Network


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

Hotspot


Dive into the research topics where Russell K. Hales is active.

Publication


Featured researches published by Russell K. Hales.


Cancer Biology & Therapy | 2013

Novel Hsp90 inhibitor NVP-AUY922 radiosensitizes prostate cancer cells

Nishant Gandhi; Aaron T. Wild; Sivarajan T. Chettiar; Khaled Aziz; Yoshinori Kato; Rajendra P. Gajula; Russell Williams; Jessica Cades; Anvesh Annadanam; Danny Y. Song; Yonggang Zhang; Russell K. Hales; Joseph M. Herman; Elwood Armour; Theodore L. DeWeese; Edward M. Schaeffer; Phuoc T. Tran

Outcomes for poor-risk localized prostate cancers treated with radiation are still insufficient. Targeting the “non-oncogene” addiction or stress response machinery is an appealing strategy for cancer therapeutics. Heat-shock-protein-90 (Hsp90), an integral member of this machinery, is a molecular chaperone required for energy-driven stabilization and selective degradation of misfolded “client” proteins, that is commonly overexpressed in tumor cells. Hsp90 client proteins include critical components of pathways implicated in prostate cancer cell survival and radioresistance, such as androgen receptor signaling and the PI3K-Akt-mTOR pathway. We examined the effects of a novel non-geldanamycin Hsp90 inhibitor, AUY922, combined with radiation (RT) on two prostate cancer cell lines, Myc-CaP and PC3, using in vitro assays for clonogenic survival, apoptosis, cell cycle distribution, γ-H2AX foci kinetics and client protein expression in pathways important for prostate cancer survival and radioresistance. We then evaluated tumor growth delay and effects of the combined treatment (RT-AUY922) on the PI3K-Akt-mTOR and AR pathways in a hind-flank tumor graft model. We observed that AUY922 caused supra-additive radiosensitization in both cell lines at low nanomolar doses with enhancement ratios between 1.4–1.7 (p < 0.01). RT-AUY922 increased apoptotic cell death compared with either therapy alone, induced G2-M arrest and produced marked changes in client protein expression. These results were confirmed in vivo, where RT-AUY922 combination therapy produced supra-additive tumor growth delay compared with either therapy by itself in Myc-CaP and PC3 tumor grafts (both p < 0.0001). Our data suggest that combined RT-AUY922 therapy exhibits promising activity against prostate cancer cells, which should be investigated in clinical studies.


Molecular Cancer Research | 2013

The Twist box domain is required for Twist1-induced prostate cancer metastasis

Rajendra P. Gajula; Sivarajan T. Chettiar; Russell Williams; Saravanan Thiyagarajan; Yoshinori Kato; Khaled Aziz; Ruoqi Wang; Nishant Gandhi; Aaron T. Wild; Farhad Vesuna; Jinfang Ma; Tarek Salih; Jessica Cades; Elana J. Fertig; Shyam Biswal; Timothy F. Burns; Christine H. Chung; Charles M. Rudin; Joseph M. Herman; Russell K. Hales; Venu Raman; Steven S. An; Phuoc T. Tran

Twist1, a basic helix-loop-helix transcription factor, plays a key role during development and is a master regulator of the epithelial–mesenchymal transition (EMT) that promotes cancer metastasis. Structure–function relationships of Twist1 to cancer-related phenotypes are underappreciated, so we studied the requirement of the conserved Twist box domain for metastatic phenotypes in prostate cancer. Evidence suggests that Twist1 is overexpressed in clinical specimens and correlated with aggressive/metastatic disease. Therefore, we examined a transactivation mutant, Twist1-F191G, in prostate cancer cells using in vitro assays, which mimic various stages of metastasis. Twist1 overexpression led to elevated cytoskeletal stiffness and cell traction forces at the migratory edge of cells based on biophysical single-cell measurements. Twist1 conferred additional cellular properties associated with cancer cell metastasis including increased migration, invasion, anoikis resistance, and anchorage-independent growth. The Twist box mutant was defective for these Twist1 phenotypes in vitro. Importantly, we observed a high frequency of Twist1-induced metastatic lung tumors and extrathoracic metastases in vivo using the experimental lung metastasis assay. The Twist box was required for prostate cancer cells to colonize metastatic lung lesions and extrathoracic metastases. Comparative genomic profiling revealed transcriptional programs directed by the Twist box that were associated with cancer progression, such as Hoxa9. Mechanistically, Twist1 bound to the Hoxa9 promoter and positively regulated Hoxa9 expression in prostate cancer cells. Finally, Hoxa9 was important for Twist1-induced cellular phenotypes associated with metastasis. These data suggest that the Twist box domain is required for Twist1 transcriptional programs and prostate cancer metastasis. Implications: Targeting the Twist box domain of Twist1 may effectively limit prostate cancer metastatic potential. Mol Cancer Res; 11(11); 1387–400. ©2013 AACR.


International Journal of Radiation Oncology Biology Physics | 2013

Hedgehog Pathway Inhibition Radiosensitizes Non-Small Cell Lung Cancers

Jing Zeng; Khaled Aziz; Sivarajan T. Chettiar; Blake T. Aftab; Michael Armour; Rajendra P. Gajula; Nishant Gandhi; Tarek Salih; Joseph M. Herman; John Wong; Charles M. Rudin; Phuoc T. Tran; Russell K. Hales

PURPOSE Despite improvements in chemoradiation, local control remains a major clinical problem in locally advanced non-small cell lung cancer. The Hedgehog pathway has been implicated in tumor recurrence by promoting survival of tumorigenic precursors and through effects on tumor-associated stroma. Whether Hedgehog inhibition can affect radiation efficacy in vivo has not been reported. METHODS AND MATERIALS We evaluated the effects of a targeted Hedgehog inhibitor (HhAntag) and radiation on clonogenic survival of human non-small cell lung cancer lines in vitro. Using an A549 cell line xenograft model, we examined tumor growth, proliferation, apoptosis, and gene expression changes after concomitant HhAntag and radiation. In a transgenic mouse model of Kras(G12D)-induced and Twist1-induced lung adenocarcinoma, we assessed tumor response to radiation and HhAntag by serial micro-computed tomography (CT) scanning. RESULTS In 4 human lung cancer lines in vitro, HhAntag showed little or no effect on radiosensitivity. By contrast, in both the human tumor xenograft and murine inducible transgenic models, HhAntag enhanced radiation efficacy and delayed tumor growth. By use of the human xenograft model to differentiate tumor and stromal effects, mouse stromal cells, but not human tumor cells, showed significant and consistent downregulation of Hedgehog pathway gene expression. This was associated with increased tumor cell apoptosis. CONCLUSIONS Targeted Hedgehog pathway inhibition can increase in vivo radiation efficacy in lung cancer preclinical models. This effect is associated with pathway suppression in tumor-associated stroma. These data support clinical testing of Hedgehog inhibitors as a component of multimodality therapy for locally advanced non-small cell lung cancer.


International Journal of Radiation Oncology Biology Physics | 2014

Patterns of Care Among Patients Receiving Radiation Therapy for Bone Metastases at a Large Academic Institution

Susannah G. Ellsworth; S.R. Alcorn; Russell K. Hales; T.R. McNutt; Theodore L. DeWeese; Thomas J. Smith

PURPOSE This study evaluates outcomes and patterns of care among patients receiving radiation therapy (RT) for bone metastases at a high-volume academic institution. METHODS AND MATERIALS Records of all patients whose final RT course was for bone metastases from April 2007 to July 2012 were identified from electronic medical records. Chart review yielded demographic and clinical data. Rates of complicated versus uncomplicated bone metastases were not analyzed. RESULTS We identified 339 patients whose final RT course was for bone metastases. Of these, 52.2% were male; median age was 65 years old. The most common primary was non-small-cell lung cancer (29%). Most patients (83%) were prescribed ≤10 fractions; 8% received single-fraction RT. Most patients (52%) had a documented goals of care (GOC) discussion with their radiation oncologist; hospice referral rates were higher when patients had such discussions (66% with vs 50% without GOC discussion, P=.004). Median life expectancy after RT was 96 days. Median survival after RT was shorter based on inpatient as opposed to outpatient status at the time of consultation (35 vs 136 days, respectively, P<.001). Hospice referrals occurred for 56% of patients, with a median interval between completion of RT and hospice referral of 29 days and a median hospice stay of 22 days. CONCLUSIONS These data document excellent adherence to American Society for Radiation Oncolology Choosing Wisely recommendation to avoid routinely using >10 fractions of palliative RT for bone metastasis. Nonetheless, single-fraction RT remains relatively uncommon. Participating in GOC discussions with a radiation oncologist is associated with higher rates of hospice referral. Inpatient status at consultation is associated with short survival.


PLOS ONE | 2013

Concurrent versus sequential sorafenib therapy in combination with radiation for hepatocellular carcinoma.

Aaron T. Wild; Nishant Gandhi; Sivarajan T. Chettiar; Khaled Aziz; Rajendra P. Gajula; Russell Williams; Rachit Kumar; Kekoa Taparra; Jing Zeng; Jessica Cades; Esteban Velarde; Siddharth Menon; J.F. Geschwind; David Cosgrove; Timothy M. Pawlik; Anirban Maitra; John Wong; Russell K. Hales; Michael Torbenson; Joseph M. Herman; Phuoc T. Tran

Sorafenib (SOR) is the only systemic agent known to improve survival for hepatocellular carcinoma (HCC). However, SOR prolongs survival by less than 3 months and does not alter symptomatic progression. To improve outcomes, several phase I-II trials are currently examining SOR with radiation (RT) for HCC utilizing heterogeneous concurrent and sequential treatment regimens. Our study provides preclinical data characterizing the effects of concurrent versus sequential RT-SOR on HCC cells both in vitro and in vivo. Concurrent and sequential RT-SOR regimens were tested for efficacy among 4 HCC cell lines in vitro by assessment of clonogenic survival, apoptosis, cell cycle distribution, and γ-H2AX foci formation. Results were confirmed in vivo by evaluating tumor growth delay and performing immunofluorescence staining in a hind-flank xenograft model. In vitro, concurrent RT-SOR produced radioprotection in 3 of 4 cell lines, whereas sequential RT-SOR produced decreased colony formation among all 4. Sequential RT-SOR increased apoptosis compared to RT alone, while concurrent RT-SOR did not. Sorafenib induced reassortment into less radiosensitive phases of the cell cycle through G1-S delay and cell cycle slowing. More double-strand breaks (DSBs) persisted 24 h post-irradiation for RT alone versus concurrent RT-SOR. In vivo, sequential RT-SOR produced the greatest tumor growth delay, while concurrent RT-SOR was similar to RT alone. More persistent DSBs were observed in xenografts treated with sequential RT-SOR or RT alone versus concurrent RT-SOR. Sequential RT-SOR additionally produced a greater reduction in xenograft tumor vascularity and mitotic index than either concurrent RT-SOR or RT alone. In conclusion, sequential RT-SOR demonstrates greater efficacy against HCC than concurrent RT-SOR both in vitro and in vivo. These results may have implications for clinical decision-making and prospective trial design.


International Journal of Molecular Sciences | 2013

Molecularly Targeted Agents as Radiosensitizers in Cancer Therapy—Focus on Prostate Cancer

S.R. Alcorn; Amanda J. Walker; Nishant Gandhi; Amol K. Narang; Aaron T. Wild; Russell K. Hales; Joseph M. Herman; Danny Y. Song; Theodore L. DeWeese; Emmanuel S. Antonarakis; Phuoc T. Tran

As our understanding of the molecular pathways driving tumorigenesis improves and more druggable targets are identified, we have witnessed a concomitant increase in the development and production of novel molecularly targeted agents. Radiotherapy is commonly used in the treatment of various malignancies with a prominent role in the care of prostate cancer patients, and efforts to improve the therapeutic ratio of radiation by technologic and pharmacologic means have led to important advances in cancer care. One promising approach is to combine molecularly targeted systemic agents with radiotherapy to improve tumor response rates and likelihood of durable control. This review first explores the limitations of preclinical studies as well as barriers to successful implementation of clinical trials with radiosensitizers. Special considerations related to and recommendations for the design of preclinical studies and clinical trials involving molecularly targeted agents combined with radiotherapy are provided. We then apply these concepts by reviewing a representative set of targeted therapies that show promise as radiosensitizers in the treatment of prostate cancer.


Cancer Biology & Therapy | 2011

Nelfinavir induces radiation sensitization in pituitary adenoma cells

Jing Zeng; Alfred P. See; Khaled Aziz; Saravanan Thiyagarajan; Tarek Salih; Rajendra P. Gajula; Michael Armour; Jillian Phallen; Stephanie A. Terezakis; Lawrence Kleinberg; Kristen Redmond; Russell K. Hales; Roberto Salvatori; Alfredo Quinones-Hinojosa; Phuoc T. Tran; Michael Lim

Pituitary adenomas with local invasion and high secretory activity remain a therapeutic challenge. The HIV protease inhibitor nelfinavir is a radiosensitizer in multiple tumor models. We tested nelfinavir as a radiosensitizer in pituitary adenoma cells in vitro and in vivo. We examined the effect of nelfinavir with radiation on in vitro cell viability, clonogenic survival, apoptosis, prolactin secretion, cell cycle distribution, and the PI3K-AKT-mTOR pathway. We evaluated tumor growth delay and confirmed nelfinavirs effect on the PI3K-AKT-mTOR pathway in a hind-flank model. Nelfinavir sensitized pituitary adenoma cells to ionizing radiation as shown by viability assays and clonogenic assay with an enhancement ratio of 1.2 (p


Chest | 2013

Novel Applications of an Injectable Radiopaque Hydrogel Tissue Marker for Management of Thoracic Malignancies

Lana de Souza Lawrence; Eric W. Ford; Christopher R. Gilbert; Lonny Yarmus; Avedis Meneshian; David Feller-Kopman; Russell K. Hales

BACKGROUND Radiopaque markers (otherwise known as fiducials) are used clinically to mark sites of biopsy or resection, which aids with targeting of local therapy, including surgery and radiation therapy. We performed a human cadaveric imaging series with a novel, injectable, radiopaque, absorbable hydrogel marker to demonstrate its potential in the management of thoracic malignancies. METHODS Baseline CT imaging was performed on three unfixed cadaveric specimens. Hydrogel marker implants were placed in the submucosa of the esophagus, the mediastinum, and lung parenchyma by an endoscopic approach with real-time endobronchial and esophageal ultrasound guidance. Subpleural implants in peripheral lung parenchyma were also performed through an anterolateral thoracotomy. Postimplant simulation CT imaging, T2-weighted MRI, and cone-beam CT imaging were performed. Gross dissection of the lung parenchyma was used to evaluate localization of the hydrogel. RESULTS Transthoracic and endoscopic marker placement was readily achieved. The hydrogel appeared hyperechoic by ultrasound, hyperenhancing on T2-weighted MRI, and demonstrated radiopacity of ~300 Hounsfield Units in simulation CT imaging and cone-beam CT imaging. Gross dissection of the lung revealed well-localized blebs of hydrogel marker within lung parenchyma. CONCLUSIONS This cadaveric series demonstrates the excellent visibility of a radiopaque injectable hydrogel marker in the human thorax by multiple common imaging techniques. The hydrogel marker forms a well-localized bleb within tissue, which can assist with triangulation of disease during minimally invasive thoracic surgery. Esophageal applications include radiographic delineation of tumor defined by endoscopy and image guidance for radiotherapy. Future in vivo studies are warranted because radiopaque injectable compounds are promising alternatives to metal fiducials.


Current Molecular Medicine | 2012

Tissue Biomarkers for Prostate Cancer Radiation Therapy

Phuoc T. Tran; Russell K. Hales; Jing Zeng; Khaled Aziz; Tarek Salih; Rajendra P. Gajula; Sivarajan T. Chettiar; Nishant Gandhi; Aaron T. Wild; Rachit Kumar; Joseph M. Herman; Danny Y. Song; Theodore L. DeWeese

Prostate cancer is the most common cancer and second leading cause of cancer deaths among men in the United States. Most men have localized disease diagnosed following an elevated serum prostate specific antigen test for cancer screening purposes. Standard treatment options consist of surgery or definitive radiation therapy directed by clinical factors that are organized into risk stratification groups. Current clinical risk stratification systems are still insufficient to differentiate lethal from indolent disease. Similarly, a subset of men in poor risk groups need to be identified for more aggressive treatment and enrollment into clinical trials. Furthermore, these clinical tools are very limited in revealing information about the biologic pathways driving these different disease phenotypes and do not offer insights for novel treatments which are needed in men with poor-risk disease. We believe molecular biomarkers may serve to bridge these inadequacies of traditional clinical factors opening the door for personalized treatment approaches that would allow tailoring of treatment options to maximize therapeutic outcome. We review the current state of prognostic and predictive tissue-based molecular biomarkers which can be used to direct localized prostate cancer treatment decisions, specifically those implicated with definitive and salvage radiation therapy.


Clinical Cancer Research | 2016

Androgen deprivation followed by acute androgen stimulation selectively sensitizes AR-positive prostate cancer cells to ionizing radiation

Mohammad Hedayati; Michael C. Haffner; Jonathan B. Coulter; Raju R. Raval; Yonggang Zhang; Haoming Zhou; O.Y. Mian; Emma J. Knight; Nina Razavi; Susan L. Dalrymple; John T. Isaacs; Aileen Santos; Russell K. Hales; William G. Nelson; Srinivasan Yegnasubramanian; Theodore L. DeWeese

Purpose: The current standard of care for patients with locally advanced prostate cancer is a combination of androgen deprivation and radiation therapy. Radiation is typically given with androgen suppression when testosterone levels are at their nadir. Recent reports have shown that androgen stimulation of androgen-deprived prostate cancer cells leads to formation of double-strand breaks (DSB). Here, we exploit this finding and investigate the extent and timing of androgen-induced DSBs and their effect on tumor growth following androgen stimulation in combination with ionizing radiation (IR). Experimental Design: Androgen-induced DNA damage was assessed by comet assays and γH2A.X foci formation. Effects of androgen stimulation and radiation were determined in vitro and in vivo with xenograft models. Results: We document that androgen treatment of androgen-deprived prostate cancer cell lines resulted in a dose- and time-dependent induction of widespread DSBs. Generation of these breaks was dependent on androgen receptor and topoisomerase II beta but not on cell-cycle progression. In vitro models demonstrated a synergistic interaction between IR and androgen stimulation when IR is given at a time point corresponding with high levels of androgen-induced DSB formation. Furthermore, in vivo studies showed a significant improvement in tumor growth delay when radiation was given shortly after androgen repletion in castrated mice. Conclusions: These results suggest a potential cooperative effect and improved tumor growth delay with androgen-induced DSBs and radiation with implications for improving the therapeutic index of prostate cancer radiation therapy. Clin Cancer Res; 22(13); 3310–9. ©2016 AACR. See related commentary by Chua and Bristow, p. 3124

Collaboration


Dive into the Russell K. Hales's collaboration.

Top Co-Authors

Avatar

Phuoc T. Tran

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

T.R. McNutt

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

S.R. Alcorn

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph M. Herman

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Khaled Aziz

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

John Wong

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Nishant Gandhi

Johns Hopkins University

View shared research outputs
Researchain Logo
Decentralizing Knowledge