Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
JAMA Oncol ; 3(8): 1094-1101, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28426845

ABSTRACT

IMPORTANCE: Glioblastoma is an incurable tumor, and the therapeutic options for patients are limited. OBJECTIVE: To determine whether the systemic administration of HER2-specific chimeric antigen receptor (CAR)-modified virus-specific T cells (VSTs) is safe and whether these cells have antiglioblastoma activity. DESIGN, SETTING, AND PARTICIPANTS: In this open-label phase 1 dose-escalation study conducted at Baylor College of Medicine, Houston Methodist Hospital, and Texas Children's Hospital, patients with progressive HER2-positive glioblastoma were enrolled between July 25, 2011, and April 21, 2014. The duration of follow-up was 10 weeks to 29 months (median, 8 months). INTERVENTIONS: Monotherapy with autologous VSTs specific for cytomegalovirus, Epstein-Barr virus, or adenovirus and genetically modified to express HER2-CARs with a CD28.ζ-signaling endodomain (HER2-CAR VSTs). MAIN OUTCOMES AND MEASURES: Primary end points were feasibility and safety. The key secondary end points were T-cell persistence and their antiglioblastoma activity. RESULTS: A total of 17 patients (8 females and 9 males; 10 patients ≥18 years [median age, 60 years; range, 30-69 years] and 7 patients <18 years [median age, 14 years; range, 10-17 years]) with progressive HER2-positive glioblastoma received 1 or more infusions of autologous HER2-CAR VSTs (1 × 106/m2 to 1 × 108/m2) without prior lymphodepletion. Infusions were well tolerated, with no dose-limiting toxic effects. HER2-CAR VSTs were detected in the peripheral blood for up to 12 months after the infusion by quantitative real-time polymerase chain reaction. Of 16 evaluable patients (9 adults and 7 children), 1 had a partial response for more than 9 months, 7 had stable disease for 8 weeks to 29 months, and 8 progressed after T-cell infusion. Three patients with stable disease are alive without any evidence of progression during 24 to 29 months of follow-up. For the entire study cohort, median overall survival was 11.1 months (95% CI, 4.1-27.2 months) from the first T-cell infusion and 24.5 months (95% CI, 17.2-34.6 months) from diagnosis. CONCLUSIONS AND RELEVANCE: Infusion of autologous HER2-CAR VSTs is safe and can be associated with clinical benefit for patients with progressive glioblastoma. Further evaluation of HER2-CAR VSTs in a phase 2b study is warranted as a single agent or in combination with other immunomodulatory approaches for glioblastoma.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , T-Lymphocytes/transplantation , Adenoviridae/immunology , Adolescent , Adult , Aged , Child , Cytomegalovirus/immunology , Female , Herpesvirus 4, Human/immunology , Humans , Male , Middle Aged , Receptor, ErbB-2 , Receptors, Antigen, T-Cell , T-Lymphocytes/immunology , Treatment Outcome
2.
Front Neurol ; 6: 234, 2015.
Article in English | MEDLINE | ID: mdl-26617566

ABSTRACT

Gait abnormalities, such as shuffling steps, start hesitation, and freezing, are common and often incapacitating symptoms of Parkinson's disease (PD) and other parkinsonian disorders. Pharmacological and surgical approaches have only limited efficacy in treating these gait disorders. Rhythmic auditory stimulation (RAS), such as playing marching music and dance therapy, has been shown to be a safe, inexpensive, and an effective method in improving gait in PD patients. However, RAS that adapts to patients' movements may be more effective than rigid, fixed-tempo RAS used in most studies. In addition to auditory cueing, immersive virtual reality technologies that utilize interactive computer-generated systems through wearable devices are increasingly used for improving brain-body interaction and sensory-motor integration. Using multisensory cues, these therapies may be particularly suitable for the treatment of parkinsonian freezing and other gait disorders. In this review, we examine the affected neurological circuits underlying gait and temporal processing in PD patients and summarize the current studies demonstrating the effects of RAS on improving these gait deficits.

3.
J Neurooncol ; 125(2): 307-15, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26341370

ABSTRACT

While the 5-year overall survival is better in pediatric than in adult patients diagnosed with glioblastoma (GBM), outcomes in children remain very poor. Understanding the mechanisms of tumorigenesis and tumor propagation can identify therapeutic targets to improve these outcomes. Human cytomegalovirus (CMV) proteins and nucleic acids are present in the majority of adult GBM. Indeed, CMV is emerging as a potential glioma-associated target for anti-CMV agents and cellular therapeutics. Furthermore, CMV appears to contribute to GBM's malignant phenotype, although its role in tumorigenesis is less certain. In this cohort of 25 serially diagnosed pediatric GBMs, the largest described cohort to date, we used immunohistochemical staining and in situ hybridization to show the presence of CMV antigens pp65 and IE1-72 as well as CMV nucleic acids, respectively. Our cohort indicated either CMV antigen pp65 or IE1-72 was present in approximately 67 % of pediatric GBM samples. The majority of samples stained positive for either CMV antigen showing a cytoplasmic pattern in 25-50 % of cells within the sample at a moderate intensity, while a few samples showed nuclear staining and higher grade/intensity. Of 16 samples where in situ hybridization was performed, 13 (81 %) showed specific staining using a CMV genome specific probe cocktail. ISH positive samples showed high concordance with being pp65 or IE1-72 positive. These findings, paired with the association of CMV expression with poor prognosis and overall survival, indicate the need to further investigate how these antigens are promoting tumor growth and preventing cell death. Also, the expression of these antigens in a majority of tumor tissues should be considered for immunotherapeutic targets in cases of pediatric GBM.


Subject(s)
Brain Neoplasms/diagnosis , Glioblastoma/diagnosis , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Adolescent , Brain Neoplasms/virology , Child , Child, Preschool , Cohort Studies , Cytomegalovirus/metabolism , Cytomegalovirus/pathogenicity , Female , Glioblastoma/virology , Humans , Infant , Male
4.
J Clin Oncol ; 33(15): 1688-96, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25800760

ABSTRACT

PURPOSE: The outcome for patients with metastatic or recurrent sarcoma remains poor. Adoptive therapy with tumor-directed T cells is an attractive therapeutic option but has never been evaluated in sarcoma. PATIENTS AND METHODS: We conducted a phase I/II clinical study in which patients with recurrent/refractory human epidermal growth factor receptor 2 (HER2) -positive sarcoma received escalating doses (1 × 10(4)/m(2) to 1 × 10(8)/m(2)) of T cells expressing an HER2-specific chimeric antigen receptor with a CD28.ζ signaling domain (HER2-CAR T cells). RESULTS: We enrolled 19 patients with HER2-positive tumors (16 osteosarcomas, one Ewing sarcoma, one primitive neuroectodermal tumor, and one desmoplastic small round cell tumor). HER2-CAR T-cell infusions were well tolerated with no dose-limiting toxicity. At dose level 3 (1 × 10(5)/m(2)) and above, we detected HER2-CAR T cells 3 hours after infusion by quantitative polymerase chain reaction in 14 of 16 patients. HER2-CAR T cells persisted for at least 6 weeks in seven of the nine evaluable patients who received greater than 1 × 10(6)/m(2) HER2-CAR T cells (P = .005). HER2-CAR T cells were detected at tumor sites of two of two patients examined. Of 17 evaluable patients, four had stable disease for 12 weeks to 14 months. Three of these patients had their tumor removed, with one showing ≥ 90% necrosis. The median overall survival of all 19 infused patients was 10.3 months (range, 5.1 to 29.1 months). CONCLUSION: This first evaluation of the safety and efficacy of HER2-CAR T cells in patients with cancer shows the cells can persist for 6 weeks without evident toxicities, setting the stage for studies that combine HER2-CAR T cells with other immunomodulatory approaches to enhance their expansion and persistence.


Subject(s)
Bone Neoplasms/therapy , Immunotherapy/methods , Receptor, ErbB-2/metabolism , Sarcoma/therapy , T-Lymphocytes/immunology , Adolescent , Adult , Bone Neoplasms/metabolism , Child , Female , Humans , Kaplan-Meier Estimate , Magnetic Resonance Imaging , Male , Maximum Tolerated Dose , Neoplasm Metastasis , Neuroectodermal Tumors/metabolism , Neuroectodermal Tumors/therapy , Osteosarcoma/metabolism , Osteosarcoma/therapy , Positron-Emission Tomography , Receptor, ErbB-2/genetics , Receptors, Antigen, T-Cell/chemistry , Recurrence , Sarcoma/metabolism , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/therapy , Tomography, X-Ray Computed , Treatment Outcome , Young Adult
5.
Mol Ther ; 21(11): 2087-101, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23939024

ABSTRACT

Preclinical and early clinical studies have demonstrated that chimeric antigen receptor (CAR)-redirected T cells are highly promising in cancer therapy. We observed that targeting HER2 in a glioblastoma (GBM) cell line results in the emergence of HER2-null tumor cells that maintain the expression of nontargeted tumor-associated antigens. Combinational targeting of these tumor-associated antigens could therefore offset this escape mechanism. We studied the single-cell coexpression patterns of HER2, IL-13Rα2, and EphA2 in primary GBM samples using multicolor flow cytometry and immunofluorescence, and applied a binomial routine to the permutations of antigen expression and the related odds of complete tumor elimination. This mathematical model demonstrated that cotargeting HER2 and IL-13Rα2 could maximally expand the therapeutic reach of the T cell product in all primary tumors studied. Targeting a third antigen did not predict an added advantage in the tumor cohort studied. We therefore generated bispecific T cell products from healthy donors and from GBM patients by pooling T cells individually expressing HER2 and IL-13Rα2-specific CARs and by making individual T cells to coexpress both molecules. Both HER2/IL-13Rα2-bispecific T cell products offset antigen escape, producing enhanced effector activity in vitro immunoassays (against autologous glioma cells in the case of GBM patient products) and in an orthotopic xenogeneic murine model. Further, T cells coexpressing HER2 and IL-13Rα2-CARs exhibited accentuated yet antigen-dependent downstream signaling and a particularly enhanced antitumor activity.


Subject(s)
Adoptive Transfer , Antigens, Neoplasm/metabolism , Glioblastoma/therapy , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Line, Tumor , Combined Modality Therapy , Glioblastoma/immunology , Glioblastoma/pathology , HEK293 Cells , Humans , Interleukin-13 Receptor alpha2 Subunit/genetics , Interleukin-13 Receptor alpha2 Subunit/immunology , Interleukin-13 Receptor alpha2 Subunit/metabolism , Mice , Mice, SCID , Models, Biological , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured , Tumor Escape , Xenograft Model Antitumor Assays
6.
J Immunother ; 35(2): 159-68, 2012.
Article in English | MEDLINE | ID: mdl-22306904

ABSTRACT

Glioblastoma (GBM) is the most common primary brain cancer in adults and is virtually incurable. Recent studies have shown that cytomegalovirus (CMV) is present in majority of GBMs. To evaluate whether the CMV antigens pp65 and IE1, which are expressed in GBMs, could be targeted by CMV-specific T cells, we measured the frequency of T cells targeting pp65 and IE1 in the peripheral blood of a cohort of 11 sequentially diagnosed CMV-seropositive GBM patients, and evaluated whether it was feasible to expand autologous CMV-specific T cells for future clinical studies. All 11 CMV-seropositive GBM patients had T cells specific for pp65 and IE1 in their peripheral blood assessed by IFNγ enzyme-linked immunospot assay. However, the precursor frequency of pp65-specific T cells was decreased in comparison with healthy donors (P=0.001). We successfully reactivated and expanded CMV-specific T cells from 6 out of 6 GBM patients using antigen-presenting cells transduced with an adenoviral vector encoding pp65 and IE1. CMV-specific T-cell lines contained CD4 as well as CD8 T cells, recognized pp65 and IE1 targets and killed CMV-infected autologous GBM cells. Infusion of such CMV-specific T-cell lines may extend the benefits of T-cell therapy to patients with CMV GBMs.


Subject(s)
Antigens, Viral/immunology , Brain Neoplasms/therapy , Glioblastoma/therapy , Immunotherapy, Adoptive/methods , T-Lymphocytes/immunology , Adult , Aged , Brain Neoplasms/immunology , Brain Neoplasms/virology , Cell Separation , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Glioblastoma/immunology , Glioblastoma/virology , Humans , In Situ Hybridization , Male , Middle Aged
7.
J Palliat Med ; 14(1): 45-50, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21194303

ABSTRACT

To understand the needs of patients and family members as physicians communicate their expectations about patients admitted to the intensive care unit (ICU), we evaluated the demographic and clinical determinants of having a Do Not Resuscitate (DNR) order for adults with cancer. Patients included were admitted from June 16, 2008-August 16, 2008, to the ICU in a comprehensive cancer center. We conducted a prospective chart review and collected data on patient demographics, length of stay, advance directives, clinical characteristics, and DNR orders. A total of 362 patients met the inclusion criteria; only 15.2% had DNR orders before ICU discharge. In the multivariate analysis, we found that medical admission was an independent predictor of having a DNR order during the ICU stay (odds ratio = 3.65; 95% confidence interval, 1.44-9.28); we also found a significant two-way interaction between race/ethnicity and type of admission (medical vs. surgical) with having a DNR order (p = .04). Although medical admissions were associated with significantly more DNR orders than were surgical admissions, we observed that the subgroup of non-white patients admitted for medical reasons was significantly less likely to have DNR orders. This finding could reflect different preferences for aggressive care by race/ethnicity in patients with cancer, and deserves further investigation.


Subject(s)
Cancer Care Facilities , Intensive Care Units , Resuscitation Orders , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Medical Audit , Middle Aged , Prospective Studies , Texas , Young Adult
8.
Mov Disord ; 23(14): 1957-65, 2008 Oct 30.
Article in English | MEDLINE | ID: mdl-18785647

ABSTRACT

The focus of this article is to review the epidemiology, phenomenology, pathophysiology, genetics, and treatment of movement disorders, particularly task-specific dystonia, in musicians. The goal is to draw attention to this group of neurological disorders among musicians, music teachers, and healthcare professionals and to highlight the importance of early diagnosis, therapeutic options, and preventive measures. To increase professional and public awareness and to facilitate the recognition of music-related neurological problems, we suggest that "medical problems of musicians" be included in curriculum of music schools and medical schools.


Subject(s)
Movement Disorders , Music , Humans , Movement Disorders/epidemiology , Movement Disorders/genetics , Movement Disorders/physiopathology , Movement Disorders/therapy , Occupational Diseases
9.
J Neurol Neurosurg Psychiatry ; 78(11): 1171-5, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17940168

ABSTRACT

In this review, we intend to explore the often asked question: "Did Mozart have Tourette's syndrome?" Although there are numerous reports attributing Mozart's peculiar personality and behaviour to a spectrum of neurobehavioural disorders such as Tourette's syndrome, autistic disorder, Asperger's syndrome, attention deficit hyperactivity disorder, obsessive-compulsive disorder and paediatric autoimmune neuropsychiatric disorders associated with streptococcal infection, the evidence for any of these disorders is lacking. Whether Mozart's behaviour was nothing more than a reflection of his unique personality or a more complex neurological disorder, aggravated later in life by enormous demands by his father and society, his behaviour has been the subject of many biographies. It will also remain unknown to what extent his accomplishments and failures were shaped by his childhood experiences, pressured lifestyle, and his innate genius and extraordinary talent. Lessons from his life may have important implications for other gifted individuals and savants whose special attributes may lead them to succeed or, on the other hand, suppress their emotional growth and make them more vulnerable to stress and failure.


Subject(s)
Famous Persons , Music/history , Tourette Syndrome/history , Austria , History, 18th Century , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL