Network


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

Hotspot


Dive into the research topics where Tarun K. Garg is active.

Publication


Featured researches published by Tarun K. Garg.


Molecular Cancer Therapeutics | 2009

Combinatorial efficacy of anti-CS1 monoclonal antibody elotuzumab (HuLuc63) and bortezomib against multiple myeloma

Frits van Rhee; Susann Szmania; Myles Dillon; Anne van Abbema; Xin Li; Mary K. Stone; Tarun K. Garg; Jumei Shi; Amberly Moreno-Bost; Rui Yun; Balaji Balasa; Bishwa Ganguly; Debra T. Chao; Audie Rice; Fenghuang Zhan; John D. Shaughnessy; Bart Barlogie; Shmuel Yaccoby; Daniel E. H. Afar

Monoclonal antibody (mAb) therapy for multiple myeloma, a malignancy of plasma cells, has not been clinically efficacious in part due to a lack of appropriate targets. We recently reported that the cell surface glycoprotein CS1 (CD2 subset 1, CRACC, SLAMF7, CD319) was highly and universally expressed on myeloma cells while having restricted expression in normal tissues. Elotuzumab (formerly known as HuLuc63), a humanized mAb targeting CS1, is currently in a phase I clinical trial in relapsed/refractory myeloma. In this report we investigated whether the activity of elotuzumab could be enhanced by bortezomib, a reversible proteasome inhibitor with significant activity in myeloma. We first showed that elotuzumab could induce patient-derived myeloma cell killing within the bone marrow microenvironment using a SCID-hu mouse model. We next showed that CS1 gene and cell surface protein expression persisted on myeloma patient-derived plasma cells collected after bortezomib administration. In vitro bortezomib pretreatment of myeloma targets significantly enhanced elotuzumab-mediated antibody-dependent cell-mediated cytotoxicity, both for OPM2 myeloma cells using natural killer or peripheral blood mononuclear cells from healthy donors and for primary myeloma cells using autologous natural killer effector cells. In an OPM2 myeloma xenograft model, elotuzumab in combination with bortezomib exhibited significantly enhanced in vivo antitumor activity. These findings provide the rationale for a clinical trial combining elotuzumab and bortezomib, which will test the hypothesis that combining both drugs would result in enhanced immune lysis of myeloma by elotuzumab and direct targeting of myeloma by bortezomib. [Mol Cancer Ther 2009;8(9):2616–24]


British Journal of Haematology | 2008

Infusion of haplo-identical killer immunoglobulin-like receptor ligand mismatched NK cells for relapsed myeloma in the setting of autologous stem cell transplantation

Jumei Shi; Guido Tricot; Susann Szmania; Nancy Rosen; Tarun K. Garg; Priyangi A Malaviarachchi; Amberly Moreno; Bo Dupont; Katharine C. Hsu; Lee Ann Baxter-Lowe; Michele Cottler-Fox; John D. Shaughnessy; Bart Barlogie; Frits van Rhee

Killer immunoglobulin‐like receptor (KIR)‐ligand mismatched natural killer (NK) cells play a key role in achieving durable remission after haplo‐identical transplantation for acute myeloid leukaemia. We investigated the feasibility of transfusing haplo‐identical, T‐cell depleted, KIR‐ligand mismatched NK cells, after conditioning therapy with melphalan and fludarabine, to patients with advanced multiple myeloma (MM) followed by delayed rescue with autologous stem cells. No graft‐versus‐host disease or failure of autologous stem cells to engraft was observed. There was significant variation in the number of allo‐reactive NK cells transfused. However, all NK products containing allo‐reactive NK cells killed the NK cell target K562, the MM cell line U266, and recipient MM cells when available. Post NK cell infusion there was a rise in endogenous interleukin‐15 accompanied by increasing donor chimaerism. Donor chimaerism was eventually lost, which correlated with the emergence of potent host anti‐donor responses indicating that the immunosuppressive properties of the conditioning regimen require further optimization. Further, blocking of inhibitory KIR‐ligands with anti‐human leucocyte antigen antibody substantially enhanced killing of MM cells thus highlighting the potential for modulating NK/MM cell interaction. Encouragingly, 50% of patients achieved (near) complete remission. These data set the stage for future studies of KIR‐ligand mismatched NK cell therapy in the autologous setting.


Cytotherapy | 2012

Large-scale ex vivo expansion and characterization of natural killer cells for clinical applications

Natalia Lapteva; April G. Durett; Jiali Sun; Lisa Rollins; Leslie L. Huye; Jian Fang; Varada Dandekar; Zhuyong Mei; Kimberley Jackson; Juan F. Vera; Jun Ando; Minhtran Ngo; Elaine Coustan-Smith; Dario Campana; Susann Szmania; Tarun K. Garg; Amberly Moreno-Bost; Frits Vanrhee; Adrian P. Gee; Cliona M. Rooney

BACKGROUND AIMS Interest in natural killer (NK) cell-based immunotherapy has resurged since new protocols for the purification and expansion of large numbers of clinical-grade cells have become available. METHODS We have successfully adapted a previously described NK expansion method that uses K562 cells expressing interleukin (IL)-15 and 4-1 BB Ligand (BBL) (K562-mb15-41BBL) to grow NK cells in novel gas-permeable static cell culture flasks (G-Rex). RESULTS Using this system we produced up to 19 × 10(9) functional NK cells from unseparated apheresis products, starting with 15 × 10(7) CD3(-) CD56 (+) NK cells, within 8-10 days of culture. The G-Rex yielded a higher fold expansion of NK cells than conventional gas-permeable bags and required no cell manipulation or feeding during the culture period. We also showed that K562-mb15-41BBL cells up-regulated surface HLA class I antigen expression upon stimulation with the supernatants from NK cultures and stimulated alloreactive CD8 (+) T cells within the NK cultures. However, these CD3 (+) T cells could be removed successfully using the CliniMACS system. We describe our optimized NK cell cryopreservation method and show that the NK cells are viable and functional even after 12 months of cryopreservation. CONCLUSIONS We have successfully developed a static culture protocol for large-scale expansion of NK cells in the gas permeable G-Rex system under good manufacturing practice (GMP) conditions. This strategy is currently being used to produce NK cells for cancer immunotherapy.


BMC Ophthalmology | 2003

Oxidative stress causes ERK phosphorylation and cell death in cultured retinal pigment epithelium: Prevention of cell death by AG126 and 15-deoxy-delta 12, 14-PGJ2

Tarun K. Garg; Jason Y. Chang

BackgroundThe retina, which is exposed to both sunlight and very high levels of oxygen, is exceptionally rich in polyunsaturated fatty acids, which makes it a favorable environment for the generation of reactive oxygen species. The cytotoxic effects of hydrogen peroxide (H2O2) induced oxidative stress on retinal pigment epithelium were characterized in this study.MethodsThe MTT cell viability assay, Texas-Red phalloidin staining, immunohistochemistry and Western blot analysis were used to assess the effects of oxidative stress on primary human retinal pigment epithelial cell cultures and the ARPE-19 cell line.ResultsThe treatment of retinal pigment epithelial cells with H2O2 caused a dose-dependent decrease of cellular viability, which was preceded by a significant cytoskeletal rearrangement, activation of the Extracellular signal-Regulated Kinase, lipid peroxidation and nuclear condensation. This cell death was prevented partially by the prostaglandin derivative, 15d-PGJ2 and by the protein kinase inhibitor, AG126.Conclusion15d-PGJ2 and AG126 may be useful pharmacological tools in the future capable of preventing oxidative stress induced RPE cell death in human ocular diseases.


Haematologica | 2012

Highly activated and expanded natural killer cells for multiple myeloma immunotherapy

Tarun K. Garg; Susann Szmania; Junaid Khan; Antje Hoering; Paul Malbrough; Amberly Moreno-Bost; Amy D Greenway; Joshuah D Lingo; Xin Li; Shmuel Yaccoby; Larry J. Suva; Brian Storrie; Guido Tricot; Dario Campana; John D. Shaughnessy; Bijay Nair; William T. Bellamy; Joshua Epstein; Bart Barlogie; Frits van Rhee

Background Patients with gene expression profiling-defined high-risk myeloma in relapse have poor outcomes with current therapies. We tested whether natural killer cells expanded by co-culture with K562 cells transfected with 41BBL and membrane-bound interleukin-15 could kill myeloma cells with a high-risk gene expression profile in vitro and in a unique model which recapitulates human myeloma. Design and Methods OPM2 and high-risk primary myeloma tumors were grown in human fetal bone implanted into non-obese diabetic severe combined immunodeficiency mice with a deficient interleukin-2 receptor gamma chain. These mice are devoid of endogenous natural killer and T-cell activity and were used to determine whether adoptively transferred expanded natural killer cells could inhibit myeloma growth and myeloma-associated bone destruction. Results Natural killer cells from healthy donors and myeloma patients expanded a median of 804- and 351-fold, respectively, without significant T-cell expansion. Expanded natural killer cells killed both allogeneic and autologous primary myeloma cells avidly via a perforin-mediated mechanism in which the activating receptor NKG2D, natural cytotoxicity receptors, and DNAX-accessory molecule-1 played a central role. Adoptive transfer of expanded natural killer cells inhibited the growth of established OPM2 and high-risk primary myeloma tumors grown in the murine model. The transferred, expanded natural killer cells proliferated in vivo in an interleukin-2 dose-dependent fashion, persisted up to 4 weeks, were readily detectable in the human bone, inhibited myeloma growth and protected bone from myeloma-induced osteolysis. Conclusions These studies provide the rationale for testing expanded natural killer cells in humans.


Journal of Neuroimmunology | 2006

Methylmercury causes oxidative stress and cytotoxicity in microglia: Attenuation by 15-deoxy-delta 12, 14-Prostaglandin J2

Tarun K. Garg; Jason Y. Chang

Methylmercury (MeHg) causes severe neurological disorders in the central nervous system. This study focused on the effects of MeHg on microglia, macrophage-like cells that reside in the CNS important in neuro-immune interactions. The murine N9 microglial cell line was used in this set of study. MeHg caused reactive oxygen species generation, mitochondrial depolarization and aconitase inactivation, all of which were signs of cellular oxidative stress. MeHg greatly increased microglial IL-6 secretion despite the fact that it severely inhibited protein synthesis. The concentration that caused 50% cell death in 24 h was approximately 9 microM. Pretreatment of microglia with the prostaglandin derivative, 15-deoxy-delta 12, 14-Prostaglandin J2 attenuated MeHg induced cell death. The saving effect did not appear to be mediated through activation of peroxisome proliferator activated receptors (PPAR) since other agonists of these receptors did not prevent MeHg induced microglial death.


Journal of Immunotherapy | 2015

Ex vivo-expanded natural killer cells demonstrate robust proliferation in vivo in high-risk relapsed multiple myeloma patients.

Susann Szmania; Natalia Lapteva; Tarun K. Garg; Amy D Greenway; Joshuah D Lingo; Bijay Nair; Katie L. Stone; Emily Woods; Junaid Khan; Justin Stivers; Susan Panozzo; Dario Campana; William T. Bellamy; Molly Robbins; Joshua Epstein; Shmuel Yaccoby; Sarah Waheed; Adrian P. Gee; Michele Cottler-Fox; Cliona M. Rooney; Bart Barlogie; Frits van Rhee

Highly activated/expanded natural killer (NK) cells can be generated by stimulation with the human leukocyte antigen-deficient cell line K562, genetically modified to express 41BB-ligand and membrane-bound interleukin (IL)15. We tested the safety, persistence, and activity of expanded NK cells generated from myeloma patients (auto-NK) or haploidentical family donors (allo-NK) in heavily pretreated patients with high-risk relapsing myeloma. The preparative regimen comprised bortezomib only or bortezomib and immunosuppression with cyclophosphamide, dexamethasone, and fludarabine. NK cells were shipped overnight either cryopreserved or fresh. In 8 patients, up to 1×108 NK cells/kg were infused on day 0 and followed by daily administrations of IL2. Significant in vivo expansion was observed only in the 5 patients receiving fresh products, peaking at or near day 7, with the highest NK-cell counts in 2 subjects who received cells produced in a high concentration of IL2 (500 U/mL). Seven days after infusion, donor NK cells comprised >90% of circulating leukocytes in fresh allo-NK cell recipients, and cytolytic activity against allogeneic myeloma targets was retained in vitro. Among the 7 evaluable patients, there were no serious adverse events that could be related to NK-cell infusion. One patient had a partial response and in another the tempo of disease progression decreased; neither patient required further therapy for 6 months. In the 5 remaining patients, disease progression was not affected by NK-cell infusion. In conclusion, infusion of large numbers of expanded NK cells was feasible and safe; infusing fresh cells was critical to their expansion in vivo.


BMC Pharmacology | 2004

15-deoxy-delta 12, 14-Prostaglandin J2 prevents reactive oxygen species generation and mitochondrial membrane depolarization induced by oxidative stress

Tarun K. Garg; Jason Y. Chang

BackgroundWith the use of cultured human retinal pigment epithelial cells, we have previously described a number of cellular responses to oxidative stress caused by H2O2. We also demonstrated that the cytotoxicity caused by H2O2 could be prevented by the prostaglandin derivative, 15-deoxy-delta 12, 14-Prostaglandin J2 (15d-PGJ2).ResultsFurther characterization of the experimental system indicated that the half-life of H2O2 in cultures was ~1 hour. At a fixed H2O2 concentration, the cytotoxicity was dependent on the volume of H2O2 solution used in the culture, such that higher volume caused more cytotoxicity. Most cells were committed to die if the culture was treated for 2 hours with a cytotoxic concentration of H2O2. The prostaglandin derivative, 15d-PGJ2, could prevent oxidative damage caused by t-butyl hydroperoxide, in addition to H2O2. Further studies indicated that both H2O2 and tBH caused an increase in reactive oxygen species and depolarization of mitochondrial membrane potential. Pretreatment of cells with 1 μM 15d-PGJ2 led to a modest decrease in reactive oxygen species generation, and a significant restoration of mitochondrial membrane potential.ConclusionThis agent may be used in the future as a pharmacological tool for preventing cellular damage caused by oxidative stress.


PLOS ONE | 2013

Interleukin-6 Receptor Polymorphism Is Prevalent in HIV-negative Castleman Disease and Is Associated with Increased Soluble Interleukin-6 Receptor Levels

Katie L. Stone; Emily Woods; Susann Szmania; Owen Stephens; Tarun K. Garg; Bart Barlogie; John D. Shaughnessy; Brett Hall; Manjula Reddy; Antje Hoering; Emily Hansen; Frits van Rhee

Multicentric Castleman Disease is largely driven by increased signaling in the pathway for the plasma cell growth factor interleukin-6. We hypothesized that interleukin-6/interleukin-6 receptor/gp130 polymorphisms contribute to increased interleukin-6 and/or other components of the interleukin-6 signaling pathway in HIV-negative Castleman Disease patients. The study group was composed of 58 patients and 50 healthy donors of a similar racial/ethnic profile. Of seven polymorphisms chosen for analysis, we observed an increased frequency between patients and controls of the minor allele of interleukin-6 receptor polymorphism rs4537545, which is in linkage disequilibrium with interleukin-6 receptor polymorphism rs2228145. Further, individuals possessing at least one copy of the minor allele of either polymorphism expressed higher levels of soluble interleukin-6 receptor. These elevated interleukin-6 receptor levels may contribute to increased interleukin-6 activity through the trans-signaling pathway. These data suggest that interleukin-6 receptor polymorphism may be a contributing factor in Castleman Disease, and further research is warranted.


Alcohol | 2008

Ethanol exposure of neonatal rats does not increase biomarkers of oxidative stress in isolated cerebellar granule neurons

Cynthia J.M. Kane; Jason Y. Chang; Paula K. Roberson; Tarun K. Garg; L. Han

Oxidative stress is a candidate mechanism for ethanol neuropathology in fetal alcohol spectrum disorders. Oxidative stress often involves production of reactive oxygen species (ROS), deterioration of the mitochondrial membrane potential (MMP), and cell death. Previous studies have produced conflicting results regarding the role of oxidative stress and the benefit of antioxidants in ethanol neuropathology in the developing brain. This study investigated the hypothesis that ethanol neurotoxicity involves production of ROS with negative downstream consequences for MMP and neuron survival. This was modeled in neonatal rats at postnatal day 4 (P4) and P14. It is well established that granule neurons in the rat cerebellar cortex are more vulnerable to ethanol neurotoxicity on P4 than at later ages. Thus, it was hypothesized that ethanol produces more oxidative stress and its negative consequences on P4 than on P14. A novel experimental approach was used in which ethanol was administered to animals in vivo (gavage 6g/kg), granule neurons were isolated 2-24h post-treatment, and ROS production and relative MMP were immediately assessed in the viable cells. Cells were also placed in culture and survival was measured 24h later. The results revealed that ethanol did not induce granule cells to produce ROS, cause deterioration of neuronal MMP, or cause neuron death when compared to vehicle controls. Further, granule neurons from neither P4 nor P14 animals mounted an oxidative response to ethanol. These findings do not support the hypothesis that oxidative stress is obligate to granule neuron death after ethanol exposure in the neonatal rat brain. Other investigators have reached a similar conclusion using either brain homogenates or cell cultures. In this context, it is likely that oxidative stress is not the sole and perhaps not the principal mechanism of ethanol neurotoxicity for cerebellar granule neurons during this stage of brain development.

Collaboration


Dive into the Tarun K. Garg's collaboration.

Top Co-Authors

Avatar

Bart Barlogie

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Frits van Rhee

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Susann Szmania

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Katie L. Stone

California Pacific Medical Center

View shared research outputs
Top Co-Authors

Avatar

Joshua Epstein

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

John D. Shaughnessy

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Amy D Greenway

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amberly Moreno-Bost

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Joshuah D Lingo

University of Arkansas for Medical Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge