Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Adv Radiat Oncol ; 6(4): 100711, 2021.
Article in English | MEDLINE | ID: mdl-34195498

ABSTRACT

PURPOSE: Leptomeningeal disease in prostate adenocarcinoma is very rare. Solitary leptomeningeal recurrence from prostate adenocarcinoma has only been previously reported once in the published literature. METHODS AND MATERIALS: A 63-year-old man with high-risk prostate cancer was treated in a phase I-II trial with androgen deprivation, radiation therapy, and cytotoxic gene therapy. He initially had biochemical control but experienced solitary leptomeningeal recurrence 47 months after diagnosis. RESULTS: He received androgen deprivation, radiation therapy to the lumbar and sacral spine, and stereotactic radiosurgery to 3 intracranial foci of disease. He died 14 months after leptomeningeal recurrence. Autopsy showed diffuse spinal leptomeningeal disease, leptomeningeal based intracranial lesions, and no other metastasis. CONCLUSIONS: The cause for solitary leptomeningeal recurrence in this patient is unknown. Although there may be many possible mechanisms, we speculate that it could be related to his initial treatment with cytotoxic gene therapy along with radiation therapy and androgen deprivation.

3.
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
4.
Front Neurol ; 7: 121, 2016.
Article in English | MEDLINE | ID: mdl-27531989

ABSTRACT

Dynamic susceptibility contrast (DSC) perfusion-weighted imaging (PWI) is widely used in clinical settings for the radiological diagnosis of brain tumor. The signal change in brain tissue in gradient echo-based DSC PWI is much higher than in spin echo-based DSC PWI. Due to its exquisite sensitivity, gradient echo-based sequence is the preferred method for imaging of all tumors except those near the base of the skull. However, high sensitivity also comes with a dynamic range problem. It is not unusual for blood volume to increase in gene-mediated cytotoxic immunotherapy-treated glioblastoma patients. The increase of fractional blood volume sometimes saturates the MRI signal during first-pass contrast bolus arrival and presents signal truncation artifacts of various degrees in the tumor when a significant amount of blood exists in the image pixels. It presents a hidden challenge in PWI, as this signal floor can be either close to noise level or just above and can go no lower. This signal truncation in the signal intensity time course is a significant issue that deserves attention in DSC PWI. In this paper, we demonstrate that relative cerebral blood volume and relative cerebral blood flow (rCBF) are underestimated due to signal truncation in DSC perfusion, in glioblastoma patients. We propose the use of second-pass tissue residue function in rCBF calculation using least-absolute-deviation deconvolution to avoid the underestimation problem.

5.
Neuro Oncol ; 18(8): 1137-45, 2016 08.
Article in English | MEDLINE | ID: mdl-26843484

ABSTRACT

BACKGROUND: Despite aggressive standard of care (SOC) treatment, survival of malignant gliomas remains very poor. This Phase II, prospective, matched controlled, multicenter trial was conducted to assess the safety and efficacy of aglatimagene besadenovec (AdV-tk) plus valacyclovir (gene-mediated cytotoxic immunotherapy [GMCI]) in combination with SOC for newly diagnosed malignant glioma patients. METHODS: Treatment cohort patients received SOC + GMCI and were enrolled at 4 institutions from 2006 to 2010. The preplanned, matched-control cohort included all concurrent patients meeting protocol criteria and SOC at a fifth institution. AdV-tk was administered at surgery followed by SOC radiation and temozolomide. Subset analyses were preplanned, based on prognostic factors: pathological diagnosis (glioblastoma vs others) and extent of resection. RESULTS: Forty-eight patients completed SOC + GMCI, and 134 met control cohort criteria. Median overall survival (OS) was 17.1 months for GMCI + SOC versus 13.5 months for SOC alone (P = .0417). Survival at 1, 2, and 3 years was 67%, 35%, and 19% versus 57%, 22%, and 8%, respectively. The greatest benefit was observed in gross total resection patients: median OS of 25 versus 16.9 months (P = .0492); 1, 2, and 3-year survival of 90%, 53%, and 32% versus 64%, 28% and 6%, respectively. There were no dose-limiting toxicities; fever, fatigue, and headache were the most common GMCI-related symptoms. CONCLUSIONS: GMCI can be safely combined with SOC in newly diagnosed malignant gliomas. Survival outcomes were most notably improved in patients with minimal residual disease after gross total resection. These data should help guide future immunotherapy studies and strongly support further evaluation of GMCI for malignant gliomas. CLINICAL TRIAL REGISTRY: ClinicalTrials.gov NCT00589875.


Subject(s)
Brain Neoplasms/drug therapy , Genetic Therapy/adverse effects , Genetic Therapy/methods , Glioma/drug therapy , Immunotherapy/adverse effects , Immunotherapy/methods , Acyclovir/adverse effects , Acyclovir/analogs & derivatives , Acyclovir/therapeutic use , Adenoviridae , Adult , Aged , Antiviral Agents/adverse effects , Antiviral Agents/therapeutic use , Brain Neoplasms/surgery , Chemotherapy, Adjuvant/adverse effects , Chemotherapy, Adjuvant/methods , Genetic Vectors/therapeutic use , Glioma/surgery , Humans , Middle Aged , Simplexvirus/genetics , Survival Analysis , Thymidine Kinase/genetics , Treatment Outcome , Valacyclovir , Valine/adverse effects , Valine/analogs & derivatives , Valine/therapeutic use
6.
J Neurooncol ; 116(3): 609-16, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24390465

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are rare soft tissue sarcomas that arise predominantly from Schwann cells. Despite the fact that MPNSTs have high local recurrence rates and are generally associated with poor prognosis, little is known about prognostic factors or effective clinical management for this tumor type. The purpose of this study was to describe the distributions of patient and tumor characteristics and to identify predictors of cause-specific survival among MPNST cases reported to SEER between 1973 and 2008. Patient and tumor characteristics were compared between pediatric and adult MPNST cases. Cox regression and tree-based survival analysis were used to examine factors associated with MPNST-related mortality separately among adults and children. A total of 1,315 MPNST cases were isolated from the 1973-2008 SEER dataset. Among pediatric cases, sex, race, and radiation therapy predicted MPNST survival, whereas among adults, tumor site, tumor grade, number of primary tumors, and tumor size were significant predictors. As tumor size at diagnosis/resection may be the only somewhat "modifiable" prognostic factor, future studies should aim to identify biological and social attributes associated with tumor size at diagnosis, separately among individuals with and without NF-1, in order to help identify earlier opportunities for clinical intervention.


Subject(s)
Nerve Sheath Neoplasms/epidemiology , Neurilemmoma/epidemiology , SEER Program , Sarcoma/epidemiology , Adolescent , Adult , Age Factors , Aged , Child , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Retrospective Studies , United States/epidemiology , Young Adult
7.
J Clin Oncol ; 29(27): 3611-9, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21844505

ABSTRACT

PURPOSE: Despite aggressive therapies, median survival for malignant gliomas is less than 15 months. Patients with unmethylated O(6)-methylguanine-DNA methyltransferase (MGMT) fare worse, presumably because of temozolomide resistance. AdV-tk, an adenoviral vector containing the herpes simplex virus thymidine kinase gene, plus prodrug synergizes with surgery and chemoradiotherapy, kills tumor cells, has not shown MGMT dependency, and elicits an antitumor vaccine effect. PATIENTS AND METHODS: Patients with newly diagnosed malignant glioma received AdV-tk at 3 × 10(10), 1 × 10(11), or 3 × 10(11) vector particles (vp) via tumor bed injection at time of surgery followed by 14 days of valacyclovir. Radiation was initiated within 9 days after AdV-tk injection to overlap with AdV-tk activity. Temozolomide was administered after completing valacyclovir treatment. RESULTS: Accrual began December 2005 and was completed in 13 months. Thirteen patients were enrolled and 12 completed therapy, three at dose levels 1 and 2 and six at dose level 3. There were no dose-limiting or significant added toxicities. One patient withdrew before completing prodrug because of an unrelated surgical complication. Survival at 2 years was 33% and at 3 years was 25%. Patient-reported quality of life assessed with the Functional Assessment of Cancer Therapy-Brain (FACT-Br) was stable or improved after treatment. A significant CD3(+) T-cell infiltrate was found in four of four tumors analyzed after treatment. Three patients with MGMT unmethylated glioblastoma multiforme survived 6.5, 8.7, and 46.4 months. CONCLUSION: AdV-tk plus valacyclovir can be safely delivered with surgery and accelerated radiation in newly diagnosed malignant gliomas. Temozolomide did not prevent immune responses. Although not powered for efficacy, the survival and MGMT independence trends are encouraging. A phase II trial is ongoing.


Subject(s)
Adenoviridae/genetics , Brain Neoplasms/therapy , Cancer Vaccines/therapeutic use , Glioma/therapy , Immunotherapy/methods , Thymidine Kinase/genetics , Acyclovir/administration & dosage , Acyclovir/analogs & derivatives , Adjuvants, Immunologic , Adult , Aged , Antineoplastic Agents, Alkylating/administration & dosage , Antiviral Agents/administration & dosage , Brain Neoplasms/mortality , Combined Modality Therapy , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Genetic Therapy , Genetic Vectors , Glioma/mortality , Herpesvirus 1, Human/enzymology , Humans , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase , Temozolomide , Treatment Outcome , Valacyclovir , Valine/administration & dosage , Valine/analogs & derivatives
8.
J Neurooncol ; 105(2): 375-81, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21547395

ABSTRACT

COL-3 is a chemically modified tetracycline that targets multiple aspects of matrix metalloproteinase regulation. This phase I clinical trial was conducted to determine the maximum tolerated dose (MTD) of COL-3 in adults with recurrent high-grade glioma, to describe the effects of enzyme-inducing antiseizure drugs (EIADs) on its pharmacokinetics, and to obtain preliminary evidence of activity. Adults with recurrent high-grade glioma were stratified by EIAD use. COL-3 was given orally daily without interruption until disease progression or treatment-related dose-limiting toxicity (DLT). Three patients in each EIAD group were evaluated at each dose level beginning with 25 mg/m(2)/day and escalated by 25 mg/m(2)/day. Toxicity, response, and pharmacokinetics were assessed. Thirty-three patients were evaluated. The MTD was 75 mg/m(2)/day in the -EIAD patients while one was not determined in +EIAD patients. The common toxicities observed were anemia, ataxia, diarrhea, hypokalemia, CNS hemorrhage, and myalgia. One partial response was observed. -EIAD patients tended to have a higher steady-state trough concentration that was apparent only at the 100 mg/m(2)/day dose level (P = 0.01). This study suggests that: (a) EIAD use does affect the pharmacokinetics of COL-3 at higher doses; and (b) there was not enough suggestion of single-agent activity to warrant further study in recurrent high-grade gliomas.


Subject(s)
Anticonvulsants/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Tetracyclines/pharmacokinetics , Tetracyclines/therapeutic use , Adult , Aged , Brain Neoplasms/mortality , Female , Follow-Up Studies , Glioma/mortality , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/mortality , Prognosis , Tissue Distribution
9.
Neuro Oncol ; 7(3): 246-53, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16053699

ABSTRACT

Penicillamine is an oral agent used to treat intracerebral copper overload in Wilson's disease. Copper is a known regulator of angiogenesis; copper reduction inhibits experimental glioma growth and invasiveness. This study examined the feasibility, safety, and efficacy of creating a copper deficiency in human glioblastoma multiforme. Forty eligible patients with newly diagnosed glioblastoma multiforme began radiation therapy (6000 cGy in 30 fractions) in conjunction with a low-copper diet and escalating doses of penicillamine. Serum copper was measured at baseline and monthly. The primary end point of this study was overall survival compared to historical controls within the NABTT CNS Consortium database. The 25 males and 15 females who were enrolled had a median age of 54 years and a median Karnofsky performance status of 90. Surgical resection was performed in 83% of these patients. Normal serum copper levels at baseline (median, 130 microg/dl; range, 50-227 microg/dl) fell to the target range of <50 microg/dl (median, 42 microg/dl; range, 12-118 microg/dl) after two months. Penicillamine-induced hypocupremia was well tolerated for months. Drug-related myelosuppression, elevated liver function tests, and skin rash rapidly reversed with copper repletion. Median survival was 11.3 months, and progression-free survival was 7.1 months. Achievement of hypocupremia did not significantly increase survival. Although serum copper was effectively reduced by diet and penicillamine, this antiangiogenesis strategy did not improve survival in patients with glioblastoma multiforme.


Subject(s)
Brain Neoplasms/therapy , Chelating Agents/toxicity , Copper/blood , Glioblastoma/therapy , Neovascularization, Pathologic/drug therapy , Penicillamine/toxicity , Brain Neoplasms/mortality , Diet Therapy , Female , Glioblastoma/mortality , Humans , Male , Middle Aged , Survival Analysis
10.
J Pharm Biomed Anal ; 37(4): 751-6, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15797797

ABSTRACT

COL-3, 6-deoxy-6-desmethyl-4-desdimethylamino-tetracycline, is a matrix metalloproteinase inhibitor currently in clinical development. A HPLC-UV method to quantitate COL-3 in human plasma was developed. COL-3 was extracted from plasma using solid-phase extraction cartridges. COL-3 is separated on a Waters Symmetry Shield RP8 (3.9 mm x150 mm, 5 microm) column with EDTA (0.001 M) in sodium acetate (0.01 M, pH 3.5)-acetonitrile mobile phase using a gradient profile at a flow rate of 1 ml/min for 22 min. Carryover was eliminated by using an extended needle wash of methanol:acetonitrile:dichloromethane (1:1:1, v/v/v). Detection of COL-3 and the internal standard, chrysin, was observed at 350 nm. COL-3 and chrysin elute at 8.9 and 9.9 min, respectively. The lower limit of quantitation in human plasma of COL-3 was 75 ng/ml, linearity was observed from 75 to 10,000 ng/ml. A 30,000 ng/ml sample that was diluted 1:50 with plasma was accurately quantitated. This method is rapid, widely applicable, and suitable for quantifying COL-3 in patient samples enabling further clinical pharmacology characterization of COL-3.


Subject(s)
Tetracyclines/blood , Analysis of Variance , Calibration , Chromatography, High Pressure Liquid , Drug Stability , Drug Storage , Humans , Indicators and Reagents , Quality Control , Reference Standards , Spectrophotometry, Ultraviolet , Tetracyclines/analysis
11.
Cancer Control ; 11(3): 152-64, 2004.
Article in English | MEDLINE | ID: mdl-15153839

ABSTRACT

BACKGROUND: High-grade glioma remains one of the most difficult cancers to treat. Recent studies in oncology have identified a role of the ubiquitous enzyme, cyclooxygenase (Cox), especially cyclooxygenase-2 (COX-2) in cell proliferation, and its inhibition in cancer control, apoptosis, as well as synergy with other forms of therapy. The inhibitors of the Cox enzyme are well known as members of the nonsteroidal anti-inflammatory drug (NSAID) class of pharmaceuticals. METHODS: In vitro and in vivo studies of different cancers expressing COX-2, including glioma studies, along with the few clinical trials that have been reported are reviewed to specifically identify the actions of these agents. RESULTS: The anticancer effect of the COX-2 inhibitors may occur irrelevant of COX-2 expression, and it appears to be drug-specific, as well as dose-specific in different cancers. In combination with chemotherapeutic agents, the COX-2 inhibitors may have an additive, synergistic, or inhibitory effect on tumor growth. CONCLUSIONS: As evaluations of this class of drugs begin in glioma, in vitro and in vivo data should be acquired to accurately predict which compounds will have an effect in controlling tumor growth and at which doses these should be used. The actual expression and inhibition of COX-2 may not always be relevant to the effects on tumor growth.


Subject(s)
Cyclooxygenase 2 Inhibitors/therapeutic use , Glioma/drug therapy , Glioma/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents/therapeutic use , Aspirin/therapeutic use , Cell Proliferation/drug effects , Combined Modality Therapy , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Drug Therapy, Combination , Glioma/physiopathology , Glioma/radiotherapy , Humans , Ibuprofen/therapeutic use , Salicylic Acid/therapeutic use , Signal Transduction/drug effects , Treatment Outcome
12.
J Clin Oncol ; 21(6): 1044-9, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12637469

ABSTRACT

PURPOSE: A multicenter, phase II study of single-agent, intravenous methotrexate in newly diagnosed non-AIDS-related primary CNS lymphoma was conducted in the New Approaches to Brain Tumor Therapy (NABTT) CNS Consortium. METHODS: Methotrexate (8 g/m(2)) was initially administered every 2 weeks. The primary end point was radiographic CR or PR, as defined by standard radiographic criteria, and secondary end points were survival and drug-related toxicity. RESULTS: Twenty-five patients were enrolled with a mean age of 60 years and median Karnofsky Performance Score of 80. Three of 14 patients who underwent lumbar puncture had malignant cells on CSF cytopathology, and five of 25 patients had ocular involvement. Two patients could not be evaluated for the primary end point because of the absence of measurable disease in one and death before radiologic imaging in another. All patients have completed the treatment program or progressed. Among 23 patients, there were 12 CR (52%), five PR (22%), one (4%) with stable disease, and five progressions (22%) while on therapy. Seven patients died of tumor progression, and two died of other causes. Median progression-free survival was 12.8 months. Median overall survival for the entire group had not been reached at 22.8+ months. The toxicity of this regimen was modest, with no grade 3 or 4 toxicity in 13 of 25 patients, grade 3 toxicity in eight of 25 patients, and grade 4 toxicity in four of 25 patients after 287 cycles of chemotherapy. CONCLUSION: These results indicate that high-dose methotrexate is associated with modest toxicity and a radiographic response proportion (74%) comparable to more toxic regimens.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Lymphoma/drug therapy , Lymphoma/radiotherapy , Methotrexate/therapeutic use , Adult , Aged , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Humans , Lymphoma/mortality , Lymphoma/pathology , Magnetic Resonance Imaging , Male , Mental Status Schedule , Middle Aged , Radiotherapy, Adjuvant , Survival Analysis , Treatment Outcome
13.
J Neurooncol ; 57(3): 231-9, 2002 May.
Article in English | MEDLINE | ID: mdl-12125986

ABSTRACT

Drugs currently available for intrathecal administration are cleared rapidly from the CSF. DepoCyt is a slow-release formulation of cytarabine that maintains cytotoxic concentrations of free cytarabine in the CSF for >14 days following a single injection. DepoCyt was administered to 110 patients with a diagnosis of neoplastic meningitis based on either a positive CSF cytology (76) or neurologic and CT or MRI scan findings sufficient to document neoplastic meningitis (34). Patients were treated with DepoCyt 50mg every 2 weeks for 1 month of induction therapy by either lumbar puncture (LP) or intraventricular (IVT) injection. Patients without neurologic progression were candidates to receive an additional 3 months of consolidation therapy. All patients received dexamethasone 4 mg BID on days 1-5 of each cycle. Median time to neurologic progression was 55 days; median overall survival was 95 days. Among the 76 patients with a positive CSF cytology at baseline, 70 were evaluable for response, and of this group 19 (27%) attained the criteria for response (cytologic response in the absence of neurologic progression). The most important adverse events were headache and arachnoiditis. When drug-related, these were largely low grade, transient, and reversible. Drug-related grade 3 headache occurred on 4% of cycles; grade 3 or 4 arachnoiditis occurred on 6% of cycles. No cumulative toxicity was observed. DepoCyt injected once every 2 weeks produced a response-rate comparable to that previously reported for methotrexate given twice a week. The once in every 2-week-dosing interval offers an advantage over conventional schedules (2-3 doses/week) used for other agents available for intrathecal injection.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , Cytarabine/administration & dosage , Meningeal Neoplasms/drug therapy , Adult , Aged , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Delayed-Action Preparations , Disease Progression , Female , Humans , Injections, Spinal , Male , Meningeal Neoplasms/mortality , Meningeal Neoplasms/secondary , Middle Aged , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL