Network


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

Hotspot


Dive into the research topics where Joshua D. Schoenfeld is active.

Publication


Featured researches published by Joshua D. Schoenfeld.


Redox biology | 2014

Ketogenic diets as an adjuvant cancer therapy: History and potential mechanism

Bryan G. Allen; Sudershan K. Bhatia; Carryn M. Anderson; Julie M. Eichenberger-Gilmore; Zita A. Sibenaller; Kranti A. Mapuskar; Joshua D. Schoenfeld; John M. Buatti; Douglas R. Spitz; Melissa A. Fath

Cancer cells, relative to normal cells, demonstrate significant alterations in metabolism that are proposed to result in increased steady-state levels of mitochondrial-derived reactive oxygen species (ROS) such as O2•−and H2O2. It has also been proposed that cancer cells increase glucose and hydroperoxide metabolism to compensate for increased levels of ROS. Given this theoretical construct, it is reasonable to propose that forcing cancer cells to use mitochondrial oxidative metabolism by feeding ketogenic diets that are high in fats and low in glucose and other carbohydrates, would selectively cause metabolic oxidative stress in cancer versus normal cells. Increased metabolic oxidative stress in cancer cells would in turn be predicted to selectively sensitize cancer cells to conventional radiation and chemotherapies. This review summarizes the evidence supporting the hypothesis that ketogenic diets may be safely used as an adjuvant therapy to conventional radiation and chemotherapies and discusses the proposed mechanisms by which ketogenic diets may enhance cancer cell therapeutic responses.


Cancer Cell | 2017

O2⋅− and H2O2-Mediated Disruption of Fe Metabolism Causes the Differential Susceptibility of NSCLC and GBM Cancer Cells to Pharmacological Ascorbate

Joshua D. Schoenfeld; Zita A. Sibenaller; Kranti A. Mapuskar; Brett A. Wagner; Kimberly Cramer-Morales; Muhammad Furqan; Sonia Sandhu; Thomas L. Carlisle; Mark C. Smith; Taher Abu Hejleh; Daniel J. Berg; Jun Zhang; John Keech; Kalpaj R. Parekh; Sudershan K. Bhatia; Varun Monga; Kellie L. Bodeker; Logan Ahmann; Sandy Vollstedt; Heather Brown; Erin P.Shanahan Kauffman; Mary E. Schall; Raymond J. Hohl; Gerald H. Clamon; Jeremy D. W. Greenlee; Matthew A. Howard; Michael K. Schultz; Brian J. Smith; Dennis Riley; Frederick E. Domann

Pharmacological ascorbate has been proposed as a potential anti-cancer agent when combined with radiation and chemotherapy. The anti-cancer effects of ascorbate are hypothesized to involve the autoxidation of ascorbate leading to increased steady-state levels of H2O2; however, the mechanism(s) for cancer cell-selective toxicity remain unknown. The current study shows that alterations in cancer cell mitochondrial oxidative metabolism resulting in increased levels of O2⋅- and H2O2 are capable of disrupting intracellular iron metabolism, thereby selectively sensitizing non-small-cell lung cancer (NSCLC) and glioblastoma (GBM) cells to ascorbate through pro-oxidant chemistry involving redox-active labile iron and H2O2. In addition, preclinical studies and clinical trials demonstrate the feasibility, selective toxicity, tolerability, and potential efficacy of pharmacological ascorbate in GBM and NSCLC therapy.


Cancer Cell | 2017

Erratum: O2 ⋅− and H2O2-Mediated Disruption of Fe Metabolism Causes the Differential Susceptibility of NSCLC and GBM Cancer Cells to Pharmacological Ascorbate (Cancer Cell (2017) 32 (2)(268) (S1535610817300624) (10.1016/j.ccell.2017.02.018))

Joshua D. Schoenfeld; Zita A. Sibenaller; Kranti A. Mapuskar; Brett A. Wagner; Kimberly Cramer-Morales; Muhammad Furqan; Sonia Sandhu; Thomas L. Carlisle; Mark C. Smith; Taher Abu Hejleh; Daniel J. Berg; Jun Zhang; John Keech; Kalpaj R. Parekh; Sudershan K. Bhatia; Varun Monga; Kellie L. Bodeker; Logan Ahmann; Sandy Vollstedt; Heather Brown; Erin P.Shanahan Kauffman; Mary E. Schall; Raymond J. Hohl; Gerald H. Clamon; Jeremy D. W. Greenlee; Matthew A. Howard; Michael K. Schultz; Brian J. Smith; Dennis P. Riley; Frederick E. Domann

Joshua D. Schoenfeld1, Zita A. Sibenaller1, Kranti A. Mapuskar1, Brett A. Wagner1, Kimberly L. Cramer-Morales1, Muhammad Furqan2, Sonia Sandhu2, Thomas L. Carlisle2, Mark C. Smith1, Taher Abu Hejleh2, Daniel J. Berg2, Jun Zhang2, John Keech3, Kalpaj R. Parekh3, Sudershan Bhatia1, Varun Monga2, Kellie L. Bodeker1, Logan Ahmann1, Sandy Vollstedt1, Heather Brown1, Erin P. Shanahan Kauffman2, Mary E. Schall2, Ray J. Hohl2, Gerald H. Clamon2, Jeremy D. Greenlee4, Matthew A. Howard4, Michael K. Shultz5, Brian J. Smith6, Dennis P. Riley7, Frederick E. Domann1, Joseph J. Cullen3, Garry R. Buettner1, John M. Buatti1, Douglas R. Spitz1,*,#, and Bryan G. Allen1,* 1Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, 52242, USA


Redox biology | 2018

Augmentation of intracellular iron using iron sucrose enhances the toxicity of pharmacological ascorbate in colon cancer cells

Kristin E. Brandt; Kelly C. Falls; Joshua D. Schoenfeld; Samuel N. Rodman; Zhimin Gu; Fenghuang Zhan; Joseph J. Cullen; Brett A. Wagner; Garry R. Buettner; Bryan G. Allen; Daniel J. Berg; Douglas R. Spitz; Melissa A. Fath

Pharmacological doses (> 1 mM) of ascorbate (a.k.a., vitamin C) have been shown to selectively kill cancer cells through a mechanism that is dependent on the generation of H2O2 at doses that are safely achievable in humans using intravenous administration. The process by which ascorbate oxidizes to form H2O2 is thought to be mediated catalytically by redox active metal ions such as iron (Fe). Because intravenous iron sucrose is often administered to colon cancer patients to help mitigate anemia, the current study assessed the ability of pharmacological ascorbate to kill colon cancer cells in the presence and absence of iron sucrose. In vitro survival assays showed that 10 mM ascorbate exposure (2 h) clonogenically inactivated 40–80% of exponentially growing colon cancer cell lines (HCT116 and HT29). When the H2O2 scavenging enzyme, catalase, was added to the media, or conditionally over-expressed using a doxycycline inducible vector, the toxicity of pharmacological ascorbate was significantly blunted. When colon cancer cells were treated in the presence or absence of 250 µM iron sucrose, then rinsed, and treated with 10 mM ascorbate, the cells demonstrated increased levels of labile iron that resulted in significantly increased clonogenic cell killing, compared to pharmacological ascorbate alone. Interestingly, when colon cancer cells were treated with iron sucrose for 1 h and then 10 mM ascorbate was added to the media in the continued presence of iron sucrose, there was no enhancement of toxicity despite similar increases in intracellular labile iron. The combination of iron chelators, deferoxamine and diethylenetriaminepentaacetic acid, significantly inhibited the toxicity of either ascorbate alone or ascorbate following iron sucrose. These observations support the hypothesis that increasing intracellular labile iron pools, using iron sucrose, can be used to increase the toxicity of pharmacological ascorbate in human colon cancer cells by a mechanism involving increased generation of H2O2.


Cancer Research | 2017

Mitochondrial Superoxide Increases Age-Associated Susceptibility of Human Dermal Fibroblasts to Radiation and Chemotherapy

Kranti A. Mapuskar; Kyle H. Flippo; Joshua D. Schoenfeld; Dennis Riley; Stefan Strack; Taher Abu-Hejleh; Muhammad Furqan; Varun Monga; Frederick E. Domann; John M. Buatti; Prabhat C. Goswami; Douglas R. Spitz; Bryan G. Allen

Elderly cancer patients treated with ionizing radiation (IR) or chemotherapy experience more frequent and greater normal tissue toxicity relative to younger patients. The current study demonstrates that exponentially growing fibroblasts from elderly (old) male donor subjects (70, 72, and 78 years) are significantly more sensitive to clonogenic killing mediated by platinum-based chemotherapy and IR (∼70%-80% killing) relative to young fibroblasts (5 months and 1 year; ∼10%-20% killing) and adult fibroblasts (20 years old; ∼10%-30% killing). Old fibroblasts also displayed significantly increased (2-4-fold) steady-state levels of O2•-, O2 consumption, and mitochondrial membrane potential as well as significantly decreased (40%-50%) electron transport chain (ETC) complex I, II, IV, V, and aconitase (70%) activities, decreased ATP levels, and significantly altered mitochondrial structure. Following adenoviral-mediated overexpression of SOD2 activity (5-7-fold), mitochondrial ETC activity and aconitase activity were restored, demonstrating a role for mitochondrial O2•- in these effects. Old fibroblasts also demonstrated elevated levels of endogenous DNA damage that were increased following treatment with IR and chemotherapy. Most importantly, treatment with the small-molecule, superoxide dismutase mimetic (GC4419; 0.25 μmol/L) significantly mitigated the increased sensitivity of old fibroblasts to IR and chemotherapy and partially restored mitochondrial function without affecting IR or chemotherapy-induced cancer cell killing. These results support the hypothesis that age-associated increased O2•- and resulting DNA damage mediate the increased susceptibility of old fibroblasts to IR and chemotherapy that can be mitigated by GC4419. Cancer Res; 77(18); 5054-67. ©2017 AACR.


Free Radical Biology and Medicine | 2017

D-penicillamine combined with inhibitors of hydroperoxide metabolism enhances lung and breast cancer cell responses to radiation and carboplatin via H2O2-mediated oxidative stress

Sebastian Sciegienka; Shane R. Solst; Kelly C. Falls; Joshua D. Schoenfeld; Adrienne R. Klinger; Natalie L. Ross; Samuel N. Rodman; Douglas R. Spitz; Melissa A. Fath

Abstract D‐penicillamine (DPEN), a copper chelator, has been used in the treatment of Wilsons disease, cystinuria, and rheumatoid arthritis. Recent evidence suggests that DPEN in combination with biologically relevant copper (Cu) concentrations generates H2O2 in cancer cell cultures, but the effects of this on cancer cell responses to ionizing radiation and chemotherapy are unknown. Increased steady‐state levels of H2O2 were detected in MB231 breast and H1299 lung cancer cells following treatment with DPEN (100 &mgr;M) and copper sulfate (15 &mgr;M). Clonogenic survival demonstrated that DPEN‐induced cancer cell toxicity was dependent on Cu and was significantly enhanced by depletion of glutathione [using buthionine sulfoximine (BSO)] as well as inhibition of thioredoxin reductase [using Auranofin (Au)] prior to exposure. Treatment with catalase inhibited DPEN toxicity confirming H2O2 as the toxic species. Furthermore, pretreating cancer cells with iron sucrose enhanced DPEN toxicity while treating with deferoxamine, an Fe chelator that inhibits redox cycling, inhibited DPEN toxicity. Importantly, DPEN also demonstrated selective toxicity in human breast and lung cancer cells, relative to normal untransformed human lung or mammary epithelial cells and enhanced cancer cell killing when combined with ionizing radiation or carboplatin. Consistent with the selective cancer cell toxicity, normal untransformed human lung epithelial cells had significantly lower labile iron pools than lung cancer cells. These results support the hypothesis that DPEN mediates selective cancer cell killing as well as radio‐chemo‐sensitization by a mechanism involving metal ion catalyzed H2O2‐mediated oxidative stress and suggest that DPEN could be repurposed as an adjuvant in conventional cancer therapy. Graphical abstract Figure. No Caption available. HighlightsDPEN+Cu at physiologic concentrations increase H2O2 levels in cancer cells.DPENs clonogenic toxicity is enhanced using auranofin and buthionine sulfoximine.DPEN+Cu treatment is more toxic to cancer cells than to normal epithelial cells.DPEN toxicity in correlated with intracellular labile iron pools.Labile iron pools are higher in cancer cells verses normal lung epithelial cells.


Redox biology | 2018

Redox active metals and H2O2 mediate the increased efficacy of pharmacological ascorbate in combination with gemcitabine or radiation in pre-clinical sarcoma models

Joshua D. Schoenfeld; Zita A. Sibenaller; Kranti A. Mapuskar; Megan D. Bradley; Brett A. Wagner; Garry R. Buettner; Varun Monga; Mohammed M. Milhem; Douglas R. Spitz; Bryan G. Allen

Soft tissue sarcomas are a histologically heterogeneous group of rare mesenchymal cancers for which treatment options leading to increased overall survival have not improved in over two decades. The current study shows that pharmacological ascorbate (systemic high dose vitamin C achieving ≥ 20 mM plasma levels) is a potentially efficacious and easily integrable addition to current standard of care treatment strategies in preclinical models of fibrosarcoma and liposarcoma both in vitro and in vivo. Furthermore, enhanced ascorbate-mediated toxicity and DNA damage in these sarcoma models were found to be dependent upon H2O2 and intracellular labile iron. Together, these data support the hypothesis that pharmacological ascorbate may represent an easily implementable and non-toxic addition to conventional sarcoma therapies based on taking advantage of fundamental differences in cancer cell oxidative metabolism.


Redox biology | 2018

Corrigendum to: Augmentation of intracellular iron using iron sucrose enhances the toxicity of pharmacological ascorbate in colon cancer cells [Redox Biol. (2018) 82–87]

Kristin E. Brandt; Kelly C. Falls; Joshua D. Schoenfeld; Samuel N. Rodman; Zhimin Gu; Fenghuang Zhan; Joseph J. Cullen; Brett A. Wagner; Garry R. Buettner; Bryan G. Allen; Daniel J. Berg; Douglas R. Spitz; Melissa A. Fath


Free Radical Biology and Medicine | 2016

Pharmacological Ascorbate in Combination with Standard-Of-Care Radio-Chemotherapy Enhances Tumor Response in an Orthotopic Sarcoma Model

Joshua D. Schoenfeld; Zita A. Sibenaller; Kranti A. Mapuskar; Brett A. Wagner; Benjamin J. Miller; Varun Monga; Mohammed M. Milhem; Douglas R. Spitz; Bryan G. Allen


Free Radical Biology and Medicine | 2015

150 – The Selective Toxicity of Pharmacological Ascorbate Is Mediated by Alterations in Iron Metabolism

Joshua D. Schoenfeld; Zita A. Sibenaller; Claire M. Doskey; Kranti A. Mapuskar; Kalpaj R. Parekh; John Keech; Joseph J. Cullen; Garry R. Buettner; Douglas R. Spitz; Bryan G. Allen

Collaboration


Dive into the Joshua D. Schoenfeld's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge