Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
J Immunother Cancer ; 7(1): 40, 2019 02 11.
Article in English | MEDLINE | ID: mdl-30744692

ABSTRACT

BACKGROUND: Several predictive biomarkers are currently approved or are under investigation for the selection of patients for checkpoint blockade. Tumor PD-L1 expression is used for stratification of non-small cell lung (NSCLC) patients, with tumor mutational burden (TMB) also being explored with promising results, and mismatch-repair deficiency is approved for tumor site-agnostic disease. While tumors with high PD-L1 expression, high TMB, or mismatch repair deficiency respond well to checkpoint blockade, tumors with lower PD-L1 expression, lower mutational burdens, or mismatch repair proficiency respond much less frequently. CASE PRESENTATION: We studied two patients with unexpected responses to checkpoint blockade monotherapy: a patient with PD-L1-negative and low mutational burden NSCLC and one with mismatch repair proficient colorectal cancer (CRC), both of whom lack the biomarkers associated with response to checkpoint blockade, yet achieved durable clinical benefit. Both maintained T-cell responses in peripheral blood to oncogenic driver mutations - BRAF-N581I in the NSCLC and AKT1-E17K in the CRC - years after treatment initiation. Mutation-specific T cells were also found in the primary tumor and underwent dynamic perturbations in the periphery upon treatment. CONCLUSIONS: These findings suggest that T cell responses to oncogenic driver mutations may be more prevalent than previously appreciated and could be harnessed in immunotherapeutic treatment, particularly for patients who lack the traditional biomarkers associated with response. Comprehensive studies are warranted to further delineate additional predictive biomarkers and populations of patients who may benefit from checkpoint blockade.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Colorectal Neoplasms/immunology , Lung Neoplasms/drug therapy , Nivolumab/therapeutic use , T-Lymphocytes/immunology , Aged , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Mutation , Oncogenes , Treatment Outcome
3.
Science ; 357(6349): 409-413, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28596308

ABSTRACT

The genomes of cancers deficient in mismatch repair contain exceptionally high numbers of somatic mutations. In a proof-of-concept study, we previously showed that colorectal cancers with mismatch repair deficiency were sensitive to immune checkpoint blockade with antibodies to programmed death receptor-1 (PD-1). We have now expanded this study to evaluate the efficacy of PD-1 blockade in patients with advanced mismatch repair-deficient cancers across 12 different tumor types. Objective radiographic responses were observed in 53% of patients, and complete responses were achieved in 21% of patients. Responses were durable, with median progression-free survival and overall survival still not reached. Functional analysis in a responding patient demonstrated rapid in vivo expansion of neoantigen-specific T cell clones that were reactive to mutant neopeptides found in the tumor. These data support the hypothesis that the large proportion of mutant neoantigens in mismatch repair-deficient cancers make them sensitive to immune checkpoint blockade, regardless of the cancers' tissue of origin.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Neoplastic Syndromes, Hereditary/immunology , Neoplastic Syndromes, Hereditary/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/immunology , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Cell Cycle Checkpoints/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , DNA Mismatch Repair , Disease-Free Survival , Female , Humans , Male , Middle Aged , Mutation , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/mortality , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Young Adult
4.
Science ; 352(6283): 366-70, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27081073

ABSTRACT

Immune-mediated tissue regeneration driven by a biomaterial scaffold is emerging as an innovative regenerative strategy to repair damaged tissues. We investigated how biomaterial scaffolds shape the immune microenvironment in traumatic muscle wounds to improve tissue regeneration. The scaffolds induced a pro-regenerative response, characterized by an mTOR/Rictor-dependent T helper 2 pathway that guides interleukin-4-dependent macrophage polarization, which is critical for functional muscle recovery. Manipulating the adaptive immune system using biomaterials engineering may support the development of therapies that promote both systemic and local pro-regenerative immune responses, ultimately stimulating tissue repair.


Subject(s)
Biocompatible Materials , Muscle, Skeletal/injuries , Muscle, Skeletal/physiology , Tissue Scaffolds , Wound Healing/immunology , Adaptive Immunity , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Disease Models, Animal , Homeostasis/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Macrophages/immunology , Mice, Inbred C57BL , Rapamycin-Insensitive Companion of mTOR Protein , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Th2 Cells/immunology , Tissue Engineering
5.
J Nucl Med ; 57(5): 735-40, 2016 May.
Article in English | MEDLINE | ID: mdl-26795289

ABSTRACT

UNLABELLED: The aim of this study was to assess the prognostic and predictive value of early quantitative (18)F-FDG PET to monitor therapy with an antibody to the insulinlike growth factor 1 receptor (IGF-1R antibody) in patients with Ewing sarcoma family of tumors (ESFT). METHODS: (18)F-FDG PET images at baseline and approximately 9 d after initiation of IGF-1R antibody therapy in 115 patients with refractory or relapsed ESFT were prospectively obtained as part of the Sarcoma Alliance for Research through Collaboration trial. Responses were centrally evaluated by PERCIST 1.0 in 93 patients. The 9-d PET responses were correlated to overall survival (OS), progression-free survival (PFS), and clinical benefit after 6 wk of therapy based on clinical observation and CT response by World Health Organization anatomic criteria. RESULTS: The median OS was 8.1 mo (95% confidence interval, 6.4-10.0 mo). When PERCIST was used, patients with progressive metabolic disease showed shorter OS (median, 4.7 mo) than patients without progression (median, 10.0 mo; P = 0.001). Progressive metabolic disease on day-9 PET was associated with a significantly higher risk of death (hazard ratio, 2.8; 95% confidence interval, 1.5-5.5). Changes in (18)F-FDG uptake after 9 d of therapy had an area under the curve of receiver-operating characteristic of 0.71 to predict 1-y OS. The area under the curve was 0.63 to predict progression at 3 mo and 0.79 to predict clinical benefit after 6 wk of therapy. CONCLUSION: Treatment response by quantitative (18)F-FDG PET assessed by PERCIST 1.0 as early as 9 d into IGF-1R antibody therapy in patients with ESFT can predict the OS, PFS, and clinical response to therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bone Neoplasms/diagnostic imaging , Fluorodeoxyglucose F18 , Positron-Emission Tomography , Receptor, IGF Type 1/immunology , Sarcoma, Ewing/drug therapy , Adolescent , Adult , Aged , Antibodies, Monoclonal/immunology , Bone Neoplasms/drug therapy , Child , Child, Preschool , Disease-Free Survival , Female , Follow-Up Studies , Glucose/metabolism , Humans , Infant , Male , Middle Aged , Sarcoma, Ewing/diagnostic imaging , Sarcoma, Ewing/metabolism , Time Factors , Treatment Outcome , Young Adult
6.
N Engl J Med ; 372(26): 2509-20, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26028255

ABSTRACT

BACKGROUND: Somatic mutations have the potential to encode "non-self" immunogenic antigens. We hypothesized that tumors with a large number of somatic mutations due to mismatch-repair defects may be susceptible to immune checkpoint blockade. METHODS: We conducted a phase 2 study to evaluate the clinical activity of pembrolizumab, an anti-programmed death 1 immune checkpoint inhibitor, in 41 patients with progressive metastatic carcinoma with or without mismatch-repair deficiency. Pembrolizumab was administered intravenously at a dose of 10 mg per kilogram of body weight every 14 days in patients with mismatch repair-deficient colorectal cancers, patients with mismatch repair-proficient colorectal cancers, and patients with mismatch repair-deficient cancers that were not colorectal. The coprimary end points were the immune-related objective response rate and the 20-week immune-related progression-free survival rate. RESULTS: The immune-related objective response rate and immune-related progression-free survival rate were 40% (4 of 10 patients) and 78% (7 of 9 patients), respectively, for mismatch repair-deficient colorectal cancers and 0% (0 of 18 patients) and 11% (2 of 18 patients) for mismatch repair-proficient colorectal cancers. The median progression-free survival and overall survival were not reached in the cohort with mismatch repair-deficient colorectal cancer but were 2.2 and 5.0 months, respectively, in the cohort with mismatch repair-proficient colorectal cancer (hazard ratio for disease progression or death, 0.10 [P<0.001], and hazard ratio for death, 0.22 [P=0.05]). Patients with mismatch repair-deficient noncolorectal cancer had responses similar to those of patients with mismatch repair-deficient colorectal cancer (immune-related objective response rate, 71% [5 of 7 patients]; immune-related progression-free survival rate, 67% [4 of 6 patients]). Whole-exome sequencing revealed a mean of 1782 somatic mutations per tumor in mismatch repair-deficient tumors, as compared with 73 in mismatch repair-proficient tumors (P=0.007), and high somatic mutation loads were associated with prolonged progression-free survival (P=0.02). CONCLUSIONS: This study showed that mismatch-repair status predicted clinical benefit of immune checkpoint blockade with pembrolizumab. (Funded by Johns Hopkins University and others; ClinicalTrials.gov number, NCT01876511.).


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , DNA Mismatch Repair , Neoplasm Metastasis/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents/adverse effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Metastasis/genetics
7.
Cancer Discov ; 5(1): 43-51, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25358689

ABSTRACT

UNLABELLED: We examined the immune microenvironment of primary colorectal cancer using immunohistochemistry, laser capture microdissection/qRT-PCR, flow cytometry, and functional analysis of tumor-infiltrating lymphocytes. A subset of colorectal cancer displayed high infiltration with activated CD8(+) cytotoxic T lymphocyte (CTL) as well as activated Th1 cells characterized by IFNγ production and the Th1 transcription factor TBET. Parallel analysis of tumor genotypes revealed that virtually all of the tumors with this active Th1/CTL microenvironment had defects in mismatch repair, as evidenced by microsatellite instability (MSI). Counterbalancing this active Th1/CTL microenvironment, MSI tumors selectively demonstrated highly upregulated expression of multiple immune checkpoints, including five-PD-1, PD-L1, CTLA-4, LAG-3, and IDO-currently being targeted clinically with inhibitors. These findings link tumor genotype with the immune microenvironment, and explain why MSI tumors are not naturally eliminated despite a hostile Th1/CTL microenvironment. They further suggest that blockade of specific checkpoints may be selectively efficacious in the MSI subset of colorectal cancer. SIGNIFICANCE: The findings reported in this article are the first to demonstrate a link between a genetically defined subtype of cancer and its corresponding expression of immune checkpoints in the tumor microenvironment. The mismatch repair-defective subset of colorectal cancer selectively upregulates at least five checkpoint molecules that are targets of inhibitors currently being clinically tested.


Subject(s)
Cell Cycle Checkpoints , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Microsatellite Instability , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Colonic Neoplasms/pathology , Humans , Immunohistochemistry , Immunophenotyping , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Phenotype , Stromal Cells/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
8.
Cancer Immunol Res ; 3(2): 110-5, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25370533

ABSTRACT

PD-L1 expression in melanoma correlates with response to PD-1 pathway-blocking antibodies. Aberrant tumor-cell PD-L1 expression may be oncogene driven and/or induced by IFNγ. Melanomas express PD-L1 in association with tumor-infiltrating lymphocytes (TIL), but the potential contribution of the BRAF V600E mutation (BRAFmut) to induced PD-L1 expression has not been determined. Fifty-two archival melanocytic lesions were assessed for PD-L1 expression, TIL infiltration, and BRAFmut simultaneously. IFNγ-induced PD-L1 expression in cultured melanomas was assessed in parallel according to BRAF status. Melanocyte PD-L1 expression was observed in 40% of specimens, and BRAFmut was observed in 42% of specimens, but no significant concordance was found between these variables. Almost all melanocytes displaying PD-L1 expression were observed to be adjacent to TILs, irrespective of BRAF status. TIL(-) lesions were not more likely to be associated with BRAFmut, when compared with TIL(+) lesions. Baseline expression of PD-L1 by melanoma cell lines was virtually nil, regardless of BRAFmut status, and the intensity of IFN-induced PD-L1 expression in melanoma cell lines likewise did not correlate with BRAF mutational status. PD-L1 expression in melanocytic lesions does not correlate with the BRAFmut. Thus, distinct populations of melanoma patients will likely benefit from BRAF inhibitors versus PD-1 pathway blockade.


Subject(s)
B7-H1 Antigen/metabolism , Melanoma/genetics , Mutation , Nevus/genetics , Proto-Oncogene Proteins B-raf/genetics , Humans , Interferon-gamma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Melanocytes/immunology , Melanoma/immunology , Melanoma/secondary , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nevus/immunology , Tumor Cells, Cultured
9.
J Oncol Pract ; 10(5): e299-306, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24986113

ABSTRACT

BACKGROUND: Patient-reported outcome (PRO) measures are increasingly being used in clinical practice to inform individual patient management, but evidence is needed on which PROs are best suited for clinical use. METHODS: This controlled trial randomly assigned patients with breast and prostate cancer undergoing treatment to complete one of three PRO measures: European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire-Core 30 (QLQ-C30), Supportive Care Needs Survey-Short Form (SCNS-SF34), or six domains from the Patient-Reported Outcomes Measurement Information System (PROMIS). Patients completed the PRO measures before clinic visits, and the results were provided to both the patient and clinician. At treatment completion, patients and clinicians completed brief feedback forms on the intervention's usefulness and value. Exit interviews were conducted with patients (at end of treatment) and clinicians (at end of study). The primary outcome was the proportion of patients in each arm who either strongly agreed or agreed to all feedback form items. RESULTS: Of 294 eligible patients invited to participate, 224 (76%) enrolled (median age 66 years, 78% white, 72% prostate). Of the 181 patients (81%) who completed at least one feedback form item, participants in the QLQ-C30 study arm were most likely to strongly agree/agree to all items (74%) followed by PROMIS (61%) and SCNS-SF34 (52%; P = .03). Of the 116 participants (52%) who completed all feedback form items, the results were similar: 82% for the QLQ-C30, 62% for PROMIS, and 56% for SCNS-SF34 (P = .05). Clinicians did not prefer one questionnaire over the others. CONCLUSION: These results suggest that, when using PROs in clinical practice for patient management, the measure matters in terms of usefulness to patients.


Subject(s)
Breast Neoplasms/therapy , Medical Oncology/standards , Prostatic Neoplasms/therapy , Self Report , Adult , Aged , Aged, 80 and over , Algorithms , Breast Neoplasms/psychology , Female , Humans , Male , Medical Oncology/methods , Middle Aged , Patient Preference , Patient Satisfaction , Prostatic Neoplasms/psychology , Quality of Life , Research Design , Surveys and Questionnaires , Treatment Outcome
10.
Cancer Immunol Res ; 1(1): 54-63, 2013 Jul.
Article in English | MEDLINE | ID: mdl-24416729

ABSTRACT

Merkel cell carcinoma (MCC) is a lethal, virus-associated cancer that lacks effective therapies for advanced disease. Agents blocking the PD-1/PD-L1 pathway have demonstrated objective, durable tumor regressions in patients with advanced solid malignancies and efficacy has been linked to PD-L1 expression in the tumor microenvironment. To investigate whether MCC might be a target for PD-1/PD-L1 blockade, we examined MCC PD-L1 expression, its association with tumor-infiltrating lymphocytes (TILs), Merkel cell polyomavirus (MCPyV), and overall survival. Sixty-seven MCC specimens from 49 patients were assessed with immunohistochemistry for PD-L1 expression by tumor cells and TILs, and immune infiltrates were characterized phenotypically. Tumor cell and TIL PD-L1 expression were observed in 49% and 55% of patients, respectively. In specimens with PD-L1(+) tumor cells, 97% (28/29) demonstrated a geographic association with immune infiltrates. Among specimens with moderate-severe TIL intensities, 100% (29/29) demonstrated PD-L1 expression by tumor cells. Significant associations were also observed between the presence of MCPyV DNA, a brisk inflammatory response, and tumor cell PD-L1 expression: MCPyV(-) tumor cells were uniformly PD-L1(-). Taken together, these findings suggest that a local tumor-specific and potentially MCPyV-specific immune response drives tumor PD-L1 expression, similar to previous observations in melanoma and head and neck squamous cell carcinomas. In multivariate analyses, PD-L1(-) MCCs were independently associated with worse overall survival (hazard ratio 3.12; 95% CI, 1.28-7.61; p=0.012). These findings suggest that an endogenous immune response promotes PD-L1 expression in the MCC microenvironment when MCPyV is present, and provide a rationale for investigating therapies blocking PD-1/PD-L1 for patients with MCC.


Subject(s)
B7-H1 Antigen/biosynthesis , Carcinoma, Merkel Cell/metabolism , Inflammation/metabolism , Merkel cell polyomavirus/isolation & purification , Skin Neoplasms/metabolism , Aged , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Carcinoma, Merkel Cell/immunology , Carcinoma, Merkel Cell/virology , Female , Humans , Immunohistochemistry , Inflammation/immunology , Inflammation/virology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Neoplasm Staging , Skin Neoplasms/immunology , Skin Neoplasms/virology , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...