67 results on '"Christopher Duma"'
Search Results
2. 952 Phase II trial of AV-GBM-1: dendritic cell vaccine pulsed with lysate enriched for autologous tumor-initiating cell antigens in the treatment of patients with newly diagnosed glioblastoma
- Author
-
Gabriel Nistor, Robert Dillman, Daniela Bota, David Piccioni, Christopher Duma, Renato LaRocca, Santosh Kesari, Jose Carrillo, Robert Aiken, Frank Hsu, Thomas Taylor, Mehrdad Abedi, Xiaotang Kong, and Candace Hsieh
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
3. 335 Leukaphereses to obtain monocytes to produce dendritic cells in manufacturing of personal autologous AV-GBM-1 vaccines in a phase II trial in patients with newly diagnosed glioblastoma
- Author
-
Gabriel Nistor, Robert Dillman, Daniela Bota, Christopher Duma, Renato LaRocca, Santosh Kesari, Jose Carrillo, Robert Aiken, Frank Hsu, Thomas Taylor, Mehrdad Abedi, Candace Hsieh, David Piccione, and Xiang-Tang Kong
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
4. 331 Tumor markers associated with increased survival in a phase II trial of dendritic cell/tumor-initiating-cell vaccine AV-GBM-1 in patients with newly diagnosed glioblastoma
- Author
-
Gabriel Nistor, Robert Dillman, Daniela Bota, David Piccioni, Christopher Duma, Renato LaRocca, Santosh Kesari, Jose Carrillo, Robert Aiken, Frank Hsu, Xiao-Tang Kong, Thomas Taylor, Mehrdad Abedi, and Candace Hsieh
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
5. 319 Phase II trial of immunotherapy in primary glioblastoma: antigens from self-renewing autologous tumor cells presented by autologous dendritic cell vaccine
- Author
-
Gabriel Nistor, Robert Dillman, Daniela Bota, David Piccioni, Christopher Duma, Renato LaRocca, Santosh Kesari, Jose Carrillo, Robert O’Donnell, Robert Aiken, Frank Hsu, Xiao-Tang Kong, and Thomas Taylor
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
6. First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma
- Author
-
Linda M. Liau, Keyoumars Ashkan, David D. Tran, Jian L. Campian, John E. Trusheim, Charles S. Cobbs, Jason A. Heth, Michael Salacz, Sarah Taylor, Stacy D. D’Andre, Fabio M. Iwamoto, Edward J. Dropcho, Yaron A. Moshel, Kevin A. Walter, Clement P. Pillainayagam, Robert Aiken, Rekha Chaudhary, Samuel A. Goldlust, Daniela A. Bota, Paul Duic, Jai Grewal, Heinrich Elinzano, Steven A. Toms, Kevin O. Lillehei, Tom Mikkelsen, Tobias Walbert, Steven R. Abram, Andrew J. Brenner, Steven Brem, Matthew G. Ewend, Simon Khagi, Jana Portnow, Lyndon J. Kim, William G. Loudon, Reid C. Thompson, David E. Avigan, Karen L. Fink, Francois J. Geoffroy, Scott Lindhorst, Jose Lutzky, Andrew E. Sloan, Gabriele Schackert, Dietmar Krex, Hans-Jorg Meisel, Julian Wu, Raphael P. Davis, Christopher Duma, Arnold B. Etame, David Mathieu, Santosh Kesari, David Piccioni, Manfred Westphal, David S. Baskin, Pamela Z. New, Michel Lacroix, Sven-Axel May, Timothy J. Pluard, Victor Tse, Richard M. Green, John L. Villano, Michael Pearlman, Kevin Petrecca, Michael Schulder, Lynne P. Taylor, Anthony E. Maida, Robert M. Prins, Timothy F. Cloughesy, Paul Mulholland, and Marnix L. Bosch
- Subjects
Glioblastoma ,Immunotherapy ,Dendritic cell ,Vaccine ,Medicine - Abstract
Abstract Background Standard therapy for glioblastoma includes surgery, radiotherapy, and temozolomide. This Phase 3 trial evaluates the addition of an autologous tumor lysate-pulsed dendritic cell vaccine (DCVax®-L) to standard therapy for newly diagnosed glioblastoma. Methods After surgery and chemoradiotherapy, patients were randomized (2:1) to receive temozolomide plus DCVax-L (n = 232) or temozolomide and placebo (n = 99). Following recurrence, all patients were allowed to receive DCVax-L, without unblinding. The primary endpoint was progression free survival (PFS); the secondary endpoint was overall survival (OS). Results For the intent-to-treat (ITT) population (n = 331), median OS (mOS) was 23.1 months from surgery. Because of the cross-over trial design, nearly 90% of the ITT population received DCVax-L. For patients with methylated MGMT (n = 131), mOS was 34.7 months from surgery, with a 3-year survival of 46.4%. As of this analysis, 223 patients are ≥ 30 months past their surgery date; 67 of these (30.0%) have lived ≥ 30 months and have a Kaplan-Meier (KM)-derived mOS of 46.5 months. 182 patients are ≥ 36 months past surgery; 44 of these (24.2%) have lived ≥ 36 months and have a KM-derived mOS of 88.2 months. A population of extended survivors (n = 100) with mOS of 40.5 months, not explained by known prognostic factors, will be analyzed further. Only 2.1% of ITT patients (n = 7) had a grade 3 or 4 adverse event that was deemed at least possibly related to the vaccine. Overall adverse events with DCVax were comparable to standard therapy alone. Conclusions Addition of DCVax-L to standard therapy is feasible and safe in glioblastoma patients, and may extend survival. Trial registration Funded by Northwest Biotherapeutics; Clinicaltrials.gov number: NCT00045968; https://clinicaltrials.gov/ct2/show/NCT00045968?term=NCT00045968&rank=1; initially registered 19 September 2002
- Published
- 2018
- Full Text
- View/download PDF
7. Correction to: First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma
- Author
-
Linda M. Liau, Keyoumars Ashkan, David D. Tran, Jian L. Campian, John E. Trusheim, Charles S. Cobbs, Jason A. Heth, Michael Salacz, Sarah Taylor, Stacy D. D’Andre, Fabio M. Iwamoto, Edward J. Dropcho, Yaron A. Moshel, Kevin A. Walter, Clement P. Pillainayagam, Robert Aiken, Rekha Chaudhary, Samuel A. Goldlust, Daniela A. Bota, Paul Duic, Jai Grewal, Heinrich Elinzano, Steven A. Toms, Kevin O. Lillehei, Tom Mikkelsen, Tobias Walbert, Steven R. Abram, Andrew J. Brenner, Steven Brem, Matthew G. Ewend, Simon Khagi, Jana Portnow, Lyndon J. Kim, William G. Loudon, Reid C. Thompson, David E. Avigan, Karen L. Fink, Francois J. Geoffroy, Scott Lindhorst, Jose Lutzky, Andrew E. Sloan, Gabriele Schackert, Dietmar Krex, Hans-Jorg Meisel, Julian Wu, Raphael P. Davis, Christopher Duma, Arnold B. Etame, David Mathieu, Santosh Kesari, David Piccioni, Manfred Westphal, David S. Baskin, Pamela Z. New, Michel Lacroix, Sven-Axel May, Timothy J. Pluard, Victor Tse, Richard M. Green, John L. Villano, Michael Pearlman, Kevin Petrecca, Michael Schulder, Lynne P. Taylor, Anthony E. Maida, Robert M. Prins, Timothy F. Cloughesy, Paul Mulholland, and Marnix L. Bosch
- Subjects
Medicine - Abstract
Following publication of the original article [1], the authors reported an error in the spelling of one of the author names. In this Correction the incorrect and correct author names are indicated and the author name has been updated in the original publication. The authors also reported an error in the Methods section of the original article. In this Correction the incorrect and correct versions of the affected sentence are indicated. The original article has not been updated with regards to the error in the Methods section.
- Published
- 2018
- Full Text
- View/download PDF
8. Early Experience With Virtual and Synchronized Augmented Reality Platform for Preoperative Planning and Intraoperative Navigation: A Case Series
- Author
-
Jonathan Pace, Walter C. Jean, Gavin W. Britz, Robert G Louis, Gary K. Steinberg, Christopher Duma, Warren R. Selman, and Vivek Mehta
- Subjects
Models, Anatomic ,medicine.medical_specialty ,Neuronavigation ,medicine.medical_treatment ,AcademicSubjects/MED00930 ,Neuros/3 ,Virtual reality ,Surgical planning ,Preoperative care ,Medical imaging ,Medicine ,Humans ,Medical physics ,Case Series ,Augmented Reality ,business.industry ,Virtual Reality ,Microsurgery ,Navigation ,Dissection ,Surgery, Computer-Assisted ,Surgery ,Augmented reality ,Neurology (clinical) ,business ,Microscopic surgery ,Craniotomy - Abstract
Background Virtual reality (VR) allows for presurgical planning. Intraoperatively, augmented reality (AR) enables integration of segmented anatomic information with neuronavigation into the microsurgical scene to provide guidance without workflow disruption. Combining VR and AR solutions may help guide microsurgical technique to improve safety, efficiency, and ergonomics. Objective To describe a VR/AR platform that provides VR planning and intraoperative guidance via microscope ocular injection of a comprehensive AR overlay of patient-specific 360°/3D anatomic model aligned and synchronized with neuronavigation. Methods Custom 360° models from preoperative imaging of 49 patients were utilized for preoperative planning using a VR-based surgical rehearsal platform. Each model was imported to SyncAR, the platform's intraoperative counterpart, which was coregistered with Medtronic StealthStation S8 and Zeiss or Leica microscope. The model was injected into the microscope oculars and referenced throughout by adjusting overlay opacity. For anatomic shifts or misalignment, the overlay was reregistered via manual realignment with known landmarks. Results No SyncAR-related complications occurred. SyncAR contributed positively to the 3D understanding of patient-specific anatomy and ability to operate. Preoperative planning and intraoperative AR with 360° models allowed for more precise craniotomy planning and execution. SyncAR was useful for guiding dissection, identifying critical structures including hidden anatomy, understanding regional anatomy, and facilitating resection. Manual realignment was performed in 48/49 surgeries. Gross total resection was achieved in 34/40 surgeries. All aneurysm clipping and microvascular decompression procedures were completed without complications. Conclusion SyncAR combined with VR planning has potential to enhance surgical performance by providing critical information in a user-friendly, continuously available, heads-up display format.
- Published
- 2021
9. Results of three or more Gamma Knife radiosurgery procedures for recurrent trigeminal neuralgia
- Author
-
Pate J Duddleston, Anudeep Yekula, Varun Sagi, Mihir Gupta, Sharona Ben-Haim, Kenneth Ott, Steven J. Goetsch, Devan Hawkins, Christopher Duma, Kathleen Thomas, Kenneth T Shimizu, Aditya Mittal, Justin Shimizu, David W. Hodgens, and John F. Alksne
- Subjects
Male ,Reoperation ,medicine.medical_specialty ,Percutaneous ,Pain relief ,Gamma knife radiosurgery ,Radiosurgery ,Refractory ,Recurrence ,Trigeminal neuralgia ,medicine ,Humans ,Adverse effect ,Aged ,Retrospective Studies ,Aged, 80 and over ,business.industry ,Multiple sclerosis ,General Medicine ,Middle Aged ,Trigeminal Neuralgia ,medicine.disease ,Surgery ,Treatment Outcome ,Facial twitching ,Female ,business - Abstract
OBJECTIVE Gamma Knife radiosurgery (GKRS) is an established surgical option for the treatment of trigeminal neuralgia (TN), particularly for high-risk surgical candidates and those with recurrent pain. However, outcomes after three or more GKRS treatments have rarely been reported. Herein, the authors reviewed outcomes among patients who had undergone three or more GKRS procedures for recurrent TN. METHODS The authors conducted a multicenter retrospective analysis of patients who had undergone at least three GKRS treatments for TN between July 1997 and April 2019 at two different institutions. Clinical characteristics, radiosurgical dosimetry and technique, pain outcomes, and complications were reviewed. Pain outcomes were scored on the Barrow Neurological Institute (BNI) scale, including time to pain relief (BNI score ≤ III) and recurrence (BNI score > III). RESULTS A total of 30 patients were identified, including 16 women and 14 men. Median pain duration prior to the first GKRS treatment was 10 years. Three patients (10%) had multiple sclerosis. Time to pain relief was longer after the third treatment (p = 0.0003), whereas time to pain recurrence was similar across each of the successive treatments (p = 0.842). Complete or partial pain relief was achieved in 93.1% of patients after the third treatment. The maximum pain relief achieved after the third treatment was significantly better among patients with no prior percutaneous procedures (p = 0.0111) and patients with shorter durations of pain before initiation of GKRS therapy (p = 0.0449). New or progressive facial sensory dysfunction occurred in 29% of patients after the third GKRS treatment and was reported as bothersome in 14%. One patient developed facial twitching, while another experienced persistent lacrimation. No statistically significant predictors of adverse effects following the third treatment were found. Over a median of 39 months of follow-up, 77% of patients maintained complete or partial pain relief. Three patients underwent a fourth GKRS treatment, including one who ultimately received five treatments; all of them reported sustained pain relief at the extended follow-up. CONCLUSIONS The authors describe the largest series to date of patients undergoing three or more GKRS treatments for refractory TN. A third treatment may produce outcomes similar to those of the first two treatments in terms of long-term pain relief, recurrence, and adverse effects.
- Published
- 2021
- Full Text
- View/download PDF
10. CTIM-33. PHASE II TRIAL OF VACCINE IMMUNOTHERAPY IN PRIMARY GLIOBLASTOMA: ADJUNCTIVE AUTOLOGOUS DENDRITIC CELLS PULSED WITH LYSATE FROM IRRADIATED SELF-RENEWING AUTOLOGOUS TUMOR CELLS (AV-GBM-1)
- Author
-
Renato V. LaRocca, Robert O Dillman, Gabriel Nistor, Mehrdad Abedi, Jose Carrillo, Thomas H. Taylor, Frank P.K. Hsu, Christopher Duma, Robert Aiken, Santosh Kesari, David Piccioni, Candace Hsieh, Xiao-Tang Kong, and Daniela A. Bota
- Subjects
Cancer Research ,medicine.medical_treatment ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Phases of clinical research ,26th Annual Meeting & Education Day of the Society for Neuro-Oncology ,Vaccine Related ,Rare Diseases ,Clinical Research ,Medicine ,Oncology & Carcinogenesis ,Progression-free survival ,Interleukin 4 ,Cancer ,Temozolomide ,business.industry ,Neurosciences ,Evaluation of treatments and therapeutic interventions ,Leukapheresis ,Immunotherapy ,Brain Disorders ,Brain Cancer ,Granulocyte macrophage colony-stimulating factor ,Oncology ,Cell culture ,6.1 Pharmaceuticals ,Cancer research ,Immunization ,Neurology (clinical) ,business ,Biotechnology ,medicine.drug - Abstract
In primary glioblastoma (GBM), overall survival (OS) is poor despite standard aggressive therapy. Adjunctive AV-GBM-1 vaccine immunotherapy may improve OS. In this multi-institutional phase II trial, key eligibility criteria for intent-to-treat (ITT) enrollment were: (1) primary GBM, (2) age < 70 years when GBM was resected, (3) successful GBM cell culture, (4) successful monocyte collection by leukapheresis, (5) KPS > 70 post-surgery, and (6) plan to treat with concurrent RT/TMZ. Dendritic cells (DC) were differentiated from monocytes by culturing in IL-4 and granulocyte-macrophage colony stimulating factor (GM-CSF). AV-GBM-1 consisted of autologous DC incubated with autologous tumor antigens contained in the lysate of irradiated cultured GBM cells. After recovery from RT/TMZ, doses were admixed with 500 mcg GM-CSF; up to 8 doses were injected subcutaneously over 6 months. Patients were not excluded by apparent progression or pseudo-progression post RT/TMZ. OS and progression-free-survival (PFS) were calculated from ITT enrollment. The success rate was 97% for both GBM cell cultures and collection of monocytes; 60/60 vaccines were successfully manufactured. Median age was 59 years. 57 patients received 392 injections. After two weekly injections there were significant increases in plasma lipocalin-2 and angiopoietin-1, and decreases in thrombospondin-5, angiotensinogen, and beta-fibroblast growth factor. The most common adverse events attributed to AV-GBM-1 were local injection site reactions (16%) and flu-like symptoms (10%). With follow up from 15.2 to 32 months, median PFS and OS were 10.3 (8.5,11.6 95% CI) and 16.0 (13.0,21.3 95% CI) months respectively. OS was better in the 25 patients who had methylguanine-methyltransferase (MGMT) methylation and/or isocitrate dehydrogenase (IDH) mutation. Age was not independently correlated with survival. From date of first injection, OS was not increased in 14 patients who were treated with alternating electrical tumor-treating fields. CONCLUSION: feasibility, safety, and PFS were encouraging. A phase III trial is in development. Clinicaltrials.gov NCT03400917.
- Published
- 2021
11. 952 Phase II trial of AV-GBM-1: dendritic cell vaccine pulsed with lysate enriched for autologous tumor-initiating cell antigens in the treatment of patients with newly diagnosed glioblastoma
- Author
-
Thomas H. Taylor, Renato V. LaRocca, Daniela A. Bota, Jose Carrillo, Mehrdad Abedi, Robert O. Dillman, David Piccioni, Xiao-Tang Kong, Candace Hsieh, Gabriel Nistor, Christopher Duma, Robert Aiken, Santosh Kesari, and Frank P.K. Hsu
- Subjects
Pharmacology ,Cancer Research ,Lysis ,business.industry ,Immunology ,Cell ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Newly diagnosed ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Antigen ,Dendritic cell vaccine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business ,RC254-282 ,Autologous tumor ,Glioblastoma - Abstract
BackgroundStandard therapy of glioblastoma (GBM), which includes maximum safe resection, concurrent radiation therapy and temozolomide chemotherapy (RT/TMZ) followed by maintenance TMZ, is associated with poor overall survival (OS). Adding treatment with AV-GBM-1, a vaccine consisting of autologous dendritic cells (DC) pulsed with autologous tumor antigens (ATA) may improve OS. A multi-center phase II clinical trial was conducted to determine feasibility, safety, and efficacy of AV-GBM-1.MethodsKey eligibility criteria for tumor collection were clinical suspicion of newly diagnosed GBM and age 18 to 70 years at the time of surgery. Prior to starting RT/TMZ, key eligibility criteria for intent-to-treat-with-AV-GBM-1 enrollment were: (1) primary GBM confirmed, (2) successful GBM cell culture, (3) collection of sufficient numbers of monocytes (MC) by leukapheresis, (4) Karnofsky Performance Status 70 or greater and (5) plan to treat with concurrent RT/TMZ. AV-GBM-1 was manufactured during RT/TMZ. Interleukin-4 and granulocyte-macrophage colony stimulating factor (GM-CSF) were used to differentiate MC into DC. AV-GBM-1 consists of autologous DC incubated with ATA from the lysate of irradiated GBM cells grown in serum-free media with factors that favor the survival and proliferation of stem cells and early progenitor cells. After recovery from RT/TMZ, over six months patients received up to 8 subcutaneous injections of AV-GBM-1 admixed with 500 μg GM-CSF. The primary objective was to determine if OS was 75% or higher 14.6 months from ITT enrollment, which ended January 2020. The minimum follow-up at the time of analysis was 15.2 months. Secondary endpoints included progression-free survival (PDS) from ITT enrollment and from the first injection.ResultsSuccess rates for cell cultures and sufficient monocyte collections were both 97%. AV-GBM-1 was manufactured for 60/60 (100%). 57 patients received 392 injections; 68% received all 8. The primary adverse events (AE) attributed to AV-GBM-1 were local injection site reactions (16%) and flu-like symptoms (10%). Progression-free survival (PFS) from ITT enrollment is 10.3 months, about 50% longer than reported in four randomized trials with comparable standard therapy arms. PFS from the first injection is 8.3 months, which is 51% and 107% longer than reported in two randomized trials with comparable standard therapy arms. OS was 72% at 12 months, but dropped to 54% at 14.6 months; median OS is 16.0 months.ConclusionsPatent-specific AV-GBM-1 was reliably manufactured and distributed for administration. AV-GBM-1 produced minimal toxicity. PFS was very encouraging but did not translate into OS, perhaps because of discontinuation of treatment after 8 months.Trial Registration[Clinicaltrials.gov NCT03400917]Ethics ApprovalWestern IRB, approval number 20182582Consent n/a
- Published
- 2021
12. 331 Tumor markers associated with increased survival in a phase II trial of dendritic cell/tumor-initiating-cell vaccine AV-GBM-1 in patients with newly diagnosed glioblastoma
- Author
-
Renato V. LaRocca, Frank P.K. Hsu, Robert O. Dillman, Christopher Duma, Candace Hsieh, Santosh Kesari, Gabriel Nistor, Mehrdad Abedi, David Piccioni, Thomas H. Taylor, Jose Carrillo, Robert Aiken, Xiao-Tang Kong, and Daniela A. Bota
- Subjects
Pharmacology ,Cancer Research ,business.industry ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Dendritic cell ,Newly diagnosed ,Tumor initiating cell ,medicine.disease ,Oncology ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,In patient ,business ,RC254-282 ,Glioblastoma - Abstract
BackgroundStandard aggressive therapy of glioblastoma (GBM), which includes maximum safe resection, concurrent radiation therapy and temozolomide chemotherapy (RT/TMZ) followed by maintenance TMZ, is associated with a 25% 2-year overall survival (OS). Adding treatment with AV-GBM-1, a vaccine consisting of autologous dendritic cells (DC) pulsed with autologous tumor antigens (ATA) may improve OS by inducing and/or enhancing the host anti-GBM immune response. Methylation of the O-6-methylguanine-DNA methyltransferase (MGMT) gene promoter, and mutation of the gene for isocitrate dehydrogenase (IDH) are favorable prognostic markers in newly diagnosed GBM. An objective of a multi-center phase II clinical trial was to determine whether these markers were still prognostic for OS in patients treated with adjunctive AV-GBM-1.MethodsKey eligibility criteria for intent-to-treat (ITT) enrollment were: (1) confirmation of primary GBM, (2) successful GBM cell culture, (3) collection of sufficient numbers of monocytes (MC) by leukapheresis, (4) Karnofsky Performance Status 70 or greater after recovery from surgery, and (5) plan to treat with concurrent RT/TMZ. AV-GBM-1 was manufactured while patients were being treated with RT/TMZ. Interleukin-4 and granulocyte-macrophage colony stimulating factor (GM-CSF) were used to differentiate DC from MC. Each vaccine consisted of autologous DC incubated with ATA from the lysate of irradiated cultured GBM cells grown in serum-free media with factors that favor survival and proliferation of stem cells and early progenitor cells (tumor-initiating cells). After recovery from RT/TMZ, intent was to vaccinate for up to six months with cryopreserved AV-GBM-1 admixed with 500 mg GM-CSF. All patients had testing for MGMT-methylation and IDH-mutation. OS was calculated from date of ITT enrollment.Results60 patients were enrolled during August 2018 to January 2020. MGMT promoter methylation was detected in 21 (35%), mutated IDH in 7 (12%), and one or both in 25 (42%). At a minimum follow-up of 15 months, median OS had not been reached for patients with a methylated MGMT promotor, IDH mutation, or one or both, compared to 14.6 months for 38 with unmethylated MGMT promotor (p=0.026), 14.7 months for 53 with IDH wild-type (p=0.044), and 14.6 months for 35 who had neither (p=0.017). 18-month OS rates were 59% vs 35% for MGMT promotor methylation, 71% vs 40% for IDH mutation and 58% vs 32% for either.ConclusionsBoth MGMT promotor methylation and IDH mutation were associated with a substantial and similar survival benefit in primary GBM patients treated with AV-GBM-1 in addition to standard aggressive therapy.Trial RegistrationClinicaltrialsgov NCT03400917Ethics ApprovalThis study was approved by the Western IRB, approval number 20182582; all participants gave written informed consent before taking part
- Published
- 2021
13. Human intracerebroventricular (ICV) injection of autologous, non-engineered, adipose-derived stromal vascular fraction (ADSVF) for neurodegenerative disorders: results of a 3-year phase 1 study of 113 injections in 31 patients
- Author
-
Elisa Singh, Oleg V. Kopyov, Anthony Bravo, Benjamin Rapaport, Michael Arata, Ami R Sanathara, Kristin Scord, Sawyer Farmer, Chad A. Caraway, Sean Berman, Mark Berman, Michael Elam, Ruslana Cannell, Rory McRory, Alex Kopyov, Christian Yassa, Christopher Duma, Samuel Khoudari, Karlyssa Chung, Lian Stemler, David Weiland, Elliot B Lander, and Chace Duma
- Subjects
Adult ,Male ,0301 basic medicine ,Traumatic brain injury ,Adipose tissue ,Mesenchymal Stem Cell Transplantation ,Transplantation, Autologous ,Ventriculoperitoneal Shunt ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Humans ,Amyotrophic lateral sclerosis ,Molecular Biology ,Stroke ,Spinal cord injury ,Dexamethasone ,Aged ,Aged, 80 and over ,business.industry ,Multiple sclerosis ,Hematopoietic Stem Cell Transplantation ,Neurodegenerative Diseases ,General Medicine ,Middle Aged ,Stromal vascular fraction ,Hematopoietic Stem Cells ,medicine.disease ,Infusions, Intraventricular ,Treatment Outcome ,030104 developmental biology ,Adipose Tissue ,030220 oncology & carcinogenesis ,Anesthesia ,Female ,business ,medicine.drug - Abstract
We have chosen to test the safety of human intracerebroventricular (ICV) brain injections of autologous non-genetically-modified adipose-derived stromal vascular fraction (ADSVF). In this IRB-approved trial, 24 patients received ICV ADSVF via an implanted reservoir between 5/22/14 and 5/22/17. Seven others were injected via their ventriculo-peritoneal shunts. Ten patients had Alzheimer’s disease (AD), 6 had amyotrophic lateral sclerosis (ALS), 6 had progressive multiple sclerosis (MS-P), 6 had Parkinson’s “Plus” (PD+), 1 had spinal cord injury, 1 had traumatic brain injury, and 1 had stroke. Median age was 74 (range 41–83). Injections were planned every 2–3 months. Thirty-one patients had 113 injections. Patients received SVF injection volumes of 3.5–20 cc (median:4 cc) containing 4.05 × 105 to 6.2 × 107 cells/cc, which contained an average of 8% hematopoietic and 7.5% adipose stem cells. Follow-up ranged from 0 to 36 months (median: 9.2 months). MRIs post injection(s) were unchanged, except for one AD patient whose hippocampal volume increased from
- Published
- 2019
- Full Text
- View/download PDF
14. Surgery, Concurrent Radiation Therapy and Chemotherapy Followed by Injections of Autologous Dendritic Cell Vaccines Pulsed With Wntigens From Self-renewing Autologous Tumor Cells in the Treatment of Primary Glioblastoma
- Author
-
Renato V. LaRocca, Adrienne M. Wang, Thomas H. Taylor, David Piccioni, Chris J. Langford, Jim Langford, Christopher Duma, Sawyer Farmer, Santosh Kesari, Jose Carrillo, Candace Hsieh, Robert T. O'Donnell, Krystal Godding, Gabriel Nistor, Daniela A. Bota, Robert O. Dillman, Robert Aiken, and Ashley Harris
- Subjects
Chemotherapy ,Temozolomide ,business.industry ,medicine.medical_treatment ,Dendritic cell ,Leukapheresis ,Chemotherapy regimen ,Radiation therapy ,Granulocyte macrophage colony-stimulating factor ,medicine ,Cancer research ,Surgery ,Neurology (clinical) ,business ,Tumor marker ,medicine.drug - Published
- 2020
- Full Text
- View/download PDF
15. Results of Multiple Gamma Knife Radiosurgery Treatments for Recurrent Trigeminal Neuralgia
- Author
-
Steven J. Goetsch, Varun Sagi, John F. Alksne, Sharona Ben-Haim, Kenneth Ott, Pate J Duddleston, Justin Shimizu, Anudeep Yekula, Mihir Gupta, Kathleen Thomas, Kenneth T Shimizu, Aditya Mittal, and Christopher Duma
- Subjects
medicine.medical_specialty ,Hypesthesia ,business.industry ,medicine.medical_treatment ,Gamma knife radiosurgery ,Gamma knife ,medicine.disease ,Radiosurgery ,SENSORY DISORDERS ,Trigeminal neuralgia ,medicine ,Surgery ,Neurology (clinical) ,Radiology ,business - Published
- 2020
- Full Text
- View/download PDF
16. CTIM-26. PATIENT-SPECIFIC DENDRITIC CELL VACCINE (DC-ATA) PULSED WITH ANTIGENS FROM SELF-RENEWING AUTOLOGOUS TUMOR CELLS IN THE TREATMENT OF NEWLY-DIAGNOSED GLIOBLASTOMA: A PHASE II TRIAL
- Author
-
Renato V. LaRocca, Daniela A. Bota, Candace Hsieh, Frank P.K. Hsu, David Piccioni, Adrienne M. Wang, Robert O. Dillman, Gabriel Nistor, Robert Aiken, Christopher Duma, Krystal Carta, Santosh Kesari, Jose Carrillo, James A. Langford, Thomas H. Taylor, Robert T. O'Donnell, Chris J. Langford, and Xiao-Tang Kong
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Oncology and Carcinogenesis ,Phases of clinical research ,Immunoglobulin E ,Clinical Trials: Immunologic ,Vaccine Related ,Rare Diseases ,Antigen ,Clinical Research ,Internal medicine ,medicine ,Oncology & Carcinogenesis ,Survival analysis ,Tumor marker ,Cancer ,biology ,business.industry ,Standard treatment ,Prevention ,Neurosciences ,Evaluation of treatments and therapeutic interventions ,Immunotherapy ,Brain Disorders ,Clinical trial ,Brain Cancer ,Good Health and Well Being ,6.1 Pharmaceuticals ,biology.protein ,Immunization ,Neurology (clinical) ,business - Abstract
Author(s): Bota, Daniela; Piccioni, David; LaRocca, R; Duma, Christopher; Kesari, Santosh; Carrillo, Jose; O’Donnell, Robert T; Aiken, Robert; Hsu, Frank; Kong, Xiao-Tang; Hsieh, Candace; Langford, Chris; Carta, Krystal; Wang, Adrienne; Langford, James; Taylor, Thomas; Nistor, Gabriel; Dillman, Robert | Abstract: Abstract GBM standard treatment is associated with poor survival. Adjunctive therapy with patient-specific vaccines may improve outcomes by enhancing anti-GBM immune responses. A multi-institutional phase II clinical trial was designed with a primary objective of 75% survival 15 months after intent-to-treat enrollment. IL-4 and GM-CSF were used to generate dendritic cells (DC) from monocytes. DC were incubated with autologous tumor antigens (ATA) from the lysate of cultured GBM cells to produce each patient-specific DC-ATA vaccine. Each dose was admixed with 500 mcg GM-CSF at the time of subcutaneous injections at weeks 1, 2, 3, 8, 12, 16, 20 and 24. Enrollment has been completed in April 2020 (n=60). Three patients withdrew from the study prior to starting treatment leaving 57 patients for whom data is available. So far 57 patients have received 344 doses; 27 have completed all 8 doses, 11 received fewer than 8 doses at the time they discontinued treatment, 19 are currently in treatment. No patient has discontinued treatment because of toxicity. 9 pt had died and the preliminary 12 months overall survival is 74%. In a preliminary serologic analysis 12 of 16 patients (75%) had an increase in markers associated with Th1/NK, Th2/immunoglobulins, and Th2 hypersensitivity (eotaxins, IgE and IL17F) by week-3; 9 of 15 (60%) had a decrease in angiogenesis factors, growth factors, and tumor markers by week-8. Immunologic data for all 55 patients who received at least two injections will be available November 2020. This patient-specific DC-ATA immunotherapy approach is feasible, is associated with changes in serologic markers, and may be increasing intratumor inflammation that may be associated with on-target toxicity and efficacy. A interim survival analysis will be conducted in mid-October 2020, 15 months after the 28th patient was enrolled; results will be available November 2020 [Clinicaltrials.gov NCT03400917].
- Published
- 2020
17. 319 Phase II trial of immunotherapy in primary glioblastoma: antigens from self-renewing autologous tumor cells presented by autologous dendritic cell vaccine
- Author
-
Christopher Duma, Santosh Kesari, Robert Aiken, Gabriel Nistor, Robert O. Dillman, Robert T. O'Donnell, Xiao-Tang Kong, Frank P.K. Hsu, Thomas H. Taylor, David Piccioni, Renato V. LaRocca, Daniela A. Bota, and Jose Carrillo
- Subjects
Oncology ,medicine.medical_specialty ,Temozolomide ,business.industry ,medicine.medical_treatment ,Autologous Monocytes ,Leukapheresis ,Immunotherapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Clinical trial ,Radiation therapy ,Internal medicine ,medicine ,Adverse effect ,business ,Survival analysis ,medicine.drug - Abstract
Background Primary glioblastoma (GBM) is associated with poor survival. Adjunctive vaccines may improve survival by inducing or enhancing anti-GBM immune responses. Methods A multi-institutional phase II clinical trial was conducted with a primary objective of 75% survival 15 months after intent-to-treat enrollment. Key eligibility criteria were: (1) primary GBM diagnosis, (2) age 70 after surgical recovery. Dendritic cells (DC) were differentiated from autologous monocytes, then incubated with autologous tumor antigens (ATA) from the GBM cell line-lysate to produce each patient-specific DC-ATA vaccine. Doses were suspended in 500 mcg granulocyte-macrophage colony-stimulating factor (GM-CSF) at the time of subcutaneous injections at weeks 1, 2, 3, 8, 12, 16, 20 and 24. Patients were enrolled just prior to starting standard concurrent temozolomide (TMZ) and radiation therapy (RT) for the intent-to-treat after recovery from RT/TMZ. Results Tumors were collected August 2018-January 2020. Cell line success rate was 71/73 (97%); monocyte collection success rate was 63/65 (97%), but 10 patients required a second leukapheresis. Patients were enrolled for in-to-treat October 2018-February 2020. The 60 patients included 42 men and 18 women with median age of 59 years (range of 27–70). Racial make-up was 43 White, 10 Hispanic, 2 Black, 1 Asian and 3 Other. KPS was 100 in 4, 90 in 25, 80 in 17 and 70 in 14 (mean 83.2). MGMT methylation was present in 13, absent in 31, and unknown in 16; IDH mutation was present in 7, absent in 50, and unknown in 3. 57 patients had received 380 doses with 9 still under treatment at time of abstract submission. 32 had completed all 8 doses; 16 had received fewer than 8 doses when they discontinued treatment. No patient discontinued treatment because of toxicity, but 28 have been hospitalized for 53 treatment-emergent central nervous system-related serious adverse events including seizures (15 episodes), falls and/or increased focal weakness (13 episodes), or severe headaches or visual changes (3 episodes). Conclusions This patient-specific DC-ATA approach is feasible and may be increasing intratumor inflammation that is associated with on-target efficacy and/or toxicity. An interim survival analysis will be conducted in October 2020, 15 months after the median patient was enrolled; results will be available November 2020 as will immunologic data for 55 patients who received at least two injections. Trial Registration Clinicaltrials. gov NCT03400917. Ethics Approval The study was approved by UCI IRB, approval number 2018-4148.
- Published
- 2020
- Full Text
- View/download PDF
18. 332 Tumor collection and establishment of tumor-initiating cell cultures as antigen source for AV-GBM-1 dendritic cell vaccines for a phase II trial in patients with newly diagnosed glioblastoma
- Author
-
Candace Hsieh, Robert O. Dillman, Frank P.K. Hsu, Gabriel Nistor, David Piccioni, Christopher Duma, Santosh Kesari, Jose Carrillo, Robert Aiken, Thomas H. Taylor, Daniela A. Bota, Xiao-Tang Kong, Renato V. LaRocca, and Mehrdad Abedi
- Subjects
Pharmacology ,Cancer Research ,business.industry ,Immunology ,Dendritic cell ,Newly diagnosed ,Tumor initiating cell ,medicine.disease ,Oncology ,Antigen ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Medicine ,In patient ,business ,Glioblastoma - Abstract
BackgroundDespite standard aggressive therapy (maximum safe surgical resection, concurrent radiation therapy and temozolomide chemotherapy (RT/TMZ), then maintenance TMZ), 2-year survival is only about 25% for patients with newly diagnosed primary glioblastoma (GBM). Adding AV-GBM-1, a vaccine consisting of autologous dendritic cells (DC) pulsed with autologous tumor antigens (ATA) may improve survival. One objective of a multi-center phase II clinical trial was to determine the feasibility of collecting fresh GBM and establishing short-term cell cultures of GBM tumor-initiating cells (TIC) to serve as ATA source.MethodsKey eligibility criteria for tumor collection were (1) clinical suspicion of new primary GBM, (2) age 18 to 70 years (3) tentative agreement to undergo a leukapheresis procedure after recovery from surgery, and (4) tentative plans for RT/TMZ. Fresh tumor was placed in media and shipped in a transport kit by overnight courier to AIVITA where a cell suspension was placed in culture and incubated in serum-free medium with factors that favor survival and proliferation of TICS (stem cells and early progenitor cells). The intent was to produce a patient-specific DC-ATA vaccine by incubating a lysate of irradiated TICs with autologous DC for subsequent subcutaneous injection.ResultsPatients were enrolled from five sites in California, one in Kentucky and one in New Jersey. Tumors were collected between August 2018 and January 2020. 106 patients consented for tumor collection, but 15 were not GBM, 4 had insufficient tissue to send, 2 patients withdrew consent, 4 were ineligible because of age, and 1 was ineligible because of autoimmune disease. Of the 80 GBM tumors that were placed into culture, 7 were discontinued because of patient withdrawal. 71/73 (97%) resulted in a successful cell culture; two were unsuccessful because of contamination. 60/71 subsequently consented for intent-to-treat ; 46/60 (77%) had cells in culture for 28 days or less, 11 were in culture for 30 to 35 days, and the remaining 3 were cultured 46, 54, and 55 days. The average number of cells per culture at the time of irradiation was 14.0 million (range 0.78 to 63.3 million). 58/60 (97%) yielded more than 1 million TICs for irradiation for the tumor cell lysate; 36/60 (60%) had more than 10 million cells irradiated. 57 patients were subsequently treated with AV-GBM-1 after recovery from RT/TMZ.ConclusionsSelf-renewing GBM TIC cultures can be reliably and rapidly established for use as the antigen source for personal DC-ATA vaccines.Trial RegistrationClinicaltrialsgov NCT03400917Ethics ApprovalThis study was approved by the Western IRB, approval number 20182582; all participants gave written informed consent before taking part
- Published
- 2021
- Full Text
- View/download PDF
19. 336 Adverse events in a phase II trial of AV-GBM-1: dendritic cell vaccine pulsed with lysate enriched for autologous tumor-initiating cell antigens for patients with newly diagnosed glioblastoma
- Author
-
David Piccioni, Daniela Bota, Renato LaRocca, Santosh Kesari, Jose Carillo, Xiao-Tang Kong, Christopher Duma, Mehrdad Abedi, Robert Aiken, Thomas Taylor, Candace Hsieh, Gabriel Nistor, and Robert Dillman
- Subjects
Pharmacology ,Cancer Research ,Lysis ,business.industry ,Immunology ,Cell ,Newly diagnosed ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Dendritic cell vaccine ,Antigen ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Adverse effect ,business ,Glioblastoma ,Autologous tumor - Abstract
BackgroundStandard glioblastoma (GBM) therapy includes maximum safe resection, concurrent radiation therapy and temozolomide chemotherapy (RT/TMZ), and maintenance TMZ, but it is associated with a 2-year survival of only about 25%. Adding treatment with AV-GBM-1, a vaccine consisting of autologous dendritic cells (DC) pulsed with autologous tumor antigens (ATA) may improve survival, but it may be associated with additional toxicity. One objective of a multi-center phase II clinical trial was to identify, characterize, and enumerate treatment-emergent adverse events (TEAE) and serious AE (SAE) that occurred during AV-GBM-1 treatment.MethodsKey eligibility criteria for enrollment prior to starting RT/TMZ, were: (1) confirmation of primary GBM, (2) successful GBM cell culture, (3) collection of sufficient numbers of monocytes (MC) from leukapheresis, (4) Karnofsky Performance Status 70 or higher, and (5) planning to treat with concurrent RT/TMZ. Interleukin-4 and granulocyte-macrophage colony stimulating factor (GM-CSF) were used to differentiate DC from MC. AV-GBM-1 was manufactured while patients were being treated with RT/TMZ. Each vaccine consisted of autologous DC incubated with ATA from the lysate of irradiated GBM cells grown in serum-free media with factors that favor survival and proliferation of tumor initiating cells, i.e., tumor stem cells and early progenitor cells. Following RT/TMZ, patients were injected subcutaneously with cryopreserved AV-GBM-1 admixed with 500 μg GM-CSF at weeks 1, 2, 3, 8, 12, 16, 20 and 24. Adverse events (AE) were identified and classified per Common Terminology Criteria for Adverse Events (CTCAE v 4.03).Results57 patients received at least one injection of AV-GBM-1 during November 2018 to October 2020. Patients received an average of 6.9 injections; 39 (68.4%) received all 8 injections. Injections generally were well-tolerated. Only 26 AE were attributed to AV-GBM-1, 24 grade-1 and 2 grade-2, including injection site reactions (16%), flu-like symptoms (10%), and bone discomfort (7%). The most frequent TEAE were fatigue (54)%, headache (37%), seizures (33%), nausea (30%), and focal weakness (28%). The frequency of seizures is higher than reported in other GBM trials; one patient discontinued AV-GBM-1 because of seizures. There were 55 SAE reported for 29 patients, including hospitalizations for 16 episodes of seizures in 13 patients, 7 falls in 6 patients, 6 episodes of focal weakness in 4 patients, and 3 for cerebral edema.ConclusionsAV-GBM-1 was well-tolerated, but it was associated with a high frequency of TEAE and SAE. The high frequency of focal neurologic events may be secondary to local inflammation induced by AV-GBM-1.Trial RegistrationClinicaltrialsgov NCT03400917Ethics ApprovalThis study was approved by the Western IRB, approval number 20182582; all participants gave written informed consent before taking part
- Published
- 2021
- Full Text
- View/download PDF
20. 333 Changes in proteomic markers after injections of personal AV-GBM-1 dendritic cell/tumor initiating cell vaccines in a phase II trial in patients with newly diagnosed glioblastoma
- Author
-
Thomas H. Taylor, Candace Hsieh, Xiao-Tang Kong, Frank P.K. Hsu, Gabriel Nistor, Christopher Duma, Santosh Kesari, Jose Carrillo, Aleksandra J. Poole, David Piccioni, Daniela A. Bota, Renato V. LaRocca, Mehrdad Abedi, Robert Aiken, and Robert O. Dillman
- Subjects
Pharmacology ,Cancer Research ,business.industry ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Newly diagnosed ,Dendritic cell ,medicine.disease ,Tumor initiating cell ,Oncology ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,In patient ,business ,RC254-282 ,Glioblastoma - Abstract
BackgroundDespite standard aggressive therapy, including maximum safe surgical resection, concurrent radiation therapy and temozolomide chemotherapy (RT/TMZ) followed by maintenance TMZ, survival is still extremely poor for patients with newly diagnosed primary glioblastoma (GBM). Adding treatment with AV-GBM-1, a personal vaccine consisting of autologous dendritic cells (DC) pulsed with autologous tumor antigens (ATA) may improve survival. One objective of a multi-center phase II clinical trial was to determine changes in blood proteomics before and after injections of AV-GBM-1.MethodsAV-GBM-1 consists of autologous DC incubated with ATA from a lysate of irradiated autologous GBM cells that had been placed in culture and incubated in serum-free medium with factors that favor the survival and proliferation of stem cells and early progenitor cells. After recovery from RT/TMZ, GBM patients were injected subcutaneously with AV-GBM-1 admixed in granulocyte-macrophage colony-stimulating factor (GM-CSF) at weeks 1, 2, 3, 8, 12, 16, 20, and 24. Blood samples obtained at baseline (week-0), just prior to the third injection (week-2) and just prior to the fourth injection (week-8), were cryopreserved and subsequently analyzed for 448 proteomic markers using quantitative, multiplex enzyme-linked immunosorbent assays (Raybiotech, Inc., Norcross, GA.). In this preliminary analysis the averages of paired samples for each time point were determined and compared using the student T-Test with a focus on differences of pResultsPatients were enrolled from five sites in California, and one each in Kentucky and New Jersey. 57 patients were treated during November 2018 to October 2020. Paired samples from all three time points were available for 49 patients. After two weekly injections there were increases in thymus-and activation-regulated chemokine (TARC, CCL17), the chemotactic protein chemerin, lipocalin-2, (expressed by macrophages and epithelium in response to inflammation) and angiopoietin-1 (suppressor of vascular inflammation), and decreases in thrombospondin-5 (possibly involved in synaptogenesis in brain repair), angiotensinogen (a precursor of all angiotensin peptides), and beta-fibroblast growth factor (important in tissue repair). The increase in TARC (pConclusionsIf there were humoral changes in proteins associated with Th1 and Th2 responses, these were no longer present after two weekly vaccinations. More sophisticated analyses of this data set, such as principal component analysis, may be needed to understand the effects of AV-GBM-1.Trial RegistrationClinicaltrialsgov NCT03400917Ethics ApprovalThis study was approved by the Western IRB, approval number 20182582; all participants gave written informed consent before taking part
- Published
- 2021
- Full Text
- View/download PDF
21. A Novel Combined Approach for Metastatic Breast Cancer with Dural and Leptomeningeal Disease with an Impressive Clinical Outcome: A Case Study
- Author
-
Julie Taguchi, M. A. Nezami, and Christopher Duma
- Subjects
Oncology ,medicine.medical_specialty ,Poor prognosis ,business.industry ,Estrogen receptor ,medicine.disease ,Metastatic breast cancer ,Combined approach ,Breast cancer ,Internal medicine ,Progesterone receptor ,medicine ,LEPTOMENINGEAL DISEASE ,business - Abstract
Concurrent dural and leptomeningeal metastatic carcinomatosis are very rare and have a poor prognosis. Here we present a woman with advanced estrogen receptor (ER) positive and progesterone receptor (PR) positive breast cancer who presented with leptomeningeal disease. Patient underwent multi targeted epigenetic therapies applied in a protocol called MTET. She continued to respond to the interval treatment, which consisted only of the nutraceutical agents. Here we discuss her case in detail and we believe that such an example might be applied to other patients in this situation resulting clinical improvement and less toxicity.
- Published
- 2018
- Full Text
- View/download PDF
22. ATIM-28. PHASE II TRIAL OF AV-GBM-1 (AUTOLOGOUS DENDRITIC CELLS LOADED WITH TUMOR ASSOCIATED ANTIGENS) AS ADJUNCTIVE THERAPY FOLLOWING SURGERY PLUS CONCURRENT CHEMORADIATION IN NEWLY DIAGNOSED GBM PATIENTS
- Author
-
Robert O. Dillman, Thomas H. Taylor, Mohamad Alsharif, Candace Hsieh, Kong Xiao-Tang, Beverly Fu, Renato V. LaRocca, Robert Aiken, Christopher Duma, Gabriel Nistor, Daniela A. Bota, and David Picconi
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Oncology and Carcinogenesis ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Clinical endpoint ,Cytotoxic T cell ,Oncology & Carcinogenesis ,Craniotomy ,biology ,business.industry ,Adult Clinical Trials–Immunologic ,Melanoma ,Neurosciences ,Immunotherapy ,Leukapheresis ,medicine.disease ,030220 oncology & carcinogenesis ,biology.protein ,Neurology (clinical) ,Antibody ,business ,030217 neurology & neurosurgery ,Progressive disease - Abstract
Newly-diagnosed glioblastoma (GBM) patients have a limited survival (18–24 months). In the last decade immunotherapy has improved survival for patients with other malignancies, but not GBM. Herein we present the design and initial enrollment results for the AV-GBM-1 single-arm phase 2 trial. The study enrolls patients with primary GBM who have undergone craniotomy, have a tumor cell culture established, and complete satisfactory leukapheresis prior to planned concurrent chemotherapy and radiation. Patients are scheduled to receive up to 8 vaccine injections at weeks 1, 2, 3, 8, 12, 16, 20 and 24. Blood samples are collected just prior to each injection. The primary endpoint is overall survival from date of enrollment for intent-to-treat with AV-GBM-1. The study has fully enrolled 26 of planned 55 patients. The cell line success rate is 30/32, with 6 in progress; successful completion of leukapheresis is 28/29. Two patients have completed all 8 doses, two discontinued after dose 3 and dose 6 because of progressive disease; 15 are currently in treatment, and 6 are about to start treatment. There have been seven SAE, all for hospitalizations related to GBM. For the first 8 treated patients, plasma samples from baseline and weeks 2, 3, and 8 have been analyzed for immune markers by RayBiotech Life Inc. using quantitative multiplex ELISA array. Markers reflecting Th1, Th2, Th17 pathways and B-cells, natural killer cells and cytotoxic T-lymphocytes increased in 7/8 patients. Principal component analysis demonstrates correlative marker groupings with early dominance of Th1/Th17 (weeks 1 and 2) followed by Th2/immunoglobulins at week 8. These findings show that these patient-specific dendritic cell vaccines are inducing pro-inflammatory responses similar to what was observed in a previous trial in melanoma. The study is progressing efficiently. Full enrollment data may be available for presentation at the time of the annual meeting.
- Published
- 2019
23. Abstract CT182: Adjunctive treatment of primary glioblastoma with patient-specific dendritic cell vaccines pulsed with antigens from self-renewing autologous tumor cells: A phase II trial
- Author
-
Robert Aiken, James A. Langford, Robert O. Dillman, Renato V. LaRocca, D. Bota, Krystal Carta, Adrienne M. Wang, Christopher Duma, Santosh Kesari, Jose Carrillo, Thomas H. Taylor, David Piccioni, Chris J. Langford, Robert T. O'Donnell, Gabriel Nistor, and Candace Hsieh
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,business.industry ,medicine.medical_treatment ,Standard treatment ,Cancer ,Immunotherapy ,medicine.disease ,Radiation therapy ,Clinical trial ,Internal medicine ,Adjunctive treatment ,medicine ,business ,Survival rate ,medicine.drug - Abstract
Standard treatment of primary glioblastoma (GBM) is associated with poor survival. Adjunctive therapy with patient-specific vaccines may improve outcomes by inducing or enhancing each patient's anti-GBM immune response. A multi-institutional phase II clinical trial was designed with a primary objective of 75% survival 15 months after intent-to-treat enrollment. Key eligibility criteria were: (1) primary GBM diagnosis, (2) age < 70 years at time of tumor resection, (3) successful cell culture of GBM cells in serum-free media, (4) successful monocyte collection by leukapheresis, and (5) KPS > 70 after recovery from surgery. IL-4 and GM-CSF were used to generate dendritic cells (DC) from monocytes. DC were incubated with autologous tumor antigens (ATA) from the lysate of cultured GBM cells to produce each patient-specific DC-ATA vaccine. Each dose was admixed with GM-CSF at the time of subcutaneous injections at weeks 1, 2, 3, 8, 12, 16, 20 and 24. At the time of this analysis, cell line success rate is 61/63 (97%); monocyte collection success rate is 53/55 (96%), but 10 patients required a second apheresis. 50 of a planned 55 patients have enrolled after recovery from surgery, just prior to starting standard concurrent radiation therapy (RT) and temozolomide (TMZ), with intent-to-treat after recovery from RT/TMZ. The 50 patients include 36 men and 14 women; median age is 58 years with a range of 27 to 70. Average KPS is 82.8. MGMT methylation has been documented in 22% of patients and IDH mutation in 14%. 37 patients have started treatment and received 231 doses. 16 have completed all 8 doses, 7 received fewer than 8 doses, and 14 are currently undergoing treatment. No patient has discontinued treatment because of toxicity, but 20 patients have experienced 35 treatment-emergent serious adverse events including hospitalizations for falls and/or increased focal weakness (12 episodes), seizures (10 episodes), or severe headaches or visual changes (3 episodes). 6-month actual survival rate is 96% for the 28 patients at risk 6 months or longer from enrollment. Serologic analyses show that 12 of 16 patients (75%) had an increase in markers associated with Th1/NK, Th2/immunoglobulins, and Th2 hypersensitivity (eotaxins, IgE and IL17F) by week-3; 9 of 15 (60%) had a decrease in angiogenesis factors, growth factors, and tumor markers by week-8. On serial MRI scans, 7 of 13 patients (54%) have exhibited an increase in T2 (tumor) and flare (edema) for several weeks after starting DC-ATA, followed by a slow gradual decrease in both. This patient-specific DC-ATA immunotherapy approach is feasible, is associated with changes in serologic markers, and may be increasing intratumor inflammation that may be associated with on-target toxicity and efficacy. Longer follow up is needed before the survival objective can be assessed. [Clinicaltrials.gov NCT03400917] Citation Format: Daniella A. Bota, Christopher M. Duma, Santosh Kesari, David E. Piccioni, Renato V. LaRocca, Robert T. O'Donnell, Robert D. Aiken, Jose A. Carrillo, Candace Hsieh, Chris J. Langford, Krystal Carta, Adrienne M. Wang, James A. Langford, Thomas H. Taylor, Gabriel I. Nistor, Robert O. Dillman. Adjunctive treatment of primary glioblastoma with patient-specific dendritic cell vaccines pulsed with antigens from self-renewing autologous tumor cells: A phase II trial [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr CT182.
- Published
- 2020
- Full Text
- View/download PDF
24. Correction to: First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma
- Author
-
Samuel Goldlust, Tom Mikkelsen, Reid C. Thompson, David Avigan, John E. Trusheim, Marnix L. Bosch, Kevin Petrecca, David S. Baskin, David Mathieu, Sarah A. Taylor, Timothy F. Cloughesy, Jian Campian, Raphael P. Davis, Christopher Duma, Lyndon Kim, Robert M. Prins, Kevin A. Walter, Keyoumars Ashkan, Scott Lindhorst, Tobias Walbert, Andrew E. Sloan, John L. Villano, Daniela A. Bota, Simon Khagi, Santosh Kesari, David Piccioni, Michael Salacz, Manfred Westphal, Steven A. Toms, Jose Lutzky, Dietmar Krex, Linda M. Liau, Timothy J. Pluard, Paul Duic, Richard M. Green, Rekha Chaudhary, Clement Pillainayagam, Andrew Brenner, Fabio M. Iwamoto, Steven Brem, Heinrich Elinzano, Edward J. Dropcho, Karen Fink, Hans Jorg Meisel, Michael Pearlman, Michel Lacroix, Jason Heth, Julian Wu, Lynne Taylor, Kevin O. Lillehei, Stacy D. D’Andre, Pamela Z. New, Gabriele Schackert, Arnold B. Etame, David Tran, Steven R. Abram, Jai Grewal, Paul Mulholland, William G. Loudon, Sven Axel May, Francois J. Geoffroy, Victor Tse, Robert Aiken, Matthew G. Ewend, Anthony E. Maida, Michael Schulder, Jana Portnow, Charles S. Cobbs, and Yaron A. Moshel
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Immunology ,lcsh:Medicine ,Phases of clinical research ,Newly diagnosed ,Medical and Health Sciences ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,business.industry ,Research ,lcsh:R ,General Medicine ,medicine.disease ,030104 developmental biology ,Dendritic cell vaccine ,030220 oncology & carcinogenesis ,Immunotherapy ,Glioblastoma ,business ,Vaccine ,Dendritic cell - Abstract
Background Standard therapy for glioblastoma includes surgery, radiotherapy, and temozolomide. This Phase 3 trial evaluates the addition of an autologous tumor lysate-pulsed dendritic cell vaccine (DCVax®-L) to standard therapy for newly diagnosed glioblastoma. Methods After surgery and chemoradiotherapy, patients were randomized (2:1) to receive temozolomide plus DCVax-L (n = 232) or temozolomide and placebo (n = 99). Following recurrence, all patients were allowed to receive DCVax-L, without unblinding. The primary endpoint was progression free survival (PFS); the secondary endpoint was overall survival (OS). Results For the intent-to-treat (ITT) population (n = 331), median OS (mOS) was 23.1 months from surgery. Because of the cross-over trial design, nearly 90% of the ITT population received DCVax-L. For patients with methylated MGMT (n = 131), mOS was 34.7 months from surgery, with a 3-year survival of 46.4%. As of this analysis, 223 patients are ≥ 30 months past their surgery date; 67 of these (30.0%) have lived ≥ 30 months and have a Kaplan-Meier (KM)-derived mOS of 46.5 months. 182 patients are ≥ 36 months past surgery; 44 of these (24.2%) have lived ≥ 36 months and have a KM-derived mOS of 88.2 months. A population of extended survivors (n = 100) with mOS of 40.5 months, not explained by known prognostic factors, will be analyzed further. Only 2.1% of ITT patients (n = 7) had a grade 3 or 4 adverse event that was deemed at least possibly related to the vaccine. Overall adverse events with DCVax were comparable to standard therapy alone. Conclusions Addition of DCVax-L to standard therapy is feasible and safe in glioblastoma patients, and may extend survival. Trial registration Funded by Northwest Biotherapeutics; Clinicaltrials.gov number: NCT00045968; https://clinicaltrials.gov/ct2/show/NCT00045968?term=NCT00045968&rank=1; initially registered 19 September 2002
- Published
- 2018
- Full Text
- View/download PDF
25. Skull base chondrosarcoma radiosurgery: report of the North American Gamma Knife Consortium
- Author
-
Christopher Duma, Zachary A. Seymour, David Mathieu, L. Dade Lunsford, Penny K. Sneed, Aditya Iyer, Michael W. McDermott, Hideyuki Kano, Byron Young, Heyoung McBride, Jason P. Sheehan, and Douglas Kondziolka
- Subjects
Adult ,Male ,Adolescent ,medicine.medical_treatment ,Chondrosarcoma ,Skull Base Tumor ,Kaplan-Meier Estimate ,Gamma knife ,Radiosurgery ,Skull Base Neoplasms ,Disease-Free Survival ,Young Adult ,Postoperative Complications ,Overall survival ,Humans ,Medicine ,Fractionated radiation ,Aged ,Postoperative Care ,business.industry ,Middle Aged ,medicine.disease ,Combined Modality Therapy ,Survival Analysis ,Skull ,Treatment Outcome ,medicine.anatomical_structure ,Tumor progression ,North America ,Female ,Nervous System Diseases ,business ,Nuclear medicine ,Follow-Up Studies - Abstract
OBJECT Stereotactic radiosurgery (SRS) is a potentially important option for patients with skull base chondrosarcomas. The object of this study was to analyze the outcomes of SRS for chondrosarcoma patients who underwent this treatment as a part of multimodality management. METHODS Seven participating centers of the North American Gamma Knife Consortium (NAGKC) identified 46 patients who underwent SRS for skull base chondrosarcomas. Thirty-six patients had previously undergone tumor resections and 5 had been treated with fractionated radiation therapy (RT). The median tumor volume was 8.0 cm3 (range 0.9–28.2 cm3), and the median margin dose was 15 Gy (range 10.5–20 Gy). Kaplan-Meier analysis was used to calculate progression-free and overall survival rates. RESULTS At a median follow-up of 75 months after SRS, 8 patients were dead. The actuarial overall survival after SRS was 89% at 3 years, 86% at 5 years, and 76% at 10 years. Local tumor progression occurred in 10 patients. The rate of progression-free survival (PFS) after SRS was 88% at 3 years, 85% at 5 years, and 70% at 10 years. Prior RT was significantly associated with shorter PFS. Eight patients required salvage resection, and 3 patients (7%) developed adverse radiation effects. Cranial nerve deficits improved in 22 (56%) of the 39 patients who deficits before SRS. Clinical improvement after SRS was noted in patients with abducens nerve paralysis (61%), oculomotor nerve paralysis (50%), lower cranial nerve dysfunction (50%), optic neuropathy (43%), facial neuropathy (38%), trochlear nerve paralysis (33%), trigeminal neuropathy (12%), and hearing loss (10%). CONCLUSIONS Stereotactic radiosurgery for skull base chondrosarcomas is an important adjuvant option for the treatment of these rare tumors, as part of a team approach that includes initial surgical removal of symptomatic larger tumors.
- Published
- 2015
- Full Text
- View/download PDF
26. Radiation necrosis presenting as pseudoprogression (PsP) during alectinib treatment of previously radiated brain metastases in ALK -positive NSCLC: Implications for disease assessment and management
- Author
-
Christopher Duma, Sai-Hong Ignatius Ou, Veronica Rausei-Mills, Samuel J. Klempner, and Michele C. Azada
- Subjects
Adult ,Male ,Pulmonary and Respiratory Medicine ,Alectinib ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,medicine.drug_class ,Biopsy ,medicine.medical_treatment ,Radiosurgery ,Necrosis ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,medicine ,Humans ,Anaplastic lymphoma kinase ,Anaplastic Lymphoma Kinase ,Pseudoprogression ,Temozolomide ,Brain Neoplasms ,business.industry ,Receptor Protein-Tyrosine Kinases ,Middle Aged ,medicine.disease ,Magnetic Resonance Imaging ,ALK inhibitor ,Disease Progression ,Female ,business ,Progressive disease ,medicine.drug ,Brain metastasis - Abstract
Objectives Radiation necrosis presenting as pseudoprogression (PsP) is relatively common after radiation and temozolomide (TMZ) treatment in glioblastoma multiforme (GBM), especially among patients with GBM that harbors intrinsic increased responsiveness to TMZ (methylated O6-methylguanine–DNA methyltransferase [MGMT] promoter). Alectinib is a second generation ALK inhibitor that has significant CNS activity against brain metastases in anaplastic lymphoma kinase (ALK)-rearranged (ALK+) non-small cell lung cancer (NSCLC) patients. Materials and methods We report 2 ALK+ NSCLC patients who met RECIST criteria for progressive disease by central radiologic review due to increased in size from increased contrast enhancement in previously stereotactically radiated brain metastases with ongoing extra-cranial response to alectinib. In both patients alectinib was started within 4 months of completing stereotactic radiosurgery (SRS). The enlarging lesions in both patients were resected and found to have undergone extensive necrosis with no residual tumor pathologically. PsP was incorrectly classified as progressive disease even by central independent imaging review. Conclusions Treatment-related necrosis of previously SRS-treated brain metastasis during alectinib treatment can present as PsP. It may be impossible to distinguish PsP from true disease progression without a pathologic examination from resected sample. High degree of clinical suspicion, close monitoring and more sensitive imaging modalities may be needed to distinguish PsP versus progression in radiated brain lesions during alectinib treatment especially if there is no progression extra-cranially.
- Published
- 2015
- Full Text
- View/download PDF
27. First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma
- Author
-
Manfred Westphal, Dietmar Krex, Rekha Chaudhary, Christopher Duma, Jana Portnow, Tom Mikkelsen, Reid C. Thompson, Edward J. Dropcho, Charles S. Cobbs, Sven-Axel May, Santosh Kesari, Michel Lacroix, Raphael P. Davis, David Avigan, Robert M. Prins, Jian Campian, Keyoumars Ashkan, John L. Villano, Yaron A. Moshel, John E. Trusheim, David Piccioni, Timothy J. Pluard, Paul Duic, Timothy F. Cloughesy, Michael Salacz, Andrew Brenner, Daniela A. Bota, Stacy D. D’Andre, Jai Grewal, Victor Tse, Steven A. Toms, Jason Heth, Lyndon Kim, Clement Pillainayagam, David S. Baskin, David Tran, Kevin A. Walter, Steven R. Abram, Andrew E. Sloan, Richard M. Green, Julian Wu, Scott Lindhorst, Simon Khagi, Heinrich Elinzano, Matthew G. Ewend, Paul Mulholland, Fabio M. Iwamoto, Anthony E. Maida, Michael Schulder, Kevin O. Lillehei, Karen Fink, Robert Aiken, Lynne Taylor, Hans-Jorg Meisel, William G. Loudon, Arnold B. Etame, Francois J. Geoffroy, Sarah A. Taylor, Pamela Z. New, Steven Brem, Gabriele Schackert, David Mathieu, Marnix L. Bosch, Tobias Walbert, Linda M. Liau, Michael Pearlman, Kevin Petrecca, Samuel Goldlust, and Jose Lutzky
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Endpoint Determination ,Immunology ,Section (typography) ,lcsh:Medicine ,Newly diagnosed ,Cancer Vaccines ,Medical and Health Sciences ,General Biochemistry, Genetics and Molecular Biology ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Medical physics ,Aged ,Brain Neoplasms ,business.industry ,Published Erratum ,lcsh:R ,Correction ,Dendritic Cells ,General Medicine ,Middle Aged ,Prognosis ,medicine.disease ,Survival Analysis ,Spelling ,Treatment Outcome ,030104 developmental biology ,Dendritic cell vaccine ,030220 oncology & carcinogenesis ,Female ,Immunotherapy ,Glioblastoma ,business ,Vaccine ,Author name ,Dendritic cell ,Sentence - Abstract
Background Standard therapy for glioblastoma includes surgery, radiotherapy, and temozolomide. This Phase 3 trial evaluates the addition of an autologous tumor lysate-pulsed dendritic cell vaccine (DCVax®-L) to standard therapy for newly diagnosed glioblastoma. Methods After surgery and chemoradiotherapy, patients were randomized (2:1) to receive temozolomide plus DCVax-L (n = 232) or temozolomide and placebo (n = 99). Following recurrence, all patients were allowed to receive DCVax-L, without unblinding. The primary endpoint was progression free survival (PFS); the secondary endpoint was overall survival (OS). Results For the intent-to-treat (ITT) population (n = 331), median OS (mOS) was 23.1 months from surgery. Because of the cross-over trial design, nearly 90% of the ITT population received DCVax-L. For patients with methylated MGMT (n = 131), mOS was 34.7 months from surgery, with a 3-year survival of 46.4%. As of this analysis, 223 patients are ≥ 30 months past their surgery date; 67 of these (30.0%) have lived ≥ 30 months and have a Kaplan-Meier (KM)-derived mOS of 46.5 months. 182 patients are ≥ 36 months past surgery; 44 of these (24.2%) have lived ≥ 36 months and have a KM-derived mOS of 88.2 months. A population of extended survivors (n = 100) with mOS of 40.5 months, not explained by known prognostic factors, will be analyzed further. Only 2.1% of ITT patients (n = 7) had a grade 3 or 4 adverse event that was deemed at least possibly related to the vaccine. Overall adverse events with DCVax were comparable to standard therapy alone. Conclusions Addition of DCVax-L to standard therapy is feasible and safe in glioblastoma patients, and may extend survival. Trial registration Funded by Northwest Biotherapeutics; Clinicaltrials.gov number: NCT00045968; https://clinicaltrials.gov/ct2/show/NCT00045968?term=NCT00045968&rank=1; initially registered 19 September 2002
- Published
- 2018
- Full Text
- View/download PDF
28. Upfront boost Gamma Knife 'leading-edge' radiosurgery to FLAIR MRI-defined tumor migration pathways in 174 patients with glioblastoma multiforme: a 15-year assessment of a novel therapy
- Author
-
Christopher Duma, Ryan M Casserly, Nathan Oh, Brian S. Kim, Marianne Plunkett, Christopher M Owen, Daniela Alexandru Abrams, Azzurra-Sky Riley, Ruslana Cannell, Alexa Smith, Ralph Mackintosh, Daniel J Furman, Robert O. Dillman, Gustavo Mendez, David Weiland, Marlon S. Mathews, Michael Brant-Zawadzki, Ami R Sanathara, Lian Stemler, Garrett Smith, Chad A. Caraway, Burton L. Eisenberg, and Peter Chen
- Subjects
Adult ,Time Factors ,medicine.medical_treatment ,Brain tumor ,Radiosurgery ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Cell Movement ,Glioma ,Multicenter trial ,Medicine ,Humans ,Aged ,Retrospective Studies ,Aged, 80 and over ,Chemotherapy ,Temozolomide ,business.industry ,Brain Neoplasms ,Astrocytoma ,General Medicine ,Middle Aged ,medicine.disease ,Magnetic Resonance Imaging ,Radiation therapy ,Treatment Outcome ,030220 oncology & carcinogenesis ,business ,Nuclear medicine ,Glioblastoma ,030217 neurology & neurosurgery ,medicine.drug ,Follow-Up Studies - Abstract
OBJECTIVEGlioblastoma multiforme (GBM) is composed of cells that migrate through the brain along predictable white matter pathways. Targeting white matter pathways adjacent to, and leading away from, the original contrast-enhancing tumor site (termed leading-edge radiosurgery [LERS]) with single-fraction stereotactic radiosurgery as a boost to standard therapy could limit the spread of glioma cells and improve clinical outcomes.METHODSBetween December 2000 and May 2016, after an initial diagnosis of GBM and prior to or during standard radiation therapy and carmustine or temozolomide chemotherapy, 174 patients treated with radiosurgery to the leading edge (LE) of tumor cell migration were reviewed. The LE was defined as a region outside the contrast-enhancing tumor nidus, defined by FLAIR MRI. The median age of patients was 59 years (range 22–87 years). Patients underwent LERS a median of 18 days from original diagnosis. The median target volume of 48.5 cm3 (range 2.5–220.0 cm3) of LE tissue was targeted using a median dose of 8 Gy (range 6–14 Gy) at the 50% isodose line.RESULTSThe median overall survival was 23 months (mean 43 months) from diagnosis. The 2-, 3-, 5-, 7-, and 10-year actual overall survival rates after LERS were 39%, 26%, 16%, 10%, and 4%, respectively. Nine percent of patients developed treatment-related imaging-documented changes due to LERS. Nineteen percent of patients were hospitalized for management of edema, 22% for resection of a tumor cyst or new tumor bulk, and 2% for shunting to treat hydrocephalus throughout the course of their disease. Of the patients still alive, Karnofsky Performance Scale scores remained stable in 90% of patients and decreased by 1–3 grades in 10% due to symptomatic treatment-related imaging changes.CONCLUSIONSLERS is a safe and effective upfront adjunctive therapy for patients with newly diagnosed GBM. Limitations of this study include a single-center experience and single-institution determination of the LE tumor target. Use of a leading-edge calculation algorithm will be described to achieve a consistent approach to defining the LE target for general use. A multicenter trial will further elucidate its value in the treatment of GBM.
- Published
- 2016
29. Gamma Knife surgery for the management of glomus tumors: a multicenter study
- Author
-
Peter A. Weisskopf, Michael J. Link, Zhiyuan Xu, A. Byron Young, Hideyuki Kano, Huai-Che Yang, Anthony M. Kaufmann, Douglas Kondziolka, L. Dade Lunsford, Shota Tanaka, David Mathieu, Jason P. Sheehan, Christopher Duma, Heyoung McBride, and Bruce E. Pollock
- Subjects
medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Cranial nerves ,General Medicine ,medicine.disease ,Radiosurgery ,Surgery ,Glomus tumor ,Skull Base Neoplasm ,Medicine ,Cranial nerve disease ,Trigeminal Nerve Diseases ,External beam radiotherapy ,medicine.symptom ,business ,Tinnitus - Abstract
Object Glomus tumors are rare skull base neoplasms that frequently involve critical cerebrovascular structures and lower cranial nerves. Complete resection is often difficult and may increase cranial nerve deficits. Stereotactic radiosurgery has gained an increasing role in the management of glomus tumors. The authors of this study examine the outcomes after radiosurgery in a large, multicenter patient population. Methods Under the auspices of the North American Gamma Knife Consortium, 8 Gamma Knife surgery centers that treat glomus tumors combined their outcome data retrospectively. One hundred thirty-four patient procedures were included in the study (134 procedures in 132 patients, with each procedure being analyzed separately). Prior resection was performed in 51 patients, and prior fractionated external beam radiotherapy was performed in 6 patients. The patients' median age at the time of radiosurgery was 59 years. Forty percent had pulsatile tinnitus at the time of radiosurgery. The median dose to the tumor margin was 15 Gy. The median duration of follow-up was 50.5 months (range 5–220 months). Results Overall tumor control was achieved in 93% of patients at last follow-up; actuarial tumor control was 88% at 5 years postradiosurgery. Absence of trigeminal nerve dysfunction at the time of radiosurgery (p = 0.001) and higher number of isocenters (p = 0.005) were statistically associated with tumor progression–free tumor survival. Patients demonstrating new or progressive cranial nerve deficits were also likely to demonstrate tumor progression (p = 0.002). Pulsatile tinnitus improved in 49% of patients who reported it at presentation. New or progressive cranial nerve deficits were noted in 15% of patients; improvement in preexisting cranial nerve deficits was observed in 11% of patients. No patient died as a result of tumor progression. Conclusions Gamma Knife surgery was a well-tolerated management strategy that provided a high rate of long-term glomus tumor control. Symptomatic tinnitus improved in almost one-half of the patients. Overall neurological status and cranial nerve function were preserved or improved in the vast majority of patients after radiosurgery.
- Published
- 2012
- Full Text
- View/download PDF
30. Intralesional Lymphokine-activated Killer Cells as Adjuvant Therapy for Primary Glioblastoma
- Author
-
Andrew N. Cornforth, Madeline Carol DePriest, Robin Anne Ellis, Shari Lynn Sharp, Robert O. Dillman, Patric M. Schiltz, and Christopher Duma
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Immunology ,chemical and pharmacologic phenomena ,Immunotherapy, Adoptive ,Gastroenterology ,Internal medicine ,medicine ,Adjuvant therapy ,Humans ,Immunology and Allergy ,Killer Cells, Lymphokine-Activated ,Survival rate ,Aged ,Pharmacology ,Chemotherapy ,Lymphokine-activated killer cell ,Temozolomide ,Brain Neoplasms ,business.industry ,Cancer ,hemic and immune systems ,Immunotherapy ,Middle Aged ,medicine.disease ,Surgery ,Interleukin-2 ,Female ,Glioblastoma ,business ,Adjuvant ,medicine.drug - Abstract
Despite recent advances, median survival for patients with resectable glioblastoma multiforme (GBM) is only 12 to 15 months. We previously observed minimal toxicity and a 9.0-month median survival after treatment with intralesional autologous lymphokine-activated killer (LAK) cells in 40 patients with recurrent GBM. In this study, GBM patients were treated with adjuvant intralesional LAK cells. Eligible patients had completed primary therapy for GBM without disease progression. LAK cells were produced by incubating autologous peripheral blood mononuclear cells with interleukin-2 for 3 to 7 days and then placed into the surgically exposed tumor cavity by a neurosurgeon. The 19 men and 14 women had a median age of 57 years. Prior therapy included surgical resection (97%), partial brain irradiation (97%), gamma knife radiosurgery (97%), and temozolomide chemotherapy (70%). Median time from diagnosis to LAK cell therapy was 5.3 months (range: 3.0 to 11.1 mo). LAK cell treatment was well tolerated; average length of hospitalization was 3 days. At the time of this analysis, 27 patients have died; the median survival from the date of original diagnosis is 20.5 months with a 1-year survival rate of 75%. In subset analyses, superior survival was observed for patients who received higher numbers of CD3+/CD16+/CD56+ (T-LAK) cells in the cell products, which was associated with not taking corticosteroids in the month before leukopheresis. Intralesional LAK cell therapy is safe and the survival sufficiently encouraging to warrant further evaluation in a randomized phase 2 trial of intralesional therapies with LAK or carmustine-impregnated wafers.
- Published
- 2009
- Full Text
- View/download PDF
31. Extramedullary hematopoeisis within a convexity meningioma
- Author
-
Franklin D. Westhout, Marlon S. Mathews, Denise Vanhorn, Anton N. Hasso, Michael Brant-Zawadzki, Christopher Duma, and David J. Klein
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Medullary cavity ,business.industry ,Chronic lymphocytic leukemia ,Hematopoietic Tissue ,medicine.disease ,Extramedullary hematopoiesis ,Meningioma ,Leukemia ,medicine.anatomical_structure ,Hematopoiesis, Extramedullary ,Meningeal Neoplasms ,medicine ,Humans ,Surgery ,Neurology (clinical) ,Bone marrow ,business ,Myelofibrosis ,Aged - Abstract
Background Hematopoiesis outside the bone marrow is known to occur in patients with severe anemia, leukemia, polycythemia, or myelofibrosis, and in patients affected by chronic poisoning by marrow-toxic substances. Case Description A 66-year-old right-handed man complained of 4 days of terrible right-sided, sharp, and shooting headache for which he saw his primary care provider. Routine laboratory examination showed a WBC count of 30 800/μL. Neuroimaging showed a large, right frontotemporal, extra-axial, heterogeneously enhancing, dural based mass with associated recent intramural hemorrhage with evidence of midline shift and uncal herniation. The mass was resected using a right-sided extended craniotomy with anterior fossa and middle fossa approach. A hematoxylin-eosin–stained biopsy specimen showed whorls of tumor cells, diagnostic of a meningioma. Interspersed within the tumor bulk were nucleated RBCs, representing areas of extramedullary erythropoiesis within a meningioma. Flow cytometric evaluation confirmed the clinical suspicion of an underlying chronic lymphocytic leukemia. Conclusion Occurrence of extramedullary hematopoiesis within a meningioma is extremely rare. Various theories may explain the occurrence of extramedullary hematopoiesis occurring within a meningioma in our patient, such as hematopoietic differentiation of multipotent mesenchymal tumor cells; direct extension of hematopoietic activity from the neighboring marrow cavity; displacement from bone marrow of stem cells that settle and develop in tissues where capillaries and blood vessels proliferate, such as a meningioma; or congenital heterotopia of totipotent connective tissue cells, which, under certain circumstances, may transform into hematopoietic tissue.
- Published
- 2008
- Full Text
- View/download PDF
32. Central Neurocytoma: Case Report of an 81-Year-Old and Histopathologic Findings
- Author
-
Christopher Duma, M S G Mathews, S Heinemann, Brian S. Kim, Peter Chen, Michael Brant-Zawadzki, and G Brant-Zawadzki
- Subjects
medicine.medical_specialty ,Pathology ,Ataxia ,Stereotactic biopsy ,medicine.diagnostic_test ,business.industry ,General Medicine ,medicine.disease ,030227 psychiatry ,Lesion ,03 medical and health sciences ,Lateral ventricles ,0302 clinical medicine ,medicine ,Central neurocytoma ,Radiology, Nuclear Medicine and imaging ,Histopathology ,Neurology (clinical) ,Young adult ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Depression (differential diagnoses) - Abstract
Central Neurocytomas are rare and usually benign tumors found primarily in the lateral ventricles of the brain. Central Neurocytomas are composed of uniform round cells exhibiting neuronal differentiation and are found almost exclusively in young adults between 15 and 60 years of age. The authors report the case of an 81-year-old man with a history of severe psychosis and depression, who presented with progressive confusion and ataxia likely unrelated to a central neurocytoma of his right lateral ventricle. The patient underwent a stereotactic biopsy of the lesion, followed by Gamma Knife radiosurgery. Histopathology showed immunohistological staining for synaptophysin, neuron specific enolase (NSE) and neuronal nuclear antigen (NeuN). A review of published literature on central neurocytomas revealed that this tumor occurs most frequently in young adults with a median age between 25 and 30 years. This pathology has never been reported in patients over the age of 80. Gamma Knife radiosurgery was successful in decreasing the tumor volume by 20% at the four month follow-up, but the fact that the patient died of unrelated occurrences in the elderly cannot be ruled out. Patients with central neurocytomas commonly present with obstructive hydrocephalus and immediate treatment may be necessary to tide over urgent situations.
- Published
- 2007
- Full Text
- View/download PDF
33. Gamma Knife Radiosurgery for Treatment of Multiple Brain Metastases
- Author
-
Sandra Vermeulen, Christopher Duma, Kenneth Winston, John C. Blasko, Stephan G. Pribil, Peter Grimm, Joseph Henderson, R. Young, Deane B. Jacques, Robert W. Rand, G. E. Bolles, Allen Posewitz, Brian Copcutt, Robert E. Breeze, and Steven D. Johnson
- Subjects
business.industry ,Medicine ,Gamma knife radiosurgery ,business ,Nuclear medicine - Published
- 2015
- Full Text
- View/download PDF
34. Stereotactic radiosurgery for intracranial hemangioblastomas: a retrospective international outcome study
- Author
-
Atsuya Akabane, David Mathieu, Toru Serizawa, Hideyuki Kano, Yoshiyasu Iwai, Douglas Kondziolka, Yukihiko Kohda, Jason P. Sheehan, Christopher Duma, Satoshi O. Suzuki, Akihito Moriki, Osamu Nagano, Byron Young, Heyoung McBride, Takashi Shuto, Hiroyuki Kenai, Mitsuya Sato, L. Dade Lunsford, Shoji Yomo, Yasuhiro Kikuchi, and Masaaki Yamamoto
- Subjects
Adult ,Male ,medicine.medical_specialty ,von Hippel-Lindau Disease ,Adolescent ,medicine.medical_treatment ,Planning target volume ,Gamma knife ,Radiosurgery ,Young Adult ,Hemangioblastoma ,Outcome Assessment, Health Care ,medicine ,Humans ,In patient ,Von Hippel–Lindau disease ,Aged ,Retrospective Studies ,Aged, 80 and over ,business.industry ,Brain Neoplasms ,Disease progression ,Retrospective cohort study ,Middle Aged ,medicine.disease ,Surgery ,Treatment Outcome ,Disease Progression ,Female ,Neoplasm Recurrence, Local ,business - Abstract
OBJECT The purpose of this study was to evaluate the role of stereotactic radiosurgery (SRS) in the management of intracranial hemangioblastomas. METHODS Six participating centers of the North American Gamma Knife Consortium and 13 Japanese Gamma Knife centers identified 186 patients with 517 hemangioblastomas who underwent SRS. Eighty patients had 335 hemangioblastomas associated with von Hippel–Lindau disease (VHL) and 106 patients had 182 sporadic hemangioblastomas. The median target volume was 0.2 cm3 (median diameter 7 mm) in patients with VHL and 0.7 cm3 (median diameter 11 mm) in those with sporadic hemangioblastoma. The median margin dose was 18 Gy in VHL patients and 15 Gy in those with sporadic hemangioblastomas. RESULTS At a median of 5 years (range 0.5–18 years) after treatment, 20 patients had died of intracranial disease progression and 9 patients had died of other causes. The overall survival after SRS was 94% at 3 years, 90% at 5 years, and 74% at 10 years. Factors associated with longer survival included younger age, absence of neurological symptoms, fewer tumors, and higher Karnofsky Performance Status. Thirty-three (41%) of the 80 patients with VHL developed new tumors and 17 (16%) of the106 patients with sporadic hemangioblastoma had recurrences of residual tumor from the original tumor. The 5-year rate of developing a new tumor was 43% for VHL patients, and the 5-year rate of developing a recurrence of residual tumor from the original tumor was 24% for sporadic hemangioblastoma patients. Factors associated with a reduced risk of developing a new tumor or recurrences of residual tumor from the original tumor included younger age, fewer tumors, and sporadic rather than VHL-associated hemangioblastomas. The local tumor control rate for treated tumors was 92% at 3 years, 89% at 5 years, and 79% at 10 years. Factors associated with an improved local tumor control rate included VHL-associated hemangioblastoma, solid tumor, smaller tumor volume, and higher margin dose. Thirteen patients (7%) developed adverse radiation effects (ARE) after SRS, and one of these patients died due to ARE. CONCLUSIONS When either sporadic or VHL-associated tumors were observed to grow on serial imaging studies, SRS provided tumor control in 79%–92% of tumors.
- Published
- 2015
35. Gamma Knife Radiosurgery for Meningiomas of the Convexity and Cavernous Sinus
- Author
-
L. Dade Lunsford, Jay Tassin, Douglas Kondziolka, W. Michael Shea, and Christopher Duma
- Subjects
business.industry ,medicine.medical_treatment ,Cavernous sinus ,medicine ,Gamma knife radiosurgery ,Neurology (clinical) ,Gamma knife ,Nuclear medicine ,business ,Convexity ,Radiosurgery - Published
- 2003
- Full Text
- View/download PDF
36. Spinal epidural metastasis in an endometrial carcinoma patient
- Author
-
Christopher Duma, Humberto Wong, Janet M. Stallman, John V. Brown, and Bram H. Goldstein
- Subjects
medicine.medical_specialty ,business.industry ,Incidence (epidemiology) ,Endometrial cancer ,Obstetrics and Gynecology ,Case Report ,Endometrial carcinoma ,medicine.disease ,Surgery ,Metastasis ,Treatment ,Spinal epidural ,Oncology ,Spinal epidural metastasis ,Carcinoma ,Medicine ,Radiology ,Epidural mass ,business - Abstract
► The incidence of CNS metastases from endometrial cancer is quite uncommon. ► We report on an endometrial cancer patient who developed a metastatic epidural mass. ► Oncology physicians should remain vigilant in order to effectuate prompt treatment and potentially benefit the patient's outcome.
- Published
- 2012
- Full Text
- View/download PDF
37. Immunotherapy targeting the C-terminal domain of TDP-43 decreases neuropathology and confers neuroprotection in mouse models of ALS/FTD
- Author
-
Tariq Afroz, Elodie Chevalier, Mickael Audrain, Christopher Dumayne, Tamar Ziehm, Roger Moser, Anne-Laure Egesipe, Lorène Mottier, Monisha Ratnam, Manuela Neumann, Daniel Havas, Romain Ollier, Kasia Piorkowska, Mayank Chauhan, Alberto B. Silva, Samjhana Thapa, Jan Stöhr, Andrej Bavdek, Valerie Eligert, Oskar Adolfsson, Peter T. Nelson, Sílvia Porta, Virginia M.-Y. Lee, Andrea Pfeifer, Marie Kosco-Vilbois, and Tamara Seredenina
- Subjects
TDP-43 ,Immunotherapy ,Frontotemporal dementia (FTD) ,Amyotrophic lateral sclerosis (ALS) ,Microglia ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Effective therapies are urgently needed to safely target TDP-43 pathology as it is closely associated with the onset and development of devastating diseases such as frontotemporal lobar degeneration with TDP-43 pathology (FTLD-TDP) and amyotrophic lateral sclerosis (ALS). In addition, TDP-43 pathology is present as a co-pathology in other neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Our approach is to develop a TDP-43-specific immunotherapy that exploits Fc gamma-mediated removal mechanisms to limit neuronal damage while maintaining physiological TDP-43 function. Thus, using both in vitro mechanistic studies in conjunction with the rNLS8 and CamKIIa inoculation mouse models of TDP-43 proteinopathy, we identified the key targeting domain in TDP-43 to accomplish these therapeutic objectives. Targeting the C-terminal domain of TDP-43 but not the RNA recognition motifs (RRM) reduces TDP-43 pathology and avoids neuronal loss in vivo. We demonstrate that this rescue is dependent on Fc receptor-mediated immune complex uptake by microglia. Furthermore, monoclonal antibody (mAb) treatment enhances phagocytic capacity of ALS patient-derived microglia, providing a mechanism to restore the compromised phagocytic function in ALS and FTD patients. Importantly, these beneficial effects are achieved while preserving physiological TDP-43 activity. Our findings demonstrate that a mAb targeting the C-terminal domain of TDP-43 limits pathology and neurotoxicity, enabling clearance of misfolded TDP-43 through microglia engagement, and supporting the clinical strategy to target TDP-43 by immunotherapy. Significance statement: TDP-43 pathology is associated with various devastating neurodegenerative disorders with high unmet medical needs such as frontotemporal dementia (FTD), amyotrophic lateral sclerosis (ALS) and Alzheimer's disease. Thus, safely and effectively targeting pathological TDP-43 represents a key paradigm for biotechnical research as currently there is little in clinical development. After years of research, we have determined that targeting the C-terminal domain of TDP-43 rescues multiple patho-mechanisms involved in disease progression in two animal models of FTD/ALS. In parallel, importantly, our studies establish that this approach does not alter the physiological functions of this ubiquitously expressed and indispensable protein. Together, our findings substantially contribute to the understanding of TDP-43 pathobiology and support the prioritization for clinical testing of immunotherapy approaches targeting TDP-43.
- Published
- 2023
- Full Text
- View/download PDF
38. The Treatment of Movement Disorders Using Gamma Knife Stereotactic Radiosurgery
- Author
-
Christopher Duma, Oleg V. Kopyov, and Deane 'skip' Jacques
- Subjects
medicine.medical_specialty ,Movement disorders ,medicine.diagnostic_test ,business.industry ,Thalamotomy ,medicine.medical_treatment ,Follow up studies ,Magnetic resonance imaging ,General Medicine ,Gamma knife ,Radiosurgery ,Medicine ,Surgery ,High surgical risk ,Neurology (clinical) ,Radiology ,Neurosurgery ,medicine.symptom ,business - Abstract
In this era of modern neurosurgery, we are able to provide adequate amelioration of disabling symptoms for the small subset of patients who have conditions that may make them unacceptable candidates for invasive stereotactic neurosurgical intervention. Gamma Knife radiosurgical thalamotomy is an effective and useful alternative to invasive radiofrequency techniques for patients at high surgical risk. The mechanical accuracy of the gamma unit combined with the anatomical accuracy of high-resolution magnetic resonance imaging makes radiosurgical lesioning safe and precise.
- Published
- 1999
- Full Text
- View/download PDF
39. ACTR-25. UP-FRONT BOOST GAMMA KNIFE 'LEADING EDGE' RADIOSURGERY (LERS) TO MR FLAIR-DEFINED TUMOR MIGRATION PATHWAYS IN 174 PATIENTS WITH GLIOBLASTOMA MULTIFORME: A 15-YEAR ASSESSMENT OF A NOVEL THERAPY
- Author
-
Gustavo Mendez, Burton L. Eisenberg, Peter Chen, Marlon Mathews, Garrett Smith, Marianne Plunkett, Michael Brant-Zawadzki, Ryan M Casserly, Daniel J Furman, Ralph Mackintosh, Brian S. Kim, Robert O. Dillman, Chad A. Caraway, Azzurra-Sky Riley, Christopher Duma, David Weiland, and Ruslana Cannell
- Subjects
Cancer Research ,Leading edge ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Fluid-attenuated inversion recovery ,Gamma knife ,medicine.disease ,Radiosurgery ,Oncology ,Medicine ,Neurology (clinical) ,Radiology ,Nuclear medicine ,business ,Front (military) ,Glioblastoma - Published
- 2016
- Full Text
- View/download PDF
40. Gamma Knife surgery for the management of glomus tumors: a multicenter study
- Author
-
Jason P, Sheehan, Shota, Tanaka, Michael J, Link, Bruce E, Pollock, Douglas, Kondziolka, David, Mathieu, Christopher, Duma, A Byron, Young, Anthony M, Kaufmann, Heyoung, McBride, Peter A, Weisskopf, Zhiyuan, Xu, Hideyuki, Kano, Huai-che, Yang, and L Dade, Lunsford
- Subjects
Adult ,Aged, 80 and over ,Male ,Adolescent ,Middle Aged ,Glomus Tumor ,Radiosurgery ,Skull Base Neoplasms ,Cranial Nerve Diseases ,Disease-Free Survival ,Tinnitus ,Young Adult ,Postoperative Complications ,Treatment Outcome ,Actuarial Analysis ,Trigeminal Nerve Diseases ,Humans ,Female ,Neoplasm Recurrence, Local ,Aged ,Follow-Up Studies - Abstract
Glomus tumors are rare skull base neoplasms that frequently involve critical cerebrovascular structures and lower cranial nerves. Complete resection is often difficult and may increase cranial nerve deficits. Stereotactic radiosurgery has gained an increasing role in the management of glomus tumors. The authors of this study examine the outcomes after radiosurgery in a large, multicenter patient population.Under the auspices of the North American Gamma Knife Consortium, 8 Gamma Knife surgery centers that treat glomus tumors combined their outcome data retrospectively. One hundred thirty-four patient procedures were included in the study (134 procedures in 132 patients, with each procedure being analyzed separately). Prior resection was performed in 51 patients, and prior fractionated external beam radiotherapy was performed in 6 patients. The patients' median age at the time of radiosurgery was 59 years. Forty percent had pulsatile tinnitus at the time of radiosurgery. The median dose to the tumor margin was 15 Gy. The median duration of follow-up was 50.5 months (range 5-220 months).Overall tumor control was achieved in 93% of patients at last follow-up; actuarial tumor control was 88% at 5 years postradiosurgery. Absence of trigeminal nerve dysfunction at the time of radiosurgery (p = 0.001) and higher number of isocenters (p = 0.005) were statistically associated with tumor progression-free tumor survival. Patients demonstrating new or progressive cranial nerve deficits were also likely to demonstrate tumor progression (p = 0.002). Pulsatile tinnitus improved in 49% of patients who reported it at presentation. New or progressive cranial nerve deficits were noted in 15% of patients; improvement in preexisting cranial nerve deficits was observed in 11% of patients. No patient died as a result of tumor progression.Gamma Knife surgery was a well-tolerated management strategy that provided a high rate of long-term glomus tumor control. Symptomatic tinnitus improved in almost one-half of the patients. Overall neurological status and cranial nerve function were preserved or improved in the vast majority of patients after radiosurgery.
- Published
- 2012
41. Tolerance of cranial nerves of the cavernous sinus to radiosurgery
- Author
-
Christopher Duma, Roy B. Tishler, Jay S. Loeffler, L. Dade Lunsford, Hanne M. Kooy, John C. Flickinger, and Eben Alexander
- Subjects
Adult ,Male ,Nervous system ,Cancer Research ,medicine.medical_specialty ,Adolescent ,medicine.medical_treatment ,Trochlear Nerve ,Radiosurgery ,Radiation Tolerance ,Abducens Nerve ,Oculomotor Nerve ,medicine ,Humans ,Pituitary Neoplasms ,Radiology, Nuclear Medicine and imaging ,Trigeminal Nerve ,Child ,Radiation treatment planning ,Aged ,Retrospective Studies ,Aged, 80 and over ,Radiation ,Brain Neoplasms ,business.industry ,Cranial nerves ,Cavernous Sinus Meningioma ,Optic Nerve ,Radiotherapy Dosage ,Middle Aged ,Cranial Nerve Diseases ,Surgery ,Radiation therapy ,medicine.anatomical_structure ,Oncology ,Child, Preschool ,Cavernous sinus ,Optic nerve ,Cavernous Sinus ,Female ,Radiology ,business ,Follow-Up Studies - Abstract
Purpose: Stereotactic radiosurgery is becoming a more accepted treatment option for benign, deep seated intracranial lesions. However, little is known about the effects of large single fractions of radiation on cranial nerves. This study was undertaken to assess the effect of radiosurgery on the cranial nerves of the cavernous sinus. Methods and Materials: We examined the tolerance of cranial nerves (II–VI) following radiosurgery for 62 patients (4262 with meningiomas) treated for lesions within or near the cavernous sinus. Twenty-nine patients were treated with a modified 6 MV linear accelerator (Joint Center for Radiation Therapy) and 33 were treated with the Gamma Knife (University of Pittsburgh). Three-dimensional treatment plans were retrospectively reviewed and maximum doses were calculated for the cavernous sinus and the optic nerve and chiasm. Results: Median follow-up was 19 months (range 3–49). New cranial neuropathies developed in 12 patients from 3–41 months following radiosurgery. Four of these complications involved injury to the optic system and 8 (38 transient) were the result of injury to the sensory or motor nerves of the cavernous sinus. There was no clear relationship between the maximum dose to the cavernous sinus and the development of complications for cranial nerves III–VI over the dose range used (1000–4000 cGy). For the optic apparatus, there was a significantly increased incidence of complications with dose. Four of 17 patients (24°10) receiving greater than 800 cGy to any part of the optic apparatus developed visual complications compared with 035 who received less than 800 cGy ( P = 0.009). Conclusion: Radiosurgery using tumor-controlling doses of up to 4000 cGy appears to be a relatively safe technique in treating lesions within or near the sensory and motor nerves (III–VI) of the cavernous sinus. The dose to the optic apparatus should be limited to under 800 cGy.
- Published
- 1993
- Full Text
- View/download PDF
42. Gamma knife radiosurgery for acoustic tumors: multivariate analysis of four year results
- Author
-
Mark E. Linskey, John C. Flickinger, Christopher Duma, L. Dade Lunsford, and Douglas Kondziolka
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,Hearing loss ,medicine.medical_treatment ,Acoustic neuroma ,Radiosurgery ,Postoperative Complications ,Hearing ,Risk Factors ,otorhinolaryngologic diseases ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Trigeminal Nerve ,Neurofibromatosis ,Aged ,Aged, 80 and over ,medicine.diagnostic_test ,business.industry ,Radiotherapy Dosage ,Neuroma, Acoustic ,Hematology ,Middle Aged ,medicine.disease ,Neuroma ,Cranial Nerve Diseases ,Surgery ,Radiation therapy ,Facial Nerve ,Oncology ,Hearing level ,Female ,Pure tone audiometry ,medicine.symptom ,business ,Nuclear medicine ,Follow-Up Studies - Abstract
In order to evaluate the results of radiosurgery for acoustic tumors and to identify optimum treatment parameters, an analysis of tumor control, as well as incidences of hearing loss, facial and trigeminal neuropathies was undertaken. Between August 1987 and August 1991, 134 patients with 136 acoustic tumors received stereotactic gamma knife radiosurgery at the University of Pittsburgh. Median follow-up was 24 months (range: 6-56 months). Tumor volumes ranged from 0.10 to 17.00 cm3 (median = 2.75 cm3). From one to ten isocenters were utilized per tumor treated (median = 3). Minimum tumor doses varied from 12 to 20 Gy (median = 17 Gy). The 4-year actuarial tumor control rate was 89.2 +/- 6.0%. Some degree of hearing (by pure tone audiometry) was preserved in 71.0 +/- 4.4% of patients. The actuarial rates for preservation of either pretreatment hearing level or useful hearing were 34.4 +/- 6.6% and 35.1 +/- 97% respectively. Respectively, the actuarial incidences of postradiosurgery facial and trigeminal neuropathies were 29.0 +/- 4.4% and 32.9 +/- 4.5%, respectively. No significant factors affecting tumor control were identified. Multivariate analysis identified a significantly increased risk of hearing loss in patients with neurofibromatosis (p = 0.0003) as well as decreased risks of facial and trigeminal neuropathies with both decreasing tumor diameter (p = 0.001) and increasing number of isocenters treated (p = 0.003). Radiosurgery is a safe and effective treatment for acoustic neuromas with acceptable morbidity that may be lowered by the use of multiple isocenter treatment techniques and by earlier treatment of small tumors.
- Published
- 1993
- Full Text
- View/download PDF
43. Durable complete response of refractory, progressing metastatic melanoma after treatment with a patient-specific vaccine
- Author
-
Christopher Duma, Carol DePriest, Scott T. Williams, Peter Chen, Andrew N. Cornforth, Robert O. Dillman, Andreea A. Nanci, Peter C. Wang, Senthamil R. Selvan, Richard B. Kim, Colleen Coleman, and Russell Hafer
- Subjects
Cancer Research ,medicine.medical_specialty ,Skin Neoplasms ,medicine.medical_treatment ,Rectum ,Cancer Vaccines ,Disease-Free Survival ,Refractory ,Interferon ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Neoplasm Metastasis ,Melanoma ,Pharmacology ,business.industry ,Remission Induction ,Combination chemotherapy ,General Medicine ,Middle Aged ,Autologous tumor cell ,Vaccine therapy ,Surgery ,Radiation therapy ,Axilla ,medicine.anatomical_structure ,Treatment Outcome ,Oncology ,Disease Progression ,Female ,business ,medicine.drug - Abstract
A patient with metastatic melanoma who experienced a durable complete response after treatment with a patient-specific vaccine has been described in this article. This 59-year-old woman presented with cervical spine metastases and, within the year, had experienced local disease progression and, despite various therapies, metastases to the axilla, rectum, gall bladder, and multiple soft-tissue sites. She had previously received radiation therapy, combination chemotherapy, interleukin-2 plus interferon biotherapy, and gamma knife radiosurgery, and undergone multiple surgical resections. At the time vaccine therapy was initiated, she had multiple, new, measurable, soft-tissue metastases that were increasing in size. She was treated with a vaccine consisting of autologous dendritic cells incubated with irradiated tumor cells from an autologous tumor cell line and suspended in granulocyte-macrophage colony stimulating factor (GM-CSF), with subcutaneous injections once a week for 3 weeks and monthly for 5 months. There was evidence of disease regression by the completion of therapy. A few months later a complete response was documented by radiologic scans, and subsequently reconfirmed at 6-month intervals. She remains in complete remission2.5 years after starting the vaccine, and2 years after completing the vaccine, and survives4 years after her initial presentation with bone, bowel, and lymph node metastases. This is the first time she has been in a complete remission since her initial diagnosis. Patient-specific vaccines can sometimes induce durable complete regression of progressing soft-tissue melanoma metastases.
- Published
- 2010
44. Treatment of recurrent high grade gliomas with hypofractionated stereotactic image-guided helical tomotherapy
- Author
-
Ralph Mackintosh, Brian S. Kim, Peter Chen, Marianne Plunkett, Craig Cox, Jim Cubellis, Annamarie Minion, Christopher Duma, Russell Hafer, and Emilie T. Soisson
- Subjects
Adult ,Male ,medicine.medical_treatment ,Radiography ,Brain tumor ,Video-Assisted Surgery ,Stereotactic radiation therapy ,Radiosurgery ,Tomotherapy ,Central nervous system disease ,Recurrence ,Glioma ,Medicine ,Humans ,Aged ,Salvage Therapy ,business.industry ,Brain Neoplasms ,General Medicine ,Middle Aged ,medicine.disease ,Magnetic Resonance Imaging ,Survival Analysis ,Radiation therapy ,Surgery ,Female ,Neurology (clinical) ,business ,Nuclear medicine - Abstract
Objective Salvage treatment of high grade gliomas that progress after standard therapy of resection and adjuvant chemoradiation therapy includes repeat surgical resection, second line chemotherapy, re-irradiation, or often a combination of the above. We present a series on patients treated with hypofractionated stereotactic image-guided helical tomotherapy and discuss the efficacy of this new technology in the treatment of high grade gliomas. Materials and methods Between June 2005 and August of 2008, eight patients with recurrent high grade gliomas were treated with salvage radiation therapy using hypofractionated stereotactic image-guided helical tomotherapy after image documentation of disease progression. Median age was 48.5 years with 4 females and 4 males. Median KPS at time of treatment was 65. All patients had either Grade III or IV gliomas at time of treatment with previous history of involved field fractionated radiotherapy. Median total dose given was 2500 cGy in 500 cGy fractions. Results The median planning target volume was 69.5 cm3. Five of the eight patients were alive at the time of last follow-up with a median survival of 7.6 months. Radiographic documented control was seen in six of the eight patients with median local control of 4.6 months. Acute Radiation Therapy Oncology Group (RTOG) toxicity scores measured zero in all patients with only one patient requiring a reoperation following treatment. Conclusions Hypofractionated stereotactic image-guided helical tomotherapy provides an alternative to other stereotactic radiation therapy and radiosurgery options for treatment of recurrent high grade gliomas.
- Published
- 2010
45. Image-Guided Stereotactic Management of Non-AIDS-Related Cerebral Infection
- Author
-
Christopher Duma, Douglas Kondziolka, and Lunsford Ld
- Subjects
medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,General Medicine ,medicine.disease ,Surgery ,Transplantation ,Hematoma ,Acquired immunodeficiency syndrome (AIDS) ,Stereotaxy ,Biopsy ,medicine ,Subacute bacterial endocarditis ,Neurology (clinical) ,Neurosurgery ,Radiology ,business ,Complication - Abstract
Every neurosurgeon can appreciate Dandy's recognition that the drainage of brain abscesses causes trauma to the delicate parenchyma. Over the years, brain surgery has evolved toward management of problems by using less and less invasive techniques and thus gaining ever lower morbidity. Clearly, the advent of better imaging techniques has improved the outcome in patients afflicted with intracerebral infections. The combination of stereotaxy with these imaging techniques is contributing a "zero mortality" in the treatment of these infections. In our series of 29 consecutive patients with non-AIDS-related infections, no patient died as a direct result of a stereotactic surgical procedure. Two patients (7%) had new neurologic deficits after surgery. The only patient left with a permanent disability had a kidney allograft and subacute bacterial endocarditis. His condition deteriorated 6 hours after aspiration of a sterile abscess, when an intra-abscess hematoma was diagnosed and evacuated. In retrospect, this complication may have been avoided by less vigorous aspiration. Three of the four patients with nonviral infections who died were iatrogenically immunosuppressed for their organ transplants. These patients are difficult to treat, and given the current popularity of transplantation procedures, neurosurgeons will face more and more opportunistic infections. In general, the patients with abscesses did well. On the other hand, nonoperative mortality was extremely high for patients with viral encephalitides. This high mortality may have resulted from a delay in diagnosis and treatment or from the unavailability of highly effective antiviral agents at the time the biopsies were performed. The importance of early diagnosis and treatment of infection cannot be overemphasized. T.H. Flewett's warning about the management of HSE applies to the management of all cerebral infections: "It seems clear from everybody's published results [in the papers already given] if we wait to do biopsy until the clinical indications are unmistakable, we have waited so long that the patient, if he survives, will be left a severe neurological cripple." Because it is relatively noninvasive, stereotactic neurosurgery has been used increasingly to diagnose brain masses in patients with AIDS. We recommend its use for establishing diagnoses in all suspected cases of cerebral infection. We agree with Rosenblum et al: Empiric treatment of brain infections should be regarded as "radical." Such treatment should be reserved for patients who have an identifiable source of infection and causative organism or for patients who are clinically too unstable to undergo surgery.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1992
- Full Text
- View/download PDF
46. The Role of Immediate Operative Intervention in Severely Head-Injured Children with a Glasgow Coma Scale Score of 3
- Author
-
Christopher Duma, Dennis L. Johnson, and Carlos J. Sivit
- Subjects
Male ,Child abuse ,Emergency Medical Services ,medicine.medical_specialty ,Time Factors ,Sedation ,medicine.medical_treatment ,Brain Edema ,Hematoma ,Trauma Centers ,Trephining ,Paralysis ,medicine ,Craniocerebral Trauma ,Humans ,Intubation ,Glasgow Coma Scale ,Child Abuse ,Coma ,Child ,Cerebral Hemorrhage ,Retrospective Studies ,Multiple Trauma ,business.industry ,Contraindications ,Accidents, Traffic ,Retrospective cohort study ,medicine.disease ,Surgery ,Hematoma, Subdural ,Brain Injuries ,Child, Preschool ,District of Columbia ,Female ,Neurology (clinical) ,medicine.symptom ,Tomography, X-Ray Computed ,business - Abstract
In an attempt to improve and expedite the care of head-injured children, data have been published recommending burr hole exploration in lieu of computed tomography for children with signs of brain stem compression or with a Glasgow Coma Scale score of 3. Exploratory burr holes revealed a high incidence of subdural hematomas, and removal of the hematomas improved survival. We are reporting 19 consecutive children with Glasgow Coma Scale scores of 3. Coma score evaluation was confounded by intubation, sedation, pharmacological paralysis, and posttraumatic seizures. We found no radiographical or postmortem pathological evidence of intracranial hemorrhage, which would warrant operative intervention. A high incidence of multisystem injuries and high cervical spine injuries would have made early intervention both dangerous and inappropriate. Although there is a definite role for emergency trephination, routine exploratory burr holes for children with a Coma score of 3 is not justified.
- Published
- 1992
- Full Text
- View/download PDF
47. Image-guided helical Tomotherapy for treatment of spine tumors
- Author
-
Jim Cubellis, Ralph Mackintosh, Emilie T. Soisson, Marianne Plunkett, Russell Hafer, Brian S. Kim, Craig Cox, Annamarie Minion, Christopher Duma, and Peter Chen
- Subjects
Male ,Vacuum ,medicine.medical_treatment ,Radiography ,Myelitis ,Tomotherapy ,Radiosurgery ,Immobilization ,Medicine ,Humans ,Radiation Injuries ,Rachis ,Neuronavigation ,Aged ,Spinal Neoplasms ,business.industry ,Radiotherapy Planning, Computer-Assisted ,General Medicine ,Equipment Design ,Middle Aged ,medicine.disease ,Spinal cord ,Radiation therapy ,Neuroendocrine Tumors ,medicine.anatomical_structure ,Surgery ,Female ,Neurology (clinical) ,Dose Fractionation, Radiation ,Radiotherapy, Conformal ,business ,Fiducial marker ,Nuclear medicine ,Tomography, Spiral Computed - Abstract
Objectives One of the most common indications for radiotherapy is treatment of the spine. The vast majority of cases are related to metastatic disease with primary tumors of the spine being rare. Conventional radiation therapy often plays an important role in the management of spine tumors although at times with significant side effects and disadvantages. Furthermore, retreatment of spine tumors is a challenge due to concerns over spinal cord toxicity. In this series, we examine the efficacy of using image-guided helical Tomotherapy and the possible advantages offered by this new technology. Patients and methods Eight patients at Hoag Memorial Hospital Presbyterian were treated between November 2005 and November 2006. The median age was 66 years. Of the eight patients, seven had metastatic disease with one patient having a primary neuroendocrine tumor of the spine. Five patients were previously treated to the spine with conventional radiation planning. Two patients received single fraction stereotactic radiosurgery (15 Gy) while the remaining patients received hypofractionated stereotactic radiotherapy to a median total dose of 2500 cGy in 500 cGy fractions. Results At the time of last follow-up, radiographic control was seen in all eight patients with a median local control rate of 2.5 months (range of 1–5.8 months). Four of the eight patients are still alive with median overall survival of 5.1 months (range 1.4–6.9 months). Acute toxicity ranged from Radiation Therapy Oncology Group (RTOG) score 0–2 and no patients experienced late complications of radiation myelitis. Conclusions The TomoTherapy Hi-ART system can be an alternative treatment option for upfront or retreatment of spine tumors. Minimal acute and late toxicity were seen in patients treated with Tomotherapy. Intensity-modulated radiation delivery combined with megavoltage CT image guidance offered by the TomoTherapy Hi-ART system allows for set-up and delivery accuracy that is required for stereotactic treatment of spine tumors and eliminates the need for any internal or external fiducial marker placement.
- Published
- 2007
48. Movement Disorder Radiosurgery – Planning, Physics and Complication Avoidance
- Author
-
Christopher Duma
- Subjects
medicine.medical_specialty ,Deep brain stimulation ,business.industry ,Thalamotomy ,medicine.medical_treatment ,Iron deposition ,Hypoxia (medical) ,Radiosurgery ,Surgery ,Physiological monitoring ,Medicine ,Pallidotomy ,medicine.symptom ,Complication ,business - Abstract
Gamma Knife radiosurgical thalamotomy is an effective and useful alternative to invasive radiofrequency techniques for patients at high surgical risk. The mechanical accuracy of the gamma unit combined with the anatomical accuracy of high-resolution MRI make radiosurgical lesioning safe and precise. Higher radiosurgical doses are more effective than lower ones at eliminating or reducing tremor, and are generally without complications. The results from radiosurgical pallidotomy, as opposed to those of gamma thalamotomy, have been disappointing. A 50% complication rate in the former (homonymous field cuts, hemipareses and dysphagias) combined with a poor success rate has led us to reevaluate the indications for this procedure in the face of the excellent results from radiofrequency pallidotomy with physiological monitoring and deep brain stimulation. Perhaps experience with lowered radiosurgical prescription doses will improve the complication rate. There appears to be a differential sensitivity of the pallidum to radiation, anatomically, than the thalamus. Age-related or anatomy-related susceptible blood supply to the area may lead to hypoxia after singlefraction radiosurgery, in a nuclear complex known to be especially susceptible to hypoxia. In addition, varying levels of iron deposition within the pallidum may catalyze free radical formation in the elderly only to be further exacerbated by tissue hypoxia. Although reported, the success of radiosurgical caudatotomy, subthalamotomy and lesioning of the VL nucleus remains to be further elucidated.
- Published
- 2007
- Full Text
- View/download PDF
49. 144 Chondrosarcoma Radiosurgery
- Author
-
Christopher Duma, Aditya Iyer, Hideyuki Kano, David Mathieu, Michael W. McDermott, A. Byron Young, Jason P. Sheehan, Heyoung McBride, L. Dade Lunsford, and Penny K. Sneed
- Subjects
business.industry ,medicine.medical_treatment ,medicine ,Surgery ,Neurology (clinical) ,Gamma knife ,Chondrosarcoma ,business ,Nuclear medicine ,medicine.disease ,Radiosurgery - Published
- 2013
- Full Text
- View/download PDF
50. Gamma-knife radiosurgery for the treatment of ovarian cancer metastatic to the brain
- Author
-
Christopher Duma, John P. Micha, Bram H. Goldstein, John V. Brown, and Mark A. Rettenmaier
- Subjects
Oncology ,Adult ,medicine.medical_specialty ,medicine.medical_treatment ,Central nervous system ,Gamma knife radiosurgery ,Disease ,Radiosurgery ,Internal medicine ,Long term survival ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Humans ,Ovarian malignancy ,Ovarian Neoplasms ,business.industry ,Brain Neoplasms ,Obstetrics and Gynecology ,Middle Aged ,medicine.disease ,medicine.anatomical_structure ,Female ,Cns disease ,business ,Ovarian cancer - Abstract
Background Central nervous system (CNS) metastases from an ovarian malignancy are uncommon. The long-term prognosis for these patients is poor, with studies reporting a mean survival of less than 12 months. Cases We present three ovarian cancer patients who developed metastatic disease to the brain. All patients were heavily pre-treated prior to the development of CNS disease. Following detection of CNS disease, they all were treated with multi-modality therapy including gamma-knife radiosurgery (GKRS). At this time, one patient is alive at 26 months following treatment with GKRS. The second and third patients survived for 88 and 22 months respectively, before succumbing to their disease. Conclusion Local control of ovarian cancer metastatic to the brain can be achieved in some patients with GKRS. Additional investigation into GKRS is warranted.
- Published
- 2004
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.