Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Alexander J. Annala is active.

Publication


Featured researches published by Alexander J. Annala.


Science Translational Medicine | 2013

Neural Stem Cell–Mediated Enzyme/Prodrug Therapy for Glioma: Preclinical Studies

Karen S. Aboody; Joseph Najbauer; Marianne Z. Metz; Massimo D'Apuzzo; Margarita Gutova; Alexander J. Annala; Timothy W. Synold; Larry A. Couture; Suzette Blanchard; Rex Moats; Elizabeth Garcia; Soraya Aramburo; Valenzuela Vv; Richard T. Frank; Michael E. Barish; Christine E. Brown; Seung U. Kim; Behnam Badie; Jana Portnow

Neural stem cells home to gliomas in mice where they convert a prodrug to 5-fluorouracil, leading to tumor regression. Cellular Assassins Derived from the supporting cells of the brain, gliomas are deadly tumors that can be only temporarily held at bay, but not cured. New ways to treat these cancers are needed. To get regulatory approval to test a new stem cell–based therapy in patients, Aboody et al. performed a series of preclinical experiments in mice with artificially implanted gliomas in their brains. By mimicking closely the treatments that they hoped to perform in humans, these authors were able to show to the satisfaction of the regulatory agency that the treatment was safe and effective enough in the mice to warrant a first-in-human trial in patients. The authors used a neural stem cell line carrying a v-myc gene and a gene for cytosine deaminase. These cells exhibit tropism to human glioma cells. When injected into mice with gliomas, they migrate to the site of the tumor, even when the mice are treated with steroids or radiation, as might be the case for human patients. The cytosine deaminase in the cells provides another anticancer weapon. This enzyme converts the prodrug 5-fluorocytosine (5-FC) to the toxic 5-flurouracil (5-FU), delivering a high concentration of the therapeutic agent directly in and around the tumor and causing it to shrink significantly. Injection of excess numbers of cells or increasing the dose of 5-FU did not result in any abnormalities in the animals; in fact, by 12 weeks after injection, no cells were to be seen in the brain or elsewhere, even when a highly sensitive polymerase chain reaction method was used to look for the v-myc DNA. This targeted cell-based approach to cancer therapy that concentrates the therapeutic agent in the vicinity of the tumor is expected to reduce toxicity to other tissues. Thus, a higher local dose is possible, potentially improving efficacy against the tumor. The phase 1 trial derived from these preclinical results is ongoing; its end will allow evaluation of how well these preclinical in vivo studies set the stage for humans. High-grade gliomas are extremely difficult to treat because they are invasive and therefore not curable by surgical resection; the toxicity of current chemo- and radiation therapies limits the doses that can be used. Neural stem cells (NSCs) have inherent tumor-tropic properties that enable their use as delivery vehicles to target enzyme/prodrug therapy selectively to tumors. We used a cytosine deaminase (CD)–expressing clonal human NSC line, HB1.F3.CD, to home to gliomas in mice and locally convert the prodrug 5-fluorocytosine to the active chemotherapeutic 5-fluorouracil. In vitro studies confirmed that the NSCs have normal karyotype, tumor tropism, and CD expression, and are genetically and functionally stable. In vivo biodistribution studies demonstrated NSC retention of tumor tropism, even in mice pretreated with radiation or dexamethasone to mimic clinically relevant adjuvant therapies. We evaluated safety and toxicity after intracerebral administration of the NSCs in non–tumor-bearing and orthotopic glioma–bearing immunocompetent and immunodeficient mice. We detected no difference in toxicity associated with conversion of 5-fluorocytosine to 5-fluorouracil, no NSCs outside the brain, and no histological evidence of pathology or tumorigenesis attributable to the NSCs. The average tumor volume in mice that received HB1.F3.CD NSCs and 5-fluorocytosine was about one-third that of the average volume in control mice. On the basis of these results, we conclude that combination therapy with HB1.F3.CD NSCs and 5-fluorocytosine is safe, nontoxic, and effective in mice. These data have led to approval of a first-in-human study of an allogeneic NSC-mediated enzyme/prodrug-targeted cancer therapy in patients with recurrent high-grade glioma.


ACS Nano | 2014

Neural Stem Cell-Mediated Intratumoral Delivery of Gold Nanorods Improves Photothermal Therapy

Rachael Mooney; Luella Roma; Donghong Zhao; Desiree Van Haute; Elizabeth Garcia; Seung U. Kim; Alexander J. Annala; Karen S. Aboody; Jacob M. Berlin

Plasmonic photothermal therapy utilizes biologically inert gold nanorods (AuNRs) as tumor-localized antennas that convert light into heat capable of eliminating cancerous tissue. This approach has lower morbidity than surgical resection and can potentially synergize with other treatment modalities including chemotherapy and immunotherapy. Despite these advantages, it is still challenging to obtain heating of the entire tumor mass while avoiding unnecessary collateral damage to surrounding healthy tissue. It is therefore critical to identify innovative methods to distribute an effective concentration of AuNRs throughout tumors without depositing them in surrounding healthy tissue. Here we demonstrate that AuNR-loaded, tumor-tropic neural stem cells (NSCs) can be used to improve the intratumoral distribution of AuNRs. A simple UV–vis technique for measuring AuNR loading within NSCs was established. It was then confirmed that NSC viability is unimpaired following AuNR loading and that NSCs retain AuNRs long enough to migrate throughout tumors. We then demonstrate that intratumoral injections of AuNR-loaded NSCs are more efficacious than free AuNR injections, as evidenced by reduced recurrence rates of triple-negative breast cancer (MDA-MB-231) xenografts following NIR exposure. Finally, we demonstrate that the distribution of AuNRs throughout the tumors is improved when transported by NSCs, likely resulting in the improved efficacy of AuNR-loaded NSCs as compared to free AuNRs. These findings highlight the advantage of combining cellular therapies and nanotechnology to generate more effective cancer treatments.


Stem Cells Translational Medicine | 2013

Magnetic Resonance Imaging Tracking of Ferumoxytol-Labeled Human Neural Stem Cells: Studies Leading to Clinical Use

Margarita Gutova; Joseph A. Frank; Massimo D'Apuzzo; Vazgen Khankaldyyan; Megan Gilchrist; Alexander J. Annala; Marianne Z. Metz; Yelena Abramyants; Kelsey Herrmann; Lucy Y. Ghoda; Joseph Najbauer; Christine E. Brown; M. Suzette Blanchard; Maciej S. Lesniak; Seung U. Kim; Michael E. Barish; Karen S. Aboody; Rex Moats

Numerous stem cell‐based therapies are currently under clinical investigation, including the use of neural stem cells (NSCs) as delivery vehicles to target therapeutic agents to invasive brain tumors. The ability to monitor the time course, migration, and distribution of stem cells following transplantation into patients would provide critical information for optimizing treatment regimens. No effective cell‐tracking methodology has yet garnered clinical acceptance. A highly promising noninvasive method for monitoring NSCs and potentially other cell types in vivo involves preloading them with ultrasmall superparamagnetic iron oxide nanoparticles (USPIOs) to enable cell tracking using magnetic resonance imaging (MRI). We report here the preclinical studies that led to U.S. Food and Drug Administration approval for first‐in‐human investigational use of ferumoxytol to label NSCs prior to transplantation into brain tumor patients, followed by surveillance serial MRI. A combination of heparin, protamine sulfate, and ferumoxytol (HPF) was used to label the NSCs. HPF labeling did not affect cell viability, growth kinetics, or tumor tropism in vitro, and it enabled MRI visualization of NSC distribution within orthotopic glioma xenografts. MRI revealed dynamic in vivo NSC distribution at multiple time points following intracerebral or intravenous injection into glioma‐bearing mice that correlated with histological analysis. Preclinical safety/toxicity studies of intracerebrally administered HPF‐labeled NSCs in mice were also performed, and they showed no significant clinical or behavioral changes, no neuronal or systemic toxicities, and no abnormal accumulation of iron in the liver or spleen. These studies support the clinical use of ferumoxytol labeling of cells for post‐transplant MRI visualization and tracking.


Stem Cells | 2012

Human Neural Stem Cell Tropism to Metastatic Breast Cancer

Donghong Zhao; Joseph Najbauer; Alexander J. Annala; Elizabeth Garcia; Marianne Z. Metz; Margarita Gutova; Monika D. Polewski; Megan Gilchrist; Carlotta A. Glackin; Seung U. Kim; Karen S. Aboody

Metastasis to multiple organs is the primary cause of mortality in breast cancer patients. The poor prognosis for patients with metastatic breast cancer and toxic side effects of currently available treatments necessitate the development of effective tumor‐selective therapies. Neural stem cells (NSCs) possess inherent tumor tropic properties that enable them to overcome many obstacles of drug delivery that limit effective chemotherapy strategies for breast cancer. We report that increased NSC tropism to breast tumor cell lines is strongly correlated with the invasiveness of cancer cells. Interleukin 6 (IL‐6) was identified as a major cytokine mediating NSC tropism to invasive breast cancer cells. We show for the first time in a preclinical mouse model of metastatic human breast cancer that NSCs preferentially target tumor metastases in multiple organs, including liver, lung, lymph nodes, and femur, versus the primary intramammary fat pad tumor. For proof‐of‐concept of stem cell‐mediated breast cancer therapy, NSCs were genetically modified to secrete rabbit carboxylesterase (rCE), an enzyme that activates the CPT‐11 prodrug to SN‐38, a potent topoisomerase I inhibitor, to effect tumor‐localized chemotherapy. In vitro data demonstrate that exposure of breast cancer cells to conditioned media from rCE‐secreting NSCs (NSC.rCE) increased their sensitivity to CPT‐11 by 200‐fold. In vivo, treatment of tumor‐bearing mice with NSC.rCE cells in combination with CPT‐11 resulted in reduction of metastatic tumor burden in lung and lymph nodes. These data suggest that NSC‐mediated enzyme/prodrug therapy may be more effective and less toxic than currently available chemotherapy strategies for breast cancer metastases. STEM CELLS 2012; 30:314–325.


Future Oncology | 2014

Neural stem cells improve intracranial nanoparticle retention and tumor-selective distribution

Rachael Mooney; Yiming Weng; Revathiswari Tirughana-Sambandan; Valerie Valenzuela; Soraya Aramburo; Elizabeth Garcia; Zhongqi Li; Margarita Gutova; Alexander J. Annala; Jacob M. Berlin; Karen S. Aboody

AIM The purpose of this work is to determine if tumor-tropic neural stem cells (NSCs) can improve the tumor-selective distribution and retention of nanoparticles (NPs) within invasive brain tumors. MATERIALS & METHODS Streptavidin-conjugated, polystyrene NPs are surface-coupled to biotinylated human NSCs. These NPs are large (798 nm), yet when conjugated to tropic cells, they are too large to passively diffuse through brain tissue or cross the blood-tumor barrier. NP distribution and retention was quantified 4 days after injections located either adjacent to an intracerebral glioma, in the contralateral hemisphere, or intravenously. RESULTS & CONCLUSION In all three in vivo injection paradigms, NSC-coupled NPs exhibited significantly improved tumor-selective distribution and retention over free-NP suspensions. These results provide proof-of-principle that NSCs can facilitate the tumor-selective distribution of NPs, a platform useful for improving intracranial drug delivery.


Cancer Gene Therapy | 2015

Analysis of glioblastoma tumor coverage by oncolytic virus-loaded neural stem cells using MRI-based tracking and histological reconstruction

Ramin A. Morshed; Margarita Gutova; Joseph Juliano; Michael E. Barish; Andrea Hawkins-Daarud; Diana Oganesyan; Khankaldyyan Vazgen; Tang Yang; Alexander J. Annala; Atique U. Ahmed; Karen S. Aboody; Kristin R. Swanson; Rex Moats; Maciej S. Lesniak

In preclinical studies, neural stem cell (NSC)-based delivery of oncolytic virus has shown great promise in the treatment of malignant glioma. Ensuring the success of this therapy will require critical evaluation of the spatial distribution of virus after NSC transplantation. In this study, the patient-derived GBM43 human glioma line was established in the brain of athymic nude mice, followed by the administration of NSCs loaded with conditionally replicating oncolytic adenovirus (NSC-CRAd-S-pk7). We determined the tumor coverage potential of oncolytic adenovirus by examining NSC distribution using magnetic resonance (MR) imaging and by three-dimensional reconstruction from ex vivo tissue specimens. We demonstrate that unmodified NSCs and NSC-CRAd-S-pk7 exhibit a similar distribution pattern with most prominent localization occurring at the tumor margins. We were further able to visualize the accumulation of these cells at tumor sites via T2-weighted MR imaging as well as the spread of viral particles using immunofluorescence. Our analyses reveal that a single administration of oncolytic virus-loaded NSCs allows for up to 31% coverage of intracranial tumors. Such results provide valuable insights into the therapeutic potential of this novel viral delivery platform.


Bioconjugate Chemistry | 2017

Intraperitoneal Administration of Neural Stem Cell–Nanoparticle Conjugates Targets Chemotherapy to Ovarian Tumors

Pengpeng Cao; Rachael Mooney; Revathiswari Tirughana; Wafa Abidi; Soraya Aramburo; Linda Flores; Megan Gilchrist; Ugochi Nwokafor; Tom Haber; Pamela Tiet; Alexander J. Annala; Ernest S. Han; Thanh H. Dellinger; Karen S. Aboody; Jacob M. Berlin

Ovarian cancer is particularly aggressive once it has metastasized to the abdominal cavity (stage III). Intraperitoneal (IP) as compared to intravenous (IV) administration of chemotherapy improves survival for stage III ovarian cancer, demonstrating that concentrating chemotherapy at tumor sites has therapeutic benefit; unfortunately, IP therapy also increases toxic side effects, thus preventing its completion in many patients. The ability to target chemotherapy selectively to ovarian tumors while sparing normal tissue would improve efficacy and decrease toxicities. We have previously shown that tumor-tropic neural stem cells (NSCs) dramatically improve the intratumoral distribution of nanoparticles (NPs) when given intracerebrally near an orthotopic brain tumor or into a flank xenograft tumor. Here, we show that NPs either conjugated to the surface of NSCs or loaded within the cells are selectively delivered to and distributed within ovarian tumors in the abdominal cavity following IP injection, with no evidence of localization to normal tissue. IP administration is significantly more effective than IV administration, and NPs carried by NSCs show substantially deeper penetration into tumors than free NPs. The NSCs and NPs target and localize to ovarian tumors within 1 h of administration. Pt-loaded silica NPs (SiNP[Pt]) were developed that can be transported in NSCs, and it was found that the NSC delivery of SiNP[Pt] (NSC-SiNP[Pt]) results in higher levels of Pt in tumors as compared to free drug or SiNP[Pt]. To the best of our knowledge, this work represents the first demonstration that cells given IP can target the delivery of drug-loaded NPs.


Molecular Therapy - Oncolytics | 2017

Optimization of a Neural Stem-Cell-Mediated Carboxylesterase/Irinotecan Gene Therapy for Metastatic Neuroblastoma

Margarita Gutova; Leanne Goldstein; Marianne Z. Metz; Anahit Hovsepyan; Lyudmila Tsurkan; Revathiswari Tirughana; Lusine Tsaturyan; Alexander J. Annala; Timothy W. Synold; Zesheng Wan; Robert C. Seeger; Clarke P. Anderson; Rex Moats; Philip M. Potter; Karen S. Aboody

Despite improved survival for children with newly diagnosed neuroblastoma (NB), recurrent disease is a significant problem, with treatment options limited by anti-tumor efficacy, patient drug tolerance, and cumulative toxicity. We previously demonstrated that neural stem cells (NSCs) expressing a modified rabbit carboxylesterase (rCE) can distribute to metastatic NB tumor foci in multiple organs in mice and convert the prodrug irinotecan (CPT-11) to the 1,000-fold more toxic topoisomerase-1 inhibitor SN-38, resulting in significant therapeutic efficacy. We sought to extend these studies by using a clinically relevant NSC line expressing a modified human CE (hCE1m6-NSCs) to establish proof of concept and identify an intravenous dose and treatment schedule that gave maximal efficacy. Human-derived NB cell lines were significantly more sensitive to treatment with hCE1m6-NSCs and irinotecan as compared with drug alone. This was supported by pharmacokinetic studies in subcutaneous NB mouse models demonstrating tumor-specific conversion of irinotecan to SN-38. Furthermore, NB-bearing mice that received repeat treatment with intravenous hCE1m6-NSCs and irinotecan showed significantly lower tumor burden (1.4-fold, p = 0.0093) and increased long-term survival compared with mice treated with drug alone. These studies support the continued development of NSC-mediated gene therapy for improved clinical outcome in NB patients.


Stem cell reports | 2016

L-MYC Expression Maintains Self-Renewal and Prolongs Multipotency of Primary Human Neural Stem Cells

Zhongqi Li; Diana Oganesyan; Rachael Mooney; Xianfang Rong; Matthew J. Christensen; David Shahmanyan; Patrick M. Perrigue; Joseph Benetatos; Lusine Tsaturyan; Soraya Aramburo; Alexander J. Annala; Yang Lu; Joseph Najbauer; Xiwei Wu; Michael E. Barish; David L. Brody; Karen S. Aboody; Margarita Gutova

Summary Pre-clinical studies indicate that neural stem cells (NSCs) can limit or reverse CNS damage through direct cell replacement, promotion of regeneration, or delivery of therapeutic agents. Immortalized NSC lines are in growing demand due to the inherent limitations of adult patient-derived NSCs, including availability, expandability, potential for genetic modifications, and costs. Here, we describe the generation and characterization of a new human fetal NSC line, immortalized by transduction with L-MYC (LM-NSC008) that in vitro displays both self-renewal and multipotent differentiation into neurons, oligodendrocytes, and astrocytes. These LM-NSC008 cells were non-tumorigenic in vivo, and migrated to orthotopic glioma xenografts in immunodeficient mice. When administered intranasally, LM-NSC008 distributed specifically to sites of traumatic brain injury (TBI). These data support the therapeutic development of immortalized LM-NSC008 cells for allogeneic use in TBI and other CNS diseases.


Molecular therapy. Methods & clinical development | 2018

GMP Production and Scale-Up of Adherent Neural Stem Cells with a Quantum Cell Expansion System

Revathiswari Tirughana; Marianne Z. Metz; Zhongqi Li; Christine Hall; David Hsu; Jim Beltzer; Alexander J. Annala; Diana Oganesyan; Margarita Gutova; Karen S. Aboody

Cell-based therapies hold great promise for a myriad of clinical applications. However, as these therapies move from phase I to phase II and III trials, there is a need to improve scale-up of adherent cells for the production of larger good manufacturing practice (GMP) cell banks. As we advanced our neural stem cell (NSC)-mediated gene therapy trials for glioma to include dose escalation and multiple treatment cycles, GMP production using cell factories (CellStacks) generated insufficient neural stem cell (NSC) yields. To increase yield, we developed an expansion method using the hollow fiber quantum cell expansion (QCE) system. Seeding of 5.2 × 107 NSCs in a single unit yielded up to 3 × 109 cells within 10 days. These QCE NSCs showed genetic and functional stability equivalent to those expanded by conventional flask-based methods. We then expanded the NSCs in 7 units simultaneously to generate a pooled GMP-grade NSC clinical lot of more than 1.5 × 1010 cells in only 9 days versus 8 × 109 over 6 weeks in CellStacks. We also adenovirally transduced our NSCs within the QCE. We found the QCE system enabled rapid cell expansion and increased yield while maintaining cell properties and reducing process time, labor, and costs with improved efficiency and reproducibility.

Collaboration


Dive into the Alexander J. Annala's collaboration.

Top Co-Authors

Avatar

Karen S. Aboody

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Margarita Gutova

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rachael Mooney

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Elizabeth Garcia

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jacob M. Berlin

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Marianne Z. Metz

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rex Moats

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Behnam Badie

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jana Portnow

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Megan Gilchrist

City of Hope National Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge