Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 175
Filter
Add more filters

Publication year range
1.
Ann Oncol ; 31(11): 1506-1517, 2020 11.
Article in English | MEDLINE | ID: mdl-32891793

ABSTRACT

Sarcomas are a heterogeneous group of malignancies with mesenchymal lineage differentiation. The discovery of neurotrophic tyrosine receptor kinase (NTRK) gene fusions as tissue-agnostic oncogenic drivers has led to new personalized therapies for a subset of patients with sarcoma in the form of tropomyosin receptor kinase (TRK) inhibitors. NTRK gene rearrangements and fusion transcripts can be detected with different molecular pathology techniques, while TRK protein expression can be demonstrated with immunohistochemistry. The rarity and diagnostic complexity of NTRK gene fusions raise a number of questions and challenges for clinicians. To address these challenges, the World Sarcoma Network convened two meetings of expert adult oncologists and pathologists and subsequently developed this article to provide practical guidance on the management of patients with sarcoma harboring NTRK gene fusions. We propose a diagnostic strategy that considers disease stage and histologic and molecular subtypes to facilitate routine testing for TRK expression and subsequent testing for NTRK gene fusions.


Subject(s)
Sarcoma , Tropomyosin , Adult , Gene Fusion , Humans , Oncogene Proteins, Fusion/genetics , Protein Kinase Inhibitors , Receptor, trkA/genetics , Sarcoma/diagnosis , Sarcoma/drug therapy , Sarcoma/genetics
2.
Ann Oncol ; 29(9): 1995-2002, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30084934

ABSTRACT

Background: Treatment options for soft tissue sarcoma (STS) patients aged ≥65 years (elderly) can be limited by concerns regarding the increased risk of toxicity associated with standard systemic therapies. Trabectedin has demonstrated improved disease control in a phase III trial (ET743-SAR-3007) of patients with advanced liposarcoma or leiomyosarcoma after failure of anthracycline-based chemotherapy. Since previous retrospective analyses have suggested that trabectedin has similar safety and efficacy outcomes regardless of patient age, we carried out a subgroup analysis of the safety and efficacy observed in elderly patients enrolled in this trial. Patients and methods: Patients were randomized 2 : 1 to trabectedin (n = 384) or dacarbazine (n = 193) administered intravenously every-3-weeks. The primary end point was overall survival (OS); secondary end points were progression-free survival (PFS), time-to-progression, objective response rate (ORR), duration of response, symptom severity, and safety. A post hoc analysis was conducted in the elderly patient subgroup. Results: Among 131 (trabectedin = 94; dacarbazine = 37) elderly patients, disease characteristics were well-balanced and consistent with those of the total study population. Treatment exposure was longer in patients treated with trabectedin versus dacarbazine (median four versus two cycles, respectively), with a significantly higher proportion receiving prolonged therapy (≥6 cycles) in the trabectedin arm (43% versus 23%, respectively; P = 0.04). Elderly patients treated with trabectedin showed significantly improved PFS [4.9 versus 1.5 months, respectively; hazard ratio (HR)=0.40; P = 0.0002] but no statistically significant improvement in OS (15.1 versus 8.0 months, respectively; HR = 0.72; P = 0.18) or ORR (9% versus 3%, respectively; P = 0.43). The safety profile for elderly trabectedin-treated patients was comparable to that of the overall trabectedin-treated study population. Conclusions: This subgroup analysis of the elderly population of ET743-SAR-3007 suggests that elderly patients with STS and good performance status can expect clinical benefit from trabectedin similar to that observed in younger patients. Trial registration: www.clinicaltrials.gov, NCT01343277.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , Dacarbazine/administration & dosage , Leiomyosarcoma/drug therapy , Liposarcoma/drug therapy , Trabectedin/administration & dosage , Administration, Intravenous , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/administration & dosage , Dacarbazine/adverse effects , Drug Administration Schedule , Female , Humans , Kaplan-Meier Estimate , Leiomyosarcoma/mortality , Leiomyosarcoma/pathology , Liposarcoma/mortality , Liposarcoma/pathology , Male , Middle Aged , Progression-Free Survival , Time Factors , Trabectedin/adverse effects , Young Adult
3.
Ann Oncol ; 28(10): 2595-2605, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28945830

ABSTRACT

BACKGROUND: While patient-derived xenografts (PDXs) offer a powerful modality for translational cancer research, a precise evaluation of how accurately patient responses correlate with matching PDXs in a large, heterogeneous population is needed for assessing the utility of this platform for preclinical drug-testing and personalized patient cancer treatment. PATIENTS AND METHODS: Tumors obtained from surgical or biopsy procedures from 237 cancer patients with a variety of solid tumors were implanted into immunodeficient mice and whole-exome sequencing was carried out. For 92 patients, responses to anticancer therapies were compared with that of their corresponding PDX models. RESULTS: We compared whole-exome sequencing of 237 PDX models with equivalent information in The Cancer Genome Atlas database, demonstrating that tumorgrafts faithfully conserve genetic patterns of the primary tumors. We next screened PDXs established for 92 patients with various solid cancers against the same 129 treatments that were administered clinically and correlated patient outcomes with the responses in corresponding models. Our analysis demonstrates that PDXs accurately replicate patients' clinical outcomes, even as patients undergo several additional cycles of therapy over time, indicating the capacity of these models to correctly guide an oncologist to treatments that are most likely to be of clinical benefit. CONCLUSIONS: Integration of PDX models as a preclinical platform for assessment of drug efficacy may allow a higher success-rate in critical end points of clinical benefit.


Subject(s)
Neoplasms/pathology , Neoplasms/therapy , Xenograft Model Antitumor Assays/methods , Adult , Aged , Animals , Cohort Studies , Female , Humans , Male , Mice , Middle Aged , Neoplasm Transplantation/methods , Neoplasms/genetics , Exome Sequencing
4.
ESMO Open ; 9(7): 103487, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38943735

ABSTRACT

BACKGROUND: Locally advanced (unresectable) or metastatic dedifferentiated liposarcoma (DDLPS) is a common presentation of liposarcoma. Despite established diagnostic and treatment guidelines for DDLPS, critical clinical gaps remain driven by diagnostic challenges, symptom burden and the lack of targeted, safe and effective treatments. The objective of this study was to gather expert opinions from Europe and the United States on the management, unmet needs and expectations for clinical trial design as well as the value of progression-free survival (PFS) in this disease. Other aims included raising awareness and educate key stakeholders across healthcare systems. MATERIALS AND METHODS: An international panel of 12 sarcoma key opinion leaders (KOLs) was recruited. The study consisted of two rounds of surveys with pre-defined statements. Experts scored each statement on a 9-point Likert scale. Consensus agreement was defined as ≥75% of experts scoring a statement with ≥7. Revised statements were discussed in a consensus meeting. RESULTS: Consensus was reached on 43 of 55 pre-defined statements across disease burden, treatment paradigm, unmet needs, value of PFS and its association with overall survival (OS), and cross-over trial design. Twelve statements were deprioritised or merged with other statements. There were no statements where experts disagreed. CONCLUSION: This study constitutes the first international Delphi panel on DDLPS. It aimed to explore KOL perception of the disease burden and unmet need in DDLPS, the value of PFS, and its potential translation to OS benefit, as well as the relevance of a cross-over trial design for DDLPS therapies. Results indicate an alignment across Europe and the United States regarding DDLPS management, unmet needs, and expectations for clinical trials. Raising awareness of critical clinical gaps in relation to DDLPS can contribute to improving patient outcomes and supporting the development of innovative treatments.

5.
Br J Cancer ; 109(7): 1717-24, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24022187

ABSTRACT

BACKGROUND: This retrospective pooled analysis assessed the effect of age on the efficacy and safety of trabectedin in young and elderly patients with recurrent advanced soft tissue sarcoma (STS). METHODS: Data from 350 adults with STS treated in five phase II trials with trabectedin were divided in the younger (<60 years; n=267) and the older cohort (≥60 years; n=83). RESULTS: The response rate did not differ with age (younger: 10.1% vs elderly 9.6%). No significant differences were found in median progression-free survival (PFS) in younger (2.5 months) and older (3.7 months) cohort with a comparable PFS rates at 3 (45.1% vs 55.1%) and 6 months (29.5% vs 36.4%). Similar median overall survival was observed in both cohorts (13.0 vs 14.0 months). Reversible neutropenia and aspartate aminotransferase/alanine aminotransferase elevation were the most common abnormalities. A higher incidence of grade 3/4 neutropenia (43.6% vs 60.2%) and fatigue (6.3% vs 14.4%) was observed in older patients. In 24 patients aged ≥70 years, no significant differences in efficacy or safety outcomes were found. CONCLUSION: This analysis demonstrated that trabectedin is a feasible treatment in young and elderly patients with STS, with meaningful clinical benefits and an acceptable safety profile, essential in palliative treatment of elderly patients.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Dioxoles/therapeutic use , Sarcoma/drug therapy , Sarcoma/mortality , Tetrahydroisoquinolines/therapeutic use , Adult , Age Factors , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/adverse effects , Dioxoles/adverse effects , Disease-Free Survival , Female , Humans , Male , Middle Aged , Retrospective Studies , Tetrahydroisoquinolines/adverse effects , Trabectedin , Treatment Outcome , Young Adult
6.
Ann Oncol ; 24(7): 1924-1930, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23493135

ABSTRACT

BACKGROUND: There are no data regarding the management of advanced soft-tissue sarcoma (STS) in elderly patients. PATIENTS AND METHODS: We retrospectively reviewed the charts of patients ≥75 years old diagnosed with metastatic or unresectable STS between 1991 and 2011 in 11 French and American centers. RESULTS: The study included 361 patients. Of these, 223 patients (62%) received systemic therapy, whereas 123 patients (34%) were managed with best supportive care (BSC) only. Patients who received BSC were more likely to be ≥80 years, with performance status (PS) ≥ 2, Charlson comorbidity score ≥ 10, and metastatic disease. The median progression-free survival of patients treated with systemic therapy was 4 months (95% CI: 2.9-5.1). Thirty-six patients (16%) stopped chemotherapy because of toxicity. Median overall survival (OS) of patients managed with specific therapy was 10.9 months (95% CI: 8.3-13.5) versus 5.3 months (95% CI: 3.6-7.1) for patients managed with BSC (P = 0.001). On multivariate analysis, age ≥ 80 years, PS ≥ 2, and number of metastatic sites were the only independent factors associated with OS. CONCLUSION: A high proportion of elderly patients with advanced STS were denied chemotherapy. Further efforts are needed to define better the optimal care for fit and unfit elderly patients with STS.


Subject(s)
Anthracyclines/therapeutic use , Antineoplastic Agents/therapeutic use , Sarcoma/drug therapy , Soft Tissue Neoplasms/drug therapy , Aged , Aged, 80 and over , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Palliative Care , Retrospective Studies , Sarcoma/mortality , Sarcoma/secondary , Soft Tissue Neoplasms/mortality , Soft Tissue Neoplasms/pathology , Treatment Outcome
7.
Ann Oncol ; 24(1): 252-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22898035

ABSTRACT

BACKGROUND: HSP90 inhibition leads to proteosomal degradation of activated KIT and has in vitro activity against gastrointestinal stromal tumors (GIST). BIIB021 is an oral non-ansamycin HSP90 inhibitor. We carried out a phase II study of BIIB021 in patients with GIST refractory to imatinib and sunitinib. PATIENTS AND METHODS: The primary end-point was metabolic partial response (mPR) as assessed by fluorodeoxyglucose positron emission tomography (FDG-PET). The secondary end-points were pharmacokinetic assessments of BIIB021 and pharmacodynamic assessments of HSP70. Twenty-three patients were treated on two schedules: 12 pts received 600 mg twice a week (BIW) and 11 patients received 400 mg three times a week (TIW). All had prior imatinib and sunitinib but stopped>14 days before starting BIIB021. RESULTS: The median age was 59 years (33-88 years), 61% male, 44% Eastern Cooperative Oncology Group 1 (ECOG1). The best response was PR by FDG-PET for five patients (3/12 at 600 mg BIW and 2/9 at 400 TIW) for an overall response rate of 22%. The response duration was 25-138 days. Adverse events (AEs) were mild to moderate. The mean Cmax was 1.5 µmol and the mean AUC was 2.9 µmol h. Cmax>1.5 µmol was associated with a decrease in standardized uptake value (SUVmax). HSP70 increased substantially following treatment. CONCLUSIONS: This study met its primary end-point. BIIB021 leads to objective responses in refractory GIST patients. Pharmacodynamic studies confirmed HSP90 inhibition. Further evaluation of BIIB021 in GIST is warranted.


Subject(s)
Adenine/analogs & derivatives , Gastrointestinal Stromal Tumors/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Pyridines/therapeutic use , Adenine/adverse effects , Adenine/pharmacokinetics , Adenine/pharmacology , Adenine/therapeutic use , Adult , Aged , Aged, 80 and over , Female , Fluorodeoxyglucose F18 , Gastrointestinal Stromal Tumors/diagnostic imaging , Humans , Male , Middle Aged , Positron-Emission Tomography , Pyridines/adverse effects , Pyridines/pharmacokinetics , Pyridines/pharmacology , Treatment Outcome
8.
Ann Oncol ; 24(2): 537-542, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23117071

ABSTRACT

BACKGROUND: The growth modulation index (GMI) is the ratio of time to progression with the nth line (TTP(n)) of therapy to the TTP(n)(-1) with the n-1th line. GMI >1.33 is considered as a sign of activity in phase II trials. PATIENTS AND METHODS: This retrospective analysis evaluated the concordance between the GMI and the efficacy outcomes in 279 patients with advanced soft tissue sarcoma (ASTS) treated with trabectedin 1.5 mg/m² (24-h infusion every 3 weeks) in four phase II trials. RESULTS: One hundred and forty-two (51%) patients received one prior line and 137 ≥ 2 lines. The median TTP(n) was 2.8 months (range 0.2-26.8), whereas the median TTP(n)(-1) was 4.0 months (0.3-79.5). The median GMI was 0.6 (0.0-14.4). Overall, 177 patients (63%) had a GMI <1; 21 (8%) a GMI equal to 1-1.33 and 81 (29%) a GMI >1.33, which correlated with the median overall survival in those patients (9.1, 13.9 and 23.8 months, respectively, P = 0.0005). A high concordance rate between the GMI and response rate (P < 0.0001) and progression-free survival (PFS, P < 0.0001) was observed. Good performance status (PS) was the only factor associated with GMI >1.33 (PS = 0; P < 0.04). CONCLUSIONS: A high GMI was associated with favorable efficacy outcomes in patients treated with trabectedin. Further research is needed to assess GMI as an indicator in this setting.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Cell Proliferation/drug effects , Dioxoles/therapeutic use , Sarcoma/drug therapy , Tetrahydroisoquinolines/therapeutic use , Adult , Aged , Aged, 80 and over , Antibiotics, Antineoplastic/therapeutic use , Disease-Free Survival , Doxorubicin/therapeutic use , Female , Humans , Male , Middle Aged , Retrospective Studies , Salvage Therapy , Sarcoma/metabolism , Sarcoma/mortality , Sarcoma/pathology , Trabectedin , Treatment Outcome , Young Adult
9.
Ann Oncol ; 24(11): 2916-22, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24099780

ABSTRACT

BACKGROUND: There are limited data about the role of chemotherapy in patients with advanced chondrosarcomas. METHODS: The medical charts of 180 patients with advanced chondrosarcomas having received chemotherapy in 15 participating institutions between 1988 and 2011 were reviewed. RESULTS: Median age was 52 years. Sixty-three percent of patients had conventional chondrosarcoma and 88% had metastatic disease. Combination chemotherapy was delivered in 98 cases (54.5%). One hundred and thirty-one patients (73%) received an anthracycline-containing regimen. Using RECIST, the objective response rate was significantly different according to histological subtype, being 31% for mesenchymal chondrosarcoma, 20.5% for dedifferentiated chondrosarcoma, 11.5% for conventional chondrosarcoma and 0% for clear-cell chondrosarcoma (P = 0.04). Median progression-free survival (PFS) was 4.7 months [95% confidence interval (CI) 3-6.5]. Performance status (PS) ≥2, number of metastatic sites ≥1 and single-agent regimen were independently associated with poor PFS. Median overall survival (OS) was 18 months (95% CI 14.5-21.6). PS, number of metastatic sites and palliative surgery were independently associated with OS. CONCLUSIONS: Conventional chemotherapy have very limited efficacy in patients with advanced chondrosarcoma, the highest benefit being observed in mesenchymal and dedifferentiated chondrosarcoma. These data should be used as a reference for response and outcome in the assessment of investigational drugs in advanced chondrosarcoma.


Subject(s)
Anthracyclines/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Chondrosarcoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Chondrosarcoma/mortality , Chondrosarcoma/pathology , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis
10.
Ann Oncol ; 23(5): 1335-1340, 2012 May.
Article in English | MEDLINE | ID: mdl-21994214

ABSTRACT

BACKGROUND: Gastrointestinal stromal tumors (GISTs) and desmoid tumors (DTs) are two rare mesenchymal tumor. Anecdotal reports of individuals with both diseases led us to make the hypothesis that the association is a nonrandom event as the probability would be extremely low to observe such cases if they were independent events. PATIENTS AND METHODS: We evaluated the existence of patients with GIST and DT in a large multicenter cohort at 10 institutions in the United States, Australia and Europe. Data on gender, age at diagnosis, KIT, PDGFRA, CTNNB1 mutation status and follow-up time after diagnosis were collected. RESULTS: We identified 28 patients diagnosed with both tumors. DT was diagnosed after GIST in 75% of patients and concomitantly in 21%. In only one case (4%), GIST was diagnosed after DT. KIT or PDGFRA mutations were detected in 12 of 14 GIST, 9 in KIT exon 11, 2 in KIT exon 9 and 1 in PDGFRA. CONCLUSION: A statistical analysis of these 28 cases suggests a nonrandom association between GIST and DT. Further studies may be able to elucidate the underlying biology responsible for this association.


Subject(s)
Fibromatosis, Aggressive/complications , Fibromatosis, Aggressive/epidemiology , Gastrointestinal Stromal Tumors/complications , Gastrointestinal Stromal Tumors/epidemiology , Adult , Aged , Aged, 80 and over , Australia/epidemiology , Cohort Studies , Europe/epidemiology , Female , Follow-Up Studies , Humans , Incidence , Male , Middle Aged , Retrospective Studies , United States/epidemiology
11.
Ann Oncol ; 23(6): 1601-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22039081

ABSTRACT

BACKGROUND: Data regarding the role of systemic therapy in patients with advanced well-differentiated/dedifferentiated liposarcomas (WDLPS/DDLPS) are limited. METHODS: From 2000 to 2010, 208 patients with advanced WDLPS/DDLPS received chemotherapy in 11 participating institutions. Clinical and pathological data were collected by reviewing medical records. RESULTS: Median age was 63 years (range 32-84). Combination chemotherapy was delivered in 85 cases (41%) and single agent in 123 cases (59%), respectively. One hundred and seventy-one patients (82%) received an anthracycline-containing regimen. Using RECIST, objective response was observed in 21 patients (12%), all treated with anthracyclines. Median progression-free survival (PFS) was 4.6 months [95% confidence interval (CI) 3.3-5.9]. On multivariate analysis, age and performance status (PS) were the sole factors significantly associated with poor PFS. Median overall survival (OS) was 15.2 months (95% CI 11.8 -18.7). On multivariate analysis, grade and PS were the sole factors significantly associated with OS. CONCLUSIONS: Chemotherapy was associated with clinical benefit in 46% of patients with advanced WDLPS/DDLPS. OS remains poor, even though visceral metastatic disease is less frequent than in other sarcomas.


Subject(s)
Anthracyclines/therapeutic use , Antineoplastic Agents/therapeutic use , Liposarcoma/drug therapy , Retroperitoneal Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Liposarcoma/mortality , Liposarcoma/secondary , Male , Middle Aged , Multivariate Analysis , Neoplasm Grading , Prognosis , Proportional Hazards Models , Retroperitoneal Neoplasms/mortality , Retroperitoneal Neoplasms/pathology , Retrospective Studies , Treatment Outcome
12.
Crit Rev Oncol Hematol ; 174: 103685, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35460913

ABSTRACT

Bone sarcoma are infrequent diseases, representing < 0.2% of all adult neoplasms. A multidisciplinary management within reference centers for sarcoma, with discussion of the diagnostic and therapeutic strategies within an expert multidisciplinary tumour board, is essential for these patients, given its heterogeneity and low frequency. This approach leads to an improvement in patient's outcome, as demonstrated in several studies. The Sarcoma European Latin-American Network (SELNET), aims to improve clinical outcome in sarcoma care, with a special focus in Latin-American countries. These Clinical Practice Guidelines (CPG) have been developed and agreed by a multidisciplinary expert group (including medical and radiation oncologist, surgical oncologist, orthopaedic surgeons, radiologist, pathologist, molecular biologist and representatives of patients advocacy groups) of the SELNET consortium, and are conceived to provide the standard approach to diagnosis, treatment and follow-up of bone sarcoma patients in the Latin-American context.


Subject(s)
Bone Neoplasms , Osteosarcoma , Sarcoma , Soft Tissue Neoplasms , Adult , Bone Neoplasms/diagnosis , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Humans , Osteosarcoma/diagnosis , Osteosarcoma/pathology , Osteosarcoma/therapy , Practice Guidelines as Topic , Sarcoma/diagnosis , Sarcoma/pathology , Sarcoma/therapy , Soft Tissue Neoplasms/pathology
13.
Nat Med ; 6(9): 991-7, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973318

ABSTRACT

We explored the role of hypocretins in human narcolepsy through histopathology of six narcolepsy brains and mutation screening of Hcrt, Hcrtr1 and Hcrtr2 in 74 patients of various human leukocyte antigen and family history status. One Hcrt mutation, impairing peptide trafficking and processing, was found in a single case with early onset narcolepsy. In situ hybridization of the perifornical area and peptide radioimmunoassays indicated global loss of hypocretins, without gliosis or signs of inflammation in all human cases examined. Although hypocretin loci do not contribute significantly to genetic predisposition, most cases of human narcolepsy are associated with a deficient hypocretin system.


Subject(s)
Brain Chemistry/genetics , Carrier Proteins , Intracellular Signaling Peptides and Proteins , Mutation , Narcolepsy/genetics , Neuropeptides/genetics , Receptors, Neuropeptide/genetics , Adult , Age of Onset , Aged , Aged, 80 and over , Cerebral Cortex/chemistry , Female , Genetic Testing , Humans , Hypothalamus/chemistry , Hypothalamus/cytology , Male , Middle Aged , Molecular Sequence Data , Neuropeptides/analysis , Neurotransmitter Agents/genetics , Orexin Receptors , Orexins , Pons/chemistry , Protein Processing, Post-Translational , Receptors, G-Protein-Coupled , Tissue Distribution , White People
14.
J Exp Med ; 170(3): 901-12, 1989 Sep 01.
Article in English | MEDLINE | ID: mdl-2475576

ABSTRACT

T cell hybridomas were established by fusing a CD8+ V beta 8.1+ CTL clone and a CD4+ V beta 8.1+ helper T lymphocyte (HTL) clone to the thymoma cell line BW5147. In contrast to the HTL x BW hybridomas, which retain the same antigen specificity as the original T cell clone, the CTL x BW hybridomas lost the class I MHC-restricted antigen response but acquired a new specificity to Mlsa antigen. Mlsa reactivity of CTL x BW hybridomas was shown to be mediated by the CTL TCR as assayed by inhibition using an anticlonotypic antibody to the CTL clone. Since hybridomas established with BW5147 lose CD8 expression, we have introduced the CD8 molecule into CTL x BW5147 hybridomas by gene transfection. The CD8+ V beta 8.1+ hybridoma was no longer capable of reacting to Mlsa antigen but exhibited the same antigen specificity as the parental CTL clone. Furthermore, the presence of the transfected CD8 molecule in the HTL x BW hybridomas was found to be inhibitory to class II MHC-restricted antigen reactivity. These results demonstrate that, besides its role in increasing the overall avidity of T cell-class I MHC/antigen interaction, the CD8 molecule inhibits T cell-class II MHC gene product/antigen interaction. This negative effect of the CD8 molecule on a class II MHC-restricted response may account for the failure of CD8+ T cells using either V beta 8.1 or V beta 6, which impart reactivity to the Mlsa antigen on CD4+ T cells, to respond to the Mlsa antigen.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/physiology , Histocompatibility Antigens Class II/immunology , T-Lymphocytes/immunology , Animals , Antigens, Differentiation, T-Lymphocyte/analysis , CD8 Antigens , Epitopes/analysis , Female , Hybridomas/immunology , Male , Mice , Mice, Inbred Strains , Minor Histocompatibility Loci , Phenotype , Receptors, Antigen, T-Cell/analysis , Receptors, Antigen, T-Cell/physiology
15.
J Exp Med ; 177(3): 691-8, 1993 Mar 01.
Article in English | MEDLINE | ID: mdl-8436907

ABSTRACT

Glucocorticoids are effective repressors of major histocompatibility complex (MHC) class II gene expression. The repression occurs in B cells, which constitutively express MHC class II, as well as in macrophages, which only express MHC class II after the cells are treated with interferon gamma. For the MHC class II gene IA beta, this negative regulation has been linked to the X box DNA sequence, located with the IA beta promoter. The addition of the glucocorticoid receptor was shown to inhibit the DNA binding of the X box DNA binding protein to the X box. The DNA binding of two other DNA binding proteins that recognize elements within this promoter was unaffected by the addition of glucocorticoid receptor. It is likely that the repression of IA beta gene expression by glucocorticoids occurs because the X box DNA binding protein is prevented from binding to the DNA and activating transcription.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Gene Expression/drug effects , Genes, MHC Class II/genetics , Glucocorticoids/pharmacology , Receptors, Glucocorticoid/physiology , Animals , Base Sequence , Cell Line , Electrophoresis, Polyacrylamide Gel , Gene Expression/genetics , Interferon-gamma/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/physiology , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , Transcription, Genetic/drug effects , Transfection
16.
J Exp Med ; 186(7): 1119-28, 1997 Oct 06.
Article in English | MEDLINE | ID: mdl-9314560

ABSTRACT

The induction of T cell anergy in vivo is thought to result from antigen recognition in the absence of co-stimulation and inflammation, and is associated with a block in T cell proliferation and Th1 differentiation. Here we have examined the role of interleukin (IL)-12, a potent inducer of Th1 responses, in regulating this process. T cell tolerance was induced by the administration of protein antigen without adjuvant in normal mice, and in recipients of adoptively transferred T cells from T cell receptor transgenic mice. The administration of IL-12 at the time of tolerance induction stimulates Th1 differentiation, but does not promote antigen-specific T cell proliferation. Conversely, inhibiting CTLA-4 engagement during anergy induction reverses the block in T cell proliferation, but does not promote full Th1 differentiation. T cells exposed to tolerogenic antigen in the presence of both IL-12 and anti-CTLA-4 antibody are not anergized, and behave identically to T cells which have encountered immunogenic antigen. These results suggest that two processes contribute to the induction of anergy in vivo; CTLA-4 engagement, which leads to a block in the ability of T cells to proliferate to antigen, and the absence of a prototypic inflammatory cytokine, IL-12, which prevents the differentiation of T cells into Th1 effector cells. The combination of IL-12 and anti-CTLA-4 antibody is sufficient to convert a normally tolerogenic stimulus to an immunogenic one.


Subject(s)
Antigens, Differentiation/immunology , Clonal Anergy , Immune Tolerance , Immunoconjugates , Interleukin-12/immunology , T-Lymphocytes/immunology , Th1 Cells/immunology , Abatacept , Adoptive Transfer , Animals , Antibodies/immunology , Antigens, CD , Antigens, Differentiation/metabolism , Autoimmune Diseases/immunology , CTLA-4 Antigen , Cell Differentiation , Flow Cytometry , Inflammation/immunology , Interferon-gamma/metabolism , Interleukin-12/pharmacology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
17.
J Exp Med ; 184(1): 61-9, 1996 Jul 01.
Article in English | MEDLINE | ID: mdl-8691150

ABSTRACT

PU.1 is a tissue-specific transcription factor that is expressed in cells of the hematopoietic lineage including macrophages, granulocytes, and B lymphocytes. Bone marrow-derived macrophages transfected with an antisense PU.1 expression construct or treated with antisense oligonucleotides showed a decrease in proliferation compared with controls. In contrast, bone marrow macrophages transfected with a sense PU.1 expression construct displayed enhanced macrophage colony-stimulating factor (M-CSF)-dependent proliferation. Interestingly, there was no effect of sense or antisense constructs of PU.1 on the proliferation of the M-CSF-independent cell line, suggesting that the response was M-CSF dependent. This was further supported by the finding that macrophages transfected with a sense or an antisense PU.1 construct showed, respectively, an increased or a reduced level of surface expression of receptors for M-CSF. The enhancement of proliferation seems to be selective for PU.1, since transfections with several other members of the ets family, including ets-2 and fli-1, had no effect. Various mutants of PU.1 were also tested for their ability to affect macrophage proliferation. A reduction in macrophage proliferation was found when cells were transfected with a construct in which the DNA-binding domain of PU.1 was expressed. The PEST (proline-, glutamic acid-, serine-, and threonine-rich region) sequence of the PU.1 protein, which is an important domain for protein-protein interactions in B cells, was found to have no influence on PU.1-enhanced macrophage proliferation when an expression construct containing PU.1 minus the PEST domain was transfected into bone marrow-derived macrophages. In vivo, PU.1 is phosphorylated on several serine residues. The transfection of plasmids containing PU.1 with mutations at each of five serines showed that only positions 41 and 45 are critical for enhanced macrophage proliferation. We conclude that PU.1 is necessary for the M-CSF-dependent proliferation of macrophages. One of the proliferation-relevant targets of this transcription factor could be the M-CSF receptor.


Subject(s)
Macrophage Activation , Macrophages/cytology , Proto-Oncogene Proteins/physiology , Trans-Activators , Animals , Base Sequence , Bone Marrow Cells , DNA, Antisense/chemistry , DNA-Binding Proteins/physiology , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Mice, Inbred DBA , Molecular Sequence Data
18.
J Exp Med ; 180(1): 233-40, 1994 Jul 01.
Article in English | MEDLINE | ID: mdl-8006583

ABSTRACT

Class II major histocompatibility complex (MHC) genes encode for alpha/beta chain pairs that are constitutively expressed principally on mature B cells and dendritic cells in mice. These gene products are easily induced on macrophages with cytokines, and may also aberrantly appear on the surface of epithelium during immune injury. The appearance of class II determinants in parenchymal tissue potentially renders these somatic cells capable of antigen presentation to circulating CD4+ T lymphocytes, and their absence may be protective for normal tissues expressing self-antigens. The low surface class II expression observed on parenchymal cells generally correlates with low levels of mRNA, suggesting that transcription rate is a major element in class II regulation. To understand the transcriptional mechanism maintaining low basal surface expression of class II in somatic cells, we transiently transfected mini-gene reporter constructs to study the regulation of the murine A beta promoter in a cultured renal epithelial cell line. We describe here a negative cis-acting regulatory region located between -552 and -489 bp upstream of the A beta cap site that silences the transcriptional activity of the A beta promoter in epithelial cells in an orientation-dependent manner, and is also able to silence a heterologous promoter. This region is not active in class II-expressing B cells (BAL-17) in culture, but is functional in two other murine class II-negative cell lines, fibroblasts and thymoma T cells. Using competition electrophoretic mobility shift assays, we have localized the core protein binding site within this region to an 8-10-bp response element, designated A beta NRE, at -543 to -534 bp. A nuclear extract from BAL-17 cells does not bind to this element. Mutation of this site abrogates the transcriptional silencing activity of the region. We conclude that the transcription of class II-A beta in parenchymal cells, and some lymphocytes, can be actively repressed by an upstream silencing element.


Subject(s)
Gene Expression Regulation , Genes, MHC Class II , Genes, Regulator , Animals , Base Sequence , Binding Sites , Cell Line , Epithelium/metabolism , Kidney Tubules/metabolism , Mice , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Messenger/analysis , Transcription, Genetic
19.
Science ; 209(4463): 1366-9, 1980 Sep 19.
Article in English | MEDLINE | ID: mdl-6774416

ABSTRACT

Two types of immature B cells, namely fetal liver hybridomas and the leukemic cell line 70Z/3, both of which have cytoplasmic mu chains but no light chains, were examined for DNA rearrangements of their light chain and heavy chain immunoglobulin genes. In the fetal liver hybridomas, which were constructed from fetal liver cells and a tumor cell, no light chain gene rearrangement was observed, whereas in the 70Z/3 cell line a kappa light chain rearrangement probably occurred. The results suggest that, although the lack of light chain synthesis can be due to a lack of gene rearrangement, there may also be transcriptional regulation, which may also be important for the expression of light chain immunoglobulins in immature B cells.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/genetics , Immunoglobulin kappa-Chains/genetics , Immunoglobulin mu-Chains/genetics , Leukemia, Experimental/immunology , Liver/embryology , Animals , Genes , Hybrid Cells/immunology , Immunoglobulin Constant Regions/genetics , Immunoglobulin Variable Region/genetics , Mice , Recombination, Genetic , Transcription, Genetic
20.
Science ; 261(5117): 82-6, 1993 Jul 02.
Article in English | MEDLINE | ID: mdl-8316859

ABSTRACT

The enhancer for the immunoglobulin mu heavy chain gene (IgH) activates a heterologous gene at the pre-B cell stage of B lymphocyte differentiation. A lymphoid-specific element, microB, is necessary for enhancer function in pre-B cells. A microB binding protein is encoded by the PU.1/Spi-1 proto-oncogene. Another sequence element, microA, was identified in the mu enhancer that binds the product of the ets-1 proto-oncogene. The microA motif was required for microB-dependent enhancer activity, which suggests that a minimal B cell-specific enhancer is composed of both the PU.1 and Ets-1 binding sites. Co-expression of both PU.1 and Ets-1 in nonlymphoid cells trans-activated reporter plasmids that contained the minimal mu enhancer. These results implicate two members of the Ets family in the activation of IgH gene expression.


Subject(s)
B-Lymphocytes/metabolism , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Immunoglobulin mu-Chains/genetics , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , Animals , B-Lymphocytes/cytology , Base Sequence , Binding Sites , Cell Differentiation , Cell Line , DNA-Binding Proteins/metabolism , Female , Genes, Immunoglobulin , Humans , Molecular Sequence Data , Mutation , Proto-Oncogene Mas , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , Retroviridae Proteins, Oncogenic , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL