Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Neurooncol ; 155(3): 297-306, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34689306

ABSTRACT

PURPOSE: Survival is dismal for bevacizumab refractory high-grade glioma patients. We prospectively investigated the efficacy of re-irradiation, bevacizumab, and temozolomide in bevacizumab-naïve and bevacizumab-exposed recurrent high-grade glioma, without volume limitations, in a single arm trial. METHODS: Recurrent high-grade glioma patients were stratified based on WHO grade (4 vs. < 4) and prior exposure to bevacizumab (yes vs. no). Eligible patients received radiation using a simultaneous integrated boost technique (55 Gy to enhancing disease, 45 Gy to non-enhancing disease in 25 fractions) with bevacizumab 10 mg/kg every 2 weeks IV and temozolomide 75 mg/m2 daily followed by maintenance bevacizumab 10 mg/kg every 2 weeks and temozolomide 50 mg/m2 daily for 6 weeks then a 2 week holiday until progression. Primary endpoint was overall survival. Quality of life was studied using FACT-Br and FACT-fatigue scales. RESULTS: Fifty-four patients were enrolled. The majority (n = 36, 67%) were bevacizumab pre-exposed GBM. Median OS for all patients was 8.5 months and 7.9 months for the bevacizumab pre-exposed GBM group. Patients ≥ 36 months from initial radiation had a median OS of 13.3 months compared to 7.5 months for those irradiated < 36 months earlier (p < 0.01). FACT-Br and FACT-Fatigue scores initially declined during radiation but returned to pretreatment baseline. Treatment was well tolerated with 5 patients experiencing > grade 3 lymphopenia and 2 with > grade 3 thrombocytopenia. No radiographic or clinical radiation necrosis occurred. CONCLUSIONS: Re-irradiation with bevacizumab and temozolomide is a safe and feasible salvage treatment for patients with large volume bevacizumab-refractory high-grade glioma. Patients further from their initial radiotherapy may derive greater benefit with this regimen.


Subject(s)
Brain Neoplasms , Chemoradiotherapy , Glioma , Re-Irradiation , Bevacizumab/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Fatigue , Glioma/drug therapy , Glioma/radiotherapy , Humans , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/radiotherapy , Prospective Studies , Quality of Life , Temozolomide/therapeutic use
2.
Cancer ; 126(12): 2821-2828, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32154928

ABSTRACT

BACKGROUND: Targeting vascular endothelial growth factor (VEGF) alone does not improve overall survival (OS) in recurrent glioblastoma (rGBM). The angiopoiein (Ang)-TIE2 system may play a role in tumor survival under VEGF inhibition. We conducted a phase 2, double-blinded, placebo-controlled trial of bevacizumab plus trebananib (a novel Fc fusion protein that sequesters Ang1/Ang2) over bevacizumab alone in rGBM. METHODS: Patients ≥18 years of age with a Karnofsky performance status ≥70 and GBM or variants in first or second relapse were randomized to bevacizumab 10 mg/kg every 2 weeks plus trebananib 15 mg/kg every week or bevacizumab plus placebo. The primary endpoint was 6-month progression-free survival (PFS). RESULTS: After an initial 6-patient lead-in cohort confirmed the safety of combining bevacizumab and trebananib, 115 eligible patients were randomized to the control (n = 58) or experimental treatment (n = 57). In the control arm, 6-month PFS was 41.1%, median survival time was 11.5 months (95% CI, 8.4-14.2 months), median PFS was 4.8 months (95% CI, 3.8-7.1 months), and radiographic response (RR) was 5.9%. In the experimental arm, 6-month PFS was 22.6%, median survival time was 7.5 months (95% CI, 6.8-10.1 months), median PFS was 4.2 months (95% CI, 3.7-5.6 months), and RR was 4.2%. The rate of severe toxicities was not significantly different between arms. CONCLUSION: The combination of bevacizumab and trebananib was well tolerated but did not significantly improve 6-month PFS rate, PFS, or OS for patients with rGBM over bevacizumab alone. The shorter PFS in the experimental arm with a hazard ratio of 1.51 (P = .04) suggests that the addition of trebananib to bevacizumab is detrimental.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Glioblastoma/drug therapy , Gliosarcoma/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab/administration & dosage , Double-Blind Method , Female , Glioblastoma/mortality , Glioblastoma/pathology , Gliosarcoma/mortality , Gliosarcoma/pathology , Humans , Male , Middle Aged , Placebos , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Treatment Outcome
3.
Oncology (Williston Park) ; 32(1): 17-22, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29447417

ABSTRACT

Primary central nervous system (CNS) lymphoma is a rare CNS neoplasm. Its highest incidence is in the elderly and the immunocompromised. The initial steps in establishing a diagnosis involve CNS imaging. Familiarity with the clinical presentation is important in order to limit the risk of a nondiagnostic biopsy. In addition to confirming the diagnosis, it is wise to evaluate for extra-CNS disease. There are important differences in the presentation and evaluation of immunocompetent patients and those of immunocompromised patients; we will delineate these in this review. Appropriate initial clinical evaluations facilitate optimal therapeutic management for patients with primary CNS lymphoma. This is of particular importance because primary CNS lymphoma is a potentially curable disease, despite the high likelihood of recurrence.


Subject(s)
Central Nervous System Neoplasms/diagnosis , Lymphoma, Non-Hodgkin/diagnosis , Central Nervous System Neoplasms/epidemiology , Central Nervous System Neoplasms/mortality , Central Nervous System Neoplasms/pathology , Humans , Lymphoma, Non-Hodgkin/epidemiology , Lymphoma, Non-Hodgkin/mortality , Lymphoma, Non-Hodgkin/pathology , Neoplasm Staging , Prognosis
4.
Oncology (Williston Park) ; 32(2): e11-e19, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29492949

ABSTRACT

Primary central nervous system (CNS) lymphoma, a rare CNS neoplasm associated with high mortality, is responsive to therapeutic interventions. In Part 1 of our two-part coverage of this entity, we provided an overview of the epidemiology of primary CNS lymphoma, followed by a discussion of the diagnostic and staging evaluation, and a review of current prognostication systems. In Part 2, we discuss the management of primary CNS lymphoma, focusing in particular on systemic therapies and radiation. With respect to systemic therapies, we provide details of a variety of regimens built around a backbone of high-dose methotrexate. Future directions for the treatment of primary CNS lymphoma are reviewed as well. These include optimization of consolidation regimens and the pursuit of novel agents.


Subject(s)
Central Nervous System Neoplasms/therapy , Lymphoma, Non-Hodgkin/therapy , Central Nervous System Neoplasms/mortality , Cranial Irradiation , Hematopoietic Stem Cell Transplantation , Humans , Lymphoma, Non-Hodgkin/mortality , Methotrexate/therapeutic use , Transplantation, Autologous
5.
J Neurooncol ; 129(1): 139-46, 2016 08.
Article in English | MEDLINE | ID: mdl-27300524

ABSTRACT

One resistance mechanism in malignant gliomas (MG) involves nuclear factor-κB (NF-κB) activation. Bortezomib prevents proteasomal degradation of NF-κB inhibitor α (NFKBIA), an endogenous regulator of NF-κB signaling, thereby limiting the effects of NF-κB on tumor survival and resistance. A presurgical phase II trial of bortezomib in recurrent MG was performed to determine drug concentration in tumor tissue and effects on NFKBIA. Patients were enrolled after signing an IRB approved informed consent. Treatment was bortezomib 1.7 mg/m(2) IV on days 1, 4 and 8 and then surgery on day 8 or 9. Post-operatively, treatment was Temozolomide (TMZ) 75 mg/m(2) PO on days 1-7 and 14-21 and bortezomib 1.7 mg/m(2) on days 7 and 21 [1 cycle was (1) month]. Ten patients were enrolled (8 M and 2 F) with 9 having surgery. Median age and KPS were 50 (42-64) and 90 % (70-100). The median cycles post-operatively was 2 (0-4). The trial was stopped as no patient had a PFS-6. All patients are deceased. Paired plasma and tumor bortezomib concentration measurements revealed higher drug concentrations in tumor than in plasma; NFKBIA protein levels were similar in drug-treated vs. drug-naïve tumor specimens. Nuclear 20S proteasome was less in postoperative samples. Postoperative treatment with TMZ and bortezomib did not show clinical activity. Bortezomib appears to sequester in tumor but pharmacological effects on NFKBIA were not seen, possibly obscured due to downregulation of NFKBIA during tumor progression. Changes in nuclear 20S could be marker of bortezomib effect on tumor.


Subject(s)
Antineoplastic Agents/therapeutic use , Bortezomib/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adult , Antineoplastic Agents/pharmacokinetics , Bortezomib/blood , Bortezomib/pharmacokinetics , Brain Neoplasms/metabolism , Combined Modality Therapy , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Drug Therapy, Combination , Female , Glioblastoma/metabolism , Humans , Male , Middle Aged , NF-KappaB Inhibitor alpha/metabolism , Neoplasm Recurrence, Local/metabolism , Proteasome Endopeptidase Complex/drug effects , Temozolomide , Treatment Outcome
6.
J Natl Compr Canc Netw ; 13(10): 1191-202, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26483059

ABSTRACT

The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Central Nervous System (CNS) Cancers provide interdisciplinary recommendations for managing adult CNS cancers. Primary and metastatic brain tumors are a heterogeneous group of neoplasms with varied outcomes and management strategies. These NCCN Guidelines Insights summarize the NCCN CNS Cancers Panel's discussion and highlight notable changes in the 2015 update. This article outlines the data and provides insight into panel decisions regarding adjuvant radiation and chemotherapy treatment options for high-risk newly diagnosed low-grade gliomas and glioblastomas. Additionally, it describes the panel's assessment of new data and the ongoing debate regarding the use of alternating electric field therapy for high-grade gliomas.


Subject(s)
Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/radiotherapy , Practice Guidelines as Topic , Adult , Central Nervous System Neoplasms/pathology , Humans , Neoplasm Metastasis
7.
Curr Treat Options Oncol ; 16(8): 38, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26143268

ABSTRACT

OPINION STATEMENT: Treating patients with brain tumors can be divided into tumor-directed therapies, the management of tumor-related symptoms and complications and the psychosocial aspect of patient care. In this review, we will discuss the management of disease and treatment-related complications, which can negatively impact patient quality of life and functional status. Brain edema is a common complication or brain tumors and often causes more symptoms than the tumor itself. Treatment options are limited to the use of corticosteroids, which although effective have a plethora of side effects, so the goal should be the lowest dose that maximizes symptoms. Seizures are more common in lower grade brain tumors and treatment should be limited to patients who have seizures using agents that do not affect the metabolism of other drugs, especially chemotherapies. Blood clots are also common in patients and although there is a "fear" of tumoral bleeding, this is not a frequent occurrence; hence, using anticoagulants should be routinely used in patients who experience this complication.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Central Nervous System Neoplasms/complications , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Edema/diagnosis , Brain Edema/etiology , Brain Edema/therapy , Central Nervous System Neoplasms/therapy , Disease Management , Humans , Seizures/diagnosis , Seizures/etiology , Seizures/therapy , Venous Thromboembolism/diagnosis , Venous Thromboembolism/etiology , Venous Thromboembolism/therapy
8.
J Natl Compr Canc Netw ; 12(11): 1517-23, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25361798

ABSTRACT

The NCCN Guidelines for Central Nervous System Cancers provide multidisciplinary recommendations for the clinical management of patients with cancers of the central nervous system. These NCCN Guidelines Insights highlight recent updates regarding the management of metastatic brain tumors using radiation therapy. Use of stereotactic radiosurgery (SRS) is no longer limited to patients with 3 or fewer lesions, because data suggest that total disease burden, rather than number of lesions, is predictive of survival benefits associated with the technique. SRS is increasingly becoming an integral part of management of patients with controlled, low-volume brain metastases.


Subject(s)
Central Nervous System Neoplasms/secondary , Central Nervous System Neoplasms/surgery , Humans , Radiosurgery/methods
9.
J Neurooncol ; 118(1): 147-53, 2014 May.
Article in English | MEDLINE | ID: mdl-24574050

ABSTRACT

Bevacizumab has been reported to cause diffusion restriction in the tumor bed of patients with malignant gliomas. This study evaluated prolonged diffusion restriction, in the corpus callosum (CC), of patients with malignant brain tumors treated with bevacizumab. We retrospectively reviewed our database of patients treated with bevacizumab for malignant brain tumors looking for those with restricted diffusion in the CC. CC ADC ratio measurements were obtained prior to and following treatment. Correlation was made with biopsy (n = 3) and MR perfusion (n = 7) and PET (n = 4). The temporal evolution of these changes relative to therapy was examined with mixed effects regression analysis. Nine patients (eight malignant gliomas, one malignant meningioma) out of 146 patients were found to have developed areas of diffusion restriction in the CC. These areas tended to enlarge and coalesce over serial MRIs and persisted for up to 22 months. Hypoperfusion was demonstrated in MR perfusion in 7/7. PET was hypometabolic in all 4. Biopsy of the CC showed no tumor in 3/3. ADC ratio measurements indicated a significant overall effect of time (F(16,60) = 11.2; p < 0.0001), consistent with persistent diffusion restriction over the measured time periods. Bevacizumab causes prolonged diffusion restriction in the CC. The negative MR perfusion, FDG PET and histopathology suggest this is a toxicity of bevacizumab and not active tumor. Awareness of these changes can assist in patient care.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Brain Neoplasms/drug therapy , Corpus Callosum/pathology , Aged , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/therapeutic use , Bevacizumab , Brain Neoplasms/pathology , Diffusion Magnetic Resonance Imaging , Female , Humans , Image Processing, Computer-Assisted , Male , Middle Aged , Retrospective Studies
10.
J Neurooncol ; 117(1): 93-101, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24449400

ABSTRACT

When surgery and radiation are no longer treatment options, salvage systemic therapy has been used for recurrent meningiomas with little compelling evidence to suggest effectiveness. Patients with surgery and radiation refractory recurrent meningiomas were treated with the oral multifunctional tyrosine kinase inhibitor PTK787/ZK 222584 (PTK787) at a dose of 500 mg twice a day. Each treatment cycle was 4 weeks with MRI done every 8 weeks. Twenty-five patients (14 men; 11 women) with a median age of 59 years and KPS of 80 were treated. Meningioma WHO Grade was I in 2 patients, II in 14 patients and III in 8 patients; 1 patient had a hemangiopericytoma. All patients had prior surgery, external beam radiation therapy or radiosurgery and 11 patients prior systemic chemotherapy. Median number of cycles of PTK 787 administered was 4 (range <1-22). Best response in the 22 evaluable patients was stable disease in 15 (68.2 %). Predominant PTK787 related toxicities included fatigue (60 %), hypertension (24 %) and elevated transaminases (24 %). Grade II patients had a progression free survival (PFS)-6 of 64.3 %, a median PFS of 6.5 months and an overall survival (OS) of 26.0 months; grade III patients had a PFS-6 of 37.5 %, median PFS of 3.6 months and OS 23 months. PTK787 was modestly toxic at the dose of 500 mg administered twice per day. Activity as determined by PFS-6 suggests that targeting PDGF/VEGF pathway warrants further investigation.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Meningioma/drug therapy , Phthalazines/administration & dosage , Pyridines/administration & dosage , Salvage Therapy , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Brain/drug effects , Brain/pathology , Brain/radiation effects , Brain/surgery , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Brain Neoplasms/surgery , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Magnetic Resonance Imaging , Male , Meningioma/pathology , Meningioma/radiotherapy , Meningioma/surgery , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/drug therapy , Phthalazines/adverse effects , Pyridines/adverse effects , Time Factors
11.
Value Health ; 17(1): 62-9, 2014.
Article in English | MEDLINE | ID: mdl-24438718

ABSTRACT

OBJECTIVES: This study's primary goals included identifying the highest priority symptoms of patients with advanced brain tumors on treatment, comparing patient priority ratings with those of oncology experts, and constructing a brief symptom index using combined input to assess these symptoms and concerns. METHODS: Fifty patients with advanced primary brain tumors and 10 physician experts were recruited from the National Comprehensive Cancer Network institutions and community support agencies. By using a 40-item symptom checklist, patients first selected up to 10 of the most important symptoms/concerns to monitor when assessing the value of drug treatment for brain tumors, then nominated up to 5 of the very most important concerns, and finally generated additional symptoms/concerns. By using the same checklist as patients, physicians rated each symptom/concern as disease- or treatment-related. RESULTS: By using the combined input, a 24-item National Comprehensive Cancer Network/Functional Assessment of Cancer Therapy-Brain Symptom Index (NFBrSI-24) was developed. The NFBrSI-24 showed good internal consistency (α = 0.84), significantly differentiated patients with different levels of functional status (F2,47 = 8.21; P < .001), and demonstrated good convergent validity with the Functional Assessment of Cancer Therapy-General functional, physical, social, emotional, and brain tumor-specific concerns (ρ = 0.59, 0.57, 0.40, 0.35, and 0.50, respectively; Ps < 0.05). CONCLUSIONS: The NFBrSI-24, an index of the symptoms in advanced brain tumors perceived as most important by both patients and clinicians, improves upon existing measures of brain tumor symptoms through better satisfaction of regulatory requirements for measure development. The findings suggest good reliability and validity, indicating that the NFBrSI-24 is a promising brief assessment of high-priority advanced brain tumor symptoms for research and clinical settings.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/psychology , Health Status Indicators , Adult , Aged , Brain Neoplasms/pathology , Checklist , Female , Humans , Interviews as Topic , Male , Middle Aged , Neoplasm Staging , Quality of Life , Surveys and Questionnaires
12.
Cancers (Basel) ; 16(11)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38893252

ABSTRACT

Patients with solid tumor brain metastases that progress after whole-brain radiation have limited options. This prospective trial investigated the efficacy, safety, and tolerability of bevacizumab as salvage therapy in this population. Eligible patients received bevacizumab 10 mg/kg intravenously every 2 weeks until progression. The primary endpoint was radiologic response using Response Assessment in Neuro-Oncology (RANO) criteria. The secondary endpoints were progression-free survival (PFS), overall survival (OS), duration of response, and safety. Quality of life (QOL) was studied using the Functional Assessment of Cancer Therapy-Brain (FACT-Br) scale. Twenty-seven patients were enrolled, with twenty-four having evaluable data for response. The majority of histologies (n = 21, 78%) were breast cancer. The remaining histologies were non-small-cell lung cancer (n = 4, 15%), neuroendocrine cancer (n = 1, 3%), and papillary fallopian serous adenocarcinoma (n = 1, 3%). Eighteen patients had radiologic response, with two patients demonstrating partial response (8.33%) and sixteen patients demonstrating stable disease (66.7%). The median duration of response was 203 days. PFS at 6 months was 46%, median PFS was 5.3 m, and median OS was 9.5 m. Treatment was well tolerated, with six patients experiencing grade 3 lymphopenia and hypertension. There was one grade 3 thromboembolism. QOL was not negatively impacted. Bevacizumab is a safe and feasible salvage treatment with durable response and favorable overall survival for patients with progressive brain metastases after whole-brain radiation.

13.
J Natl Compr Canc Netw ; 11(9): 1114-51, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-24029126

ABSTRACT

Primary and metastatic tumors of the central nervous system are a heterogeneous group of neoplasms with varied outcomes and management strategies. Recently, improved survival observed in 2 randomized clinical trials established combined chemotherapy and radiation as the new standard for treating patients with pure or mixed anaplastic oligodendroglioma harboring the 1p/19q codeletion. For metastatic disease, increasing evidence supports the efficacy of stereotactic radiosurgery in treating patients with multiple metastatic lesions but low overall tumor volume. These guidelines provide recommendations on the diagnosis and management of this group of diseases based on clinical evidence and panel consensus. This version includes expert advice on the management of low-grade infiltrative astrocytomas, oligodendrogliomas, anaplastic gliomas, glioblastomas, medulloblastomas, supratentorial primitive neuroectodermal tumors, and brain metastases. The full online version, available at NCCN. org, contains recommendations on additional subtypes.


Subject(s)
Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/therapy , Humans
15.
J Neurooncol ; 112(2): 247-55, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23354655

ABSTRACT

Brain metastases (BM) and leptomeningeal metastases (LM) are devastating neurologic complications. Pemetrexed is a multi-targeted anti-folate agent approved for treatment of nonsquamous non-small cell lung cancer but has anti-tumor activity in other solid tumors. We performed two trials using pemetrexed in patients with BM and LM to assess CSF penetration and anti-tumor activity. Patients were treated with intravenous pemetrexed at doses of 500 (n = 3), 750 (n = 3), 900 (n = 12) or 1,050 mg/m(2) (n = 3) every 3 weeks. Neuro-imaging was done every 6 weeks. Matched CSF and plasma samples were obtained serially from three patients with Ommaya reservoirs; the remaining patients had a single paired collection. Twenty-one patients (15 women and six men) with median age of 50 years and median KPS of 90 were treated. Primary tumors included breast (13), lung (4), colorectal (1), endometrial (1), esophageal (1) and pinealoblastoma (1). Nine patients had prior whole brain RT and median number of prior chemotherapies was two including prior methotrexate in four patients. Median pemetrexed doses administered was three (range 1-14). Responses included one partial response, ten stable disease and ten progressive disease. Median time to progression and survival was 2.7 and 7.3 months; PFS six was 22 %. No major toxicities were seen. Pemetrexed distributed from the plasma to the CSF within 1-4 h with the resulting CSF concentrations < 5 % of plasma. Pemetrexed was tolerated in solid tumor patients with CNS metastases. Limited anti-tumor activity was seen, which might have been due to low CSF concentrations, although some patients displayed prolonged benefit.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Brain Neoplasms/drug therapy , Glutamates/pharmacokinetics , Guanine/analogs & derivatives , Meningeal Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Neoplasms/drug therapy , Adult , Aged , Antimetabolites, Antineoplastic/blood , Antimetabolites, Antineoplastic/cerebrospinal fluid , Antimetabolites, Antineoplastic/therapeutic use , Brain Neoplasms/mortality , Brain Neoplasms/secondary , Disease Progression , Female , Follow-Up Studies , Glutamates/blood , Glutamates/cerebrospinal fluid , Glutamates/therapeutic use , Guanine/blood , Guanine/cerebrospinal fluid , Guanine/pharmacokinetics , Guanine/therapeutic use , Humans , Meningeal Neoplasms/mortality , Meningeal Neoplasms/secondary , Middle Aged , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Neoplasms/mortality , Neoplasms/pathology , Pemetrexed , Pilot Projects , Prognosis , Survival Rate , Tissue Distribution
16.
Neuro Oncol ; 25(3): 557-565, 2023 03 14.
Article in English | MEDLINE | ID: mdl-35948282

ABSTRACT

BACKGROUND: Patients with human epidermal growth factor receptor 2-positive (HER2-positive) cancers have a high incidence of central nervous system (CNS) spread, but unfortunately systemic trastuzumab which targets the HER2 receptor has little CNS penetration. The purpose of this study was to determine the maximum-tolerated dose of intrathecal trastuzumab and its efficacy in patients with HER2-positive leptomeningeal disease (LMD). METHODS: This multicenter study enrolled 34 LMD patients in a combined phase I/II study in treating patients with intrathecal trastuzumab. Any HER2-positive histology was allowed in the phase I; the phase II was limited to HER2-positive breast cancer. RESULTS: Intrathecal trastuzumab was well-tolerated, with one dose limiting toxicity of grade 4 (arachnoiditis) occurring at the 80 mg twice weekly dose. The recommended phase II dose was 80 mg intrathecally twice weekly. Twenty-six patients at dose level 80 mg were included in evaluation for efficacy: partial response was seen in 5 (19.2%) patients, stable disease was observed in 13 (50.0%), and 8 (30.8%) of the patients had progressive disease. Median overall survival (OS) for phase II dose treated patients was 8.3 months (95% CI 5.2-19.6). The phase II HER2-positive breast cancer patients median OS was 10.5 months (95% CI 5.2-20.9). Pharmacokinetic (PK) studies were limited in the setting of concurrent systemic trastuzumab administration, however, did show stable cerebrospinal fluid (CSF) concentrations with repeated dosing suggest that trastuzumab does not accumulate in the CSF in toxic concentrations. CONCLUSION: This study suggests promise for potentially improved outcomes of HER-positive LMD patients when treated with intrathecal trastuzumab while remaining safe and well-tolerated for patients.


Subject(s)
Breast Neoplasms , Meningeal Carcinomatosis , Humans , Female , Trastuzumab/adverse effects , Receptor, ErbB-2/metabolism , Breast Neoplasms/pathology , Meningeal Carcinomatosis/drug therapy , Meningeal Carcinomatosis/pathology , Antineoplastic Combined Chemotherapy Protocols/adverse effects
17.
J Clin Oncol ; 41(6): 1285-1295, 2023 02 20.
Article in English | MEDLINE | ID: mdl-36260832

ABSTRACT

PURPOSE: To assess whether reirradiation (re-RT) and concurrent bevacizumab (BEV) improve overall survival (OS) and/or progression-free survival (PFS), compared with BEV alone in recurrent glioblastoma (GBM). The primary objective was OS, and secondary objectives included PFS, response rate, and treatment adverse events (AEs) including delayed CNS toxicities. METHODS: NRG Oncology/RTOG1205 is a prospective, phase II, randomized trial of re-RT and BEV versus BEV alone. Stratification factors included age, resection, and Karnofsky performance status (KPS). Patients with recurrent GBM with imaging evidence of tumor progression ≥ 6 months from completion of prior chemo-RT were eligible. Patients were randomly assigned 1:1 to re-RT, 35 Gy in 10 fractions, with concurrent BEV IV 10 mg/kg once in every 2 weeks or BEV alone until progression. RESULTS: From December 2012 to April 2016, 182 patients were randomly assigned, of whom 170 were eligible. Patient characteristics were well balanced between arms. The median follow-up for censored patients was 12.8 months. There was no improvement in OS for BEV + RT, hazard ratio, 0.98; 80% CI, 0.79 to 1.23; P = .46; the median survival time was 10.1 versus 9.7 months for BEV + RT versus BEV alone. The median PFS for BEV + RT was 7.1 versus 3.8 months for BEV, hazard ratio, 0.73; 95% CI, 0.53 to 1.0; P = .05. The 6-month PFS rate improved from 29.1% (95% CI, 19.1 to 39.1) for BEV to 54.3% (95% CI, 43.5 to 65.1) for BEV + RT, P = .001. Treatment was well tolerated. There were a 5% rate of acute grade 3+ treatment-related AEs and no delayed high-grade AEs. Most patients died of recurrent GBM. CONCLUSION: To our knowledge, NRG Oncology/RTOG1205 is the first prospective, randomized multi-institutional study to evaluate the safety and efficacy of re-RT in recurrent GBM using modern RT techniques. Overall, re-RT was shown to be safe and well tolerated. BEV + RT demonstrated a clinically meaningful improvement in PFS, specifically the 6-month PFS rate but no difference in OS.


Subject(s)
Brain Neoplasms , Glioblastoma , Re-Irradiation , Humans , Bevacizumab , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Re-Irradiation/adverse effects , Prospective Studies , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects
18.
Clin Cancer Res ; 29(19): 3882-3891, 2023 10 02.
Article in English | MEDLINE | ID: mdl-37490286

ABSTRACT

PURPOSE: Zolbetuximab, an IgG1 monoclonal antibody, binds to claudin 18.2 (CLDN18.2) and mediates tumor cell death through antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. We sought to examine zolbetuximab combinations in CLDN18.2-positive HER2-negative gastric/gastroesophageal junction (G/GEJ) adenocarcinoma. PATIENTS AND METHODS: This phase II study assessed efficacy and safety of zolbetuximab, alone or with modified FOLFOX6 (mFOLFOX6) or pembrolizumab, in CLDN18.2-positive advanced/metastatic G/GEJ adenocarcinoma. Patients received zolbetuximab as monotherapy in third/later-line (Cohort 1A, n = 30), with mFOLFOX6 in first-line (Cohort 2, n = 21), or with pembrolizumab in third/later-line (Cohort 3A, n = 3) treatment. The primary endpoint for Cohort 1A was objective response rate (ORR). Key secondary endpoints were ORR (Cohorts 2 and 3A), overall survival (OS; Cohort 1A), and progression-free survival (PFS) and safety (all cohorts). RESULTS: ORR was 0% in Cohorts 1A and 3A, and 71.4% [95% confidence interval (CI), 47.82-88.72] in Cohort 2. Median PFS was 1.54 months (95% CI, 1.31-2.56) in Cohort 1A, 2.96 months (95% CI, 1.48-4.44) in Cohort 3A, and 17.8 months (95% CI, 8.05-25.69) in Cohort 2. Median OS in Cohort 1A was 5.62 months (95% CI, 2.27-11.53). Gastrointestinal adverse events occurred across cohorts [nausea, 63%-90% (grade ≥ 3, 4.8%-6.7%) and vomiting, 33%-67% (grade ≥ 3, 6.7%-9.5%)]. CONCLUSIONS: Zolbetuximab plus mFOLFOX6 demonstrated promising efficacy in previously untreated patients with CLDN18.2-positive G/GEJ adenocarcinoma. These data support the first-line development of zolbetuximab in patients whose tumors are CLDN18.2-positive. Across cohorts, zolbetuximab treatment was tolerable with no new safety signals.


Subject(s)
Adenocarcinoma , Antibodies, Monoclonal , Claudins , Esophageal Neoplasms , Stomach Neoplasms , Humans , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Claudins/metabolism , Esophageal Neoplasms/pathology , Esophageal Neoplasms/therapy , Esophagogastric Junction/pathology , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy
19.
Cancer ; 118(15): 3743-8, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22179954

ABSTRACT

BACKGROUND: Despite initial treatment with high-dose methotrexate-based regimens, many patients with primary central nervous system lymphoma (PCNSL) relapse and die from their disease. No standard of care exists at progression or relapse, but chemotherapy and in some cases radiation are usually used. Pemetrexed is a multitargeted antifolate, similar to methotrexate, but with a broader spectrum of activity. Because methotrexate is an integral part of PCSNL treatment, the authors assessed the antitumor activity and safety of pemetrexed in recurrent PCNSL. METHODS: Patients with relapsed/refractory PCNSL were enrolled in this trial. Treatment consisted of pemetrexed 900 mg/m(2) given every 3 weeks with low-dose dexamethasone, folate, and B12 supplementation. Each cycle was 6 weeks, and follow-up imaging was done before each new cycle. Treatment was continued until complete remission, progression, or toxicity. RESULTS: Eleven patients were treated, with a median age of 69.8 years and Karnofsky performance status of 70%; 10 of 11 patients had failed prior high-dose methotrexate. The median number of pemetrexed cycles given was 5, with an associated overall response rate of 55% and disease control rate of 91%. The 6-month progression-free survival (PFS) was 45%, median PFS was 5.7 months, and median overall survival was 10.1 months. Toxicities were primarily hematologic and infectious. CONCLUSIONS: Pemetrexed has single-agent activity in relapsed/refractory PCNSL. Toxicities were seen likely because of the higher than standard dose used. Further investigation of this agent or other multitargeted antifolates in PCNSL is warranted to determine optimal dose and efficacy in a more homogeneous population.


Subject(s)
Antineoplastic Agents/therapeutic use , Central Nervous System Neoplasms/drug therapy , Glutamates/administration & dosage , Guanine/analogs & derivatives , Lymphoma/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols , Disease-Free Survival , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Glutamates/adverse effects , Guanine/administration & dosage , Guanine/adverse effects , Humans , Male , Middle Aged , Pemetrexed , Recurrence , Salvage Therapy
20.
J Neurooncol ; 107(1): 21-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21964740

ABSTRACT

Extracranial metastases from glioblastoma (GBM) are uncommon with an estimated incidence of less than 2%. We report two cases of metastatic GBM seen within an 8-week period followed by a literature review. We attempted to identify common factors or a causative mechanism. Factors that predominated among the reviewed cases included male gender, tumor location, and younger age. Causative mechanisms were not apparent. While metastatic disease remains rare, it might be occurring with increasing frequency. This trend might be due to increased diagnosis, better imaging, a more extensive physician workup, or an increase in survival. Metastatic GBM can present and progress quite rapidly, and repeat evaluations of persistent or worsening complaints among GBM patients are warranted. Early diagnosis of metastatic disease spread can help to expedite alleviation of patients' discomfort, in an already aggressive disease process.


Subject(s)
Bone Neoplasms/secondary , Glioblastoma/pathology , Adult , Aged , Bone Neoplasms/therapy , Female , Glioblastoma/therapy , Humans , Male , Review Literature as Topic
SELECTION OF CITATIONS
SEARCH DETAIL