Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Rev Cancer ; 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38867074

ABSTRACT

Although more than a decade has passed since the approval of immune checkpoint inhibitors (ICIs) for the treatment of melanoma and non-small-cell lung, breast and gastrointestinal cancers, many patients still show limited response. US Food and Drug Administration (FDA)-approved biomarkers include programmed cell death 1 ligand 1 (PDL1) expression, microsatellite status (that is, microsatellite instability-high (MSI-H)) and tumour mutational burden (TMB), but these have limited utility and/or lack standardized testing approaches for pan-cancer applications. Tissue-based analytes (such as tumour gene signatures, tumour antigen presentation or tumour microenvironment profiles) show a correlation with immune response, but equally, these demonstrate limited efficacy, as they represent a single time point and a single spatial assessment. Patient heterogeneity as well as inter- and intra-tumoural differences across different tissue sites and time points represent substantial challenges for static biomarkers. However, dynamic biomarkers such as longitudinal biopsies or novel, less-invasive markers such as blood-based biomarkers, radiomics and the gut microbiome show increasing potential for the dynamic identification of ICI response, and patient-tailored predictors identified through neoadjuvant trials or novel ex vivo tumour models can help to personalize treatment. In this Perspective, we critically assess the multiple new static, dynamic and patient-specific biomarkers, highlight the newest consortia and trial efforts, and provide recommendations for future clinical trials to make meaningful steps forwards in the field.

2.
Radiother Oncol ; 165: 44-51, 2021 12.
Article in English | MEDLINE | ID: mdl-34695520

ABSTRACT

BACKGROUND: Trimodality therapy (TMT) with preoperative chemoradiation followed by surgical resection is used for locally-advanced non-small-cell lung cancer (LA-NSCLC). Traditionally, preoperative radiation doses ≤54 Gy are used due to concerns regarding excess morbidity, but little is known about outcomes and toxicities after TMT with intensity-modulated radiotherapy (IMRT) to higher doses. METHODS: A retrospective analysis of patients who received planned TMT with IMRT for LA-NSCLC at Brigham and Women's Hospital/Dana-Farber Cancer Institute between 2008 and 2017 was performed. Clinical and treatment characteristics, pathologic response, and surgical toxicity were assessed. Kaplan-Meier method and log-rank test was used for survival outcomes. Cox proportional-hazards regression was used for multivariable analysis. RESULTS: Forty-six patients received less than definitive doses of <60 Gy and 30 patients received definitive doses ≥60 Gy. Surgical outcomes, pathologic complete response, and postoperative toxicity did not differ significantly between the groups. With median follow-up of 3.6 years (range: 0.4-11.4), three-year locoregional recurrence-free survival (78.0% vs. 68.3%, p = 0.51) and overall survival (OS) (61.0% vs. 69.4%, p = 0.32) was not significantly different between patients receiving <60 Gy and ≥60 Gy, respectively. On multivariable analysis, older age, clinical stage, and length of hospital stay (LOS) >7 days were associated with OS. CONCLUSIONS: With IMRT, there was no increased rate of surgical complications in patients receiving higher doses of radiation. Survival outcomes or LOS did not differ based on radiation dose, but increased LOS was associated with worse OS. Larger prospective studies are needed to further examine outcomes after IMRT in patients with LA-NSCLC receiving TMT.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Radiotherapy, Intensity-Modulated , Aged , Carcinoma, Non-Small-Cell Lung/therapy , Chemoradiotherapy , Disease-Free Survival , Female , Humans , Lung Neoplasms/drug therapy , Radiotherapy Dosage , Radiotherapy, Intensity-Modulated/adverse effects , Retrospective Studies
3.
J Surg Case Rep ; 2020(2): rjz379, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32047592

ABSTRACT

Desmoid tumors are rare malignancies derived from myofibroblasts, which can cause significant morbidity due to life-threatening invasion of local structures. Risk factors include familial adenomatous polyposis, antecedent surgical trauma and estrogen exposure. We described a previously healthy 27-year-old female in whom a desmoid tumor developed 2 years after a Roux-en-Y gastric bypass was performed for the treatment of obesity. Computed tomography scan demonstrated a 16-cm complex density intra-abdominal mass. Exploratory laparotomy was performed, revealing a mass firmly adherent to the Roux limb, as well as the jejunojejunostomy and distal portion of the bilopancreatic limb. En bloc resection of the mass and the Roux limb was required, as well as reconstruction of the Roux-en-Y anatomy. This case describes a unique, long-term complication of bariatric surgery, in light of a growing population of patients with altered gastric anatomy.

5.
Nat Med ; 22(6): 614-23, 2016 06.
Article in English | MEDLINE | ID: mdl-27158907

ABSTRACT

An attenuated Plasmodium falciparum (Pf) sporozoite (SPZ) vaccine, PfSPZ Vaccine, is highly protective against controlled human malaria infection (CHMI) 3 weeks after immunization, but the durability of protection is unknown. We assessed how vaccine dosage, regimen, and route of administration affected durable protection in malaria-naive adults. After four intravenous immunizations with 2.7 × 10(5) PfSPZ, 6/11 (55%) vaccinated subjects remained without parasitemia following CHMI 21 weeks after immunization. Five non-parasitemic subjects from this dosage group underwent repeat CHMI at 59 weeks, and none developed parasitemia. Although Pf-specific serum antibody levels correlated with protection up to 21-25 weeks after immunization, antibody levels waned substantially by 59 weeks. Pf-specific T cell responses also declined in blood by 59 weeks. To determine whether T cell responses in blood reflected responses in liver, we vaccinated nonhuman primates with PfSPZ Vaccine. Pf-specific interferon-γ-producing CD8 T cells were present at ∼100-fold higher frequencies in liver than in blood. Our findings suggest that PfSPZ Vaccine conferred durable protection to malaria through long-lived tissue-resident T cells and that administration of higher doses may further enhance protection.


Subject(s)
Antibodies, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Immunogenicity, Vaccine/immunology , Liver/immunology , Malaria Vaccines/therapeutic use , Malaria, Falciparum/prevention & control , Parasitemia/prevention & control , Plasmodium falciparum/immunology , Administration, Intravenous , Adolescent , Adult , Animals , Enzyme-Linked Immunosorbent Assay , Female , Healthy Volunteers , Humans , Immunoglobulin G/immunology , Interferon-gamma/immunology , Liver/cytology , Macaca mulatta , Malaria Vaccines/immunology , Male , Middle Aged , Parasitemia/immunology , Sporozoites/immunology , T-Lymphocytes/immunology , Young Adult
6.
N Engl J Med ; 374(17): 1635-46, 2016 Apr 28.
Article in English | MEDLINE | ID: mdl-25629663

ABSTRACT

BACKGROUND: The West African outbreak of Ebola virus disease that peaked in 2014 has caused more than 11,000 deaths. The development of an effective Ebola vaccine is a priority for control of a future outbreak. METHODS: In this phase 1 study, we administered a single dose of the chimpanzee adenovirus 3 (ChAd3) vaccine encoding the surface glycoprotein of Zaire ebolavirus (ZEBOV) to 60 healthy adult volunteers in Oxford, United Kingdom. The vaccine was administered in three dose levels--1×10(10) viral particles, 2.5×10(10) viral particles, and 5×10(10) viral particles--with 20 participants in each group. We then assessed the effect of adding a booster dose of a modified vaccinia Ankara (MVA) strain, encoding the same Ebola virus glycoprotein, in 30 of the 60 participants and evaluated a reduced prime-boost interval in another 16 participants. We also compared antibody responses to inactivated whole Ebola virus virions and neutralizing antibody activity with those observed in phase 1 studies of a recombinant vesicular stomatitis virus-based vaccine expressing a ZEBOV glycoprotein (rVSV-ZEBOV) to determine relative potency and assess durability. RESULTS: No safety concerns were identified at any of the dose levels studied. Four weeks after immunization with the ChAd3 vaccine, ZEBOV-specific antibody responses were similar to those induced by rVSV-ZEBOV vaccination, with a geometric mean titer of 752 and 921, respectively. ZEBOV neutralization activity was also similar with the two vaccines (geometric mean titer, 14.9 and 22.2, respectively). Boosting with the MVA vector increased virus-specific antibodies by a factor of 12 (geometric mean titer, 9007) and increased glycoprotein-specific CD8+ T cells by a factor of 5. Significant increases in neutralizing antibodies were seen after boosting in all 30 participants (geometric mean titer, 139; P<0.001). Virus-specific antibody responses in participants primed with ChAd3 remained positive 6 months after vaccination (geometric mean titer, 758) but were significantly higher in those who had received the MVA booster (geometric mean titer, 1750; P<0.001). CONCLUSIONS: The ChAd3 vaccine boosted with MVA elicited B-cell and T-cell immune responses to ZEBOV that were superior to those induced by the ChAd3 vaccine alone. (Funded by the Wellcome Trust and others; ClinicalTrials.gov number, NCT02240875.).


Subject(s)
Ebola Vaccines/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Adenoviruses, Simian/immunology , Adult , Animals , Antibodies, Viral/blood , B-Lymphocytes/physiology , Cytokines/blood , Ebola Vaccines/administration & dosage , Female , Hemorrhagic Fever, Ebola/immunology , Humans , Immunity, Cellular , Immunization, Secondary , Male , Middle Aged , Pan troglodytes , T-Lymphocytes/physiology , Vaccinia , Young Adult
7.
Vaccine ; 33(52): 7444-51, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26476366

ABSTRACT

Viral vectors used in heterologous prime-boost regimens are one of very few vaccination approaches that have yielded significant protection against controlled human malaria infections. Recently, protection induced by chimpanzee adenovirus priming and modified vaccinia Ankara boosting using the ME-TRAP insert has been correlated with the induction of potent CD8(+) T cell responses. This regimen has progressed to field studies where efficacy against infection has now been reported. The same vectors have been used pre-clinically to identify preferred protective antigens for use in vaccines against the pre-erythrocytic, blood-stage and mosquito stages of malaria and this work is reviewed here for the first time. Such antigen screening has led to the prioritization of the PfRH5 blood-stage antigen, which showed efficacy against heterologous strain challenge in non-human primates, and vectors encoding this antigen are in clinical trials. This, along with the high transmission-blocking activity of some sexual-stage antigens, illustrates well the capacity of such vectors to induce high titre protective antibodies in addition to potent T cell responses. All of the protective responses induced by these vectors exceed the levels of the same immune responses induced by natural exposure supporting the view that, for subunit vaccines to achieve even partial efficacy in humans, "unnatural immunity" comprising immune responses of very high magnitude will need to be induced.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Vaccinia virus/genetics , Adenoviruses, Human/genetics , Animals , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Clinical Trials as Topic , Humans , Immunization, Secondary , Malaria Vaccines/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Vaccination , Vaccines, Subunit/immunology
8.
Sci Transl Med ; 4(123): 123ra25, 2012 Feb 29.
Article in English | MEDLINE | ID: mdl-22378925

ABSTRACT

Early immunological events during acute HIV infection are thought to fundamentally influence long-term disease outcome. Whereas the contribution of HIV-specific CD8 T cell responses to early viral control is well established, the role of HIV-specific CD4 T cell responses in the control of viral replication after acute infection is unknown. A growing body of evidence suggests that CD4 T cells-besides their helper function-have the capacity to directly recognize and kill virally infected cells. In a longitudinal study of a cohort of individuals acutely infected with HIV, we observed that subjects able to spontaneously control HIV replication in the absence of antiretroviral therapy showed a significant expansion of HIV-specific CD4 T cell responses-but not CD8 T cell responses-compared to subjects who progressed to a high viral set point (P = 0.038). Markedly, this expansion occurred before differences in viral load or CD4 T cell count and was characterized by robust cytolytic activity and expression of a distinct profile of perforin and granzymes at the earliest time point. Kaplan-Meier analysis revealed that the emergence of granzyme A(+) HIV-specific CD4 T cell responses at baseline was highly predictive of slower disease progression and clinical outcome (average days to CD4 T cell count <350/µl was 575 versus 306, P = 0.001). These data demonstrate that HIV-specific CD4 T cell responses can be used during the earliest phase of HIV infection as an immunological predictor of subsequent viral set point and disease outcome. Moreover, these data suggest that expansion of granzyme A(+) HIV-specific cytolytic CD4 T cell responses early during acute HIV infection contributes substantially to the control of viral replication.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , HIV Infections/immunology , HIV Infections/virology , HIV-1/growth & development , Biomarkers/blood , Boston , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/enzymology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Cytotoxicity, Immunologic , Disease Progression , Germany , Granzymes/metabolism , HIV Infections/diagnosis , HIV Infections/enzymology , HIV-1/genetics , Humans , Kaplan-Meier Estimate , Longitudinal Studies , Lymphocyte Activation , Macrophages/immunology , Macrophages/virology , Phenotype , Prognosis , RNA, Viral/blood , Time Factors , Viral Load , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...