54 results on '"Laura K Aguilar"'
Search Results
2. Perturbing DDR signaling enhances cytotoxic effects of local oncolytic virotherapy and modulates the immune environment in glioma
- Author
-
Marilin S. Koch, Mykola Zdioruk, Michal O. Nowicki, Alec M. Griffith, Estuardo Aguilar-Cordova, Laura K. Aguilar, Brian W. Guzik, Francesca Barone, Paul Peter Tak, Katharina Schregel, Michael S. Hoetker, James A. Lederer, E. Antonio Chiocca, Ghazaleh Tabatabai, and Sean E. Lawler
- Subjects
DDR signaling ,ATR ,CAN-2409 ,glioblastoma ,oncolytic therapy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
CAN-2409 is a replication-deficient adenovirus encoding herpes simplex virus (HSV) thymidine kinase (tk) currently in clinical trials for treatment of glioblastoma. The expression of tk in transduced cancer cells results in conversion of the pro-drug ganciclovir into a toxic metabolite causing DNA damage, inducing immunogenic cell death and immune activation. We hypothesize that CAN-2409 combined with DNA-damage-response inhibitors could amplify tumor cell death, resulting in an improved response. We investigated the effects of ATR inhibitor AZD6738 in combination with CAN-2409 in vitro using cytotoxicity, cytokine, and fluorescence-activated cell sorting (FACS) assays in glioma cell lines and in vivo with an orthotopic syngeneic murine glioma model. Tumor immune infiltrates were analyzed by cytometry by time of flight (CyTOF). In vitro, we observed a significant increase in the DNA-damage marker γH2AX and decreased expression of PD-L1, pro-tumorigenic cytokines (interleukin-1β [IL-1β], IL-4), and ligand NKG2D after combination treatment compared with monotherapy or control. In vivo, long-term survival was increased after combination treatment (66.7%) compared with CAN-2409 (50%) and control. In a tumor re-challenge, long-term immunity after combination treatment was not improved. Our results suggest that ATR inhibition could amplify CAN-2409’s efficacy in glioblastoma through increased DNA damage while having complex immunological ramifications, warranting further studies to determine the ideal conditions for maximized therapeutic benefit.
- Published
- 2022
- Full Text
- View/download PDF
3. Neoadjuvant Gene-Mediated Cytotoxic Immunotherapy for Non-Small-Cell Lung Cancer: Safety and Immunologic Activity
- Author
-
Mitchell G. Bryski, Jason Stadanlick, Brian W. Guzik, Patrick Woodruff, Lydia G. Frenzel-Sulyok, Edmund K. Moon, Laura K. Aguilar, Charuhas Deshpande, Estuardo Aguilar-Cordova, Steven M. Albelda, Evgeniy Eruslanov, Andrea G. Manzanera, Corey J. Langer, Jarrod D. Predina, Sunil Singhal, Marina Martinez, Andrew R. Haas, Christopher Corbett, and Shaun O'Brien
- Subjects
PD-L1 ,Cytotoxicity, Immunologic ,Lung Neoplasms ,medicine.medical_treatment ,Genetic enhancement ,gene-mediated cytotoxic immunotherapy ,Genetic Vectors ,CD8-Positive T-Lymphocytes ,Thymidine Kinase ,Adenoviridae ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Carcinoma, Non-Small-Cell Lung ,Drug Discovery ,PD-1 ,Genetics ,medicine ,Cytotoxic T cell ,Humans ,Lung cancer ,Molecular Biology ,030304 developmental biology ,Pharmacology ,0303 health sciences ,biology ,business.industry ,Immunotherapy ,adenovirus ,neoadjuvant clinical trial ,Genetic Therapy ,medicine.disease ,gene therapy ,Neoadjuvant Therapy ,lung cancer ,intratumoral immunotherapy ,checkpoint inhibitor ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Molecular Medicine ,Original Article ,business ,Cell activation ,CD8 - Abstract
Gene-mediated cytotoxic immunotherapy (GMCI) is an immuno-oncology approach involving local delivery of a replication-deficient adenovirus expressing herpes simplex thymidine kinase (AdV-tk) followed by anti-herpetic prodrug activation that promotes immunogenic tumor cell death, antigen-presenting cell activation, and T cell stimulation. This phase I dose-escalation pilot trial assessed bronchoscopic delivery of AdV-tk in patients with suspected lung cancer who were candidates for surgery. A single intra-tumoral AdV-tk injection in three dose cohorts (maximum 1012 viral particles) was performed during diagnostic staging, followed by a 14-day course of the prodrug valacyclovir, and subsequent surgery 1 week later. Twelve patients participated after appropriate informed consent. Vector-related adverse events were minimal. Immune biomarkers were evaluated in tumor and blood before and after GMCI. Significantly increased infiltration of CD8+ T cells was found in resected tumors. Expression of activation, inhibitory, and proliferation markers, such as human leukocyte antigen (HLA)-DR, CD38, Ki67, PD-1, CD39, and CTLA-4, were significantly increased in both the tumor and peripheral CD8+ T cells. Thus, intratumoral AdV-tk injection into non-small-cell lung cancer (NSCLC) proved safe and feasible, and it effectively induced CD8+ T cell activation. These data provide a foundation for additional clinical trials of GMCI for lung cancer patients with potential benefit if combined with other immune therapies., Graphical Abstract, Predina and colleagues describe a phase I dose-escalation trial using bronchoscopic delivery of a replication-deficient adenovirus expressing herpes simplex thymidine kinase (AdV-tk) in lung cancer patients who underwent surgery. AdV-tk injection proved safe, feasible, and effectively induced CD8+ T cell activation in blood and in resected tumor tissues.
- Published
- 2020
4. Systemic high-dose dexamethasone treatment may modulate the efficacy of intratumoral viral oncolytic immunotherapy in glioblastoma models
- Author
-
Marilin S Koch, Mykola Zdioruk, Michal O Nowicki, Alec M Griffith, Estuardo Aguilar, Laura K Aguilar, Brian W Guzik, Francesca Barone, Paul P Tak, Ghazaleh Tabatabai, James A Lederer, E Antonio Chiocca, and Sean Lawler
- Subjects
Pharmacology ,Oncolytic Virotherapy ,Cancer Research ,brain neoplasms ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Dexamethasone ,Mice ,Oncolytic and Local Immunotherapy ,Oncology ,translational medical research ,Tumor Microenvironment ,Molecular Medicine ,Immunology and Allergy ,Animals ,Humans ,Female ,Immunotherapy ,Glioblastoma ,Glucocorticoids ,RC254-282 - Abstract
BackgroundIntratumoral viral oncolytic immunotherapy is a promising new approach for the treatment of a variety of solid cancers. CAN-2409 is a replication-deficient adenovirus that delivers herpes simplex virus thymidine kinase to cancer cells, resulting in local conversion of ganciclovir or valacyclovir into a toxic metabolite. This leads to highly immunogenic cell death, followed by a local immune response against a variety of cancer neoantigens and, next, a systemic immune response against the injected tumor and uninjected distant metastases. CAN-2409 treatment has shown promising results in clinical studies in glioblastoma (GBM). Patients with GBM are usually given the corticosteroid dexamethasone to manage edema. Previous work has suggested that concurrent dexamethasone therapy may have a negative effect in patients treated with immune checkpoint inhibitors in patients with GBM. However, the effects of dexamethasone on the efficacy of CAN-2409 treatment have not been explored.MethodsIn vitro experiments included cell viability and neurosphere T-cell killing assays. Effects of dexamethasone on CAN-2409 in vivo were examined using a syngeneic murine GBM model; survival was assessed according to Kaplan-Meier; analyses of tumor-infiltrating lymphocytes were performed with mass cytometry (CyTOF - cytometry by time-of-flight). Data were analyzed using a general linear model, with one-way analysis of variance followed by Dunnett’s multiple comparison test, Kruskal-Wallis test, Dunn’s multiple comparison test or statistical significance analysis of microarrays.ResultsIn a mouse model of GBM, we found that high doses of dexamethasone combined with CAN-2409 led to significantly reduced median survival (29.0 days) compared with CAN-2409 treatment alone (39.5 days). CyTOF analyses of tumor-infiltrating immune cells demonstrated potent immune stimulation induced by CAN-2409 treatment. These effects were diminished when high-dose dexamethasone was used. Functional immune cell characterization suggested increased immune cell exhaustion and tumor promoting profiles after dexamethasone treatment.ConclusionOur data suggest that concurrent high-dose dexamethasone treatment may impair the efficacy of oncolytic viral immunotherapy of GBM, supporting the notion that dexamethasone use should be balanced between symptom control and impact on the therapeutic outcome.
- Published
- 2021
5. CTIM-09. ENRICHED TCR/BCR VDJ REARRANGEMENTS CORRELATE WITH MRI AND SURVIVAL OUTCOMES IN PATIENTS WITH RECURRENT HIGH-GRADE GLIOMA TREATED WITH CAN-3110
- Author
-
E Antonio Chiocca, Hiroshi Nakashima, Xiaokui Mo, Isaac Solomon, Alexander Ling, Jared Woods, Joshua Bernstock, Genaro Villa, Raziye Piranlioglu, Ana Montalvo Landivar, Nafisa Masud, Daniel Triggs, James Grant, Patrick Y Wen, Eudocia Lee, Lakshmi Nayak, Ugonma Chukwueke, Tracy Batchelor, David Krisky, Estuardo Aguilar-Cordova, Laura K Aguilar, Soledad Fernandez, Christopher Matheny, Andrea Manzanera, Francesca Barone, Paul Peter Tak, Keith Ligon, and David A Reardon
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BACKGROUND CAN-3110 (rQNestin34.5v2) is an HSV-1 oncolytic viral immunotherapy with one copy of the inflammatory ICP34.5 gene under transcriptional control of the Nestin glioma-specific promoter. We completed a phase 1 sequential dose-escalation trial of CAN-3110 in recurrent high-grade glioma (rHGG). METHODS CAN-3110 was injected intratumorally starting at 1x106 plaque forming units (pfu) and dose- escalated by half log up to 1x1010 pfu in biopsy confirmed rHGG. An expansion cohort of 12 patients was then accrued at 1x109 pfus. Blood and post-injection rHGG were collected. RESULTS 41 rHGG patients were treated (42 separate interventions): median age 56 years (range 27-74); 21 females, 20 males; median baseline KPS 90 (range 70-100). CAN-3110 administration was well-tolerated with no dose limiting toxicities. Median overall survival (mOS) was 11.9 months. Histologic and molecular analyses showed significantly increased T cell infiltration in post treatment samples with elevated T cell and/or B cell receptor (TCR/BCR) transcripts which correlated with patient survival (HR 0.26 for patients with elevated TCR/BCR rearrangements as compared to patients with low). Volumetric analyses of MRI suggest a trend between reduction in the relative change in tumor growth, TCR/BCRs enrichment and survival in CAN-3110 treated patients. CLINICAL IMPLICATIONS Administration of CAN-3110 into rHGG was well tolerated. OS of CAN-3110 treated subjects compare favorably to historical controls. The association of increased TCR/BCR transcripts with survival suggests that CAN-3110 induces T cell responses against rHGG, supporting further clinical development of CAN-3110 viral immunotherapy.
- Published
- 2022
- Full Text
- View/download PDF
6. Phase I study of gene-mediated cytotoxic immunotherapy with AdV-tk as adjuvant to surgery and radiation for pediatric malignant glioma and recurrent ependymoma
- Author
-
Liliana Goumnerova, Stewart Goldman, Rishi Lulla, Estuardo Aguilar-Cordova, Susan N. Chi, Peter E. Manley, Andrea G. Manzanera, Mark W. Kieran, Tadanori Tomita, Karen J. Marcus, Laura K. Aguilar, and Arthur J DiPatri
- Subjects
0301 basic medicine ,Ependymoma ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,03 medical and health sciences ,0302 clinical medicine ,Monitoring, Immunologic ,Glioma ,medicine ,Humans ,Child ,Chemotherapy ,Temozolomide ,Brain Neoplasms ,business.industry ,Common Terminology Criteria for Adverse Events ,Genetic Therapy ,medicine.disease ,Chemotherapy regimen ,Surgery ,Radiation therapy ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Immunotherapy ,Neurology (clinical) ,business ,Pediatric Neuro-Oncology ,medicine.drug ,Anaplastic astrocytoma - Abstract
Background Gene-mediated cytotoxic immunotherapy (GMCI) is a tumor-specific immune stimulatory strategy implemented through local delivery of aglatimagene besadenovec (AdV-tk) followed by anti-herpetic prodrug. GMCI induces T-cell dependent tumor immunity and synergizes with radiotherapy. Clinical trials in adult malignant gliomas demonstrated safety and potential efficacy. This is the first trial of GMCI in pediatric brain tumors. Methods This phase I dose escalation study was conducted to evaluate GMCI in patients 3 years of age or older with malignant glioma or recurrent ependymoma. AdV-tk at doses of 1 × 1011 and 3 × 1011 vector particles (vp) was injected into the tumor bed at the time of surgery followed by 14 days of valacyclovir. Radiation started within 8 days of surgery, and if indicated, chemotherapy began after completion of valacyclovir. Results Eight patients (6 glioblastoma, 1 anaplastic astrocytoma, 1 recurrent ependymoma) were enrolled and completed therapy: 3 on dose level 1 and 5 on dose level 2. Median age was 12.5 years (range 7-17) and Lansky/Karnofsky performance scores were 60-100. Five patients had multifocal/extensive tumors that could not be resected completely and 3 had gross total resection. There were no dose-limiting toxicities. The most common possibly GMCI-related adverse events included Common Terminology Criteria for Adverse Events grade 1-2 fever, fatigue, and nausea/vomiting. Three patients, in dose level 2, lived more than 24 months, with 2 alive without progression 37.3 and 47.7 months after AdV-tk injection. Conclusions GMCI can be safely combined with radiation therapy with or without temozolomide in pediatric patients with brain tumors and the present results strongly support further investigation. Clinical trial registry ClinicalTrials.gov NCT00634231.
- Published
- 2019
- Full Text
- View/download PDF
7. First report of safety/tolerability and preliminary antitumor activity of CAN-2409 in inadequate responders to immune checkpoint inhibitors for stage III/IV NSCLC
- Author
-
Charu Aggarwal, Andrew Haas, Sarah W. Gordon, Ranee Mehra, Peter M Lee, Christine M. Bestvina, Fabien Maldonado, Vamsidhar Velcheti, Roy S. Herbst, Susan D. Bell, Rachael Gillmor, Andrea Manzanera, Christopher Jon Matheny, Estuardo Aguilar-Cordova, Laura K. Aguilar, Francesca Barone, Paul Peter Tak, and Daniel Sterman
- Subjects
Cancer Research ,Oncology - Abstract
9037 Background: Immune checkpoint inhibitors (ICI) are standard of care for advanced NSCLC. Even among patients with initial response, a majority ultimately progress, and rational combination approaches are needed to improve outcomes. CAN-2409 is a replication-deficient adenoviral gene construct that delivers the thymidine kinase gene, resulting in local conversion of a prodrug (valacyclovir) into a toxic metabolite. This leads to tumor cell lysis and immunization against the injected tumor and uninjected metastases. We have previously shown that monotherapy intra-tumoral (IT) delivery of CAN-2409 followed by oral valacyclovir in NSCLC patients is safe and results in CD8+ T cell infiltration in the injected tumor and activation of this cell population in tissue and peripheral blood. Methods: This open-label Ph2 experimental medicine clinical trial evaluates safety and clinical activity of IT CAN-2409 combined with ICI (± chemo) for stage III/IV NSCLC. Three cohorts are defined based on response to ICI at enrollment: stable disease (SD; Cohort 1; C1), progressive disease (PD) after ≥18 weeks (w) of ICI (Cohort 2; C2), or ICI refractory disease (RD; Cohort 3; C3). Two doses of CAN-2409 (5x1011 vp) are given 5-7w apart via bronchoscopic or percutaneous injection into a lung tumor, disease-positive lymph node or peripheral metastasis, followed by valacyclovir. Patients are assessed for safety, immunologic biomarkers (analysis in progress), and clinical response. Results: As of data cutoff (10Jan22), 28 patients received ≥1 dose of CAN-2409 (safety population). Median age was 70 years; 86% stage IV; 32% squamous; 11% PD-L1 >50%; 82% receiving pembrolizumab and 18% nivolumab. Study treatment and procedures were generally well tolerated. The most common TRAEs were Gr1/2, with fatigue, fever, and chills in 18-39% of patients; 1 patient had Gr3 fever. Twenty-two patients are alive and 6 patients died due to disease. Of the 14 RECIST evaluable patients who received 2 doses of CAN-2409, clinical response was seen in 4 patients (Table 1). Two PRs are ongoing (6w, 24w) and reduction in tumor size was observed in non-injected lesions. In C2, 6 of 7 patients achieving SD are ongoing with median duration of 13w (range 10-40w). Conclusions: The addition of CAN-2409 for patients with advanced NSCLC and inadequate response to front-line ICI (± chemo) appears to be well tolerated. Preliminary clinical data suggest that CAN-2409 induced a clinical response in 4/14 evaluable patients and produced disease stabilization in most patients entering the trial with PD, with evidence of abscopal effect in a subset of patients. Clinical trial information: NCT04495153. [Table: see text]
- Published
- 2022
- Full Text
- View/download PDF
8. Therapeutic Endoscopic Ultrasonography: Intratumoral Injection for Pancreatic Adenocarcinoma
- Author
-
Lawrence A. Shirley, Laura K. Aguilar, Estuardo Aguilar-Cordova, Mark Bloomston, and Jon P. Walker
- Subjects
Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Pancreatic adenocarcinoma is an aggressive disease that has poor outcomes despite maximal traditional therapies. Thus, treatment of this cancer demands innovative strategies to be used in addition to standing therapies in order to provide new avenues of care. Here, we describe the technique of using endoscopic ultrasound in order to directly inject both novel and conventional therapies into pancreatic tumors. We detail the rationale behind this strategy and the many benefits it provides. We then describe our technique in detail, including our experience injecting the AdV-tk adenoviral vector to create an in situ vaccine effect.
- Published
- 2013
- Full Text
- View/download PDF
9. 468 Enhancers and repressors of immunotherapy: translational perspectives on gene-mediated cytotoxic immunotherapy in glioblastoma
- Author
-
Laura K. Aguilar, Ghazaleh Tabatabai, Marilin Koch, Brian W. Guzik, Michał Nowicki, Estuardo Aguilar-Cordova, Sean E. Lawler, E. Antonio Chiocca, and Mikolay Zdioruk
- Subjects
0301 basic medicine ,Cell growth ,business.industry ,medicine.medical_treatment ,Immunotherapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,lcsh:RC254-282 ,Immune checkpoint ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Aglatimagene Besadenovec ,In vivo ,030220 oncology & carcinogenesis ,Glioma ,medicine ,Cancer research ,Cytotoxic T cell ,business - Abstract
Background Gene-mediated cytotoxic immunotherapy (GMCI) is a local tumor immunotherapy that uses aglatimagene besadenovec (a non-replicating serotype 5 adenovirus, expressing HSV1 thymidine kinase) with the prodrug ganciclovir to induce DNA double strand breaks (DSB), leading to immunogenic tumor cell death and intratumoral immune cell invasion. Here we investigate potential repressors and enhancers of GMCI’s effectiveness. GMCI is currently in clinical trials in combination with immune checkpoint blockade in glioblastoma. Thus we set out to identify potential areas to improve this approach for future application. Dexamethasone is used in symptomatic treatment of glioma patients, although it is known to cause immune suppression. However, the influence of dexamethasone on the efficacy of GMCI has not been explored. In contrast, DNA damage response inhibitors like the ATR inhibitor (ATRi) AZD6738 might not only amend the cytotoxic but also the immunogenic profile of GMCI, rendering it an attractive combination partner. Methods We investigated the effects of ATR-inhibition and dexamethasone on GMCI in vitro using cytotoxicity, flow cytometry and T-cell-killing assays in glioblastoma cell lines. The impact of dexamethasone and ATRi in vivo was assessed in an orthotopic syngeneic murine glioblastoma model. Tumor immune infiltrates were analyzed with flow cytometry. Results Cytotoxicity assays showed that dexamethasone has a slight impact on GMCI in vitro. In T-cell-functional assays, we observed a significantly impaired tumor cell killing. Immune cell response assays revealed a reduced immune cell proliferation after co-culture with supernatant from dexamethasone or combination treated glioblastoma cells. In vivo, while treatment with GMCI alone resulted in longer median symptom-free survival (39.5d) versus no treatment (23d), the combination of GMCI and dexamethasone resulted in the significant reduction of this effect (29d vs 39.5d ; p = 0.0184). The combination of ATRi with GMCI proved to be synergistic in cytotoxicity assays. Flow cytometry revealed a significant increase in DSB-associated H2AX foci as well as an improved immune profile by downregulation of GMCI-induced PD-L1 expression. In vivo, the combination with ATRi led to an increase in long-term surviving animals (66.7%) compared to GMCI (50%) and proved to be highly significant compared to the untreated control (p=0.0022). Conclusions Our data suggest that dexamethasone may decrease the efficacy of immunotherapy for glioma through impaired T cell function: this emphasizes the need in identifying alternatives to dexamethasone to prevent attenuated responses in immunotherapies. The combination of GMCI with ATRi however points to additional therapeutic benefit through enhanced cytotoxic efficacy, improved immunogenicity in vitro and increased long-term survival in vivo, making it a promising future approach for the treatment of glioblastoma. Acknowledgements This was supported by NCI P01CA069246 (Chiocca)
- Published
- 2020
- Full Text
- View/download PDF
10. CTIM-13. PHASE 1 CLINICAL TRIAL OF ONCOLYTIC VIRAL IMMUNOTHERAPY WITH CAN-2409 + VALACYCLOVIR IN COMBINATION WITH NIVOLUMAB AND STANDARD OF CARE (SOC) IN NEWLY DIAGNOSED HIGH-GRADE GLIOMA (HGG)
- Author
-
Estuardo Aguilar-Cordova, Serena Desideri, Pierpaolo Peruzzi, Patrick Y. Wen, Wenya Linda Bi, Frank S. Lieberman, Ian Lee, James Snyder, Steven Brem, Paul Peter Tak, Sean E. Lawler, Susan Bell, Stephen J Bagley, Nirav J. Patel, Andrea G. Manzanera, E. Antonio Chiocca, Francesca Barone, Nduka Amankulor, Lenika Lopez, L. Burt Nabors, Megan Mantica, Neeraja Danda, Brian W. Guzik, Stephen B. Tatter, Stuart A. Grossman, Tobias Walbert, Laura K. Aguilar, Roy E. Strowd, Lixian Jin, Arati Desai, William Timmer, Xiaobu Ye, and David A. Reardon
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Standard of care ,Temozolomide ,business.industry ,medicine.medical_treatment ,Phases of clinical research ,Immunotherapy ,medicine.disease ,Oncolytic virus ,Internal medicine ,Glioma ,medicine ,Neurology (clinical) ,Nivolumab ,Adverse effect ,business ,medicine.drug - Abstract
BACKGROUND CAN-2409 is a replication-deficient adenovirus that delivers HSV thymidine kinase to cancer cells, resulting in local conversion of orally administered valacyclovir into a toxic metabolite. Previously, a phase 1b/2 clinical trial of CAN-2409 combined with standard-of-care (SOC) demonstrated safety and improved survival in HGG patients. Addition of CAN-2409 to nivolumab has the potential to activate locally recruited lymphocytes and teach them to recognize tumor neoantigens, changing the ‘cold’ immunosuppressive tumor microenvironment, and synergizing with the activity mediated by immune checkpoint inhibitors. This notion is supported by preclinical experiments showing that the combination of CAN-2409 with anti-PD1 therapy was more effective than either treatment alone. METHODS This ongoing phase 1 clinical trial evaluates safety and initial efficacy of CAN-2409 combined with nivolumab and SOC in newly diagnosed HGG. CAN-2409 is injected during neurosurgery into the tumor bed, followed by 14-days of valacyclovir. Radiation starts within 8 (+/-4) days of surgery. Temozolomide is administered to MGMT-methylation positive patients only. Nivolumab is given every 2 weeks, up to 52-weeks. Deep immune profiling studies are ongoing and initial results will be available shortly. RESULTS From February 2019 to March 2021, 41 patients were enrolled and 35 were evaluable for safety from the combination of CAN-2409, nivolumab and SOC: 24 male and 11 female; 34 glioblastoma, 1 diffuse astrocytoma; 33 IDH-wildtype, 2 IDH-mutant; 15 MGMT-methylated, 20 unmethylated. Median age was 62-years (range 28-79), median KPS 90 (range 80-100). With 13 months median follow-up, no unexpected serious adverse events were observed, and 23 patients are still alive. The most frequent possibly related adverse events (>10%) were nausea, fatigue, fever, headache, and increased ALT. CONCLUSIONS The combination of CAN-2409 + nivolumab + SOC was well tolerated. Clinical follow-up and extensive biomarker analyses will provide a better understanding of the therapeutic potential of this approach.
- Published
- 2021
- Full Text
- View/download PDF
11. 395 Detection of viral antigen and immune activation after intra-tumor injection of CAN-3110 (ICP-34.5 expressing HSV-1 oncolytic virus) in patients with recurrent high-grade glioma
- Author
-
Jared Woods, Hiroshi Nakashima, David A. Reardon, Sean E. Lawler, Sascha Marx, Anita Giobbie-Hurder, Yu Zeng, Patrick Y. Wen, E. Antonio Chiocca, Jessica Dwyer, Daniel Triggs, Mario L. Suvà, Isaac Soloman, Scott J. Rodig, Kai W. Wucherpfennig, Francesca Barone, Simon Gritsch, William Pisano, Eudocia Q. Lee, Nathan Mathewson, Paul Peter Tak, Abigail Tianai Zhang, Estuardo Aguilar-Cordova, Brian W. Guzik, James Grant, Mariano Severgnini, Keith L. Ligon, Laura K. Aguilar, and David Krisky
- Subjects
Pharmacology ,Cancer Research ,business.industry ,T cell ,Immunology ,T-cell receptor ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease_cause ,medicine.disease ,Peripheral blood mononuclear cell ,Oncolytic virus ,Herpes simplex virus ,medicine.anatomical_structure ,Oncology ,Antigen ,Glioma ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business ,RC254-282 ,CD8 - Abstract
BackgroundRecurrent high-grade glioma (HGG) represents a significant clinical unmet need with expected survival between 6 to 9 months. Oncolytic viruses are a new therapeutic approach for solid tumors that deploy oncolytic activity combined with local and systemic immune activation. CAN-3110 (rQNestin34.5v2) is an oncolytic herpes simplex virus (HSV), modified to encode the HSV1 ICP34.5 protein under control of the nestin promoter. Selective expression of nestin in brain tumors confers tumor-restricted replication of CAN-3110. We conducted an open-label dose-escalation phase 1 clinical trial in patients with recurrent HGG to evaluate safety, tolerability, and immunological changes after CAN-3110 treatment.MethodsThirty patients with biopsy-confirmed recurrent HGG were enrolled from September 2017 to February 2020. CAN-3110 was injected intratumorally starting at 1x106 plaque forming units (pfu) and dose-escalated by half log to 1x1010 pfu. Patients also received standard of care. Peripheral blood mononuclear cells (PBMCs), plasma and tumor samples were collected for analysis at different time-points post treatment. We evaluated HSV antigen expression in tumor tissue. RNA sequencing and T cell receptor (TCR) rearrangement analysis was performed in matched tissue and PBMCs. Cytokine profiling was completed in 29 patients at baseline, day 2, and day 28 post treatment.ResultsEighteen patients were recruited at their first recurrence and 12 at the second recurrence. Three patients presented with multifocal disease. Tumor volume ranged from 357.4 to and 54,036.1mm3 (median 7,733.9mm3, SDV 15,610.2). CAN-3110 was well-tolerated with no dose-limiting toxicity. Median overall survival was 11.7 months. We demonstrated persistence of HSV antigen and CD8+ T cell infiltrates at the site of injected tumor. Preliminary analysis revealed expansion of shared TCR clonotypes and upregulation of pro-inflammatory genes in post-treatment tumors and peripheral blood samples. Longitudinal modeling of cytokine profiling demonstrated increased levels of IL-6, VEGF alpha, CCL2 and IL1-RA and a decrease in GCP-2 levels at day 2 post-treatment (p ConclusionsIntratumoral administration of CAN-3110 appears well-tolerated in recurrent HGG. Histologic, molecular, and cytokine analyses demonstrate persistence of viral antigen as well as local and systemic immune activation after treatment.Ethics ApprovalThe study was approved by the Office for Human Research Studies at Dana-Farber Cancer Institute, Protocol Number 16–557.
- Published
- 2021
- Full Text
- View/download PDF
12. First-in-human CAN-3110 (ICP-34.5 expressing HSV-1 oncolytic virus) in patients with recurrent high-grade glioma
- Author
-
Isaac H. Solomon, Estuardo Aguilar-Cordova, E. Antonio Chiocca, Abigail Tianai Zhang, Simon Gritsch, Nathan Mathewson, Hiroshi Nakashima, Patrick Y. Wen, Sean E. Lawler, William Pisano, Eudocia Q. Lee, Mario L. Suvà, James Grant, Daniel Triggs, Scott J. Rodig, Keith L. Ligon, David A. Reardon, Kai W. Wucherpfennig, David Krisky, and Laura K. Aguilar
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,First in human ,HSL and HSV ,Recurrent Glioma ,Oncolytic virus ,Patient population ,Internal medicine ,medicine ,In patient ,business ,High-Grade Glioma - Abstract
2009 Background: Recurrent glioma patients have few therapeutic options and an expected survival of only 7 to 10 months. New treatments to improve the prognosis of this patient population are a dire medical need. Oncolytic viruses (OVs) are emerging as important new agents for cancer treatment. The first FDA approved OV was talimogene laherparepvec (Imlygic, T-Vec) for treatment of melanoma. T-Vec, as most other clinical HSV-1 based OVs, is deleted in the ICP34.5 gene, which is responsible for HSV-1 neurovirulence. However, deletion of ICP34.5 also impedes efficient viral replication. CAN-3110 (rQNestin34.5v2) maintains a copy of the HSV1 ICP34.5 gene under transcriptional control of the tumor-specific promoter for nestin to drive robust tumor-selective replication. CAN-3110 replicates in malignant glioma cells far above levels seen with ICP34.5 deleted viruses. This potency also created the hypothetical risk for increased neurovirulence, thus the regulatory advice to conduct a cautious nine-dose-level Phase-1 dose escalation study in patients with recurrent high-grade glioma (HGG). Methods: From September 2017 to February 2020, thirty patients with biopsy-confirmed recurrent high-grade glioma were treated in an open label clinical trial. Patients with multifocal, multicentric, tumors larger than 5 cm, and tumors that had recurred multiple times were eligible. All patients received best standard of care treatments as indicated by their physician. CAN-3110 was injected intratumorally starting at 1x106 plaque forming units (pfu) and dose-escalating (3+3 design) by half log increments up to 1x1010 pfu. Tissue (when possible) and blood samples were obtained before and during treatment for experimental medicine analyses. Results: CAN-3110 was well tolerated with no dose limiting toxicity observed. The initial tissue diagnosis of the recurrent tumor for the 30 subjects was 26 glioblastoma, 3 anaplastic oligodendroglioma, and 1 anaplastic astrocytoma. The median overall survival (mOS) of the entire study group is 13.25 months. Post-treatment tissue is available for 18/30 subjects and revealed persistence of HSV antigen and CD8+ T cell infiltrates. Additional response, immunologic (including T cell receptor repertoire), transcriptomic and single cell RNA sequencing analyses are ongoing. Conclusions: Administration of CAN-3110 into recurrent glioma was well tolerated without evidence of ICP34.5-induced encephalitis/meningitis. Histological and molecular analyses showed evidence of biological activity and that CAN-3110 injection was associated with immune activation and viral antigen persistence. Although definitive clinical efficacy cannot be determined in this small phase 1 study, OS of CAN-3110 treated subjects compares favorably to historical reports and warrants further clinical studies. Clinical trial information: NCT03152318.
- Published
- 2021
- Full Text
- View/download PDF
13. Phase II multicenter study of gene-mediated cytotoxic immunotherapy as adjuvant to surgical resection for newly diagnosed malignant glioma
- Author
-
Behnam Badie, Lee A. Wheeler, Susan Bell, Bin S. Teh, Estuardo Aguilar-Cordova, Jana Portnow, Simon S. Lo, Laura K. Aguilar, John C. Grecula, Pamela Z. New, E. Antonio Chiocca, David S. Baskin, Todd Trask, Herbert B. Newton, Robert Cavaliere, Andrea G. Manzanera, and John M. McGregor
- Subjects
Adult ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Genetic Vectors ,Clinical Investigations ,Acyclovir ,Antiviral Agents ,Thymidine Kinase ,Adenoviridae ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Glioma ,medicine ,Humans ,Simplexvirus ,Survival rate ,Survival analysis ,Aged ,Temozolomide ,Brain Neoplasms ,business.industry ,Hazard ratio ,Valine ,Genetic Therapy ,Middle Aged ,medicine.disease ,Debulking ,Survival Analysis ,Minimal residual disease ,Confidence interval ,Surgery ,Treatment Outcome ,030104 developmental biology ,Chemotherapy, Adjuvant ,Valacyclovir ,030220 oncology & carcinogenesis ,Immunotherapy ,Neurology (clinical) ,business ,medicine.drug - Abstract
BACKGROUND: Gene Mediated Cytotoxic Immunotherapy (GMCI) generates a polyvalent anti-tumor immune response through local delivery of aglatimagene besadenovec (AdV-tk) which synergizes with standard of care (SOC). METHODS: A Phase II trial was designed to evaluate overall survival (OS) after GMCI + SOC compared to a concurrent matched control group meeting protocol criteria and SOC at an institution not active in the treatment trial. Primary efficacy analysis was planned on the null hypothesis of no improvement in the 2-yr survival over the SOC group with planned subset analysis of significant disease prognostic factors. RESULTS: From 2006 to 2010, 48 patients completed SOC + GMCI and 134 SOC in the matched cohort. There were no dose-limiting toxicities. Fever, fatigue and headache were the most common GMCI-related symptoms. Median OS was 17.1 and 13.5 months for GMCI + SOC and SOC, respectively (hazard ratio [HR] 0.72, 95% confidence interval [CI] 0.52-0.99, p = 0.0417). Median PFS was 8.1 and 6.5 months for GMCI + SOC and SOC, respectively (HR 0.66, 95% CI 0.48-0.91, p = 0.0100). OS at 1- 2- and 3-years increased from 57%, 22% and 8% to 67%, 35% and 19%, respectively. The improvement was mostly in patients that underwent gross total resection: median OS increased from 16.9 to 25 months (p = 0.0492); 1- 2- and 3-year survival rates from 64%, 28% and 6% to 90%, 53% and 32%. CONCLUSIONS: GMCI can be safely combined with SOC in newly diagnosed malignant gliomas. Survival rates compared favorably to historical reports and a matched control group. Survival outcomes were significantly improved in patients with minimal residual disease after total resection. The 2-yr survival rate met the planned statistical threshold for significance. This is the first study to demonstrate a correlation between maximum debulking and a survival advantage using immunotherapy. These data strongly support further evaluation of GMCI for malignant gliomas.
- Published
- 2016
- Full Text
- View/download PDF
14. Neoadjuvant endobronchial delivery of gene mediated cytotoxic immunotherapy (GMCI) for non-small cell lung cancer (NSCLC): Safety and immunologic activity
- Author
-
Andrew R. Haas, Steven M. Albelda, Laura K. Aguilar, Evgeniy Eruslanov, Marina Martinez, Mitchell G. Bryski, Christopher Corbett, Charuhas Deshpande, Brian W. Guzik, Andrea G. Manzanera, Shaun O'Brien, Edmund K. Moon, Jarrod D. Predina, Estuardo Aguilar-Cordova, Sunil Singhal, and Lydia Frenzel Sulyok
- Subjects
Cancer Research ,business.industry ,medicine.medical_treatment ,non-small cell lung cancer (NSCLC) ,Immunotherapy ,Prodrug ,medicine.disease ,Aglatimagene Besadenovec ,Oncology ,Cancer research ,Medicine ,Cytotoxic T cell ,business ,Gene - Abstract
9050 Background: GMCI is a tumor-specific immuno-oncology approach implemented through local delivery of aglatimagene besadenovec(AdV-tk) followed by anti-herpetic prodrug. This leads to immunogenic tumor cell death, antigen presenting cell activation, and T cell stimulation resulting in CD8+ T cell dependent protection, as demonstrated in preclinical models and clinical trials in other tumor types. This is the first study to assess endobronchial delivery of AdV-tk for NSCLC. Methods: This Phase I dose escalation trial enrolled patients with suspected NSCLC who were candidates for surgery. A single AdV-tk injection was performed by endobronchial ultrasound (n = 11) or mediastinoscopy (n = 1) during the diagnostic staging procedure 3 weeks prior to surgery. Three dose levels were evaluated: 2.5x 1011, 5x1011, and 1x1012 vector particles (vp) in a 3+3 design. Valacyclovir was administered for 14 days, starting the day after AdV-tk injection. To assess the local and systemic effects of GMCI, immune biomarkers were evaluated in blood and tumor samples before and after GMCI. Results: From 2017-2019, 12 patients (9 men, 3 women, median age 65 [range 55-80]) received GMCI followed by surgery. Average tumor size was 5.1 cm (largest diameter) and final pathologic stage was I (n = 4), II (n = 3), and III (n = 5). Treatment-related adverse events were CTC grade 1 fever (n = 1), flu-like symptoms (n = 1) and nausea/vomiting/diarrhea (n = 1). The only > grade 2 lab abnormality was transient grade 3 lymphopenia (n = 2). A measurable reduction in tumor size was observed in one patient. The average amount of tumor necrosis was 29.4%. Significant infiltration of CD8+T cells (5.2-fold compared to baseline, p = 0.001) was found in tumor 19-22 days after AdV-tk injection. Within the CD8+tumor infiltrating lymphocytes, there was increased expression of CD38 (2.5-fold, p = 0.002), Ki67 (4.8-fold, p = 0.02), PD1 (1.9-fold, p = 0.002), CD39 (2.9-fold, p = 0.04) and CTLA-4 (4.8-fold, p < 0.001), without significant detected differences in Tim3 or TIGIT. Simultaneously, peripheral blood CD8+ cells displayed significant increases in CD38 (3.4-fold, p = 0.006), HLA-DR (4.2-fold, p = 0.002), and Ki67 (5.8-fold, p = 0.017). Conclusions: Intratumoral injection of AdV-tk into lung tumors was safe and feasible. Further, AdV-tk effectively induced peripheral blood and intra-tumoral CD8 T cell activation. Consequent upregulation of inhibitory receptors suggests a potential benefit for combination therapies. Clinical trial information: NCT03131037.
- Published
- 2020
- Full Text
- View/download PDF
15. Gene-mediated cytotoxic immunotherapy as adjuvant to surgery or chemoradiation for pancreatic adenocarcinoma
- Author
-
Tanios Bekaii-Saab, Jon P. Walker, Walter J. Coyle, Mark Bloomston, Gregory B. Lesinski, Lawrence A. Shirley, Christopher L. Marsh, Daniel Sanchez, Estuardo Aguilar-Cordova, Andrea G. Manzanera, Howard Marx, Vincent Chung, Laura K. Aguilar, and Benjamin Swanson
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Genetic enhancement ,Genetic Vectors ,Immunology ,Acyclovir ,Adenocarcinoma ,Thymidine Kinase ,Adenoviridae ,Internal medicine ,Pancreatic cancer ,PD-L1 ,medicine ,Humans ,Immunology and Allergy ,Combined Modality Therapy ,Aged ,Dose-Response Relationship, Drug ,biology ,business.industry ,Valine ,Chemoradiotherapy ,Genetic Therapy ,Immunotherapy ,Middle Aged ,medicine.disease ,Immunohistochemistry ,Surgery ,Pancreatic Neoplasms ,Valacyclovir ,biology.protein ,Female ,Neoplasm Recurrence, Local ,business ,Adjuvant - Abstract
While surgical resection of pancreatic adenocarcinoma provides the only chance of cure, long-term survival remains poor. Immunotherapy may improve outcomes, especially as adjuvant to local therapies. Gene-mediated cytotoxic immunotherapy (GMCI) generates a systemic anti-tumor response through local delivery of an adenoviral vector expressing the HSV-tk gene (aglatimagene besadenovec, AdV-tk) followed by anti-herpetic prodrug. GMCI has demonstrated synergy with standard of care (SOC) in other tumor types. This is the first application in pancreatic cancer.Four dose levels (3 × 10(10) to 1 × 10(12) vector particles) were evaluated as adjuvant to surgery for resectable disease (Arm A) or to 5-FU chemoradiation for locally advanced disease (Arm B). Each patient received two cycles of AdV-tk + prodrug.Twenty-four patients completed therapy, 12 per arm, with no dose-limiting toxicities. All Arm A patients were explored, eight were resected, one was locally advanced and three had distant metastases. CD8(+) T cell infiltration increased an average of 22-fold (range sixfold to 75-fold) compared with baseline (p = 0.0021). PD-L1 expression increased in 5/7 samples analyzed. One node-positive resected patient is alive66 months without recurrence. Arm B RECIST response rate was 25 % with a median OS of 12 months and 1-year survival of 50 %. Patient-reported quality of life showed no evidence of deterioration.AdV-tk can be safely combined with pancreatic cancer SOC without added toxicity. Response and survival compare favorably to expected outcomes and immune activity increased. These results support further evaluation of GMCI with more modern chemoradiation and surgery as well as PD-1/PD-L1 inhibitors in pancreatic cancer.
- Published
- 2015
- Full Text
- View/download PDF
16. Phase I Study of Intrapleural Gene-Mediated Cytotoxic Immunotherapy in Patients with Malignant Pleural Effusion
- Author
-
Tracey L. Evans, Andrew R. Haas, Sharyn I. Katz, Joshua Bauml, Steven M. Albelda, Daniel H. Sterman, Gregoria Gómez-Hernández, Charu Aggarwal, Susan Metzger, Estuardo Aguilar-Cordova, Andrea G. Manzanera, Roger B. Cohen, Evan W. Alley, Corey J. Langer, and Laura K. Aguilar
- Subjects
0301 basic medicine ,Oncology ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Genetic Vectors ,Adenoviridae ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,Positron Emission Tomography Computed Tomography ,Drug Discovery ,Genetics ,Clinical endpoint ,Medicine ,Malignant pleural effusion ,Humans ,Mesothelioma ,Lung cancer ,Molecular Biology ,Aged ,Pharmacology ,Aged, 80 and over ,Chemotherapy ,business.industry ,Immunotherapy ,Genetic Therapy ,Middle Aged ,medicine.disease ,Combined Modality Therapy ,Pleural Effusion, Malignant ,Respiratory Function Tests ,Cytokine release syndrome ,030104 developmental biology ,Treatment Outcome ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,Original Article ,business ,Biomarkers ,T-Lymphocytes, Cytotoxic - Abstract
Gene-mediated cytotoxic immunotherapy (GMCI) is an immune strategy implemented through local delivery of an adenovirus-based vector expressing the thymidine kinase gene (aglatimagene besadenovec, AdV-tk) followed by anti-herpetic prodrug valacyclovir. A phase I dose escalation trial of GMCI followed by chemotherapy was conducted in patients with malignant pleural effusion (MPE). AdV-tk was administered intrapleurally (IP) in three cohorts at a dose of 1 × 10(12) to 10(13) vector particles. Primary endpoint was safety; secondary endpoints included response rate, progression-free survival, and overall survival. Nineteen patients were enrolled: median age 67 years; 14 with malignant mesothelioma, 4 non-small-cell lung cancer (NSCLC), and 1 breast cancer. There were no dose limiting toxicities. All 3 patients in cohort 2 experienced transient cytokine release syndrome (CRS). Addition of celecoxib in cohort 3 reduced the incidence and severity of CRS (none > grade 2). Three patients are alive (23–33 months after GMCI), and 3 of 4 NSCLC patients had prolonged disease stabilization; one is alive 29 months after GMCI, 3.6 years after initial diagnosis. GMCI was safe and well tolerated in combination with chemotherapy in patients with MPE and showed encouraging response. Further studies are warranted to determine efficacy.
- Published
- 2017
17. Preclinical investigation of combined gene-mediated cytotoxic immunotherapy and immune checkpoint blockade in glioblastoma
- Author
-
Hiroshi Nakashima, Patrick Y. Wen, David A. Reardon, Michał Nowicki, Brian W. Guzik, Maria Carmela Speranza, Laura K. Aguilar, Gordon J. Freeman, Johanna K. Kaufmann, Franz Ricklefs, Xandra O. Breakefield, Ralph Weissleder, Agnieszka Bronisz, Sarah R. Klein, Carmela Passaro, Prisca Obi, Kazue Kasai, Sean E. Lawler, Abdul-Kareem Ahmed, E. Antonio Chiocca, and Estuardo Aguilar-Cordova
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,T-Lymphocytes ,Programmed Cell Death 1 Receptor ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,In vivo ,Cell Line, Tumor ,medicine ,Cytotoxic T cell ,Animals ,Humans ,Interferon gamma ,Microglia ,business.industry ,Brain Neoplasms ,Antibodies, Monoclonal ,Immunotherapy ,Combined Modality Therapy ,Immune checkpoint ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,Basic and Translational Investigations ,Cancer research ,Neurology (clinical) ,business ,Glioblastoma ,CD8 ,medicine.drug - Abstract
Background Combined immunotherapy approaches are promising cancer treatments. We evaluated anti-programmed cell death protein 1 (PD-1) treatment combined with gene-mediated cytotoxic immunotherapy (GMCI) performed by intratumoral injection of a prodrug metabolizing nonreplicating adenovirus (AdV-tk), providing in situ chemotherapy and immune stimulation. Methods The effects of GMCI on PD ligand 1 (PD-L1) expression in glioblastoma were investigated in vitro and in vivo. The efficacy of the combination was investigated in 2 syngeneic mouse glioblastoma models (GL261 and CT-2A). Immune infiltrates were analyzed by flow cytometry. Results GMCI upregulated PD-L1 expression in vitro and in vivo. Both GMCI and anti-PD-1 increased intratumoral T-cell infiltration. A higher percentage of long-term survivors was observed in mice treated with combined GMCI/anti-PD-1 relative to single treatments. Long-term survivors were protected from tumor rechallenge, demonstrating durable memory antitumor immunity. GMCI led to elevated interferon gamma positive T cells and a lower proportion of exhausted double positive PD1+TIM+CD8+ T cells. GMCI also increased PD-L1 levels on tumor cells and infiltrating macrophages/microglia. Our data suggest that anti-PD-1 treatment improves the effectiveness of GMCI by overcoming interferon-induced PD-L1-mediated inhibitory signals, and GMCI improves anti-PD-1 efficacy by increasing tumor-infiltrating T-cell activation. Conclusions Our data show that the GMCI/anti-PD-1 combination is well tolerated and effective in glioblastoma mouse models. These results support evaluation of this combination in glioblastoma patients.
- Published
- 2017
18. EPT-14PHASE I STUDY OF GENE MEDIATED CYTOTOXIC IMMUNOTHERAPY WITH AdV-tk AS ADJUVANT TO SURGERY AND RADIATION THERAPY FOR PEDIATRIC MALIGNANT GLIOMA AND RECURRENT EPENDYMOMA
- Author
-
Mark W. Kieran, Liliana Goumnerova, Peter Manley, Susan N. Chi, Karen Marcus, Andrea G. Manzanera, Estuardo Aguilar-Cordova, Arthur J. DiPatri, Tadanori Tomita, Rishi Lulla, Laura K. Aguilar, and Stewart Goldman
- Subjects
Cancer Research ,Abstracts ,Oncology ,Neurology (clinical) - Published
- 2016
19. The Spectrum of Vaccine Therapies for Patients With Glioblastoma Multiforme
- Author
-
Laura K. Aguilar, Mariel Arvizu, E. Antonio Chiocca, and Estuardo Aguilar-Cordova
- Subjects
Oncology ,medicine.medical_specialty ,Dacarbazine ,medicine.medical_treatment ,Acyclovir ,Autoantigens ,Article ,Central Nervous System Neoplasms ,Internal medicine ,Temozolomide ,medicine ,Overall survival ,Humans ,Combined Modality Therapy ,Pharmacology (medical) ,Chemotherapy ,business.industry ,Immunotherapy, Active ,Valine ,Dendritic Cells ,Immunotherapy ,medicine.disease ,Valacyclovir ,Immunology ,Glioblastoma ,business ,Median survival ,Signal Transduction ,medicine.drug - Abstract
Glioblastoma multiforme (GBM) is the most common primary malignant tumor of the central nervous system (CNS) and one of the most lethal cancers in adults and children. Despite aggressive treatment with surgery, radiation, and chemotherapy, median survival is less than 15 months and overall survival is less than 10 % at 5 years. Development of therapeutics for malignant gliomas has been hampered by their natural complexity as well as protective mechanisms unique to the CNS. Better understanding of the pathogenesis of GBM is opening the path to novel, specific-targeted therapies. Recently, multiple immunotherapy approaches have been acquiring substantial indication of therapeutic efficacy with a very safe profile. Examples of the leading clinical approaches for GBM will be discussed in detail in this review.
- Published
- 2012
- Full Text
- View/download PDF
20. Phase IB Study of Gene-Mediated Cytotoxic Immunotherapy Adjuvant to Up-Front Surgery and Intensive Timing Radiation for Malignant Glioma
- Author
-
Estuardo Aguilar-Cordova, E. Antonio Chiocca, Kimbra S. Harris, John M. McGregor, Susan Bell, Simon S. Lo, Abhik Ray-Chaudhuri, Laura K. Aguilar, Balveen Kaur, David S. Baskin, Todd Trask, Jayson Hardcastle, Pamela Z. New, Carissa Monterroso, Arnab Chakravarti, Herbert B. Newton, John C. Grecula, Robert Cavaliere, Andrea G. Manzanera, and Robert G. Grossman
- Subjects
Adult ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Dacarbazine ,Genetic Vectors ,Acyclovir ,Herpesvirus 1, Human ,Antiviral Agents ,Cancer Vaccines ,Thymidine Kinase ,Adenoviridae ,O(6)-Methylguanine-DNA Methyltransferase ,Adjuvants, Immunologic ,Glioma ,Temozolomide ,medicine ,Humans ,Antineoplastic Agents, Alkylating ,Aged ,Brain Neoplasms ,business.industry ,O-6-methylguanine-DNA methyltransferase ,Valine ,Genetic Therapy ,Immunotherapy ,Middle Aged ,medicine.disease ,Combined Modality Therapy ,Surgery ,Treatment Outcome ,Oncology ,Thymidine kinase ,Valacyclovir ,business ,Adjuvant ,Chemoradiotherapy ,medicine.drug - Abstract
Purpose Despite aggressive therapies, median survival for malignant gliomas is less than 15 months. Patients with unmethylated O6-methylguanine–DNA methyltransferase (MGMT) fare worse, presumably because of temozolomide resistance. AdV-tk, an adenoviral vector containing the herpes simplex virus thymidine kinase gene, plus prodrug synergizes with surgery and chemoradiotherapy, kills tumor cells, has not shown MGMT dependency, and elicits an antitumor vaccine effect. Patients and Methods Patients with newly diagnosed malignant glioma received AdV-tk at 3 × 1010, 1 × 1011, or 3 × 1011 vector particles (vp) via tumor bed injection at time of surgery followed by 14 days of valacyclovir. Radiation was initiated within 9 days after AdV-tk injection to overlap with AdV-tk activity. Temozolomide was administered after completing valacyclovir treatment. Results Accrual began December 2005 and was completed in 13 months. Thirteen patients were enrolled and 12 completed therapy, three at dose levels 1 and 2 and six at dose level 3. There were no dose-limiting or significant added toxicities. One patient withdrew before completing prodrug because of an unrelated surgical complication. Survival at 2 years was 33% and at 3 years was 25%. Patient-reported quality of life assessed with the Functional Assessment of Cancer Therapy-Brain (FACT-Br) was stable or improved after treatment. A significant CD3+ T-cell infiltrate was found in four of four tumors analyzed after treatment. Three patients with MGMT unmethylated glioblastoma multiforme survived 6.5, 8.7, and 46.4 months. Conclusion AdV-tk plus valacyclovir can be safely delivered with surgery and accelerated radiation in newly diagnosed malignant gliomas. Temozolomide did not prevent immune responses. Although not powered for efficacy, the survival and MGMT independence trends are encouraging. A phase II trial is ongoing.
- Published
- 2011
- Full Text
- View/download PDF
21. Neoadjuvant in situ gene-mediated cytotoxic immunotherapy improves postoperative outcomes in novel syngeneic esophageal carcinoma models
- Author
-
Laura K. Aguilar, Jarrod D. Predina, Anil K. Rustgi, H Nakagawa, Benjamin Laguna, Louis A. Aliperti, Zvi G. Fridlender, Aaron Blouin, Estuardo Aguilar-Cordova, Veena Kapoor, Sunil Singhal, Steven M. Albelda, and Brendan F. Judy
- Subjects
Cytotoxicity, Immunologic ,Cancer Research ,Esophageal Neoplasms ,viruses ,medicine.medical_treatment ,surgery ,Mice ,0302 clinical medicine ,Simplexvirus ,Medicine ,Cytotoxic T cell ,Cyclin D1 ,Neoadjuvant therapy ,0303 health sciences ,Esophageal cancer ,animal models ,3. Good health ,Treatment Outcome ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,Original Article ,immunotherapy ,medicine.drug ,Ganciclovir ,Cell Survival ,Genetic Vectors ,Thymidine Kinase ,esophageal carcinoma ,03 medical and health sciences ,Cell Line, Tumor ,Carcinoma ,Animals ,Humans ,neoadjuvant therapy ,Molecular Biology ,030304 developmental biology ,Cisplatin ,Chemotherapy ,business.industry ,Genetic Therapy ,Neoplasms, Experimental ,Immunotherapy ,medicine.disease ,Mice, Inbred C57BL ,Genes, ras ,Immunology ,Cancer research ,Neoplasm Recurrence, Local ,Tumor Suppressor Protein p53 ,business - Abstract
Esophageal carcinoma is the most rapidly increasing tumor in the United States and has a dismal 15% 5-year survival. Immunotherapy has been proposed to improve patient outcomes; however, no immunocompetent esophageal carcinoma model exists to date to test this approach. We developed two mouse models of esophageal cancer by inoculating immunocompetent mice with syngeneic esophageal cell lines transformed by cyclin-D1 or mutant HRAS(G12V) and loss of p53. Similar to humans, surgery and adjuvant chemotherapy (cisplatin and 5-fluorouracil) demonstrated limited efficacy. Gene-mediated cyototoxic immunotherapy (adenoviral vector carrying the herpes simplex virus thymidine kinase gene in combination with the prodrug ganciclovir; AdV-tk/GCV) demonstrated high levels of in vitro transduction and efficacy. Using in vivo syngeneic esophageal carcinoma models, combining surgery, chemotherapy and AdV-tk/GCV improved survival (P=0.007) and decreased disease recurrence (P0.001). Mechanistic studies suggested that AdV-tk/GCV mediated a direct cytotoxic effect and an increased intra-tumoral trafficking of CD8 T cells (8.15% vs 14.89%, P=0.02). These data provide the first preclinical evidence that augmenting standard of care with immunotherapy may improve outcomes in the management of esophageal carcinoma.
- Published
- 2011
- Full Text
- View/download PDF
22. Phase I–II trial evaluating combined intensity-modulated radiotherapy and in situ gene therapy with or without hormonal therapy in treatment of prostate cancer—interim report on PSA response and biopsy data
- Author
-
J. Kam Chiu, Laura K. Aguilar, Timothy C. Thompson, Brian J. Miles, E. Brian Butler, Wei Yuan Mai, Shiao Y. Woo, James Caillouet, Terry L. Timme, Hsin H. Lu, Dov Kadmon, Gustavo Ayala, L. Steven Carpenter, Maria T. Vlachaki, Maria Davis, Estuardo Aguilar-Cordova, Thomas M. Wheeler, and Bin S. Teh
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Antineoplastic Agents, Hormonal ,Biopsy ,medicine.medical_treatment ,Genetic Vectors ,Urology ,Acyclovir ,Antiviral Agents ,Thymidine Kinase ,Adenoviridae ,Management of prostate cancer ,Prostate cancer ,Prostate ,PSA Failure ,medicine ,Carcinoma ,Humans ,Prodrugs ,Radiology, Nuclear Medicine and imaging ,Aged ,Aged, 80 and over ,Radiation ,medicine.diagnostic_test ,business.industry ,Prostatic Neoplasms ,Androgen Antagonists ,Radiotherapy Dosage ,Valine ,Genetic Therapy ,Middle Aged ,Prostate-Specific Antigen ,medicine.disease ,Combined Modality Therapy ,Flutamide ,Surgery ,Radiation therapy ,medicine.anatomical_structure ,Oncology ,Valacyclovir ,Hormonal therapy ,Leuprolide ,Radiotherapy, Conformal ,business ,Follow-Up Studies - Abstract
There is an evolving role for combining radiotherapy (RT) with gene therapy in the management of prostate cancer. However, the clinical results of this combined approach are much needed. The preliminary results addressing the safety of this Phase I-II study combining RT and gene therapy (adenovirus/herpes simplex virus-thymidine kinase gene/valacyclovir with or without hormonal therapy) in the treatment of prostate cancer have been previously reported. We now report the prostate-specific antigen (PSA) response and biopsy data.This trial was composed of three separate arms. Arm A consisted of low-risk patients (Stage T1-T2a, Gleason score7, pretreatment PSA10 ng/mL) treated with combined RT-gene therapy. A mean dose of 76 Gy was delivered to the prostate with intensity-modulated RT. They also received adenovirus/herpes simplex virus-thymidine kinase/valacyclovir gene therapy. Arm B consisted of high-risk patients (Stage T2b-T3, Gleason score6, pretreatment PSA level10 ng/mL) treated with combined RT-gene therapy and hormonal therapy (luteinizing hormone-releasing hormone agonist [30-mg Lupron, 4-month depot] and an antiandrogen [flutamide, 250 mg t.i.d. for 14 days]). Arm C consisted of patients with Stage D1 (positive pelvic lymph nodes) who received the same regimen as Arm B with the addition of 45 Gy to the pelvic lymphatics. PSA determination and biopsy were performed before, during, and after treatment. The American Society for Therapeutic Radiology and Oncology consensus definition (three consecutive rises in PSA level) was used to denote PSA failure.Fifty-nine patients (29 in Arm A, 26 in Arm B, and 4 in Arm C) completed the trial. The median age was 68 years (range, 39-85 years). The median follow-up for the entire group was 13.5 months (range, 1.4-27.8 months). Only Arm A patients were observed to have an increase in PSA on Day 14. The PSA then declined appropriately. All patients in Arm A (median follow-up, 13.4 months) and Arm B (median follow-up, 13.9 months) had biochemical control at last follow-up. Three patients in Arm C (with pretreatment PSA of 335, 19.6, and 2.5 ng/mL and a combined Gleason score of 8, 9, and 9 involving all biopsy cores) had biochemical failure at 3, 3, and 7.7 months. Two patients had distant failure in bone and 1 patient in the para-aortic lymph nodes outside the RT portal. Six to twelve prostate biopsies performed in these 3 patients revealed no evidence of residual carcinoma. In Arm A, biopsy showed no evidence of carcinoma in 66.7% (18 of 27), 92.3% (24 of 26), 91.7% (11 of 12), 100% (8 of 8), and 100% (6 of 6) at 6 weeks, 4 months, 12 months, 18 months, and 24 months after treatment, respectively. In Arm B, no evidence of carcinoma on biopsy was noted in 96% (24 of 25), 90.5% (19 of 21), 100% (14 of 14), 100% (7 of 7), and 100% (2 of 2), respectively, in the same interval after treatment.This is the first reported trial of its kind in the field of prostate cancer that aims to expand the therapeutic index of RT by combining it with in situ gene therapy. The initial transient PSA rise in the Arm A patients may have been a result of local immunologic response or inflammation elicited by in situ gene therapy. Additional investigation to elucidate the mechanisms is needed. Hormonal therapy may have obliterated this rise in Arm B and C patients. The biopsy data were encouraging and appeared to show no evidence of malignancy earlier than historical data. Combined RT, short-course hormonal therapy, and in situ therapy appeared to provide good locoregional control but inadequate systemic control in patients with positive pelvic lymph nodes. Longer term use of hormonal therapy in addition to gene therapy and RT has been adopted for this group of patients to maximize both locoregional and systemic control.
- Published
- 2004
- Full Text
- View/download PDF
23. Evolution of a Gene Therapy Clinical Trial
- Author
-
Laura K. Aguilar and Estuardo Aguilar-Cordova
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,Process (engineering) ,Genetic enhancement ,Therapy Trial ,Bench to bedside ,Viral vector ,Surgery ,Clinical trial ,Neurology ,Oncology ,medicine ,Neurology (clinical) ,Intensive care medicine ,business ,Gene ,Toxicity profile - Abstract
Developing and conducting gene therapy clinical trials poses unique challenges which must be addressed to satisfy regulatory requirements and, most importantly, to protect human subjects. Experimental products used for gene transfer studies, such as viral vectors, are often complex and cannot be sterilized or completely characterized to the extent of a typical pharmaceutical. Thus, quality and characterization must be built into the production process. Extensive preclinical studies must be performed to determine the feasibility of the approach, the safety of the product, and the appropriate dose range to evaluate in humans. Once a clinical trial is initiated, subjects must be followed carefully for short- and long-term toxicity especially since preclinical studies may not adequately predict the toxicity profile of these novel, complicated products. Results of early phase studies in gene therapy have often sent the investigators back to the laboratory to improve the delivery vector or identify a more potent or less toxic gene. This circular developmental process is expected for the early stages of a new technology such as gene therapy. Although these hurdles appear extensive, they can be overcome, as evidenced by the initiation of more than 500 clinical gene therapy trials in the United States to date, and are imperative for the maintenance of high-quality studies and public trust. This article describes the step-by-step process for developing a gene therapy trial incorporating specific examples relevant to neuro-oncology.
- Published
- 2003
- Full Text
- View/download PDF
24. Cirrhotic rat livers with extensive fibrosis can be safely transduced with clinical-grade adenoviral vectors. Evidence of cirrhosis reversion
- Author
-
Alejandra Miranda, Estuardo Aguilar-Cordova, Juan Armendáriz-Borunda, Jesús García-Bañuelos, Laura K. Aguilar, and Fernando Siller-López
- Subjects
Alcoholic liver disease ,Pathology ,medicine.medical_specialty ,DNA, Complementary ,Cirrhosis ,Genetic enhancement ,Biliary cirrhosis ,Genetic Vectors ,Liver Cirrhosis, Experimental ,MMP8 ,Adenoviridae ,Viral vector ,Transduction, Genetic ,Fibrosis ,Genetics ,Animals ,Medicine ,Tissue Distribution ,Rats, Wistar ,Molecular Biology ,business.industry ,Neutrophil collagenase ,Genetic Therapy ,medicine.disease ,Rats ,Matrix Metalloproteinase 8 ,Treatment Outcome ,Molecular Medicine ,business - Abstract
Adenoviral vectors efficiently target normal liver cells; however, a clear-cut description of the safety boundaries for using adenovectors in hepatic cirrhosis has not been settled. With this in mind, we used a first-generation, replication-deficient adenoviral vector carrying the E. coli lacZ gene (Ad5betaGal) to monitor therapeutic range, biodistribution, toxicity and transduction efficiency in Wistar rats made cirrhotic by two different experimental approaches resembling alcoholic cirrhosis and biliary cirrhosis in humans. Further, we show proof of concept on fibrosis reversion by a 'therapeutic' Ad-vector (AdMMP8) carrying a gene coding for a collagen-degrading enzyme. Dose-response experiments with Ad5betaGal ranging from 1 x 10(8)-3 x 10(12) viral particles (vp) per rat (250 g), demonstrated that adenovirus-mediated gene transfer via iliac vein at 3 x 10(11 )vp/rat, resulted in an approximately 40% transduction in livers of rats made cirrhotic by chronic intoxication with carbon tetrachloride, compared with approximately 80% in control non-cirrhotic livers. In rats made cirrhotic by bile-duct obstruction only, 10% efficiency of transduction was observed. Biodistribution analyses showed that vector expression was detected primarily in liver and at a low level in spleen and kidney. Although there was an important increase in liver enzymes between the first 48 h after adenovirus injection in cirrhotic animals compared to non-transduced cirrhotic rats, this hepatic damage was resolved after 72-96 h. Then, the cDNA for neutrophil collagenase, also known as Matrix Metalloproteinase 8 (MMP8), was cloned in an Ad-vector and delivered to cirrhotic rat livers being able to reverse fibrosis in 44%. This study demonstrates the potential use of adenoviral vectors in safe transient gene therapy strategies for human liver cirrhosis.
- Published
- 2002
- Full Text
- View/download PDF
25. Population based PSA screening in Mexico City
- Author
-
Eduardo Gonzalez-Cuenca, Ricardo Castillejos-Molina, Guillermo Feria Bernal, Jaime O. Herrera-Caceres, Andrea G. Manzanera, Estuardo Aguilar-Cordova, and Laura K. Aguilar
- Subjects
Gerontology ,Cancer Research ,Pediatrics ,medicine.medical_specialty ,Psa screening ,business.industry ,Incidence (epidemiology) ,Early detection ,Cancer ,Population based ,urologic and male genital diseases ,medicine.disease ,Standardized mortality ratio ,medicine.anatomical_structure ,Oncology ,Prostate ,Mexico city ,medicine ,business - Abstract
92 Background: Since the U.S. Preventive Task Force recommended against PSA screening in 2012, the polemic between the benefit of early detection and the risk of early overtreatment, has been heightened. Furthermore, in countries where diagnosis is delayed even in symptomatic men and resources are limited for treating advanced disease, the impact of limiting PSA screening may be greater resulting in a shorter incidence/mortality ratio. In Mexico PSA screening is not widely used and many men are never diagnosed with PCa. Thus, the true incidence of PCa is unknown and yet it is the number one cause of cancer morbidity and mortality in men. The current study was designed to evaluate the incidence of PCa in a random population of men from Mexico City using PSA screening. Methods: Prospective enrollment of 3,837 men > 50 years old without previous history of PCa underwent PSA testing. Men with a PSA > 4ng/ml were selected for a second analysis and for completion of a questionnaire on prostate health history. Men with confirmed elevated PSA are being invited to further evaluation and possible biopsy by a urologist. Results: Median age was 59 years (interquartile range 56 to 63 years). In this cohort, 412 (10.8%) men had a total PSA > 4ng/ml (5% of those < 55 years vs. 26% of those > 70 years). From these men, 284 had a repeat PSA approximately 3 months after the first evaluation (median 98 days). A total of 229 men had a 2nd PSA above 4mg/ml, 25 had PSA > 20ng/ml. During evaluations, 309 men completed the questionnaire showing that 29% had previous PSA testing, 11% had a previous DRE, 4% had history of a previous biopsy and 2% identified having a family history of PCa. Conclusions: Greater than 10% (n = 412/3837) of men from a random screen in Mexico City were found to have elevated PSA ( > 4ng/ml). Of these, 12% (n = 48) and 6% (n = 25) had levels consistent with intermediate- and high-risk disease, respectively. Most of these ”high-PSA” patients did not have a previous PSA evaluation despite symptoms, representing a considerable number of patients in whom screening could be life-saving intervention. A low awareness of family history of PCa could be representative of both the lack of diagnosis of PCa and general ignorance regarding this disease. We continue evaluating and doing biopsies to patients with indication.
- Published
- 2017
- Full Text
- View/download PDF
26. Enhanced therapeutic effect of mutiple injections of HSV-TK + GCV gene therapy in combination with ionizing radiation in a mouse mammary tumor model
- Author
-
Bin S. Teh, Shiao Y. Woo, Laura K. Aguilar, Maria T. Vlachaki, Timothy C. Thompson, Kam J. Chiu, Xiaohong Zhu, Estuardo Aguilar-Cordova, Madhu Chhikara, and E. Brian Butler
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,Radiobiology ,Genetic enhancement ,medicine.medical_treatment ,Genetic Vectors ,Antiviral Agents ,Thymidine Kinase ,Adenoviridae ,Mice ,Breast cancer ,Tumor Cells, Cultured ,medicine ,Animals ,Combined Modality Therapy ,Radiology, Nuclear Medicine and imaging ,Ganciclovir ,Mice, Inbred BALB C ,Mammary tumor ,Radiation ,business.industry ,Therapeutic effect ,Mammary Neoplasms, Experimental ,Cancer ,Herpes Simplex ,Radiotherapy Dosage ,Genetic Therapy ,medicine.disease ,Survival Analysis ,Radiation therapy ,Oncology ,Cancer research ,business - Abstract
Purpose: Standard therapies for breast cancer lack tumor specificity and have significant risk for recurrence and toxicities. Herpes simplex virus-thymidine kinase (HSV-tk) gene therapy combined with radiation therapy (XRT) may be effective because of complementary mechanisms and distinct toxicity profiles. HSV-tk gene therapy followed by systemic administration of ganciclovir (GCV) enhances radiation-induced DNA damage by generating high local concentrations of phosphorylated nucleotide analogs that increase radiation-induced DNA breaks and interfere with DNA repair mechanisms. In addition, radiation-induced membrane damage enhances the “bystander effect” by facilitating transfer of nucleotide analogs to neighboring nontransduced cells and by promoting local and systemic immune responses. This study assesses the effect of single and multiple courses of HSV-tk gene therapy in combination with ionizing radiation in a mouse mammary cancer model. Methods and Materials: Mouse mammary TM40D tumors transplanted s.c. in syngeneic immunocompetent BALB-c mice were treated with either adenoviral-mediated HSV-tk gene therapy or local radiation or the combination of gene and radiation therapy. A vector consisting of a replication-deficient (E1-deleted) adenovirus type 5 was injected intratumorally to administer the HSV-tk gene, and GCV was initiated 24 h later for a total of 6 days. Radiation was given as a single dose of 5 Gy 48 h after the HSV-tk injection. A metastatic model was developed by tail vein injection of TM40D cells on the same day that the s.c. tumors were established. Systemic antitumor effect was evaluated by counting the number of lung nodules after treating only the primary tumors with gene therapy, radiation, or the combination of gene and radiation therapy. To assess the therapeutic efficacy of multiple courses of this combinatorial approach, one, two, and three courses of HSV-tk + GCV gene therapy, in combination with radiation, were compared to HSV-tk or XRT alone and to sham-treated animals. (Treatments were repeated at 7-day intervals from the HSV-tk injection.) Results: Both single-therapy modalities reduced tumor growth by 11% compared to controls, while the combined therapy resulted in a decrease of 29%. Median survival was 36 days in the combined therapy group, compared to 33 days in the monotherapy groups and 26 days in the control group. In the metastatic model, the number of lung nodules was reduced by 59.5% after HSV-tk gene therapy, whereas radiotherapy had no effect on metastatic growth. Combined therapy led to an additional 66.7% reduction in lung colonization. Compared to controls, local tumor growth was maximally suppressed by three courses of combined therapy (51.5%), followed by two courses of combined therapy (37.2%), and three sessions of XRT alone (35.6%). Median survival was also significantly prolonged to 58 days with the three courses of combined therapy, followed by two courses, to 45 days. All other treatment groups demonstrated median survival times between 26 and 35 days, while controls had a median survival of 24 days. Conclusions: These results indicate that multiple courses of HSV-tk therapy in combination with radiation improve the therapeutic efficacy of this approach and may provide therapeutic implications for the treatment of human breast cancer and other solid tumors.
- Published
- 2001
- Full Text
- View/download PDF
27. Liver Cirrhosis Is Reverted by Urokinase-Type Plasminogen Activator Gene Therapy
- Author
-
Miriam Bueno, Silvia Salgado, Rogelio Hernández-Pando, Estuardo Aguilar-Cordova, Jose Vera, Alejandra Miranda, Laura K. Aguilar, Juan Armendáriz-Borunda, Guillermo Grijalva, Fernando Siller, Mary Fafutis, Jorge Segura, Jesus Garcia, Héctor Fabio Bermúdez Orozco, and Aida Segura
- Subjects
Cirrhosis ,Enzyme-Linked Immunosorbent Assay ,Biology ,Liver Cirrhosis, Experimental ,Fibrosis ,Drug Discovery ,Genetics ,medicine ,Animals ,Rats, Wistar ,Carbon Tetrachloride ,Molecular Biology ,DNA Primers ,Pharmacology ,Urokinase ,Base Sequence ,Genetic Therapy ,Hepatitis B ,medicine.disease ,Urokinase-Type Plasminogen Activator ,Liver Regeneration ,Rats ,medicine.anatomical_structure ,Hepatocyte ,Immunology ,Cancer research ,Molecular Medicine ,Hepatocyte growth factor ,Liver function ,Plasminogen activator ,medicine.drug - Abstract
Liver cirrhosis represents a worldwide health problem and is a major cause of mortality. Cirrhosis is the result of extensive hepatocyte death and fibrosis induced by chronic alcohol abuse and hepatitis B and C viruses. Successful gene therapy approaches to this disease may require both reversal of fibrosis and stimulation of hepatocyte growth. Urokinase-type plasminogen activator (uPA) may serve this function, as it is an initiator of the matrix proteolysis cascade and induces hepatocyte growth factor expression. In a rat cirrhosis model, a single iv administration of a replication-deficient adenoviral vector encoding a nonsecreted form of human uPA resulted in high production of functional uPA protein in the liver. This led to induction of collagenase expression and reversal of fibrosis with concomitant hepatocyte and improved liver function. Thus, uPA gene therapy may be an effective strategy for treating cirrhosis in humans.
- Published
- 2000
- Full Text
- View/download PDF
28. Reporting Adverse Events in Gene Therapy Studies
- Author
-
Bambi Grilley, Laura K. Aguilar, and Estuardo Aguilar-Cordova
- Subjects
Pharmacology ,medicine.medical_specialty ,business.industry ,Public concern ,General Medicine ,Unexpected adverse drug reaction ,medicine.disease ,Pharmacotherapy ,medicine ,Pharmacology (medical) ,Medical emergency ,medicine.symptom ,Intensive care medicine ,Adverse effect ,business ,Adverse drug reaction ,Biotechnology ,Confusion - Abstract
The reporting of adverse events occurring in gene therapy studies is currently under discussion. There are many agencies involved in the reporting of adverse events, often with reporting guidelines that are unique to each organisation. Guidelines differ according to the type of event (adverse event, adverse drug reaction, unexpected adverse drug reaction, serious adverse event and serious adverse drug reaction). While there is a move toward making guidelines more stringent in the US, there is concern that this will increase the number of reports of unrelated events, create confusion from a global perspective, and increase public concern needlessly.
- Published
- 2000
- Full Text
- View/download PDF
29. A Prescription for Gene Therapy
- Author
-
John Nemunaitis, Barrie J. Carter, Douglas J. Jolly, Estuardo Aguilar-Cordova, Dale Ando, Juan Armendáriz-Borunda, Steve Scherer, Joseph C. Glorioso, Michelle Rives, Parris R. Burd, Fintan R. Steele, Daniel R. Henderson, E. Brian Butler, Gerald P. McKenny, Yawen L. Chiang, Laura K. Aguilar, and Savio L. C. Woo
- Subjects
Pharmacology ,medicine.medical_specialty ,business.industry ,Group (mathematics) ,Genetic enhancement ,Internal medicine ,Drug Discovery ,Genetics ,medicine ,Molecular Medicine ,Medical prescription ,business ,Molecular Biology - Published
- 2000
- Full Text
- View/download PDF
30. Administration of Neomycin Resistance Gene Marked EBV Specific Cytotoxic T-Lymphocytes to Patients with Relapsed EBV-Positive Hodgkin Disease. Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Author
-
Bambi Grilley, Malcolm K. Brenner, Robert A. Krance, Laura K. Aguilar, Helen E. Heslop, Cliona M. Rooney, Mary V. Gresik, Elizabeth Rob, George Carrum, Kenneth L. McClain, and Marie A. Roskrow
- Subjects
business.industry ,Genetic enhancement ,Cell ,Drug resistance ,Disease ,EBV-Specific Cytotoxic T-Lymphocyte ,medicine.anatomical_structure ,Cell culture ,Immunology ,Genetics ,medicine ,Molecular Medicine ,Cytotoxic T cell ,business ,Molecular Biology ,Gene - Published
- 1998
- Full Text
- View/download PDF
31. Intraprostatic distribution and long-term follow-up after AdV-tk immunotherapy as neoadjuvant to surgery in patients with prostate cancer
- Author
-
Hugo A. Barrera-Saldaña, Laura K. Aguilar, Lauro S. Gómez-Guerra, Augusto Rojas-Martinez, Juan Pablo Flores-Gutiérrez, Ivan Delgado-Enciso, Rocio Ortiz-Lopez, J. Esteban-María, S. W. Sukin, Estuardo Aguilar-Cordova, G Elizondo Riojas, Andrea G. Manzanera, Edward Brian Butler, Juan Francisco González-Guerrero, and Raquel Garza-Guajardo
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Genetic Vectors ,androgen deprivation therapy ,Thymidine Kinase ,Article ,Adenoviridae ,Androgen deprivation therapy ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Prostate ,medicine ,Humans ,Simplexvirus ,Molecular Biology ,Neoadjuvant therapy ,030304 developmental biology ,Aged ,Oncolytic Virotherapy ,0303 health sciences ,Prostatectomy ,business.industry ,Gene Transfer Techniques ,Prostatic Neoplasms ,Genetic Therapy ,Middle Aged ,Prostate-Specific Antigen ,medicine.disease ,Neoadjuvant Therapy ,3. Good health ,Surgery ,Gene-Mediated Cytotoxic Immunotherapy ,Prostate-specific antigen ,medicine.anatomical_structure ,Tumor progression ,030220 oncology & carcinogenesis ,intra-tumor vector distribution ,Molecular Medicine ,Adenocarcinoma ,Kallikreins ,Immunotherapy ,business ,long-term safety ,Follow-Up Studies - Abstract
A phase I-II study to evaluate gene-mediated cytotoxic immunotherapy in newly diagnosed prostate cancer before radical prostatectomy was conducted in Monterrey, Mexico. First, to investigate delivery of adenovirus to the prostate, fluorescently labeled vector was injected into fresh prostatectomy specimens and distribution was visually analyzed. The optimal volume and site instillation was then used for transrectal ultrasound guided intraprostatic injection in 10 patients with adenocarcinoma scheduled for radical prostatectomy. Each received two apical and two basal 0.5 ml injections of AdV-tk for a total of 1 × 10(11) vp followed by 14 days of prodrug. Nine patients continued to tumor resection: six high risk, one intermediate and two low risk. In vivo vector distribution was analyzed from the resected tissue of four patients. Patients were monitored for tumor progression and acute and long-term safety. For vector delivery, two apical and two basal injections of 0.5 ml led to optimal organ-wide distribution ex vivo and in vivo. Cytotoxicity was evidenced by transient rise in PSA and tumor histology. There were no significant adverse events deemed related to the treatment and no late toxicities after median follow-up of 11.3 years. All six high-risk patients had positive surgical margins and one had seminal vesicle involvement. Despite slow PSA rise post surgery in three of these patients, none developed metastases. The intermediate- and low-risk patients had complete resections and none have progressed. In conclusion, in vivo transrectal ultrasound guided instillation of an adenoviral vector into four sites in the prostate was practical as an outpatient procedure, well tolerated and led to distribution throughout the intraprostatic tumor mass. AdV-tk demonstrated no significant acute or late toxicities. Trends in PSA and disease progression conveyed the possibility of a sustained immune response against residual disease.
- Published
- 2013
32. Therapeutic Endoscopic Ultrasonography: Intratumoral Injection for Pancreatic Adenocarcinoma
- Author
-
Laura K. Aguilar, Estuardo Aguilar-Cordova, Jon P. Walker, Lawrence A. Shirley, and Mark Bloomston
- Subjects
Oncology ,Endoscopic ultrasound ,medicine.medical_specialty ,Hepatology ,medicine.diagnostic_test ,Methodology Report ,business.industry ,Gastroenterology ,Cancer ,Aggressive disease ,Endoscopic ultrasonography ,Bioinformatics ,medicine.disease ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,Medicine ,Adenocarcinoma ,030211 gastroenterology & hepatology ,lcsh:Diseases of the digestive system. Gastroenterology ,lcsh:RC799-869 ,business - Abstract
Pancreatic adenocarcinoma is an aggressive disease that has poor outcomes despite maximal traditional therapies. Thus, treatment of this cancer demands innovative strategies to be used in addition to standing therapies in order to provide new avenues of care. Here, we describe the technique of using endoscopic ultrasound in order to directly inject both novel and conventional therapies into pancreatic tumors. We detail the rationale behind this strategy and the many benefits it provides. We then describe our technique in detail, including our experience injecting the AdV-tk adenoviral vector to create an in situ vaccine effect.
- Published
- 2013
33. Abstract CT110: Phase II multicenter study of gene-mediated cytotoxic immunotherapy as an adjuvant to surgical resection for newly diagnosed malignant glioma: lessons learned from pseudo-progression and a nine-year survivor
- Author
-
David S. Baskin, Estuardo Aguilar-Cordova, John M. McGregor, E. Antonio Chiocca, Laura K. Aguilar, Pamela Z. New, Todd Trask, Behnam Badie, Susan Bell, Bin S. Teh, Simon S. Lo, John C. Grecula, Lee A. Wheeler, Jana Portnow, Herbert B. Newton, Robert Cavaliere, Andrea G. Manzanera, and Edward Brian Butler
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,education.field_of_study ,business.industry ,medicine.medical_treatment ,Population ,Cancer ,Immunotherapy ,medicine.disease ,Minimal residual disease ,Surgery ,Radiation therapy ,Glioma ,Internal medicine ,Multicenter trial ,medicine ,education ,business ,Pseudoprogression - Abstract
Gene Mediated Cytotoxic Immunotherapy (GMCI) generates a polyvalent anti-tumor immune response via local delivery of aglatimagene besadenovec (AdV-tk) plus prodrug. The resulting cytotoxic nucleotide analog is toxic to cells as it halts DNA replication and repair. Tumor cell death releases tumor associated antigens, which are taken up by antigen presenting cells. There is a simultaneous stimulation of pro-inflammatory cytokines by thymidine kinase. The treatment mounts an immune response that is ongoing for many months to years. Extensive preclinical studies were performed, followed by a Phase II multicenter trial assess safety and overall survival (OS) after GMCI+SOC compared to a concurrent matched control group meeting protocol criteria and undergoing SOC. Ten 100 microliter injections of AdV-tk were made into the tumor resection bed followed by oral valacyclovir for 14 days. Results: Preclinical studies have documented that response to treatment is dependent on upregulation of antigen presenting cells and an immuno-modulating effect from interleukins, including IL-2 and IL-12. The response is greatly enhanced by the synergistic effect of radiation therapy initiated on day 3 after injection of therapy. CD4 helper cells did not appear to play an important role. Survival was greatly enhanced in those in whom an immune response can be documented either radiographically or histologically. 48 patients completed SOC+GMCI and 134 SOC in the matched cohort. There were no dose-limiting toxicities. Median OS increased by 3.6 months, from 13.5 for SOC to 17.1 months for GMCI+SOC (p = 0.0417). Survival at 1- 2- and 3-years increased from 57%, 22%, and 8% to 67%, 35%, and 19%, respectively. Improvement was mostly in patients who underwent gross total resection: median OS increased from 16.9 to 25 months (p = 0.0492); 1- 2- and 3-year survival rates from 64%, 28% and 6% to 90%, 53% and 32%. A number of patients had marked pseudoprogression, well beyond the RANO or iRANO criteria. The most robust response was in a nine year survivor who demonstrated a doubling of apparent enhancing tumor at three months after treatment. Enhancing tumor slowly resolved over a five year period, with complete resolution only seen in an MRI scan made nine years later. Many other patients had similar responses, with effects of treatment ongoing for many years, and an initial response falsely suggesting treatment failure. Conclusions: GMCI can be safely combined with SOC in newly diagnosed malignant gliomas. Survival rates compared favorably to historical reports and a matched control group. Survival outcomes were significantly improved in patients with minimal residual disease after total resection. The most robust responders had dramatic worsening of contrast enhancing lesions at three and six months after intervention, well beyond standard criteria utilized to distinguish between true disease and pseudoprogression. The results document efficacy of treatment, and illustrate the need for new tools for evaluation of pseudoprogression in CNS immunotherapy trials. Characteristics and factors predicting a robust response to treatment will be discussed, as well as important clinical findings in the responder population. Citation Format: David S. Baskin, Laura K. Aguilar, Lee A. Wheeler, Andrea G. Manzanera, Susan D. Bell, Robert Cavaliere, John M. McGregor, Simon S. Lo, John C. Grecula, Herbert B. Newton, Behnam Badie, Bin S. Teh, Edward Brian Butler, Todd W. Trask, Jana Portnow, Pamela Z. New, Estuardo Aguilar-Cordova, E. Antonio Chiocca. Phase II multicenter study of gene-mediated cytotoxic immunotherapy as an adjuvant to surgical resection for newly diagnosed malignant glioma: lessons learned from pseudo-progression and a nine-year survivor. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr CT110.
- Published
- 2016
- Full Text
- View/download PDF
34. Phase I study of gene mediated cytotoxic immunotherapy (GMCI) for patients with malignant pleural effusion (MPE)
- Author
-
Andrew R. Haas, Joshua Bauml, Andrea G. Manzanera, Laura K. Aguilar, Corey J. Langer, Daniel H. Sterman, Evan W. Alley, Charu Aggarwal, Tracey L. Evans, Susan Metzger, Roger B. Cohen, Estuardo Aguilar-Cordova, and Steven M. Albelda
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,Thymidine Kinase Gene ,animal diseases ,viruses ,medicine.medical_treatment ,Immunotherapy ,medicine.disease ,Phase i study ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Aglatimagene Besadenovec ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,medicine ,Cytotoxic T cell ,Malignant pleural effusion ,Vector (molecular biology) ,business ,Gene - Abstract
3081Background: GMCI is a tumor-specific immune-stimulator through local delivery of aglatimagene besadenovec, an adenovirus-based vector expressing the HSV-1 thymidine kinase gene (AdV-tk) followe...
- Published
- 2016
- Full Text
- View/download PDF
35. Phase II multicenter study of gene mediated cytotoxic immunotherapy as adjuvant to surgical resection for newly diagnosed malignant glioma
- Author
-
Behnam Badie, Todd Trask, Laura K. Aguilar, Simon S. Lo, Pamela Z. New, David S. Baskin, Estuardo Aguilar-Cordova, Herbert B. Newton, Robert Cavaliere, Andrea G. Manzanera, E. Antonio Chiocca, Susan Bell, John M. McGregor, Lee A. Wheeler, John C. Grecula, and Jana Portnow
- Subjects
Surgical resection ,Oncology ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,business.industry ,medicine.medical_treatment ,Newly diagnosed ,Immunotherapy ,medicine.disease ,Multicenter study ,Glioma ,Internal medicine ,medicine ,Cytotoxic T cell ,business ,Adjuvant ,medicine.drug - Abstract
2010 Background: New therapies are desperately needed for malignant gliomas since aggressive standard of care (SOC) treatment with surgery, radiation, and temozolomide leads to median survival of l...
- Published
- 2015
- Full Text
- View/download PDF
36. Cytotoxic T lymphocyte therapy for Epstein-Barr virus+ Hodgkin's disease
- Author
-
Dagmar Dilloo, Cliona M. Rooney, Karin Straathof, George Carrum, Malcolm K. Brenner, Alexandra Rousseau, Melissa M. Hudson, Benedikt Gahn, M. Helen Huls, Catherine M. Bollard, Helen E. Heslop, Adrian P. Gee, M. Victoria Gresik, John W. Sixbey, and Laura K. Aguilar
- Subjects
Adult ,Male ,Herpesvirus 4, Human ,Adolescent ,medicine.medical_treatment ,Immunology ,lymphoma ,Biology ,medicine.disease_cause ,Immunotherapy, Adoptive ,Virus ,Article ,Viral Matrix Proteins ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Cell Movement ,hemic and lymphatic diseases ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Humans ,Epstein-Barr virus ,LMP2 ,Child ,030304 developmental biology ,0303 health sciences ,Remission Induction ,Immunotherapy ,Herpesviridae Infections ,Viral Load ,medicine.disease ,Prognosis ,Epstein–Barr virus ,Hodgkin Disease ,3. Good health ,Lymphoma ,CTL ,B symptoms ,gene marking ,030220 oncology & carcinogenesis ,Female ,Tumor Escape ,immunotherapy ,medicine.symptom ,T-Lymphocytes, Cytotoxic - Abstract
Epstein Barr virus (EBV)+ Hodgkin's disease (HD) expresses clearly identified tumor antigens derived from the virus and could, in principle, be a target for adoptive immunotherapy with viral antigen–specific T cells. However, like most tumor-associated antigens in immunocompetent hosts, these potential targets are only weakly immunogenic, consisting primarily of the latent membrane protein (LMP)1 and LMP2 antigens. Moreover, Hodgkin tumors possess a range of tumor evasion strategies. Therefore, the likely value of immunotherapy with EBV-specific cytotoxic effector cells has been questioned. We have now used a combination of gene marking, tetramer, and functional analyses to track the fate and assess the activity of EBV cytotoxic T lymphocyte (CTL) lines administered to 14 patients treated for relapsed EBV+ HD. Gene marking studies showed that infused effector cells could further expand by several logs in vivo, contribute to the memory pool (persisting up to 12 mo), and traffic to tumor sites. Tetramer and functional analyses showed that T cells reactive with the tumor-associated antigen LMP2 were present in the infused lines, expanded in peripheral blood after infusion, and also entered tumor. Viral load decreased, demonstrating the biologic activity of the infused CTLs. Clinically, EBV CTLs were well tolerated, could control type B symptoms (fever, night sweats, and weight loss), and had antitumor activity. After CTL infusion, five patients were in complete remission at up to 40 mo, two of whom had clearly measurable tumor at the time of treatment. One additional patient had a partial response, and five had stable disease. The performance and fate of these human tumor antigen–specific T cells in vivo suggests that they might be of value for the treatment of EBV+ Hodgkin lymphoma.
- Published
- 2004
37. The combination of adenoviral HSV TK gene therapy and radiation is effective in athymic mouse glioblastoma xenografts without increasing toxic side effects
- Author
-
Hiroaki Wakimoto, Fred H. Hochberg, Arnab Chakravarti, Fernando Siller-López, Anat Stemmer-Rachamimov, Ulf Nestler, Estuardo Aguilar-Cordova, Alona Muzikansky, Laura K. Aguilar, and E. Antonio Chiocca
- Subjects
Ganciclovir ,Cancer Research ,Pathology ,medicine.medical_specialty ,Ratón ,Genetic enhancement ,Transplantation, Heterologous ,Athymic mouse ,Mice, Nude ,Antiviral Agents ,Thymidine Kinase ,Mice ,Breast cancer ,HSV-Tk Gene ,Prostate ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Simplexvirus ,Radiotherapy ,business.industry ,Therapeutic effect ,Genetic Therapy ,medicine.disease ,Combined Modality Therapy ,Immunohistochemistry ,medicine.anatomical_structure ,Neurology ,Oncology ,Cancer research ,Female ,Neurology (clinical) ,business ,Glioblastoma ,medicine.drug - Abstract
In mouse models of prostate and breast cancer therapeutic effects are enhanced when adenoviral HSV TK gene therapy is combined with ionizing radiation. In the present study, we adopted this approach for the treatment of human glioblastoma xenografts in an athymic mouse model and assessed treatment results as well as toxic side effects.About 72 nude mice received intracerebral inoculations of 2 x 10(5) U87deltaEGFR cells. On day 7 after tumor implantation the study population was randomized into six treatment arms: (1) intratumoral buffer inoculation on day 7, (2) intratumoral adenoviral vector injection (2 x 10(9) vp) on day 7, (3) single dose radiation (2.1 Gy) on day 9, (4) adenoviral injection + radiation, (5) adenoviral injection + ganciclovir (GCV) (20 microg/g twice daily from day 8 to 17), (6) adenoviral injection + GCV + radiation. On day 21 half of the animals were sacrificed for histological evaluation of the brain tumors, the other half was assessed for survival.This study showed significantly prolonged median survival time of 5 days for the GCV treated groups. The addition of radiation decreased the frequency of neurological symptoms and delayed the onset of deficits without altering the expression of thymidine kinase in the tumor cells.We conclude that adenoviral HSV TK gene therapy in combination with adjuvant radiotherapy does not generate increased toxic side effects in glioblastoma treatment. The prolonged survival time of animals receiving gene therapy and the reduced occurrence of neurological symptoms in irradiated mice constitute promising features of the combined treatment.
- Published
- 2004
38. Combining radiotherapy with gene therapy (from the bench to the bedside): a novel treatment strategy for prostate cancer
- Author
-
Hsin H. Lu, Gustavo Ayala, Laura K. Aguilar, Estuardo Aguilar-Cordova, J. Kam Chiu, James Caillouet, Dov Kadmon, E. Brian Butler, Walter H. Grant, Timothy C. Thompson, Shiao Y. Woo, Brian J. Miles, Wei Yuan Mai, Maria Davis, L. Steven Carpenter, Thomas M. Wheeler, Bin S. Teh, and Maria T. Vlachaki
- Subjects
Oncology ,CD4-Positive T-Lymphocytes ,Male ,Cancer Research ,medicine.medical_specialty ,Pathology ,Lung Neoplasms ,Time Factors ,medicine.medical_treatment ,Genetic enhancement ,Antiviral Agents ,Adenoviridae ,Prostate cancer ,Mice ,Internal medicine ,medicine ,Tumor Cells, Cultured ,Combined Modality Therapy ,Animals ,Humans ,Simplexvirus ,Neoplasm Metastasis ,Ganciclovir ,business.industry ,Cancer ,Prostatic Neoplasms ,Transfection ,Genetic Therapy ,medicine.disease ,Radiation therapy ,Clinical trial ,Mice, Inbred C57BL ,Treatment Outcome ,Cancer cell ,business - Abstract
Combined radiotherapy and gene therapy is a novel therapeutic approach for prostate cancer. There are various potential benefits in combining ionizing radiation with gene therapy to achieve enhanced antitumor effects: A) ionizing radiation improves transfection/ transduction efficiency, transgene integration, and possibly, the “bystander effect” of gene therapy; B) gene therapy, on the other hand, may interfere with repair of radiation-induced DNA damage and increase DNA susceptibility to radiation damage in cancer cells, and C) radiotherapy and gene therapy target at different parts of the cell cycle. Preclinical data have demonstrated the enhanced antitumor effects of this combined approach in local tumor control, prolongation of survival, as well as systemic control. This combined radio-gene therapy is under study in an ongoing clinical trial in prostate cancer. Our study adds gene therapy to the standard of care therapy (radiotherapy). These treatment modalities have different toxicity profiles. The goal of this combined approach is to enhance cancer cure without an increase in treatment-related toxicity. This approach also offers a new paradigm in spatial cooperation, whereby two local therapies are combined to elicit both local and systemic effects. Early clinical results showed the safety of this approach.
- Published
- 2002
39. Impact of preimmunization on adenoviral vector expression and toxicity in a subcutaneous mouse cancer model
- Author
-
Maria T, Vlachaki, Andres, Hernandez-Garcia, Michael, Ittmann, Madhu, Chhikara, Laura K, Aguilar, Xiaohong, Zhu, Bin S, Teh, E Brain, Butler, Shiao, Woo, Timothy C, Thompson, Hugo, Barrera-Saldana, Estuardo, Aguilar-Cordova, and Bin S, The
- Subjects
Genetic enhancement ,Genetic Vectors ,Biology ,Viral vector ,Hepatitis ,Mice ,Immune system ,Immunity ,Genes, Reporter ,Drug Discovery ,Gene expression ,Genetics ,Animals ,Molecular Biology ,Gene ,Pharmacology ,Mammary tumor ,Vaccination ,Mammary Neoplasms, Experimental ,Viral Vaccines ,Genetic Therapy ,Dependovirus ,Liver ,Toxicity ,Immunology ,Cancer research ,Molecular Medicine - Abstract
Immune responses against adenoviral vectors may influence the toxicity and therapeutic effectiveness of adenovirus-mediated gene transfer and may be a limiting factor in adenovirus-mediated gene therapy. The purpose of this study was to determine the effects of preimmunization on intratumoral adenoviral transduction and systemic spread. The hypothesis was that increased doses of adenoviral vectors could overcome local neutralization without added systemic toxicity. The level and duration of gene expression were assessed as a function of time and dose after intratumoral delivery of adenoviral vector (AdV) encoding the luciferase reporter gene (AdV-luc) in a subcutaneous mouse mammary tumor model. Preimmunization resulted in significantly decreased gene expression in tumor and normal tissues (P0.01). The decrease was significantly greater in liver than in tumor. Increased AdV doses could be used to overcome the intratumoral inhibition without a concomitant increase in liver transduction. However, preimmunized animals showed greater toxicity than nai;ve animals (P0.001). The preimmunized group developed histologic evidence of grade 2-3 hepatic toxicity and increases in the average values of hepatic enzymes. In addition, there was a significant increase in mortality (P0.01) in the preimmunized group (12 of 20 animals) compared with the naive group (3 of 20 animals). These findings suggest that although preimmunity can inhibit systemic expression from adenoviral vectors, at high vector doses it may potentiate hepatotoxicity.
- Published
- 2002
40. CASE STUDY OF COMBINED GENE AND RADIATION THERAPY AS AN APPROACH IN THE TREATMENT OF CANCER
- Author
-
BIN S. TEH, MARIA T. VLACHAKI, LAURA K. AGUILAR, BRIAN MILES, GUSTAVO AYALA, DOV KADMON, THOMAS WHEELER, TIMOTHY C. THOMPSON, E. BRIAN BUTLER, and ESTUARDO AGUILAR-CORDOVA
- Subjects
Oncology ,medicine.medical_specialty ,Pathology ,business.industry ,medicine.medical_treatment ,Genetic enhancement ,Cancer ,Tumor control ,medicine.disease ,Preclinical data ,Combined approach ,Radiation therapy ,Prostate cancer ,Internal medicine ,medicine ,business ,Gene - Abstract
This chapter discusses the case study of combined gene and radiation therapy as an approach in the treatment of cancer. Combined gene therapy and radiation therapy is a novel approach in the armament against cancer. This radio/gene therapy combination may create a new spatial cooperation whereby two local treatment modalities, radiotherapy and in situ gene therapy, can lead to improved local and systemic control. Preclinical data have demonstrated the enhanced antitumor effects of this combined approach in local tumor control, prolongation of survival, and systemic control. This combined radio/gene therapy has now entered clinical phase in prostate cancer. The current approach adds a novel therapy to the standard-of-care therapy. These treatment modalities have very distinct spectra of toxicity profiles. This combined approach enhances the cancer cure without an increase in treatment-related toxicity. Early clinical results have shown the safety of this approach.
- Published
- 2002
- Full Text
- View/download PDF
41. Contributors
- Author
-
Rafat Abonour, Scott I. Abrams, Laura K. Aguilar, Estuardo Aguilar-Cordova, Steven M. Albelda, Mark R. Albertini, Gustavo Ayala, Jacques Banchereau, Christopher Baum, Christian M. Becker, Carmela Beger, Joseph R. Bertino, Patrick Blanco, Tulin Budak-Alpdogan, E. Brian Butler, Lisa H. Butterfield, Alfred E. Chang, Saswati Chatterjee, K.V. Chin, Virginia K. Clements, Mark J. Cooper, Kenneth Cornetta, James M. Croop, Samudra Dissanayake, Stephen L. Eck, James S. Economou, Laurence C. Eisenlohr, Wafik S. El-Deiry, Filip A. Farnebo, Andrew L. Feldman, Judah Folkman, Stanton L. Gerson, Mileka Gilbert, Leonard G. Gomella, David H. Gorski, William N. Hait, Mien-Chie Hung, Kevin D. Judy, Dov Kadmon, Thomas Kearney, Edsel U. Kim, David M. King, David Kirn, Omer N. Koç, Martin Krüger, Calvin J. Kuo, C. Lampert, Edmund C. Lattime, Irina V. Lebedeva, Steven K. Libutti, H. Kim Lyerly, Michael J. Mastrangelo, Helena J. Mauceri, A.M. McCall, Kevin T. McDonagh, R. Scott McIvor, Raymond D. Meng, Brian Miles, Frederick L. Moolten, Michael A. Morse, Paula J. Mroz, James J. Mulé, Smita K. Nair, Owen A. O'Connor, Wolfram Ostertag, Suzanne Ostrand-Rosenberg, A. Karolina Palucka, Beth A. Pulaski, Ling Qi, Alexander L. Rakhmilevich, Jane S. Reese, Michael Reiss, Antoni Ribas, Isabelle Rivière, Justin C. Roth, Michel Sadelain, Ruping Shao, C.A. Stein, Daniel H. Sterman, Carol Stocking, Bin S. Teh, Timothy C. Thompson, Deborah Toppmeyer, Catherine M. Verfaillie, Maria T. Vlachaki, Dorothee von Laer, Ralph R. Weichselbaum, L.M. Weiner, Thomas Wheeler, Lee G. Wilke, K.K. Wong, Flossie Wong-Staal, Duen-Hwa Yan, Steven P. Zielske, and Robert CH Zhao
- Published
- 2002
- Full Text
- View/download PDF
42. Enhanced therapeutic effect of HSV-tk+GCV gene therapy and ionizing radiation for prostate cancer
- Author
-
Shiao Y. Woo, Madhu Chhikara, Laura K. Aguilar, Estuardo Aguilar-Cordova, Kerby C. Oberg, Barry M. Berner, Xiaohong Zhu, Hanxian Huang, Maria T. Vlachaki, E. O'Brian Smith, Bin Teh, Kam J. Chiu, E. Brian Butler, and Timothy C. Thompson
- Subjects
Oncology ,Ganciclovir ,CD4-Positive T-Lymphocytes ,Male ,medicine.medical_specialty ,Lung Neoplasms ,Time Factors ,Combination therapy ,medicine.medical_treatment ,Genetic enhancement ,Antiviral Agents ,Adenoviridae ,03 medical and health sciences ,Prostate cancer ,Mice ,0302 clinical medicine ,Internal medicine ,Drug Discovery ,medicine ,Genetics ,Tumor Cells, Cultured ,Combined Modality Therapy ,Animals ,Simplexvirus ,Neoplasm Metastasis ,Molecular Biology ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Prostatectomy ,business.industry ,Therapeutic effect ,Prostatic Neoplasms ,Genetic Therapy ,medicine.disease ,3. Good health ,Radiation therapy ,Mice, Inbred C57BL ,Treatment Outcome ,030220 oncology & carcinogenesis ,Immunology ,Molecular Medicine ,business ,medicine.drug - Abstract
Standard therapies for prostate cancer including radiation, prostatectomy, and hormone ablation have significant toxicities and recurrence risk. HSV-tk gene therapy may be effective in combination with radiation therapy due to complementary mechanisms and distinct toxicity profiles. Mouse prostate tumors transplanted subcutaneously were treated by either gene therapy involving intratumoral injection of AdV-tk followed by systemic ganciclovir or local radiation therapy or the combination of gene and radiation therapy. Both single-therapy modalities showed a 38% decrease in tumor growth compared to controls. The combined treatment resulted in a decrease of 61%. In addition the combined-therapy group had a mean survival of 22 days versus 16.6 days for single therapy and 13.8 days for nontreated controls. To analyze systemic anti-tumor activity, lung metastases were generated by tail vein injection of RM-1 prostate cancer cells on the same day that they were injected subcutaneously. The primary tumors were treated as before with AdV-tk followed by ganciclovir, radiation, or the combination. The number of lung nodules was reduced by 37% following treatment with AdV-tk, whereas radiotherapy alone had no effect on metastatic growth. The combination led to an additional 50% reduction in lung colonization. Primary tumors that received the combination therapy had a marked increase in CD4 T cell infiltrate. This is the first report showing a dramatic systemic effect following the local combination treatment of radiation and AdV-tk. A clinical study using this strategy has been initiated and patient accrual is ongoing.
- Published
- 2001
43. In vivo surgical resection plus adjuvant gene therapy in the treatment of mammary and prostate cancer
- Author
-
Brian J. Miles, Dov Kadmon, Gustavo Ayala, Timothy C. Thompson, Laura K. Aguilar, Estuardo Aguilar-Cordova, Xiaohong Zhu, Madhu Chhikara, E. O'Brian Smith, and Steven W. Sukin
- Subjects
Oncology ,Male ,medicine.medical_specialty ,Lung Neoplasms ,Time Factors ,medicine.medical_treatment ,Genetic enhancement ,Genetic Vectors ,Thymidine Kinase ,Adenoviridae ,03 medical and health sciences ,Prostate cancer ,Mice ,0302 clinical medicine ,Prostate ,In vivo ,Internal medicine ,Drug Discovery ,medicine ,Genetics ,Animals ,Neoplasm Metastasis ,Molecular Biology ,Lung ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Mice, Inbred BALB C ,business.industry ,Cancer ,Mammary Neoplasms, Experimental ,Prostatic Neoplasms ,Genetic Therapy ,medicine.disease ,Combined Modality Therapy ,3. Good health ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Treatment Outcome ,030220 oncology & carcinogenesis ,Toxicity ,Molecular Medicine ,Female ,business ,Adjuvant ,Neoplasm Transplantation - Abstract
Adenoviral-mediated gene therapy delivery, combining the herpes simplex virus thymidine kinase gene (Ad- tk ) with gancyclovir, has been evaluated as a treatment modality for numerous tumors in the laboratory and in the clinics. As a single modality, gene therapy has shown some promising local and systemic results but no curative success. Surgery is the standard of care for many solid tumors. However, minor residual tumor following surgical resection can lead to local recurrence, and surgery is neither efficient nor plausible for metastatic disease. In this study, two tumor models were used to evaluate the effects of Ad- tk gene therapy as an adjuvant to surgery. Subcutaneous mammary- and prostate-derived tumors were produced in syngeneic mice. To evaluate systemic effects, tumor cells were injected intravenously, with subsequent formation of lung nodules. The subcutaneous tumors were surgically resected and the tumor bed was bathed with saline or Ad- tk. The animals were evaluated for toxicity, local tumor recurrence, survival, and lung nodule formation. No evidence of additional toxicity was observed. In the less aggressive mammary model, the time to recurrence was increased from 11.7 (±1.0) days to 22.7 (±5.5) days. In the prostate model, recurrence went from a mean of 17.3 (±5.6) to 22.6 (±6.8) days. Survival was also improved from a mean of 19.7 (±1.1) to 32.3 (±4.8) and 26.1 (±5.0) to 34.1 (±6.1) days in the mammary and prostate models, respectively. Evidence of systemic benefits from the use of adjuvant Ad- tk therapy was demonstrated by a significant reduction in lung nodules from a mean of 17 to 3.5. These results suggest that Ad- tk gene therapy may be a useful adjuvant for patients undergoing surgery for treatment of cancer.
- Published
- 2001
44. Adoptive Immunotherapy of EBV-Associated Malignancies with EBV-Specific Cytotoxic T-Cell Lines
- Author
-
Helen E. Heslop, Cliona M. Rooney, Laura K. Aguilar, Malcolm K. Brenner, and M. H. Huls
- Subjects
biology ,medicine.medical_treatment ,chemical and pharmacologic phenomena ,Immunotherapy ,Major histocompatibility complex ,CTL ,Immune system ,Antigen ,In vivo ,Immunology ,biology.protein ,medicine ,Cytotoxic T cell ,Ex vivo - Abstract
The goal of immunotherapy is to overcome the immune response deficits of the host or the immune stimulatory deficits of the tumor and activate an effective tumor-specific immune response. The cytotoxic T-lymphocyte (CTL) arm of the cellular immune response is thought to be the most important defense against tumors and virus-infected cells. CTLs recognize short peptides derived from viral antigens that are carried to the infected cell surface in association with major histocompatibility (MHC) molecules (see Fig. 1). Epstein-Barr virus (EBV)-associated malignancies express a range of antigens against which to target CTLs. For immunotherapy, CTLs may be activated and expanded in vivo or ex vivo. In vivo strategies involve immunization with DNA, tumor vaccines, or antigen- or peptide-loaded dendritic cells. Ex vivo strategies involve exposing T cells to tumor or viral antigens expressed on antigen-presenting cells (APCs) and expanding them in T-cell growth factors in vitro. Although the ex vivo approach may be more costly in the time, effort, and expertise required to grow CTLs for patient infusion, it may be the only option in Fig.1.
- Published
- 2001
- Full Text
- View/download PDF
45. Lymphoproliferative disorders involving Epstein-Barr virus after hemopoietic stem cell transplantation
- Author
-
Cliona M. Rooney, Laura K. Aguilar, and Helen E. Heslop
- Subjects
Cancer Research ,Herpesvirus 4, Human ,medicine.medical_treatment ,T cell ,T-Lymphocytes ,Lymphoproliferative disorders ,Hematopoietic stem cell transplantation ,Major histocompatibility complex ,medicine.disease_cause ,Risk Factors ,hemic and lymphatic diseases ,medicine ,Cytotoxic T cell ,Humans ,B-Lymphocytes ,biology ,business.industry ,Hematopoietic Stem Cell Transplantation ,Immunosuppression ,medicine.disease ,Epstein–Barr virus ,Lymphoproliferative Disorders ,Transplantation ,medicine.anatomical_structure ,Oncology ,Immunology ,biology.protein ,business - Abstract
Lymphoproliferative disorders involving uncontrolled expansion of donor-derived B cells infected with Epstein-Barr virus (EBV) are a significant problem after hemopoietic stem cell transplantation. Risk factors, which include T cell depletion, major histocompatibility complex mismatch, and intensity of immunosuppression illustrate the importance of T cell immune surveillance. Recent studies have identified viral and host factors that affect the T-cell response to EBV. Monitoring EBV load in the blood by polymerase chain reaction allows early identification of high-risk patients and early institution of therapy. Adoptive immunotherapy approaches using donor T cells have proven effective and EBV-specific cytotoxic T lymphocytes have also been used successfully for prophylaxis. The simplest way of preventing EBV lymphoproliferation, however, may be to deplete B cells from the donor marrow prior to infusion to prevent the transmission of EBV-infected B cells.
- Published
- 1999
46. 298. Gene Therapy Against Prostate Cancer Using the HSV-tk/Prodrug System Followed by Prostatectomy
- Author
-
Estuardo Aguilar-Cordova, Hugo A. Barrera-Saldaña, Laura K. Aguilar, E. Brian Butler, Raquel Garza-Guajardo, Juan Pablo Flores-Gutiérrez, Augusto Rojas-Martinez, Rocio Ortiz-Lopez, Juan Francisco González-Guerrero, and Jacinto Esteban-Maria
- Subjects
Pharmacology ,medicine.medical_specialty ,business.industry ,Prostatectomy ,Genetic enhancement ,medicine.medical_treatment ,Urology ,Prodrug ,medicine.disease ,Viral vector ,Clinical trial ,Prostate cancer ,Drug Discovery ,Toxicity ,Genetics ,medicine ,Molecular Medicine ,Population study ,business ,Molecular Biology - Abstract
The authors present the first Phase I gene transfer clinical trial initiated in Latin America. The study population was low to high risk prostate cancer patients scheduled to undergo surgical tumor removal. The study was designed to evaluate the safety of the procedure, vector distribution within the tumor and pathologic effects of a four quadrant transrectal intra-prostatic injection of a non-replicating adenoviral vector carrying the HSV-tk gene (AdV-tk). Differences in these parameters after treatment with intravenous ganciclovir (GCV) vs oral valacyclovir (VCV) were also evaluated. Each subject received 1X1011 vector particles followed by 14 days of GCV or VCV prior to radical prostatectomy. No significant vector related toxicity was observed any of the ten analyzed patients; one subject abandoned the study prior to surgery for personal reasons. Vector sequences were detected in all tumors, with 30% to 85% positive distribution-cores from each gland, even in a specimen removed 15 weeks after vector injection. Pathological analyses demonstrated zones of regional tumor necrosis and inflammation of tumor tissue associated with significant CD8+ T-cell infiltration. Biological activity of the vector was evidenced by a spike in circulating PSA levels shortly after injection followed by an overall decrease from starting PSA concentration to the level observed just prior to surgery in seven of ten patients. No differences were observed between the two prodrugs. Patient follow-up has continued for an average of 52.8 months (range: 38 to 64 months) with encouraging results. These data demonstrate the safety and distribution-efficacy of simple outpatient procedure and oral prodrug for AdV-tk in prostate cancer.
- Published
- 2005
- Full Text
- View/download PDF
47. Phase I trial of gene-mediated cytotoxic immunotherapy in combination with chemoradiation for locally advanced pancreatic cancer
- Author
-
Jon P. Walker, Walter J. Coyle, Mark Bloomston, Estuardo Aguilar-Cordova, Vincent Chung, Laura K. Aguilar, Sanaa Tahiri, Christopher L. Marsh, C. Moran Cruz, and Howard Marx
- Subjects
Oncology ,Cancer Research ,Pathology ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Locally advanced ,Disease ,Immunotherapy ,medicine.disease ,Locally advanced pancreatic cancer ,Pancreatic cancer ,Internal medicine ,Medicine ,Cytotoxic T cell ,business ,Gene ,Median survival - Abstract
195 Background: More than 80% of patients with pancreatic cancer present with locally advanced or metastatic disease and have a median survival of only 6 months. Immunotherapy approaches may improve outcomes. Gene Mediated Cytotoxic Immunotherapy (GMCI) is an approach that generates a systemic anti-tumor vaccine effect through intra-tumoral delivery of an adenoviral vector expressing the HSV-thymidine kinase gene (AdV-tk) followed by anti-herpetic prodrug and synergy with chemoradiation. The mechanisms of action involve tumor cytotoxicity, activation of antigen presenting cells and stimulation of systemic anti-tumor T-cell immunity. Safety with potential efficacy has been demonstrated in multiple clinical studies. This is the first application of GMCI in pancreatic cancer. Methods: This study evaluated 4 dose levels of AdV-tk (3x1010 to 1x1012 vector particles) injected into locally advanced tumors via EUS or CT-guidance before and during week 3 of standard 5-FU-chemoradiation. Valacyclovir (Valtrex, GSK) prodrug was given for 14 days after each of 2 AdV-tk injections. Results: The study completed accrual with 13 patients enrolled and 12 completing therapy with 3 at each of the 4 dose levels. One patient refused further participation during course 1 after recovering from azotemia. Median age was 64 years (range 55-81) and median baseline CA19-9 was 1634 U/ml. No dose limiting toxicities and no injection related complications occurred. Possibly related grade 3-4 toxicities, all of which were transient, included dehydration, azotemia and worsening elevation of bilirubin and AST. Kaplan-Meier estimated median survival is 12.2 months with 6 patients still alive at 8-20 months. Two patients achieved a partial response by RECIST criteria. One occurred in week 6 despite discontinuing 5-FU/radiation during week 1. The other had gradual decrease of a 7 cm tumor over 11 months. Serum CA19-9 levels decreased in 8/8 evaluable patients by 32-91% at 3 months after treatment initiation. Conclusions: AdV-tk can be safely injected into pancreatic tumors and combined with standard chemoradiation. Early results are highly encouraging and justify further evaluation in a phase II study. [Table: see text]
- Published
- 2011
- Full Text
- View/download PDF
48. Five year follow up of a phase II study of cytotoxic immunotherapy combined with radiation in newly diagnosed prostate cancer
- Author
-
Gustavo E. Ayala, Laura K. Aguilar, Edward Brian Butler, Estuardo Aguilar-Cordova, W. Mai, J. Caillouet, and Bin S. Teh
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Incidence (epidemiology) ,Five year follow up ,Phases of clinical research ,Immunotherapy ,Newly diagnosed ,medicine.disease ,Prostate cancer ,Internal medicine ,medicine ,Cytotoxic T cell ,business - Abstract
4635 Background: In the U.S. there are about 70,000 annual prostate cancer recurrences. The purpose of this study is to evaluate a product to decrease incidence of recurrence. This study is based on objective clinical responses in Phase I studies with AdV-tk (ProstAtak™, Advantagene, Inc) as monotherapy in recurrent disease and preclinical data demonstrating synergy between AdV-tk and radiation. AdV-tk is an adenoviral vector expressing the herpes thymidine kinase gene delivered to the prostate via TRUS-guided injection followed by 14 days of oral prodrug. The mechanisms of function involve direct tumor cytotoxicity, local elicitation of danger signals, recruitment and activation of antigen presenting cells and stimulation of systemic anti-tumor T-cell immunity. Method: AdV-tk was evaluated in combination with radiation in 66 newly diagnosed patients: 33 low risk (Arm A, PSA [Table: see text]
- Published
- 2006
- Full Text
- View/download PDF
49. PSA nadir and 24 month biopsy interim analysis of AdV-tk/Valacyclovir gene therapy in combination with radiotherapy vs radiotherapy alone for prostate cancer
- Author
-
Bin S. Teh, Estuardo Aguilar-Cordova, Brian J. Miles, Thomas M. Wheeler, Gustavo E. Ayala, Laura K. Aguilar, Timothy C. Thompson, Maria T. Vlachaki, Edward Brian Butler, and Dov Kadmon
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,viruses ,medicine.medical_treatment ,Genetic enhancement ,virus diseases ,Prodrug ,medicine.disease ,Interim analysis ,Clinical trial ,Radiation therapy ,Prostate cancer ,Thymidine kinase ,Internal medicine ,Biopsy ,medicine ,business - Abstract
4571 Background: A Phase II clinical trial for newly diagnosed, non-metastatic prostate cancer is ongoing in which adenoviral-HSV thymidine kinase (AdV-tk) + prodrug (valacyclovir) is added to stan...
- Published
- 2004
- Full Text
- View/download PDF
50. Late toxicity of a phase I/II trial evaluating combined radiotherapy and in-situ gene-therapy with or without hormonal therapy in the treatment of prostate cancer
- Author
-
Laura K. Aguilar, Dov Kadmon, Gustavo Ayala, J. Caillouet, E.B. Butler, Estuardo Aguilar-Cordova, Maria T. Vlachaki, Bin S. Teh, Timothy C. Thompson, Brian J. Miles, M Davis, and W. Mai
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Radiation ,business.industry ,medicine.medical_treatment ,Genetic enhancement ,medicine.disease ,Radiation therapy ,Late toxicity ,Prostate cancer ,Phase i ii ,Combined radiotherapy ,Internal medicine ,Medicine ,Hormonal therapy ,Radiology, Nuclear Medicine and imaging ,business - Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.