL-6, IL-18, IL-17A, TNF-alpha) with increased activity of classical monocytic, T helper 17 cells (Th17), CD4 T and effector memory CD8 T subsets, whereas NK cell activation was not observed. The data suggest an important role for monocytes as initiators of this FcγR-dependent antibody-dependent cytotoxicity, leading to the overactivation of an adaptive T cell response, in which Th17 cells may act in synergy with T helper 1 cells (Th1) to drive the severe cytokine release syndrome. After discontinuation of trastuzumab/pertuzumab, hypercytokinemia and complement activity normalized along with clinical recovery. Cardiac function returned to baseline within 2 months of initial presentation, together with a resolution of the myocardial inflammation on MRI.


Subject(s)
Breast Neoplasms , Female , Humans , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/etiology , Cytokines , Shock, Cardiogenic/chemically induced
17.
J Cancer ; 14(10): 1913-1919, 2023.
Article in English | MEDLINE | ID: mdl-37476185

ABSTRACT

Background: Immune-mediated diarrhea and colitis (IMDC) frequently develop after treatment with immune checkpoint inhibitors. C-reactive protein (CRP) is a serum inflammatory biomarker used to stratify and monitor disease severity in many inflammatory conditions. However, CRP level is not specific and is widely influenced by various factors non-specific to bowel inflammation. We aimed to study the utility of CRP as a predictor of disease severity and therapy response in IMDC. Methods: We performed a retrospective analysis of patients diagnosed with IMDC who had CRP measured at IMDC onset and after treatment with selective immunosuppressive therapy (SIT: infliximab and vedolizumab), between 01/2016 and 02/2022 at MD Anderson Cancer Center. Patient demographics, clinical characteristics, and IMDC data were collected and analyzed. Results: Our sample of 128 patients had a median age of 67 years; most were white (89.8%); and male (65.6%). Prior to development of IMDC, 15 (11.7%) were initially treated with anti-CTLA-4, 42 (32.8%) with anti-PD-1 or PD-L1, and 71 (55.5%) with a combination of both. We found higher CRP level was associated with higher CTCAE grade of clinical symptoms such as diarrhea (p=0.015), colitis (p=0.013), and endoscopic findings (p=0.016). While CRP levels decreased after IMDC treatment, there was no significant association between CRP levels with clinical remission, endoscopic remission or histologic remission. There also was no significant correlation between CRP level and recurrence of IMDC, or with fecal calprotectin levels. Conclusion: CRP level may be useful to assess initial severity of IMDC, including grade of diarrhea and colitis and degree of endoscopic inflammation. However, CRP is not a robust surrogate biomarker for assessing treatment response or disease recurrence. Despite the reduction of CRP levels observed following IMDC treatment, this finding might be nonspecific and potentially confounded by concurrent clinical factors, such as underlying malignancy, other inflammatory processes, and systemic anti-cancer therapy. Further studies of the role of CRP are warranted in patients with cancer and IMDC.

18.
J Immunother Cancer ; 10(10)2022 10.
Article in English | MEDLINE | ID: mdl-36283734

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors (ICIs) have become a mainstay of cancer treatment. Their immune-boosting quality has one major drawback, their proclivity to induce a broad array of immune-related adverse events (irAEs) affecting, among others, the liver and sharing some similarities with classic autoimmune liver diseases (AILD).We aimed to compare clinical, laboratory and histological features of patients with liver-related irAEs and AILD. METHODS: We systematically compared liver irAEs with AILD, namely autoimmune hepatitis (AIH) and primary biliary cholangitis, regarding their clinical, laboratory, and histological features. RESULTS: Twenty-seven patients with liver irAEs (ICI group) and 14 patients with AILD were identified. We observed three distinct ICI-induced histological liver injury patterns: hepatitic (52%), cholangitic (19%), and mixed (29%). When comparing the ICI and AILD groups, centrilobular injury as well as granuloma formation were more prevalent in the former (p=0.067 and 0.002, respectively). CD4+/CD8+ T cell ratios were heterogeneous between the two groups, without statistically significant difference but with a trend toward increased CD8+ T cells among hepatitic irAEs as compared with AIH. Pattern of liver function test alteration was predictive for the type of irAEs but did not correlate with histological severity. CONCLUSIONS: Liver irAEs have broad clinical, laboratory and histological presentations. Histological features of irAEs and AILD are distinct, likely underpinning their different immunological mechanisms.


Subject(s)
Antineoplastic Agents, Immunological , Hepatitis, Autoimmune , Immune System Diseases , Liver Diseases , Neoplasms , Humans , Antineoplastic Agents, Immunological/adverse effects , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy/adverse effects , Immune System Diseases/chemically induced , Liver Diseases/etiology , Liver Diseases/drug therapy , Hepatitis, Autoimmune/etiology , Hepatitis, Autoimmune/drug therapy
19.
JAMA Oncol ; 8(5): e220446, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35271706

ABSTRACT

Importance: There are limited comparative data on the durability of neutralizing antibody (nAb) responses elicited by messenger RNA (mRNA) vaccines against the SARS-CoV-2 variants of concern (VOCs) in immunocompromised patients and healthy controls. Objective: To assess the humoral responses after vaccination with BNT162b2 (Pfizer-BioNTech) or mRNA-1273 (Moderna) vaccines. Design, Setting, and Participants: In this prospective, longitudinal monocentric comparative effectiveness study conducted at the Lausanne University Hospital, binding IgG anti-spike antibody and nAb levels were measured at 1 week, 1 month, 3 months, and 6 months after vaccination with mRNA-1273 (24.6% of participants) or BNT162b2 (75.3% of participants). Interventions: All participants received 2 doses of either mRNA-1273 or BNT162b2 vaccines 4 to 6 weeks apart. Main Outcomes and Measures: The primary outcome of the study was the persistence of nAb responses against the original, nonvariant SARS-CoV-2 (2019-nCoV) and different VOCs at 6 months after vaccination. Key secondary outcomes were associations of the type of mRNA vaccine, the underlying disease, and the treatment with the response to vaccination. Results: Among the 841 participants enrolled between January 14 and August 8, 2021, the patient population comprised 637 participants (mean [SD] age, 61.8 [13.7] years; 386 [60.6%] female), and the healthy control population comprised 204 participants (mean [SD] age, 45.9 [12.0] years; 144 [70.6%] female). There were 399 patients with solid cancers, 101 with hematologic cancers, 38 with solid organ transplants, 99 with autoimmune diseases, and 204 healthy controls. More than 15 000 nAb determinations were performed against the original, nonvariant 2019-nCoV and the Alpha, Beta, Gamma, and Delta variants. The proportions of nAbs and their titers decreased in all study groups at 6 months after vaccination, with the greatest decreases for the Beta and Delta variants. For Beta, the proportion decreased to a median (SE) of 39.2% (5.5%) in those with hematologic cancers, 44.8% (2.7%) in those with solid cancers, 23.1% (8.3%) in those with solid organ transplants, and 22.7% (4.8%) in those with autoimmune diseases compared with 52.1% (4.2%) in healthy controls. For Delta, the proportions decreased to 41.8% (5.6%) in participants with hematologic cancer, 51.9% (2.7%) in those with solid cancers, 26.9% (8.7%) in those with solid organ transplants, and 30.7% (5.3%) in those with autoimmune diseases compared with 56.9% (4.1%) healthy controls. Neutralizing antibody titers decreased 3.5- to 5-fold between month 1 and month 6, and the estimated duration of response was greater and more durable among those participants vaccinated with mRNA-1273. In participants with solid cancers, the estimated duration of nAbs against the Beta variant was 221 days with mRNA-1273 and 146 days with BNT162b2, and against the Delta variant, it was 226 days with mRNA-1273 and 161 with BNT162b2. The estimated duration of nAbs in participants with hematologic cancers was 113 and 127 days against Beta and Delta variants, respectively. Conclusions and Relevance: This comparative effectiveness study suggests that approximately half of patients with hematologic cancers and solid cancers, about 70% of patients with solid organ transplants or autoimmune diseases, and 40% of healthy controls have lost nAbs against the circulating VOCs at 6 months after vaccination. These findings may be helpful for developing the best boosting vaccination schedule especially in immunocompromised patients.


Subject(s)
Autoimmune Diseases , COVID-19 , Hematologic Neoplasms , Neoplasms , Aged , Antibodies, Neutralizing , Antibodies, Viral , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines , Female , Humans , Immunocompromised Host , Immunoglobulin G , Male , Middle Aged , Prospective Studies , RNA, Messenger , SARS-CoV-2 , Vaccination , Vaccines, Synthetic , mRNA Vaccines
20.
Front Oncol ; 12: 1043675, 2022.
Article in English | MEDLINE | ID: mdl-36568192

ABSTRACT

During the acute phase of the COVID-19 pandemic, hospitals faced a challenge to manage patients, especially those with other comorbidities and medical needs, such as cancer patients. Here, we use Process Mining to analyze real-world therapeutic pathways in a cohort of 1182 cancer patients of the Lausanne University Hospital following COVID-19 infection. The algorithm builds trees representing sequences of coarse-grained events such as Home, Hospitalization, Intensive Care and Death. The same trees can also show probability of death or time-to-event statistics in each node. We introduce a new tool, called Differential Process Mining, which enables comparison of two patient strata in each node of the tree, in terms of hits and death rate, together with a statistical significance test. We thus compare management of COVID-19 patients with an active cancer in the first vs. second COVID-19 waves to quantify hospital adaptation to the pandemic. We also compare patients having undergone systemic therapy within 1 year to the rest of the cohort to understand the impact of an active cancer and/or its treatment on COVID-19 outcome. This study demonstrates the value of Process Mining to analyze complex event-based real-world data and generate hypotheses on hospital resource management or on clinical patient care.

SELECTION OF CITATIONS
SEARCH DETAIL