Network


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

Hotspot


Dive into the research topics where Akm Ghulam Muhammad is active.

Publication


Featured researches published by Akm Ghulam Muhammad.


Clinical Cancer Research | 2009

Release of HMGB1 in Response to Proapoptotic Glioma Killing Strategies: Efficacy and Neurotoxicity

Marianela Candolfi; Kader Yagiz; David Foulad; Gabrielle E. Alzadeh; Matthew Tesarfreund; Akm Ghulam Muhammad; Mariana Puntel; Kurt M. Kroeger; Chunyan Liu; Sharon E. Lee; James F. Curtin; Gwendalyn D. King; Jonathan Lerner; Katsuaki Sato; Yohei Mineharu; Weidong Xiong; Pedro R. Lowenstein; Maria G. Castro

Purpose: In preparation for a phase I clinical trial using a combined cytotoxic/immunotherapeutic strategy with adenoviruses (Ad) expressing Flt3L (Ad-Flt3L) and thymidine kinase (Ad-TK) to treat glioblastoma (GBM), we tested the hypothesis that Ad-TK+GCV would be the optimal tumor-killing agent in relation to efficacy and safety when compared with other proapoptotic approaches. Experimental Design: The efficacy and neurotoxicity of Ad-TK+GCV was compared with Ads encoding the proapoptotic cytokines [tumor necrosis factor-α, tumor necrosis factor–related apoptosis-inducing factor (TRAIL), and Fas ligand (FasL)], alone or in combination with Ad-Flt3L. In rats bearing small GBMs (day 4), only Ad-TK+GCV or Ad-FasL improved survival. Results: In rats bearing large GBMs (day 9), the combination of Ad-Flt3L with Ad-FasL did not improve survival over FasL alone, whereas Ad-Flt3L combined with Ad-TK+GCV led to 70% long-term survival. Expression of FasL and TRAIL caused severe neuropathology, which was not encountered when we used Ad-TK+/−Ad-Flt3L. In vitro, all treatments elicited release of high mobility group box 1 protein (HMGB1) from dying tumor cells. In vivo, the highest levels of circulating HMGB1 were observed after treatment with Ad-TK+GCV+Ad-Flt3L; HMGB1 was necessary for the therapeutic efficacy of AdTK+GCV+Ad-Flt3L because its blockade with glycyrrhizin completely blocked tumor regression. We also showed the killing efficacy of Ad-TK+GCV in human GBM cell lines and GBM primary cultures, which also elicited release of HMGB1. Conclusions: Our results indicate that Ad-TK+GCV+Ad-Flt3L exhibit the highest efficacy and safety profile among the several proapoptotic approaches tested. The results reported further support the implementation of this combined approach in a phase I clinical trial for GBM.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Gene therapy-mediated delivery of targeted cytotoxins for glioma therapeutics

Marianela Candolfi; Weidong Xiong; Kader Yagiz; Chunyan Liu; Akm Ghulam Muhammad; Mariana Puntel; David Foulad; Ali Zadmehr; Gabrielle E. Ahlzadeh; Kurt M. Kroeger; Matthew Tesarfreund; Sharon E. Lee; Waldemar Debinski; Dhruv Sareen; Clive N. Svendsen; Ronald Rodriguez; Pedro R. Lowenstein; Maria G. Castro

Restricting the cytotoxicity of anticancer agents by targeting receptors exclusively expressed on tumor cells is critical when treating infiltrative brain tumors such as glioblastoma multiforme (GBM). GBMs express an IL-13 receptor (IL13Rα2) that differs from the physiological IL4R/IL13R receptor. We developed a regulatable adenoviral vector (Ad.mhIL-4.TRE.mhIL-13-PE) encoding a mutated human IL-13 fused to Pseudomonas exotoxin (mhIL-13-PE) that specifically binds to IL13Rα2 to provide sustained expression, effective anti-GBM cytotoxicity, and minimal neurotoxicity. The therapeutic Ad also encodes mutated human IL-4 that binds to the physiological IL4R/IL13R without interacting with IL13Rα2, thus inhibiting potential binding of mhIL-13-PE to normal brain cells. Using intracranial GBM xenografts and syngeneic mouse models, we tested the Ad.mhIL-4.TRE.mhIL-13-PE and two protein formulations, hIL-13-PE used in clinical trials (Cintredekin Besudotox) and a second-generation mhIL-13-PE. Cintredekin Besudotox doubled median survival without eliciting long-term survival and caused severe neurotoxicity; mhIL-13-PE led to ∼40% long-term survival, eliciting severe neurological toxicity at the high dose tested. In contrast, Ad-mediated delivery of mhIL-13-PE led to tumor regression and long-term survival in over 70% of the animals, without causing apparent neurotoxicity. Although Cintredekin Besudotox was originally developed to target GBM, when tested in a phase III trial it failed to achieve clinical endpoints and revealed neurotoxicity. Limitations of Cintredekin Besudotox include its short half-life, which demanded frequent or continued administration, and binding to IL4R/IL13R, present in normal brain cells. These shortcomings were overcome by our therapeutic Ad, thus representing a significant advance in the development of targeted therapeutics for GBM.


Current Gene Therapy | 2009

Gene Therapy for Brain Cancer: Combination Therapies Provide Enhanced Efficacy and Safety

Marianela Candolfi; Kurt M. Kroeger; Akm Ghulam Muhammad; Kader Yagiz; Catherine Farrokhi; Robert N. Pechnick; Pedro R. Lowenstein; Maria G. Castro

Glioblastoma multiforme (GBM) is the most common primary brain cancer in adults. Despite significant advances in treatment and intensive research, the prognosis for patients with GBM remains poor. Therapeutic challenges for GBM include its invasive nature, the proximity of the tumor to vital brain structures often preventing total resection, and the resistance of recurrent GBM to conventional radiotherapy and chemotherapy. Gene therapy has been proposed as a useful adjuvant for GBM, to be used in conjunction with current treatment. Work from our laboratory has shown that combination of conditional cytotoxic with immunotherapeutic approaches for the treatment of GBM elicits regression of large intracranial tumor masses and anti-tumor immunological memory in syngeneic rodent models of GBM. In this review we examined the currently available animal models for GBM, including rodent transplantable models, endogenous rodent tumor models and spontaneous GBM in dogs. We discuss non-invasive surrogate end points to assess tumor progression and therapeutic efficacy, such as behavioral tests and circulating biomarkers. Growing preclinical and clinical data contradict the old dogma that cytotoxic anti-cancer therapy would lead to an immune-suppression that would impair the ability of the immune system to mount an anti-tumor response. The implications of the findings reviewed indicate that combination of cytotoxic therapy with immunotherapy will lead to synergistic antitumor efficacy with reduced neurotoxicity and supports the clinical implementation of combined cytotoxic-immunotherapeutic strategies for the treatment of patients with GBM.


Molecular Therapy | 2008

Flt3L in combination with HSV1-TK-mediated gene therapy reverses brain tumor-induced behavioral deficits.

Gwendalyn D. King; Kurt M. Kroeger; Catherine Bresee; Marianela Candolfi; Chunyan Liu; Charlene M. Manalo; Akm Ghulam Muhammad; Robert N. Pechnick; Pedro R. Lowenstein; Maria G. Castro

Glioblastoma multiforme (GBM) is an invasive and aggressive primary brain tumor which is associated with a dismal prognosis. We have earlier developed a macroscopic, intracranial, syngeneic GBM model, in which treatment with adenoviral vectors (Ads) expressing herpes simplex virus type 1 thymidine kinase (HSV1-TK) plus ganciclovir (GCV) resulted in survival of approximately 20% of the animals. In this model, treatment with Ads expressing Fms-like tyrosine kinase 3 ligand (Flt3L), in combination with Ad-HSV1-TK improves the survival rate to approximately 70% and induces systemic antitumor immunity. We hypothesized that the growth of a large intracranial tumor mass would cause behavioral abnormalities that can be reversed by the combined gene therapy. We assessed the behavior and neuropathology of tumor-bearing animals treated with the combined gene therapy, 3 days after treatment, in long-term survivors, and in a recurrent model of glioma. We demonstrate that the intracranial GBM induces behavioral deficits that are resolved after treatment with Ad-Flt3L/Ad-TK (+GCV). Neuropathological analysis of long-term survivors revealed an overall recovery of normal brain architecture. The lack of long-term behavioral deficits and limited neuropathological abnormalities demonstrate the efficacy and safety of the combined Ad-Flt3L/Ad-TK gene therapy for GBM. These findings can serve to underpin further developments of this therapeutic modality, leading toward implementation of a Phase I clinical trial.Glioblastoma multiforme (GBM) is an invasive and aggressive primary brain tumor which is associated with a dismal prognosis. We have earlier developed a macroscopic, intracranial, syngeneic GBM model, in which treatment with adenoviral vectors (Ads) expressing herpes simplex virus type 1 thymidine kinase (HSV1-TK) plus ganciclovir (GCV) resulted in survival of ∼20% of the animals. In this model, treatment with Ads expressing Fms-like tyrosine kinase 3 ligand (Flt3L), in combination with Ad-HSV1-TK improves the survival rate to ∼70% and induces systemic antitumor immunity. We hypothesized that the growth of a large intracranial tumor mass would cause behavioral abnormalities that can be reversed by the combined gene therapy. We assessed the behavior and neuropathology of tumor-bearing animals treated with the combined gene therapy, 3 days after treatment, in long-term survivors, and in a recurrent model of glioma. We demonstrate that the intracranial GBM induces behavioral deficits that are resolved after treatment with Ad-Flt3L/Ad-TK (+GCV). Neuropathological analysis of long-term survivors revealed an overall recovery of normal brain architecture. The lack of long-term behavioral deficits and limited neuropathological abnormalities demonstrate the efficacy and safety of the combined Ad-Flt3L/Ad-TK gene therapy for GBM. These findings can serve to underpin further developments of this therapeutic modality, leading toward implementation of a Phase I clinical trial.


Clinical Pharmacology & Therapeutics | 2010

Study of the Efficacy, Biodistribution, and Safety Profile of Therapeutic Gutless Adenovirus Vectors as a Prelude to a Phase I Clinical Trial for Glioblastoma

Akm Ghulam Muhammad; Mariana Puntel; Marianela Candolfi; Alireza Salem; Kader Yagiz; Catherine Farrokhi; Kurt M. Kroeger; Weidong Xiong; James F. Curtin; Chunyan Liu; K Lawrence; Niyati Bondale; Jonathan Lerner; Gregory J. Baker; David Foulad; Robert N. Pechnick; Donna Palmer; Philip Ng; Pedro R. Lowenstein; Maria G. Castro

Glioblastoma multiforme (GBM) is the most common and most aggressive primary brain tumor in humans. Systemic immunity against gene therapy vectors has been shown to hamper therapeutic efficacy; however, helper‐dependent high‐capacity adenovirus (HC‐Ad) vectors elicit sustained transgene expression, even in the presence of systemic anti‐adenoviral immunity. We engineered HC‐Ads encoding the conditional cytotoxic herpes simplex type 1 thymidine kinase (TK) and the immunostimulatory cytokine fms‐like tyrosine kinase ligand 3 (Flt3L). Flt3L expression is under the control of the regulatable Tet‐ON system. In anticipation of a phase I clinical trial for GBM, we assessed the therapeutic efficacy, biodistribution, and clinical and neurotoxicity with escalating doses of HC‐Ad‐TetOn‐Flt3L + HC‐Ad‐TK in rats. Intratumoral administration of these therapeutic HC‐Ads in rats bearing large intracranial GBMs led to long‐term survival in _70% of the animals and development of antiglioma immunological memory without signs of neuropathology or systemic toxicity. Systemic anti‐adenoviral immunity did not affect therapeutic efficacy. These data support the idea that it would be useful to develop HC‐Ad vectors further as a therapeutic gene‐delivery platform to implement GBM phase I clinical trials.


Clinical Cancer Research | 2011

Engineering the brain tumor microenvironment enhances the efficacy of dendritic cell vaccination: implications for clinical trial design.

Yohei Mineharu; Gwendalyn D. King; Akm Ghulam Muhammad; Serguei Bannykh; Kurt M. Kroeger; Chunyan Liu; Pedro R. Lowenstein; Maria G. Castro

Purpose: Glioblastoma multiforme (GBM) is a deadly primary brain tumor. Clinical trials for GBM using dendritic cell (DC) vaccination resulted in antitumor immune responses. Herein, we tested the hypothesis that combining in situ (intratumoral) Ad-Flt3L/Ad-TK–mediated gene therapy with DC vaccination would increase therapeutic efficacy and antitumor immunity. Experimental Design: We first assessed the immunogenicity of tumor lysates generated by Ad-TK (+GCV), temozolomide (TMZ), or freeze/thawing cycles (FTC) in a syngeneic brain tumor model. We also assessed phenotypic markers, cytokine release, and phagocytosis of bone marrow–derived DCs generated by fms-like tyrosine kinase 3 ligand (Flt3L) + IL-6 or by granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin (IL) 4. Inhibition of tumor progression and production of anti-GBM antibodies was assessed following vaccination with (i) tumor cell lysates, (ii) DCs generated with either Flt3L/IL-6 or GM-CSF/IL-4 loaded with either Ad-TK/GCV-, TMZ-, or FTC-generated tumor lysates, or (iii) DCs in combination with in situ Ad-Flt3L/Ad-TK gene therapy. Results: DCs loaded with tumor cell lysates generated with either Ad-TK/GCV or TMZ led to increased levels of phagocytosis, therapeutic efficacy, and humoral immune response. In situ immunogene therapy in combination with DC vaccination led to brain tumor regression and long-term survival in about 90% of animals, a significant increase when compared with either therapy alone. Conclusions: Our results indicate that modifying the tumor microenvironment using intratumoral Ad-Flt3L/Ad-TK–mediated gene therapy potentiates therapeutic efficacy and antitumor immunity induced by DC vaccination. These data support novel phase I clinical trials to assess the safety and efficacy of this combined approach. Clin Cancer Res; 17(14); 4705–18. ©2011 AACR.


Neurotherapeutics | 2012

Gene Therapy-Mediated Reprogramming Tumor Infiltrating T Cells Using IL-2 and Inhibiting NF-κB Signaling Improves the Efficacy of Immunotherapy in a Brain Cancer Model

Yohei Mineharu; Akm Ghulam Muhammad; Kader Yagiz; Marianela Candolfi; Kurt M. Kroeger; Weidong Xiong; Mariana Puntel; Chunyan Liu; Eva Levy; Claudia Lugo; Adrina Kocharian; James P. Allison; Michael A. Curran; Pedro R. Lowenstein; Maria G. Castro

Immune-mediated gene therapy using adenovirus expressing Flt3 ligand and thymidine kinase followed by ganciclovir administration (Flt3/TK) effectively elicits tumor regression in preclinical glioma models. Herein, we assessed new strategies to optimize Flt3L/TK therapeutic efficacy in a refractory RG2 orthotopic glioblastoma model. Specifically, we aimed to optimize the therapeutic efficacy of Flt3L/TK treatment in the RG2 model by overexpressing the following genes within the brain tumor microenvironment: 1) a TK mutant with enhanced cytotoxicity (SR39 mutant TK), 2) Flt3L-IgG fusion protein that has a longer half-life, 3) CD40L to stimulate DC maturation, 4) T helper cell type 1 polarizing dendritic cell cytokines interleukin-12 or C-X-C motif ligand 10 chemokine (CXCL)-10, 5) C-C motif ligand 2 chemokine (CCL2) or C-C motif ligand 3 chemokine (CCL3) to enhance dendritic cell recruitment into the tumor microenvironment, 6) T helper cell type 1 cytokines interferon-γ or interleukin-2 to enhance effector T-cell functions, and 7) IκBα or p65RHD (nuclear factor kappa-B [NF-κB] inhibitors) to suppress the function of Foxp3+ Tregs and enhanced effector T-cell functions. Anti-tumor immunity and tumor specific effector T-cell functions were assessed by cytotoxic T lymphocyte assay and intracellular IFN-γ staining. Our data showed that overexpression of interferon-γ or interleukin-2, or inhibition of the nuclear factor kappa-B within the tumor microenvironment, enhanced cytotoxic T lymphocyte-mediated immune responses and successfully extended the median survival of rats bearing intracranial RG2 when combined with Flt3L/TK. These findings indicate that enhancement of T-cell functions constitutes a critical therapeutic target to overcome immune evasion and enhance therapeutic efficacy for brain cancer. In addition, our study provides novel targets to be used in combination with immune-therapeutic strategies for glioblastoma, which are currently being tested in the clinic.


Journal of Virology | 2010

A Novel Bicistronic High-Capacity Gutless Adenovirus Vector That Drives Constitutive Expression of Herpes Simplex Virus Type 1 Thymidine Kinase and Tet-Inducible Expression of Flt3L for Glioma Therapeutics

Mariana Puntel; Akm Ghulam Muhammad; Marianela Candolfi; Alireza Salem; Kader Yagiz; Catherine Farrokhi; Kurt M. Kroeger; Weidong Xiong; James F. Curtin; Chunyan Liu; Niyati Bondale; Jonathan Lerner; Robert N. Pechnick; Donna Palmer; Philip Ng; Pedro R. Lowenstein; Maria G. Castro

ABSTRACT Glioblastoma multiforme (GBM) is a deadly primary brain tumor. Conditional cytotoxic/immune-stimulatory gene therapy (Ad-TK and Ad-Flt3L) elicits tumor regression and immunological memory in rodent GBM models. Since the majority of patients enrolled in clinical trials would exhibit adenovirus immunity, which could curtail transgene expression and therapeutic efficacy, we used high-capacity adenovirus vectors (HC-Ads) as a gene delivery platform. Herein, we describe for the first time a novel bicistronic HC-Ad driving constitutive expression of herpes simplex virus type 1 thymidine kinase (HSV1-TK) and inducible Tet-mediated expression of Flt3L within a single-vector platform. We achieved anti-GBM therapeutic efficacy with no overt toxicities using this bicistronic HC-Ad even in the presence of systemic Ad immunity. The bicistronic HC-Ad-TK/TetOn-Flt3L was delivered into intracranial gliomas in rats. Survival, vector biodistribution, neuropathology, systemic toxicity, and neurobehavioral deficits were assessed for up to 1 year posttreatment. Therapeutic efficacy was also assessed in animals preimmunized against Ads. We demonstrate therapeutic efficacy, with vector genomes being restricted to the brain injection site and an absence of overt toxicities. Importantly, antiadenoviral immunity did not inhibit therapeutic efficacy. These data represent the first report of a bicistronic vector platform driving the expression of two therapeutic transgenes, i.e., constitutive HSV1-TK and inducible Flt3L genes. Further, our data demonstrate no promoter interference and optimum gene delivery and expression from within this single-vector platform. Analysis of the efficacy, safety, and toxicity of this bicistronic HC-Ad vector in an animal model of GBM strongly supports further preclinical testing and downstream process development of HC-Ad-TK/TetOn-Flt3L for a future phase I clinical trial for GBM.


Anti-cancer Agents in Medicinal Chemistry | 2011

Targeted Toxins for Glioblastoma Multiforme: pre-clinical studies and clinical implementation

Marianela Candolfi; Kurt M. Kroeger; Weidong Xiong; Chunyan Liu; Mariana Puntel; Kader Yagiz; Akm Ghulam Muhammad; Yohei Mineharu; David Foulad; Mia Wibowo; Hikmat Assi; Gregory J. Baker; Pedro R. Lowenstein; Maria G. Castro

Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults. GBM is very aggressive due to its poor cellular differentiation and invasiveness, which makes complete surgical resection virtually impossible. Therefore, GBMs invasive nature as well as its intrinsic resistance to current treatment modalities makes it a unique therapeutic challenge. Extensive examination of human GBM specimens has uncovered that these tumors overexpress a variety of receptors that are virtually absent in the surrounding non-neoplastic brain. Human GBMs overexpress receptors for cytokines, growth factors, ephrins, urokinase-type plasminogen activator (uPA), and transferrin, which can be targeted with high specificity by linking their ligands with highly cytotoxic molecules, such as Diptheria toxin and Pseudomonas exotoxin A. We review the preclinical development and clinical translation of targeted toxins for GBM. In view of the clinical experience, we conclude that although these are very promising therapeutic modalities for GBM patients, efforts should be focused on improving the delivery systems utilized in order to achieve better distribution of the immuno-toxins in the tumor/resection cavity. Delivery of targeted toxins using viral vectors would also benefit enormously from improved strategies for local delivery.


Cancer Research | 2011

Abstract 3645: Glioma-derived factors induce the expansion of myeloid derived suppressor cells which mediate immune suppression and tumor progression

Marianela Candolfi; Kader Yagiz; Hikmat Assi; Akm Ghulam Muhammad; Chunyan Liu; David Foulad; Gabrielle Alzadeh; Daniel A. Pisera; Kurt M. Kroeger; Pedro R. Lowenstein; Maria G. Castro

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL Glioblastoma multiforme (GBM) is the most common and aggressive primary brain cancer in adults. In spite of improved standard of care (surgery, chemotherapy and radiation) the median survival of GBM patients remains dismal. GBMs therapeutic challenges include its invasiveness, resistance to therapies, and immune suppression. We and others have shown that immune suppressive cells infiltrate the GBM microenvironment, i.e., regulatory T cells and myeloid derived suppressor cells (MDSC). Expansion of MDSCs can be triggered by factors produced by the tumor cells, which interfere with myelopoiesis and inhibit the maturation of myeloid precursor cells. We aimed to study whether intracranial GBMs release factors to the general circulation that induce expansion of MDSCs and their recruitment into the tumor mass. We first explored the ability of GBM cells to release soluble factors that induce MDSC expansion in vitro. When compared to control 3T3 mouse fibroblasts, conditioned media (CM) from GL26 and M7 brain tumor cells expressed high levels of ligands for the receptor for advanced glycation end products (RAGE), i.e., HMGB1, S100A8 and S100A9, which have been shown to play a critical role in the expansion of MDSCs in several cancer models. Tallying with these findings, CM from GL26 and M7 glioma cells induced an increase in the number of MDSC (Gr-1+/Cd11b+) and an inhibition of dendritic cell maturation in bone marrow cultures in vitro. We then implanted intracranial GL26 tumors in syngeneic C57/Bl6 mice and evaluated the infiltration of MDSCs into the brain tumor mass and their expansion in peripheral lymphoid organs during tumor progression, as well as the capacity of GBM cells to release immunossupressive factors in vivo. An increase in circulating RAGE ligands that correlated with tumor progression was observed in the serum of tumor-bearing mice. Expansion of MDSCs was detected in cervical lymph nodes, spleen and blood 2 weeks after tumor implantation. The presence of Gr-1+/Cd11b+ MDSCs was detected within the tumor by flow cytometry at 21 and 35 (moribund) days after implantation. These cells accounted for as much as 30% of total tumor infiltrating immune cells (CD45+). By Confocal microscopy, we found Gr-1+/CD11b+ MDSCs at the tumor border and within the brain tumor mass, as well as GBM cells expressing S1008 and S100A9 and exhibiting cytoplasmic HMGB1. Multiparameter immunophenotypic analysis of bone marrow showed an increase in hematopoietic stem cells and a reduction in common myeloid progenitors, indicating an inhibition of myeloid cells’ differentiation. Our results suggest that GBM-derived factors stimulate peripheral expansion of MDSCs and their recruitment into the tumor mass, eliciting immune-suppression and GBM progression. Thus, manipulation of MDSCs emerges as an attractive therapeutic target for brain cancer. Supported by grants from NIH/NINDS Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3645. doi:10.1158/1538-7445.AM2011-3645

Collaboration


Dive into the Akm Ghulam Muhammad's collaboration.

Top Co-Authors

Avatar

Kurt M. Kroeger

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chunyan Liu

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kader Yagiz

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

David Foulad

University of California

View shared research outputs
Top Co-Authors

Avatar

Mariana Puntel

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James F. Curtin

Dublin Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge