Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 273
Filter
1.
Lancet Oncol ; 25(5): e205-e216, 2024 05.
Article in English | MEDLINE | ID: mdl-38697166

ABSTRACT

Multiple myeloma remains an incurable disease, despite the development of numerous drug classes and combinations that have contributed to improved overall survival. Immunotherapies directed against cancer cell-surface antigens, such as chimeric antigen receptor (CAR) T-cell therapy and T-cell-redirecting bispecific antibodies, have recently received regulatory approvals and shown unprecedented efficacy. However, these immunotherapies have unique mechanisms of action and toxicities that are different to previous treatments for myeloma, so experiences from clinical trials and early access programmes are essential for providing specific recommendations for management of patients, especially as these agents become available across many parts of the world. Here, we provide expert consensus clinical practice guidelines for the use of bispecific antibodies for the treatment of myeloma. The International Myeloma Working Group is also involved in the collection of prospective real-time data of patients treated with such immunotherapies, with the aim of learning continuously and adapting clinical practices to optimise the management of patients receiving immunotherapies.


Subject(s)
Antibodies, Bispecific , Consensus , Multiple Myeloma , T-Lymphocytes , Humans , Antibodies, Bispecific/therapeutic use , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Multiple Myeloma/drug therapy , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Immunotherapy/methods , Immunotherapy/standards , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/adverse effects
2.
Zhonghua Wei Chang Wai Ke Za Zhi ; 27(4): 353-358, 2024 Apr 25.
Article in Chinese | MEDLINE | ID: mdl-38644240

ABSTRACT

Neoadjuvant immunotherapy has achieved exciting efficacy with high clinical complete response (cCR) and pathologic complete response (pCR) rates and durable long-term effects. PD-1 checkpoint blockade-based immunotherapy has been highly successful in microsatellite instability high (MSI-H)/mismatch repair deficiency (dMMR) colorectal cancer and has been recommended as the first-line treatment for metastatic colorectal cancer by domestic and international guidelines. Several studies have shown that immunotherapy can be a potentially curable treatment for MSI-H rectal cancer and has even shown promise in organ preservation in colon cancer. In this study, we first clarified the feasibility of the watch-and-wait strategy after PD-1 checkpoint blockade treatment by indirect and direct evidence. Then from the assessment tools (including digital rectal examination, endoscopy, radiology, and lymph node assessment), the viable assessment methods of cCR for immunotherapy and related difficulties are proposed. Finally, the medication choices of immunotherapy, the treatment regimen, and the follow-up strategy are further discussed. We hope that neoadjuvant immunotherapy could be appropriately applied in MSI-H/dMMR colorectal cancer so that more patients can achieve organ preservation.


Subject(s)
Colorectal Neoplasms , Immune Checkpoint Inhibitors , Immunotherapy , Microsatellite Instability , Neoadjuvant Therapy , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Immunotherapy/methods , Immunotherapy/standards , Immune Checkpoint Inhibitors/therapeutic use , Pathologic Complete Response , Watchful Waiting , Humans
4.
Hum Genomics ; 17(1): 116, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38111044

ABSTRACT

BACKGROUND: The effect of SPP1 in squamous cell carcinoma of the penis (PSCC) remained unknown. We attempted to clarify the function of the SPP1 gene in PSCC. METHOD: Eight paired penile cancer specimens (including penile cancer tissue, paracancerous tissue, and positive lymph node tissue) subjected to whole transcriptome sequencing were analysed to identify differentially expressed genes. We used immunohistochemistry to detect the expression of SPP1 protein and immune cell related proteins in penile cancer tissue. Then, we performed weighted gene coexpression network analysis (WGCNA) to identify the genes related to SPP1 in penile cancer tissue and positive lymph node tissue. Based on the GSE57955 dataset, the CIBERSORT and ssGSEA algorithms were carried out to investigate the immune environment of PSCC. GSVA analysis was conducted to identify the signaling pathways related to SPP1 subgroups. Enzyme-linked immunosorbent assay (ELISA) method was adopted to detect SPP1 level in the serum of 60 patients with penile cancer. RESULTS: Differential analysis indicated that SPP1 was the most differentially upregulated gene in both penile cancer tissues and positive lymph node tissues. Survival analysis suggested that the prognosis of the low-SPP1 group was significantly poorer than that of the high-SPP1 group. Subsequently, immune-related bioinformatics showed that SPP1 was significantly associated with B cells, CD8 + T cells, CD4 + T cells, macrophages, helper T cells, neutrophils and dendritic cells. The immunohistochemical results showed that the high-SPP1 group was characterized by relatively high expression of CD16 and relatively low expression of CD4. GSVA analysis indicated that high-SPP1 group was significantly associated with immune-related pathways such as PD-L1 expression and the PD-1 checkpoint pathway in cancer and the TNF signaling pathway. ELISA demonstrated that the serum level of SPP1 in patients with positive lymph node metastasis of penile cancer was significantly higher than that in patients with negative lymph node metastasis of penile cancer. CONCLUSION: Our study shows that the SPP1 gene might be an effective biomarker for predicting the prognosis and the efficacy of immunotherapy in PSCC patients.


Subject(s)
Carcinoma, Squamous Cell , Osteopontin , Penile Neoplasms , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Penile Neoplasms/diagnosis , Penile Neoplasms/genetics , Penile Neoplasms/pathology , Penile Neoplasms/therapy , Immunotherapy/standards , Osteopontin/blood , Osteopontin/genetics , Osteopontin/metabolism , Biomarkers, Tumor/blood , Gene Expression Profiling , Survival Analysis , Sequence Analysis, RNA
5.
J. immunotherap. cancer ; 11(10): 1-39, 20231001. tab
Article in English | BIGG - GRADE guidelines | ID: biblio-1525933

ABSTRACT

Since the first approval for immune checkpoint inhibitors (ICIs) for the treatment of cutaneous melanoma more than a decade ago, immunotherapy has completely transformed the treatment landscape of this chemotherapy-resistant disease. Combination regimens including ICIs directed against programmed cell death protein 1 (PD-1) with anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) agents or, more recently, anti-lymphocyte-activation gene 3 (LAG-3) agents, have gained regulatory approvals for the treatment of metastatic cutaneous melanoma, with long-term follow-up data suggesting the possibility of cure for some patients with advanced disease. In the resectable setting, adjuvant ICIs prolong recurrence-free survival, and neoadjuvant strategies are an active area of investigation. Other immunotherapy strategies, such as oncolytic virotherapy for injectable cutaneous melanoma and bispecific T-cell engager therapy for HLA-A*02:01 genotype-positive uveal melanoma, are also available to patients. Despite the remarkable efficacy of these regimens for many patients with cutaneous melanoma, traditional immunotherapy biomarkers (ie, programmed death-ligand 1 expression, tumor mutational burden, T-cell infiltrate and/or microsatellite stability) have failed to reliably predict response. Furthermore, ICIs are associated with unique toxicity profiles, particularly for the highly active combination of anti-PD-1 plus anti-CTLA-4 agents. The Society for Immunotherapy of Cancer (SITC) convened a panel of experts to develop this clinical practice guideline on immunotherapy for the treatment of melanoma, including rare subtypes of the disease (eg, uveal, mucosal), with the goal of improving patient care by providing guidance to the oncology community. Drawing from published data and clinical experience, the Expert Panel developed evidence- and consensus-based recommendations for healthcare professionals using immunotherapy to treat melanoma, with topics including therapy selection in the advanced and perioperative settings, intratumoral immunotherapy, when to use immunotherapy for patients with BRAFV600- mutated disease, management of patients with brain metastases, evaluation of treatment response, special patient populations, patient education, quality of life, and survivorship, among others.


Subject(s)
Humans , Immunotherapy/standards , Melanoma/immunology , Antineoplastic Agents, Immunological/therapeutic use
6.
Proc Natl Acad Sci U S A ; 119(36): e2205983119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037341

ABSTRACT

Effective antitumor immunity in mice requires activation of the type I interferon (IFN) response pathway. IFNα and IFNß therapies have proven promising in humans, but suffer from limited efficacy and high toxicity. Intratumoral IFN retention ameliorates systemic toxicity, but given the complexity of IFN signaling, it was unclear whether long-term intratumoral retention of type I IFNs would promote or inhibit antitumor responses. To this end, we compared the efficacy of IFNα and IFNß that exhibit either brief or sustained retention after intratumoral injection in syngeneic mouse tumor models. Significant enhancement in tumor retention, mediated by anchoring these IFNs to coinjected aluminum-hydroxide (alum) particles, greatly improved both their tolerability and efficacy. The improved efficacy of alum-anchored IFNs could be attributed to sustained pleiotropic effects on tumor cells, immune cells, and nonhematopoietic cells. Alum-anchored IFNs achieved high cure rates of B16F10 tumors upon combination with either anti-PD-1 antibody or interleukin-2. Interestingly however, these alternative combination immunotherapies yielded disparate T cell phenotypes and differential resistance to tumor rechallenge, highlighting important distinctions in adaptive memory formation for combinations of type I IFNs with other immunotherapies.


Subject(s)
Aluminum Hydroxide , Immunotherapy , Interferon Type I , Alum Compounds/chemistry , Aluminum Hydroxide/chemistry , Animals , Antineoplastic Agents/therapeutic use , Disease Models, Animal , Humans , Immunotherapy/methods , Immunotherapy/standards , Interferon Type I/chemistry , Interferon Type I/therapeutic use , Interferon-alpha , Interferon-beta , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice
7.
Mol Cancer ; 21(1): 2, 2022 01 02.
Article in English | MEDLINE | ID: mdl-34980132

ABSTRACT

BACKGROUND: In recent years, the application of functional genetic immuno-oncology screens has showcased the striking ability to identify potential regulators engaged in tumor-immune interactions. Although these screens have yielded substantial data, few studies have attempted to systematically aggregate and analyze them. METHODS: In this study, a comprehensive data collection of tumor immunity-associated functional screens was performed. Large-scale genomic data sets were exploited to conduct integrative analyses. RESULTS: We identified 105 regulator genes that could mediate resistance or sensitivity to immune cell-induced tumor elimination. Further analysis identified MON2 as a novel immune-oncology target with considerable therapeutic potential. In addition, based on the 105 genes, a signature named CTIS (CRISPR screening-based tumor-intrinsic immune score) for predicting response to immune checkpoint blockade (ICB) and several immunomodulatory agents with the potential to augment the efficacy of ICB were also determined. CONCLUSION: Overall, our findings provide insights into immune oncology and open up novel opportunities for improving the efficacy of current immunotherapy agents.


Subject(s)
CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Genetic Testing/methods , Genomics/methods , Medical Oncology , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/therapeutic use , Clinical Decision-Making , Computational Biology/methods , Disease Management , Gene Expression Profiling , Gene Regulatory Networks , Humans , Immunotherapy/methods , Immunotherapy/standards , Medical Oncology/methods , Medical Oncology/standards , Prognosis , Transcriptome , Treatment Outcome
10.
Indian J Pathol Microbiol ; 64(4): 664-670, 2021.
Article in English | MEDLINE | ID: mdl-34673583

ABSTRACT

BACKGROUND: The programmed cell death protein - 1 (PD-1) - programmed cell death ligand - 1 (PD-L1) axis is emerging as a promising target for immunotherapy in triple-negative breast cancers (TNBC). AIMS: We analyzed the expression of PD-L1 in TNBC cases, with special emphasis on lymphocyte-predominant tumors along with correlation of the same with clinicopathological features and outcome. SETTINGS AND DESIGN: Tissue microarrays (TMA) were prepared from resection specimens of TNBC cases diagnosed from 2004 to 2008. SUBJECTS AND METHODS: Immunohistochemical staining was performed on the TMA using the ventana PD-L1 antibody (Clone SP 263). STATISTICAL ANALYSIS: Chi-square test was used for correlation of PD-L1 positivity in tumor and immune cells with clinicopathological features. Univariate and multivariate survival analyses were carried out using the Kaplan Meir and Cox Regression methods, respectively. RESULTS: Overall, PD-L1 staining was seen in 35.9% (66 out of 184) tumors. PD-L1 positivity of tumor cells was seen in 14.7% (27 out of 184 cases), whereas stromal immune cell expression was observed in 21.2% (39 out of 184) cases. Lymphocyte-predominant tumors showed statistically significant expression of PD-L1 in both tumor (P < 0.0001) and immune cells (P 0.036). On univariate analysis, PD-L1 in immune cells was associated with good overall survival (P 0.05) as well as disease-free survival (P 0.013). On multivariate analysis, the same was associated with a significantly decreased risk for recurrence (P 0.018). CONCLUSION: PD-L1 expression in stromal immune cells proved to be a significant prognostic factor for TNBC. This data can serve as a baseline to plan clinical trials with anti-PD-L1 drugs for TNBC in the Indian setting.


Subject(s)
Apoptosis/drug effects , B7-H1 Antigen/therapeutic use , Carcinoma/drug therapy , Immunotherapy/statistics & numerical data , Immunotherapy/standards , Ligands , Triple Negative Breast Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Humans , Middle Aged , Practice Guidelines as Topic , Retrospective Studies , Young Adult
11.
J Hepatol ; 75(6): 1397-1408, 2021 12.
Article in English | MEDLINE | ID: mdl-34216724

ABSTRACT

BACKGROUND & AIMS: Intratumor molecular heterogeneity is a key feature of tumorigenesis and is linked to treatment failure and patient prognosis. Herein, we aimed to determine what drives tumor cell evolution by performing single-cell transcriptomic analysis. METHODS: We analyzed 46 hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA) biopsies from 37 patients enrolled in interventional studies at the NIH Clinical Center, with 16 biopsies collected before and after treatment from 7 patients. We developed a novel machine learning-based consensus clustering approach to track cellular states of 57,000 malignant and non-malignant cells including tumor cell transcriptome-based functional clonality analysis. We determined tumor cell relationships using RNA velocity and reverse graph embedding. We also studied longitudinal samples from 4 patients to determine tumor cellular state and its evolution. We validated our findings in bulk transcriptomic data from 488 patients with HCC and 277 patients with iCCA. RESULTS: Using transcriptomic clusters as a surrogate for functional clonality, we observed an increase in tumor cell state heterogeneity which was tightly linked to patient prognosis. Furthermore, increased functional clonality was accompanied by a polarized immune cell landscape which included an increase in pre-exhausted T cells. We found that SPP1 expression was tightly associated with tumor cell evolution and microenvironmental reprogramming. Finally, we developed a user-friendly online interface as a knowledge base for a single-cell atlas of liver cancer. CONCLUSIONS: Our study offers insight into the collective behavior of tumor cell communities in liver cancer as well as potential drivers of tumor evolution in response to therapy. LAY SUMMARY: Intratumor molecular heterogeneity is a key feature of tumorigenesis that is linked to treatment failure and patient prognosis. In this study, we present a single-cell atlas of liver tumors from patients treated with immunotherapy and describe intratumoral cell states and their hierarchical relationship. We suggest osteopontin, encoded by the gene SPP1, as a candidate regulator of tumor evolution in response to treatment.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Immunotherapy/standards , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/ultrastructure , Biopsy/methods , Biopsy/statistics & numerical data , Carcinoma, Hepatocellular/physiopathology , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/physiopathology , Humans , Immunotherapy/methods , Immunotherapy/statistics & numerical data , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Neoplastic Cells, Circulating/classification
12.
Lancet Oncol ; 22(6): e240-e253, 2021 06.
Article in English | MEDLINE | ID: mdl-34087151

ABSTRACT

Non-AIDS-defining cancers are a growing source of morbidity for people with HIV globally. Although people living with HIV have a disproportionately increased risk of developing virally mediated cancers, cancer burden for common non-AIDS-defining cancers that are not virally associated and are linked to ageing, such as prostate cancer, is becoming higher than for virally mediated cancers. Ageing, behavioural, and HIV-specific factors drive the incidence and affect the outcomes of non-AIDS-defining cancers, presenting different challenges for addressing global morbidity and mortality from non-AIDS-defining cancer. Although large population-based studies have shown that people living with HIV with non-AIDS-defining cancers have poorer cancer outcomes than do people without HIV, current guidelines emphasise that people living with HIV with non-AIDS-defining cancers should receive standard, guideline-based treatment, and infectious disease and oncology providers should work closely to address potential drug interactions between antiretroviral therapy and antineoplastic treatment. Most trials target preventive measures focusing on non-AIDS-defining cancers. However, treatment trials for the optimal management of people living with HIV and non-AIDS-defining cancer, including interventions such as immunotherapies, are needed to improve non-AIDS-defining cancer outcomes.


Subject(s)
Acquired Immunodeficiency Syndrome/therapy , HIV Infections/therapy , Neoplasms/therapy , Sarcoma, Kaposi/therapy , Acquired Immunodeficiency Syndrome/complications , Acquired Immunodeficiency Syndrome/epidemiology , Antiretroviral Therapy, Highly Active , HIV/pathogenicity , HIV Infections/epidemiology , HIV Infections/virology , Humans , Immunotherapy/standards , Neoplasms/epidemiology , Neoplasms/etiology , Risk Factors , Sarcoma, Kaposi/epidemiology , Sarcoma, Kaposi/etiology , Sarcoma, Kaposi/pathology , Treatment Outcome
13.
J Immunother Cancer ; 9(3)2021 03.
Article in English | MEDLINE | ID: mdl-33782108

ABSTRACT

Cancer immunotherapy based on the use of antibodies targeting the so-called checkpoint inhibitors, such as programmed cell death-1 receptor, its ligand, or CTLA-4, has shown durable clinical benefit and survival improvement in melanoma and other tumors. However, there are some special situations that could be a challenge for clinical management. Persons with chronic infections, such as HIV-1 or viral hepatitis, latent tuberculosis, or a history of solid organ transplantation, could be candidates for cancer immunotherapy, but their management requires a multidisciplinary approach. The Spanish Melanoma Group (GEM) panel in collaboration with experts in virology and immunology from different centers in Spain reviewed the literature and developed evidence-based guidelines for cancer immunotherapy management in patients with chronic infections and immunosuppression. These are the first clinical guidelines for cancer immunotherapy treatment in special challenging populations. Cancer immunotherapy in chronically infected or immunosuppressed patients is feasible but needs a multidisciplinary approach in order to decrease the risk of complications related to the coexistent comorbidities.


Subject(s)
Communicable Diseases/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immunocompromised Host , Immunotherapy/standards , Medical Oncology/standards , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Communicable Diseases/epidemiology , Communicable Diseases/therapy , Comorbidity , Consensus , Evidence-Based Medicine , Humans , Immune Checkpoint Inhibitors/adverse effects , Melanoma/epidemiology , Melanoma/immunology , Risk Assessment , Risk Factors , Skin Neoplasms/epidemiology , Skin Neoplasms/immunology , Treatment Outcome
14.
Medicine (Baltimore) ; 100(1): e24018, 2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33429766

ABSTRACT

INTRODUCTION: Pembrolizumab, a monoclonal antibody targeting programmed cell death-1 (PD-1), is approved as a therapy for unresectable or metastatic melanoma. Immunotherapy-associated pneumonitis is an uncommon event. PATIENT CONCERNS: A 73-year-old man was admitted to our hospital with a history of melanoma on the left side of the face (resected in December 2012) and metastasis to the left lung upper lobe (resected in November 2016). Recurrence of metastasis to the bilateral lungs and left pleura was detected in April 2018. A complete response was achieved following treatment with pembrolizumab, with lower limb rashes the only adverse events occurring during therapy. The patient was readmitted in March 2019 with a productive cough, shortness of breath, and mild fever, and sputum culture identified Escherichia coli. DIAGNOSIS: A diagnosis of pneumonia was made, and although cough and shortness of breath responded to ceftazidime and levofloxacin, but fever and poor appetite persisted. Computed tomography showed no improvement in the bilateral lower lobe lesions. Prednisone was initiated based on a clinical diagnosis of immunotherapy-related pneumonitis. The response to prednisone confirmed the diagnosis. INTERVENTIONS: The patient first received ceftazidime and levofloxacin, but the symptoms persisted. Prednisone was initiated based on a clinical diagnosis of immunotherapy-related pneumonitis. OUTCOME: Complete resolution of the bilateral lung lesions occurred after 45 days of prednisone therapy. CONCLUSION: This case report highlights that both pneumonitis and bacterial pneumonia can occur as complications of anti-PD-1 immunotherapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Immunotherapy/adverse effects , Melanoma/drug therapy , Pneumonia, Bacterial/etiology , Pneumonia/etiology , Aged , Cough/etiology , Dyspnea/etiology , Fever/etiology , Humans , Immunotherapy/methods , Immunotherapy/standards , Male , Melanoma/complications , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/physiopathology , Pneumonia/physiopathology , Pneumonia, Bacterial/physiopathology
16.
Medicine (Baltimore) ; 99(51): e23815, 2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33371161

ABSTRACT

ABSTRACT: The present study aimed to evaluate the role of early F-18 2-deoxy-2-[fluorine-18] fluoro-D-glucose positron emission tomography/computed tomography (FDG PET/CT) in non-small cell lung cancer patients undergoing immune checkpoint inhibitor (ICI) treatment.Twenty-four non-small cell lung cancer patients who received nivolumab or pembrolizumab and underwent FDG PET/CT as an interim analysis after 2 or 3 cycles of ICI treatment were retrospectively enrolled. Tumor response was assessed using the PET Response Criteria in Solid Tumors 1.0 (PERCIST) and the European Organization for Research and Treatment of Cancer (EORTC) criteria after 2 or 3 cycles of ICI treatment (SCAN-1) and after an additional 2 cycles of ICI treatment (SCAN-2). The best overall response was determined by FDG PET/CT or chest CT at ≥ 3 months after therapy initiation, and the clinical benefit was investigated. progression-free survival was investigated, and its correlation with clinicopathologic and metabolic parameters was examined using a Cox multivariate proportional hazards model.In the interim analysis, 4 patients achieved a complete metabolic response (CMR), 1 patient exhibited a partial metabolic response (PMR), and 14 patients had Progressive metabolic disease (PMD) according to the PERCIST and EORTC criteria. Four patients showed stable metabolic disease (SMD) according to the PERCIST criteria, and 2 patients showed different responses (i.e., PMR) according to the EORTC criteria. Patients with a CMR or PMR at SCAN-1 had a clinical benefit. Among the 4 patients with SMD at SCAN-1, only 1 experienced a clinical benefit regardless of the percent change in the peak standardized uptake value. Two patients with discordant response assessments between the PERCIST and EORTC criteria showed conflicting clinical benefits. Among the 14 patients with PMD, none experienced any clinical benefit. Only metabolic parameters were significant factors for predicting progression in the multivariate analysis (peak standardized uptake value and metabolic tumor volume, HRs of 1.18 and 1.00, respectively).Based on early F-18 FDG PET/CT after ICI treatment, metabolic parameters could predict post-treatment progression. Responses after ICI treatment were correctly assessed in patients with a CMR, a PMR, and PMD, but patients with SMD required a meticulous follow-up because of varying clinical benefits.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Immunotherapy/standards , Positron Emission Tomography Computed Tomography/methods , Adult , Aged , Antibodies, Monoclonal, Humanized/therapeutic use , Carcinoma, Non-Small-Cell Lung/immunology , Female , Humans , Immunotherapy/methods , Male , Middle Aged , Nivolumab/therapeutic use , Proportional Hazards Models , Retrospective Studies , Treatment Outcome
17.
Neurocirugía (Soc. Luso-Esp. Neurocir.) ; 31(6): 289-298, nov.-dic. 2020.
Article in Spanish | IBECS | ID: ibc-197923

ABSTRACT

INTRODUCCIÓN: El tratamiento de los glioblastomas (GMB) comienza en la mayor parte de los pacientes con una cirugía, ya sea para la extirpación tumoral, ya sea para la obtención de tejido con el que determinar un diagnóstico histológico. Con el fin de obtener el máximo beneficio de estos tratamientos cada paciente debe ser valorado de forma individualizada por un equipo multidisciplinar, constituido por aquellas especialidades involucradas tanto en el diagnóstico como en el tratamiento. MATERIAL Y MÉTODOS: El objetivo de este trabajo es elaborar unas recomendaciones de tratamiento para los pacientes con GBM, para lo cual un experto en cada campo ha descrito lo más relevante de dicha área basado tanto en su experiencia como en la literatura. RESULTADOS: Se han desarrollado los distintos apartados sobre el tratamiento de los GBM y al final de cada apartado se concluye la recomendación del GTNO. CONCLUSIONES: A pesar de que los GBM son tumores agresivos y el pronóstico es malo, los pacientes se pueden beneficiar de tratamientos que mejoren no solo la supervivencia global sino también la calidad de vida. El neurocirujano debe conocer las distintas opciones de tratamientos, sus indicaciones y riesgos para poder participar activamente en la toma de decisiones y ofrecer un tratamiento neuroquirúrgico oportuno a cada situación


INTRODUCTION: Glioblastoma (GBM) treatment starts in most patients with surgery, either resection surgery or biopsy, to reach a histology diagnose. Multidisciplinar team, including specialists in brain tumors diagnose and treatment, must make an individualize assessment to get the maximum benefit of the available treatments. MATERIAL AND METHODS: Experts in each GBM treatment field have briefly described it based in their experience and the reviewed of the literature. RESULTS: Each area has been summarized and the consensus of the brain tumor group has been included at the end. CONCLUSIONS: GBM are aggressive tumors with a dismal prognosis, however accurate treatments can improve overall survival and quality of life. Neurosurgeons must know treatment options, indications and risks to participate actively in the decision making and to offer the best surgical treatment in every case


Subject(s)
Humans , Consensus Development Conferences as Topic , Retinoblastoma/therapy , Patient Care Team/standards , Brain Neoplasms/surgery , Decision Making , Societies, Medical/standards , Glioma/radiotherapy , Glioma/surgery , Glioma/drug therapy , Intraoperative Neurophysiological Monitoring/standards , Immunotherapy/standards
18.
Front Immunol ; 11: 2044, 2020.
Article in English | MEDLINE | ID: mdl-33013864

ABSTRACT

Immunotherapy is a very promising therapeutic approach against cancer that is particularly effective when combined with gene therapy. Immuno-gene therapy approaches have led to the approval of four advanced therapy medicinal products (ATMPs) for the treatment of p53-deficient tumors (Gendicine and Imlygic), refractory acute lymphoblastic leukemia (Kymriah) and large B-cell lymphomas (Yescarta). In spite of these remarkable successes, immunotherapy is still associated with severe side effects for CD19+ malignancies and is inefficient for solid tumors. Controlling transgene expression through an externally administered inductor is envisioned as a potent strategy to improve safety and efficacy of immunotherapy. The aim is to develop smart immunogene therapy-based-ATMPs, which can be controlled by the addition of innocuous drugs or agents, allowing the clinicians to manage the intensity and durability of the therapy. In the present manuscript, we will review the different inducible, versatile and externally controlled gene delivery systems that have been developed and their applications to the field of immunotherapy. We will highlight the advantages and disadvantages of each system and their potential applications in clinics.


Subject(s)
Genetic Therapy , Immunotherapy , Animals , Biomarkers , Gene Expression Regulation , Genetic Therapy/methods , Genetic Therapy/standards , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Immunotherapy/standards , Molecular Targeted Therapy , Transgenes , Translational Research, Biomedical
19.
BMJ Open ; 10(10): e040989, 2020 10 26.
Article in English | MEDLINE | ID: mdl-33109677

ABSTRACT

OBJECTIVE: The spread of misinformation has accompanied the coronavirus pandemic, including topics such as immune boosting to prevent COVID-19. This study explores how immune boosting is portrayed on the internet during the COVID-19 pandemic. DESIGN: Content analysis. METHODS: We compiled a dataset of 227 webpages from Google searches in Canada and the USA using the phrase 'boost immunity' AND 'coronavirus' on 1 April 2020. We coded webpages for typology and portrayal of immune boosting and supplements. We recorded mentions of microbiome, whether the webpage was selling or advertising an immune boosting product or service, and suggested strategies for boosting immunity. RESULTS: No significant differences were found between webpages that appeared in the searches in Canada and the USA. The most common types of webpages were from news (40.5%) and commercial (24.7%) websites. The concept of immune boosting was portrayed as beneficial for avoiding COVID-19 in 85.5% of webpages and supplements were portrayed as beneficial in 40% of the webpages, but commercial sites were more likely to have these portrayals. The top immune boosting strategies were vitamin C (34.8%), diet (34.4%), sleep (34.4%), exercise (30.8%) and zinc (26.9%). Less than 10% of the webpages provide any critique of the concept of immune boosting. CONCLUSIONS: Pairing evidence-based advice for maintaining one's health (eg, healthy diet, exercise, sleep) with the phrase immune boosting and strategies lacking in evidence may inadvertently help to legitimise the concept, making it a powerful marketing tool. Results demonstrate how the spread of misinformation is complex and often more subtle than blatant fraudulent claims.


Subject(s)
Communication , Consumer Health Information , Coronavirus Infections , Immunologic Factors , Immunotherapy , Internet , Marketing , Pandemics , Pneumonia, Viral , Betacoronavirus , COVID-19 , Canada/epidemiology , Consumer Health Information/methods , Consumer Health Information/standards , Coronavirus Infections/epidemiology , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Data Accuracy , Dietary Supplements/standards , Humans , Immunologic Factors/standards , Immunologic Factors/therapeutic use , Immunotherapy/methods , Immunotherapy/standards , Information Dissemination/ethics , Information Dissemination/methods , Internet/statistics & numerical data , Internet/trends , Marketing/ethics , Marketing/methods , Pandemics/prevention & control , Pneumonia, Viral/epidemiology , Pneumonia, Viral/immunology , Pneumonia, Viral/prevention & control , Public Health , SARS-CoV-2 , United States/epidemiology
20.
Am J Clin Dermatol ; 21(6): 821-832, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32767272

ABSTRACT

Talimogene laherparepvec (T-VEC) is the first agent approved for cancer in the emerging class of oncolytic viral therapies. While T-VEC was approved for the treatment of advanced melanoma in 2015, clinical utilization has been hampered by rapid changes in the therapeutic landscape of melanoma related to advances in both immune checkpoint blockade and targeted therapy, cumbersome logistics involved in T-VEC administration, biosafety concerns, and a perception that T-VEC has limited impact on uninjected, visceral disease. However, with further survival follow-up from the phase III OPTiM (OncovexGM-CSF Pivotal Trial in Melanoma), along with new real-world data and consensus guidelines on safe administration of oncolytic viruses, a roadmap for when and how to use T-VEC has been emerging. In addition, preliminary data have demonstrated improved therapeutic responses to T-VEC in combination with immune checkpoint blockade in patients with melanoma without additive toxicity. This review provides an update on recent data with T-VEC alone and in combination with other agents. The emerging data provide guidance for how to better utilize T-VEC for patients with melanoma and identifies critical areas for clinical investigation to expand the role of T-VEC in combination strategies for the treatment of melanoma and perhaps other cancers.


Subject(s)
Biological Products/administration & dosage , Immunotherapy/standards , Melanoma/therapy , Practice Guidelines as Topic , Skin Neoplasms/therapy , Biological Products/adverse effects , Clinical Trials, Phase III as Topic , Containment of Biohazards/standards , Herpesvirus 1, Human , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Immunotherapy/trends , Injections, Intralesional , Melanoma/diagnosis , Melanoma/immunology , Melanoma/mortality , Oncolytic Virotherapy/methods , Oncolytic Virotherapy/standards , Oncolytic Virotherapy/trends , Oncolytic Viruses/immunology , Oncolytic Viruses/pathogenicity , Skin Neoplasms/diagnosis , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Survival Analysis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...