Network


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

Hotspot


Dive into the research topics where Alan A. Alfieri is active.

Publication


Featured researches published by Alan A. Alfieri.


Nitric Oxide | 2008

Sustained release nitric oxide releasing nanoparticles: Characterization of a novel delivery platform based on nitrite containing hydrogel/glass composites ☆

Adam J. Friedman; George Han; Mahantesh S. Navati; Manju Chacko; Leslie Gunther; Alan A. Alfieri; Joel M. Friedman

A new platform using biocompatible materials is presented for generating powders comprised of nanoparticles that release therapeutic levels of nitric oxide (NO) in a controlled and sustained manner. The capacity of these particles to retain and gradually release NO arises from their having combined features of both glassy matrices and hydrogels. This feature allows both for the generation of NO through the thermal reduction of added nitrite by glucose and for the retention of the generated NO within the dry particles. Exposure of these robust biocompatible nanoparticles to moisture initiates the sustained release of the trapped NO over extended time periods as determined both fluorimetrically and amperometrically. The slow sustained release is in contrast to the much faster release pattern associated with the hydration-initialed NO release in powders derived from glassy matrices. These glasses are prepared using trehalose and sucrose doped with either glucose or tagatose as the source of thermal electrons needed to convert nitrite to gNO. Significantly, the release profiles for the NO in the hydrogel/glass composite materials are found to be an easily tuned parameter that is modulated through the specific additives used in preparing the hydrogel/glass composites. The presented data raise the prospect that these new NO releasing nanoparticles can be easily formulated for use under a wide range of therapeutic circumstances.


PLOS ONE | 2009

Protective role of R-spondin1, an intestinal stem cell growth factor, against radiation-induced gastrointestinal syndrome in mice

Payel Bhanja; Subhrajit Saha; Rafi Kabarriti; L. Liu; Namita Roy-Chowdhury; Jayanta Roy-Chowdhury; Rani S. Sellers; Alan A. Alfieri; Chandan Guha

Background Radiation-induced gastrointestinal syndrome (RIGS) results from a combination of direct cytocidal effects on intestinal crypt and endothelial cells and subsequent loss of the mucosal barrier, resulting in electrolyte imbalance, diarrhea, weight loss, infection and mortality. Because R-spondin1 (Rspo1) acts as a mitogenic factor for intestinal stem cells, we hypothesized that systemic administration of Rspo1 would amplify the intestinal crypt cells and accelerate the regeneration of the irradiated intestine, thereby, ameliorating RIGS. Methods and Findings Male C57Bl/6 mice received recombinant adenovirus expressing human R-spondin1 (AdRspo1) or E.coli Lacz (AdLacz), 1–3 days before whole body irradiation (WBI) or abdominal irradiation (AIR). Post-irradiation survival was assessed by Kaplan Meier analysis. RIGS was assessed by histological examination of intestine after hematoxilin and eosin staining, immunohistochemical staining of BrdU incorporation, Lgr5 and β-catenin expression and TUNEL staining. The xylose absorption test (XAT) was performed to evaluate the functional integrity of the intestinal mucosal barrier. In order to examine the effect of R-spondin1 on tumor growth, AdRspo1 and AdLacZ was administered in the animals having palpable tumor and then exposed to AIR. There was a significant increase in survival in AdRspo1 cohorts compared to AdLacZ (p<0.003) controls, following WBI (10.4 Gy). Significant delay in tumor growth was observed after AIR in both cohorts AdRspo1 and AdLacZ but AdRspo1 treated animals showed improved survival compared to AdLacZ. Histological analysis and XAT demonstrated significant structural and functional regeneration of the intestine in irradiated animals following AdRspo1 treatment. Immunohistochemical analysis demonstrated an increase in Lgr5+ve crypt cells and the translocation of β-catenin from the cytosol to nucleus and upregulation of β-catenin target genes in AdRspo1-treated mice, as compared to AdLacz-treated mice. Conclusion Rspo1 promoted radioprotection against RIGS and improved survival of mice exposed to WBI. The mechanism was likely related to induction of the Wnt-β-catenin pathway and promotion of intestinal stem cell regeneration. Rspo1 has protective effect only on normal intestinal tissue but not in tumors after AIR and thereby may increase the therapeutic ratio of chemoradiation therapy in patients undergoing abdominal irradiation for GI malignancies.


Clinical Cancer Research | 2005

Metabolic Signatures Associated with a NAD Synthesis Inhibitor–Induced Tumor Apoptosis Identified by 1H-Decoupled-31P Magnetic Resonance Spectroscopy

Manickam Muruganandham; Alan A. Alfieri; Cornelia Matei; Yuchun Chen; George Sukenick; Isabel Schemainda; Max Hasmann; Leonard Saltz; Jason A. Koutcher

Purpose: Attempts to selectively initiate tumor cell death through inducible apoptotic pathways are increasingly being exploited as a potential anticancer strategy. Inhibition of NAD+ synthesis by a novel agent FK866 has been recently reported to induce apoptosis in human leukemia, hepatocarcinoma cells in vitro, and various types of tumor xenografts in vivo. In the present study, we used 1H-decoupled phosphorus (31P) magnetic resonance spectroscopy (MRS) to examine the metabolic changes associated with FK866 induced tumor cell death in a mouse mammary carcinoma. Experimental Design: Induction of apoptosis in FK866-treated tumors was confirmed by histology and cytofluorometric analysis. FK866-induced changes in mammary carcinoma tumor metabolism in vivo were investigated using 1H-decoupled 31P MRS. To discern further the changes in metabolic profiles of tumors observed in vivo, high-resolution in vitro 1H-decoupled 31P MRS studies were carried out with perchloric acid extracts of mammary carcinoma tumors excised after similar treatments. In addition, the effects of FK866 on mammary carcinoma tumor growth and radiation sensitivity were studied. Results: Treatment with FK866 induced a tumor growth delay and enhanced radiation sensitivity in mammary carcinoma tumors that was associated with significant increases in the 31P MR signal in the phosphomonoester region and a decrease in NAD+ levels, pH, and bioenergetic status. The 31P MRS of perchloric acid extracts of treated tumors identified the large unresolved signal in the phosphomonoester region as the resultant of resonances originating from intermediates of tumor glycolysis and guanylate synthesis in addition to alterations in pyridine nucleotide pools and phospholipid metabolism. Conclusion: The present results suggest that FK866 interferes with multiple biochemical pathways that contribute to the increased cell death (apoptosis) and subsequent radiation sensitivity observed in the mammary carcinoma that could be serially monitored by 31P MRS.


International Journal of Radiation Oncology Biology Physics | 1996

Radiotherapy for age-related macular degeneration: Preliminary results of a potentially new treatment

Anthony M. Berson; Paul T. Finger; David L. Sherr; Richard Emery; Alan A. Alfieri; Jay L. Bosworth

PURPOSE Neovascular macular degeneration is the leading cause of severe blindness in North America today. Limited treatments are available for this disease process. A Phase I/II study was performed to determine the toxicity and efficacy of external beam radiotherapy in patients with age-related subfoveal neovascularization. METHODS AND MATERIALS Between March 1994 and June 1995, 52 patients with a mean age of 80 (60-92) were enrolled. These patients were either not eligible or were poor candidates for laser photocoagulation, primarily because of the subfoveal location of the neovascularization. Initial visual acuities ranged from 20 out of 32 to finger counting at 3 feet. All patients underwent fluorescein angiographic evaluation and documentation of their neovascular disease prior to irradiation. Patients were treated with a single lateral 4- or 6-MV photon beam, to a dose of 14-15 Gy in eight fractions over 10 days. The field size averaged 5 x 3 cm. RESULTS No significant acute morbidity was noted. All patients underwent ophthalmic examinations and repeat angiography at 1 and 3 months posttreatment and then at 3-month intervals. With a mean follow-up of 7 months (3-18 months), 41 patients (79%) are within two lines of their pretreatment visual acuity. On angiographic imaging, there was stabilization of subfoveal neovascular membranes in 34 patients (65%). New neovascular membranes have been noted in five patients. CONCLUSIONS It appears that radiotherapy can affect active subretinal neovascularization, but it is unlikely to prevent new neovascular events produced by this chronic disease. Further investigation is warranted.


PLOS ONE | 2011

Bone Marrow Stromal Cell Transplantation Mitigates Radiation-Induced Gastrointestinal Syndrome in Mice

Subhrajit Saha; Payel Bhanja; Rafi Kabarriti; L. Liu; Alan A. Alfieri; Chandan Guha

Background Nuclear accidents and terrorism presents a serious threat for mass casualty. While bone-marrow transplantation might mitigate hematopoietic syndrome, currently there are no approved medical countermeasures to alleviate radiation-induced gastrointestinal syndrome (RIGS), resulting from direct cytocidal effects on intestinal stem cells (ISC) and crypt stromal cells. We examined whether bone marrow-derived adherent stromal cell transplantation (BMSCT) could restitute irradiated intestinal stem cells niche and mitigate radiation-induced gastrointestinal syndrome. Methodology/Principal Findings Autologous bone marrow was cultured in mesenchymal basal medium and adherent cells were harvested for transplantation to C57Bl6 mice, 24 and 72 hours after lethal whole body irradiation (10.4 Gy) or abdominal irradiation (16–20 Gy) in a single fraction. Mesenchymal, endothelial and myeloid population were characterized by flow cytometry. Intestinal crypt regeneration and absorptive function was assessed by histopathology and xylose absorption assay, respectively. In contrast to 100% mortality in irradiated controls, BMSCT mitigated RIGS and rescued mice from radiation lethality after 18 Gy of abdominal irradiation or 10.4 Gy whole body irradiation with 100% survival (p<0.0007 and p<0.0009 respectively) beyond 25 days. Transplantation of enriched myeloid and non-myeloid fractions failed to improve survival. BMASCT induced ISC regeneration, restitution of the ISC niche and xylose absorption. Serum levels of intestinal radioprotective factors, such as, R-Spondin1, KGF, PDGF and FGF2, and anti-inflammatory cytokines were elevated, while inflammatory cytokines were down regulated. Conclusion/Significance Mitigation of lethal intestinal injury, following high doses of irradiation, can be achieved by intravenous transplantation of marrow-derived stromal cells, including mesenchymal, endothelial and macrophage cell population. BMASCT increases blood levels of intestinal growth factors and induces regeneration of the irradiated host ISC niche, thus providing a platform to discover potential radiation mitigators and protectors for acute radiation syndromes and chemo-radiation therapy of abdominal malignancies.


Magnetic Resonance in Medicine | 2003

In vivo multiple-mouse imaging at 1.5 T

Su Xu; T. Gade; Cornelia Matei; Kristen L. Zakian; Alan A. Alfieri; X. Hu; Eric C. Holland; S. Soghomonian; Juri Gelovani Tjuvajev; Douglas Ballon; Jason A. Koutcher

A multiple‐mouse solenoidal MR coil was developed for in vivo imaging of up to 13 mice simultaneously to screen for tumors on a 1.5 T clinical scanner. For the coil to be effective as a screening tool, it should permit acquisition of MRIs in which orthotopic tumors with diameters >2 mm are detectable in a reasonable period of time (<1 hr magnet time) and their sizes accurately measured. Using a spin echo sequence, we demonstrated that this coil provides sufficient sensitivity for moderately high resolution images (156–176 μm in plane‐resolution, 1.5 mm slice thickness). This spatial resolution permitted detection of primary brain tumors in transgenic/knockout mice and orthotopic xenografts. Brain tumor size as measured by MRI was correlated with size measured by histopathology (P < 0.001). Metastatic tumors in the mouse lung were also successfully imaged in a screening setting. The multiple mouse coil is simple in construction and may be implemented without any significant modification to the hardware or software on a clinical scanner. Magn Reson Med 49:551–557, 2003.


Cancer Research | 2007

Localized Hyperthermia Combined with Intratumoral Dendritic Cells Induces Systemic Antitumor Immunity

Arunika Mukhopadhaya; Joseph Mendecki; Xinyuan Dong; L. Liu; S. Kalnicki; Madhur Garg; Alan A. Alfieri; Chandan Guha

Prostate adenocarcinoma, treated with localized tumor hyperthermia (LTH), can potentially serve as a source of tumor antigen, where dying apoptotic/necrotic cells release tumor peptides slowly over time. In addition, LTH-treated cells can release heat shock proteins that can chaperone antigenic peptides to antigen-presenting cells, such as dendritic cells. We attempted to discern whether sequential LTH and intratumoral dendritic cell and/or systemic granulocyte macrophage colony-stimulating factor (GM-CSF) would activate antitumor immune response in a syngeneic murine model of prostate cancer (RM-1). Palpable RM-1 tumors, grown in the distal appendage of C57BL/6 male mice, were subjected to LTH (43.7 degrees C for 1 h) x 2, separated by 5 days. Following the second LTH treatment, animals received either PBS or dendritic cells (2 x 10(6)) intratumorally (every 3 days for three injections). Separate cohorts also received i.v. injection of recombinant adenovirus-expressing murine GM-CSF (AdGMCSF), 1 day after LTH. Control animals received AdenoLacZ or AdenoGFP. Intratumoral dendritic cell injection induced tumor-specific T-helper cell activity (IFNgamma ELISPOTS) and CTL activity, which was further augmented by AdGMCSF, indicating amplification of tumor-specific TH1 immunity. The combination of LTH, AdGMCSF, and intratumoral dendritic cell injection resulted in significant tumor growth delays when compared with animal cohorts that received LTH alone. These results support an in situ autovaccination strategy where systemic administration of GM-CSF and/or intratumoral injection of autologous dendritic cells, when combined with LTH, could be an effective treatment for local and systemic recurrence of prostate cancer.


Nanomedicine: Nanotechnology, Biology and Medicine | 2012

Nitric oxide-releasing nanoparticles accelerate wound healing in NOD-SCID mice

Karin Blecher; Luis R. Martinez; Chaim Tuckman-Vernon; Parimala Nacharaju; David Schairer; Jason Chouake; Joel M. Friedman; Alan A. Alfieri; Chandan Guha; Joshua D. Nosanchuk; Adam J. Friedman

UNLABELLED Wound healing is a complex process, coordinated by various biological factors. In immunocompromised states wound healing can be interrupted as a result of decreased numbers of immune cells, impairing the production of effector molecules such as nitric oxide (NO). Therefore, topical NO-releasing platforms, such as diethylenetriamine (DETA NONOate), have been investigated to enhance wound healing. Recently, we demonstrated a nanoparticle platform that releases NO (NO-NPs) in a sustained manner, accelerating wound healing in both uninfected and infected murine wound models. Here, NO-NPs were investigated and compared to DETA NONOate in an immunocompromised wound model using non-obese, diabetic, severe combined immunodeficiency mice. NO-NP treatment accelerated wound closure as compared to controls and DETA NONOate treatment. In addition, histological assessment revealed that wounds treated with NO-NPs had less inflammation, more collagen deposition, and more blood vessel formation as compared to other groups, consistent with our previous data in immunocompetent animals. These data suggest that NO-NPs may serve as a novel wound-healing therapy in the setting of immunocompromised states associated with impaired wound healing. FROM THE CLINICAL EDITOR Wound healing in an immunocompromised host is often incomplete and is a source of major concern in such conditions. This work demonstrates in a murine model that in these settings NO releasing nanoparticles significantly enhance wound healing.


Oncology | 1984

Lonidamine: a hyperthermic sensitizer of HeLa cells in culture and of the Meth-A tumor in vivo.

Jae-Ho Kim; Sang Hie Kim; Alan A. Alfieri; Charles W. Young; Bruno Silvestrini

Lonidamine was studied in HeLa cell cultures and BALB/c mice with Meth-A fibrosarcoma. Lonidamine is a hyperthermia sensitizer, both in vivo and in vivo. In vitro the potentiating effects were higher at low pH conditions.


Radiology | 1974

Increased cures using fractionated exposures of x irradiation and hyperthermia in the local treatment of the Ridgway osteogenic sarcoma in mice

Eric W. Hahn; Alan A. Alfieri; Jae Ho Kim

Experiments were carried out to determine the effect of heat and/or x rays on the Ridgway osteogenic sarcoma in AKD2F1/J male mice. Tumors averaging 0.9 ± 0.2 cm in diameter were exposed to 200 rads and/or heat three times on alternate days. Tumor core temperatures of 42.5 ± 0.5 °C were maintained for 15, 30 or 45 minutes immediately before or after x irradiation. Heat alone did not effect cures whereas radiation alone cured 8 of 18 animals (44%). In contrast, 71 of the 74 animals (96%) receiving the combined treatment were cured. Radiation was equally effective whether given immediately before or after tumor hyperthermia. There were no differences in cure rates in the 15-, 30- or 45-minute heat/radiation groups.

Collaboration


Dive into the Alan A. Alfieri's collaboration.

Top Co-Authors

Avatar

Chandan Guha

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

L. Liu

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Subhrajit Saha

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jason A. Koutcher

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Madhur Garg

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Sang Hie Kim

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Payel Bhanja

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

H. Zhang

Albert Einstein College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge