455 results on '"Phuoc T. Tran"'
Search Results
2. Prostate-Specific Membrane Antigen PET Response Associates with Metastasis-Free Survival After Stereotactic Ablative Radiation in Oligometastatic Prostate Cancer
- Author
-
Philip Sutera, MD, Matthew P. Deek, MD, Rebecca A. Deek, MS, Ozan Cem Guler, MD, Pervin Hurmuz, MD, Mehmet Reyhan, MD, Steven Rowe, MD, PhD, Noura Radwan, MD, Shirl Dipasquale, MSRN, William T. Hrinivich, PhD, Kathryn Lowe, MA, Lei Ren, PhD, Biren Saraiya, MD, Ronald Ennis, MD, Lara Hathout, MD, Tina Mayer, MD, Theodore L. Deweese, MD, Daniel Y. Song, MD, Ana Kiess, MD, Ezgi Oymak, MD, Kenneth Pienta, MD, Felix Feng, MD, Martin Pomper, MD, PhD, Gokhan Ozyigit, MD, Phuoc T. Tran, MD, PhD, Cem Onal, MD, and Ryan M. Phillips, MD, PhD
- Subjects
Medical physics. Medical radiology. Nuclear medicine ,R895-920 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Purpose: Emerging data suggest that metastasis-directed therapy (MDT) improves outcomes in patients with oligometastatic castration-sensitive prostate cancer (omCSPC). Prostate-specific membrane antigen positron emission tomography (PSMA-PET) can detect occult metastatic disease, and PSMA response has been proposed as a biomarker for treatment response. Herein, we identify and validate a PSMA-PET biomarker for metastasis-free survival (MFS) following MDT in omCSPC. Methods and Materials: We performed an international multi-institutional retrospective study of patients with omCSPC, defined as ≤3 lesions, treated with metastasis-directed stereotactic ablative radiation who underwent PSMA-PET/computed tomography (CT) before and after (median, 6.2 months; range, 2.4-10.9 months) treatment. Pre- and post-MDT PSMA-PET/CT maximum standardized uptake value (SUVmax) was measured for all lesions, and PSMA response was defined as the percent change in SUVmax of the least responsive lesion. PSMA response was both evaluated as a continuous variable and dichotomized into PSMA responders, with a complete/partial response (at least a 30% reduction in SUVmax), and PSMA nonresponders, with stable/progressive disease (less than a 30% reduction in SUVmax). PSMA response was correlated with conventional imaging-defined metastasis-free survival (MFS) via Kaplan-Meier and Cox regression analysis. Results: A total of 131 patients with 261 treated metastases were included in the analysis, with a median follow-up of 29 months (IQR, 18.5-41.3 months). After stereotactic ablative radiation, 70.2% of patients were classified as PSMA responders. Multivariable analysis demonstrated that PSMA response as a continuous variable was associated with a significantly worse MFS (hazard ratio = 1.003; 95% CI, 1.001-1.006; P = .016). Patients classified as PSMA responders were found to have a significantly improved median MFS of 39.9 versus 12 months (P = .001) compared with PSMA nonresponders. Our study is limited as it is a retrospective review of a heterogenous population. Conclusions: After stereotactic ablative radiation, PSMA-PET response appears to be a radiographic biomarker that correlates with MFS in omCSPC. This approach holds promise for guiding clinical management of omCSPC and should be validated in a prospective setting.
- Published
- 2024
- Full Text
- View/download PDF
3. Evaluating potential overuse of surveillance care in cancer survivors
- Author
-
Jennifer Y. Sheng, Claire F. Snyder, Katherine C. Smith, Jennifer DeSanto, Nancy Mayonado, Susan Rall, Sharon White, Amanda L. Blackford, Fabian M. Johnston, Robert L. Joyner Jr., Joan Mischtschuk, Kimberly S. Peairs, Elissa Thorner, Phuoc T. Tran, Antonio C. Wolff, and Youngjee Choi
- Subjects
breast cancer ,clinical cancer research ,clinical observations ,colorectal cancer ,prostate cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Survivorship care plans (SCPs) communicate cancer‐related information from oncology providers to patients and primary care providers. SCPs may limit overuse testing by specifying necessary follow‐up care. From a randomized, controlled trial of SCP delivery, we examined whether cancer‐related tests not specified in SCPs, but conducted after SCP receipt, were appropriate or consistent with overuse. Methods Survivors of breast, colorectal, or prostate cancer treated at urban‐academic or rural‐community health systems were randomized to one of three SCP delivery arms. Tests during 18 months after SCP receipt were classified as consistent with overuse if they were (1) not included in SCPs and (2) on a guideline‐based predetermined list of “not recommended surveillance.” After chart abstraction, physicians performed review and adjudication of potential overuse. Descriptive analyses were conducted of tests consistent with overuse. Negative binomial regression models determined if testing consistent with overuse differed across study arms. Results Among 316 patients (137 breast, 67 colorectal, 112 prostate), 140 individual tests were identified as potential overuse. Upon review, 98 were deemed to be consistent with overuse: 78 tumor markers and 20 imaging tests. The majority of overuse testing was breast cancer‐related (95%). Across sites, 27 patients (9%) received ≥1 test consistent with overuse; most were breast cancer patients (22/27). Exploratory analyses of overuse test frequency by study arm showed no significant difference. Conclusions This analysis identified practice patterns consistent with overuse of surveillance testing and can inform efforts to improve guideline‐concordant care. Future interventions may include individual practice patterns and provider education.
- Published
- 2023
- Full Text
- View/download PDF
4. Cell-free DNA sequencing sheds additional insights on BRCA-altered metastatic castration-resistant prostate cancer
- Author
-
Soha Bazyar, Philip Sutera, Matthew P. Deek, Catherine H. Marshall, and Phuoc T. Tran
- Subjects
Medicine ,Medicine (General) ,R5-920 - Published
- 2023
- Full Text
- View/download PDF
5. Prostate cancer therapy personalization via multi-modal deep learning on randomized phase III clinical trials
- Author
-
Andre Esteva, Jean Feng, Douwe van der Wal, Shih-Cheng Huang, Jeffry P. Simko, Sandy DeVries, Emmalyn Chen, Edward M. Schaeffer, Todd M. Morgan, Yilun Sun, Amirata Ghorbani, Nikhil Naik, Dhruv Nathawani, Richard Socher, Jeff M. Michalski, Mack Roach, Thomas M. Pisansky, Jedidiah M. Monson, Farah Naz, James Wallace, Michelle J. Ferguson, Jean-Paul Bahary, James Zou, Matthew Lungren, Serena Yeung, Ashley E. Ross, NRG Prostate Cancer AI Consortium, Howard M. Sandler, Phuoc T. Tran, Daniel E. Spratt, Stephanie Pugh, Felix Y. Feng, and Osama Mohamad
- Subjects
Computer applications to medicine. Medical informatics ,R858-859.7 - Abstract
Abstract Prostate cancer is the most frequent cancer in men and a leading cause of cancer death. Determining a patient’s optimal therapy is a challenge, where oncologists must select a therapy with the highest likelihood of success and the lowest likelihood of toxicity. International standards for prognostication rely on non-specific and semi-quantitative tools, commonly leading to over- and under-treatment. Tissue-based molecular biomarkers have attempted to address this, but most have limited validation in prospective randomized trials and expensive processing costs, posing substantial barriers to widespread adoption. There remains a significant need for accurate and scalable tools to support therapy personalization. Here we demonstrate prostate cancer therapy personalization by predicting long-term, clinically relevant outcomes using a multimodal deep learning architecture and train models using clinical data and digital histopathology from prostate biopsies. We train and validate models using five phase III randomized trials conducted across hundreds of clinical centers. Histopathological data was available for 5654 of 7764 randomized patients (71%) with a median follow-up of 11.4 years. Compared to the most common risk-stratification tool—risk groups developed by the National Cancer Center Network (NCCN)—our models have superior discriminatory performance across all endpoints, ranging from 9.2% to 14.6% relative improvement in a held-out validation set. This artificial intelligence-based tool improves prognostication over standard tools and allows oncologists to computationally predict the likeliest outcomes of specific patients to determine optimal treatment. Outfitted with digital scanners and internet access, any clinic could offer such capabilities, enabling global access to therapy personalization.
- Published
- 2022
- Full Text
- View/download PDF
6. Author Correction: Prostate cancer therapy personalization via multi-modal deep learning on randomized phase III clinical trials
- Author
-
Andre Esteva, Jean Feng, Douwe van der Wal, Shih-Cheng Huang, Jeffry P. Simko, Sandy DeVries, Emmalyn Chen, Edward M. Schaeffer, Todd M. Morgan, Yilun Sun, Amirata Ghorbani, Nikhil Naik, Dhruv Nathawani, Richard Socher, Jeff M. Michalski, Mack Roach, Thomas M. Pisansky, Jedidiah M. Monson, Farah Naz, James Wallace, Michelle J. Ferguson, Jean-Paul Bahary, James Zou, Matthew Lungren, Serena Yeung, Ashley E. Ross, NRG Prostate Cancer AI Consortium, Howard M. Sandler, Phuoc T. Tran, Daniel E. Spratt, Stephanie Pugh, Felix Y. Feng, and Osama Mohamad
- Subjects
Computer applications to medicine. Medical informatics ,R858-859.7 - Published
- 2023
- Full Text
- View/download PDF
7. A phase II randomized trial of RAdium-223 dichloride and SABR Versus SABR for oligomEtastatic prostate caNcerS (RAVENS)
- Author
-
Hamza Hasan, Matthew P. Deek, Ryan Phillips, Robert F. Hobbs, Reem Malek, Noura Radwan, Ana P. Kiess, Shirl Dipasquale, James Huang, Terry Caldwell, Jessica Leitzel, Danielle Wendler, Hao Wang, Elizabeth Thompson, Jonathan Powell, Sara Dudley, Curtiland Deville, Stephen C. Greco, Daniel Y. Song, Theodore L. DeWeese, Michael A. Gorin, Steven P. Rowe, Sam Denmeade, Mark Markowski, Emmanuel S. Antonarakis, Michael A. Carducci, Mario A. Eisenberger, Martin G. Pomper, Kenneth J. Pienta, Channing J. Paller, and Phuoc T. Tran
- Subjects
Oligometastatic ,Prostate ,Cancer ,Stereotactic ablative radiation (SABR) ,Radium-223 ,Bone ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Metastasis directed therapy (MDT) for patients with oligometastatic disease is associated with improvements in progression free survival (PFS) and overall survival (OS) compared to systemic therapy alone. Additionally, within a prostate-cancer-specific cohort, MDT is able to forestall initiation of androgen deprivation therapy (ADT) in men with hormone-sensitive, oligometastatic prostate cancer (HSOPCa) compared to observation. While MDT appears to be safe and effective in HSOPCa, a large percentage of men will eventually have disease recurrence. Patterns of failure in HSOPCa demonstrate patients tend to have recurrence in the bone following MDT, raising the question of sub-clinically-apparent osseous disease. Radium-223 dichloride is a radiopharmaceutical with structural similarity to calcium, allowing it to be taken up by bone where it emits alpha particles, and therefore might have utility in the treatment of micrometastatic osseous disease. Therefore, the primary goal of the phase II RAVENS trial is to evaluate the efficacy of MDT + radium-223 dichloride in prolonging progression free survival in men with HSOPCa. Methods Patients with HSOPCa and 3 or less metastases with at least 1 bone metastasis will be randomized 1:1 to stereotactic ablative radiation (SABR, also known as stereotactic body radiation therapy (SBRT)) alone vs SABR + radium-223 dichloride with a minimization algorithm to balance assignment by institution, primary intervention, prior hormonal therapy, and PSA doubling time. SABR is delivered in one to five fractions and patients in the SABR + radium-223 dichloride arm will receive six infusions of radium-223 dichloride at four-week intervals. The primary end point is progression free survival. The secondary clinical endpoints include toxicity and quality of life assessments, local control at 12 months, locoregional progression, time to distant progression, time to new metastasis, and duration of response. Discussion The RAVENS trial will be the first described phase II, non-blinded, randomized study to compare SABR +/− radium-223 dichloride in patients with HSOPCa and 3 or less metastases with at least one bone metastasis. The primary hypothesis is that SABR + radium-223 dichloride will increase median progression-free survival from 10 months in the SABR arm to 20 months in the SABR + radium-223 dichloride arm. Trial registrations Clinicaltrials.gov. Identifier: NCT04037358 . Date of Registration: July 30, 2019. Date of First Participant Enrolled: August 9, 2019. Date of Last Approved Amendment: October 16, 2019. Protocol Version: Version 5.
- Published
- 2020
- Full Text
- View/download PDF
8. Online Prostate-Specific Membrane Antigen and Positron Emission Tomography–Guided Radiation Therapy for Oligometastatic Prostate Cancer
- Author
-
William T. Hrinivich, PhD, Ryan Phillips, MD, PhD, Angela J. Da Silva, PhD, Noura Radwan, MD, Michael A. Gorin, MD, Steven P. Rowe, MD, PhD, Kenneth J. Pienta, MD, Martin G. Pomper, MD, PhD, John Wong, PhD, Phuoc T. Tran, MD, PhD, and Ken Kang-Hsin Wang, PhD
- Subjects
Medical physics. Medical radiology. Nuclear medicine ,R895-920 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Purpose: Stereotactic ablative radiation therapy (SABR) for oligometastatic prostate cancer (OMPC) may improve clinical outcomes, but current challenges in intrafraction tracking of multiple small targets limits treatment accuracy. A biology-guided radiation therapy (BgRT) delivery system incorporating positron emission tomography (PET) detectors is being developed to use radiotracer uptake as a biologic fiducial for intrafraction tumor tracking to improve geometric accuracy. This study simulates prostate-specific membrane antigen (PSMA)-directed BgRT using a cohort from our phase II randomized trial of SABR in men with recurrent hormone sensitive OMPC and compares dose distributions to clinical SABR (CSABR). Methods and Materials: A research treatment planning system (RTPS) was used to replan 15 patients imaged with PSMA-targeted 18F-DCFPyL PET/computed tomography and previously treated with CSABR using conventional linear accelerators (linacs). The RTPS models a prototype ring-mounted linac incorporating PET and kilo-voltage computed tomography imaging subsystems and can be used to optimize BgRT plans, as well as research SABR (RSABR) plans, which use the prototype linac without radiotracer guidance. CSABR, RSABR, and BgRT plans were compared in terms of maximum planning target volume (PTV) dose (Dmax), mean dose to proximal organs at risk (DOAR), conformity index, as well as voxel-wise correlation of dose with PET specific uptake values to investigate possible dose-painting effects. Results: RSABR and BgRT plans resulted in mean ± standard deviation increases in Dmax of 4 ± 11% (P = .21) and 18 ± 15% (P < .001) and reductions in DOAR of –20 ± 19% (P
- Published
- 2020
- Full Text
- View/download PDF
9. A phase II randomized placebo-controlled double-blind study of salvage radiation therapy plus placebo versus SRT plus enzalutamide with high-risk PSA-recurrent prostate cancer after radical prostatectomy (SALV-ENZA)
- Author
-
Roche Kapoor, Matthew P. Deek, Riley McIntyre, Natasha Raman, Megan Kummerlowe, Iyah Chen, Matt Gaver, Hao Wang, Sam Denmeade, Tamara Lotan, Channing Paller, Mark Markowski, Michael Carducci, Mario Eisenberger, Tomasz M. Beer, Daniel Y. Song, Theodore L. DeWeese, Jason W. Hearn, Stephen Greco, Curtiland DeVille, Neil B. Desai, Elisabeth I. Heath, Stanley Liauw, Daniel E. Spratt, Arthur Y. Hung, Emmanuel S. Antonarakis, and Phuoc T. Tran
- Subjects
Recurrent prostate cancer ,Salvage radiation therapy (SRT) ,High-risk prostate cancer ,Enzalutamide ,Prostatectomy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background In men with a rising PSA following radical prostatectomy, salvage radiation therapy (SRT) offers a second chance for cure. Hormonal therapy can be combined with SRT in order to increase prostate tumor control, albeit with associated higher rates of treatment side effects. This trial studies the effectiveness of SRT combined with hormonal therapy using a more potent anti-androgen with a favorable side effect profile. Enzalutamide, a next generation selective androgen receptor antagonist, is approved by the Food and Drug Administration for the treatment of metastatic castrate-resistant prostate cancer (CRPC) where it has been shown to improve overall survival in combination with androgen deprivation therapy. The primary objective of this study is to evaluate the efficacy of combination SRT and enzalutamide for freedom-from-PSA-progression. Secondary objectives include time to local recurrence within the radiation field, metastasis-free survival and safety as determined by frequency and severity of adverse events. Methods/design This is a randomized, double-blind, phase II, prospective, multicenter study in adult males with biochemically recurrent prostate cancer following radical prostatectomy. Following registration, enzalutamide 160 mg or placebo by mouth (PO) once daily will be administered for 6 months. Following two months of study drug, external beam radiotherapy to 66.6–70.2 Gray (Gy) will be administered to the prostate bed over 7–8 weeks while continuing daily placebo/enzalutamide. This is followed by two additional months of placebo/enzalutamide. Discussion The SALV-ENZA trial is the first phase II placebo-controlled double-blinded randomized study to test SRT in combination with a next generation androgen receptor antagonist in men with high-risk recurrent prostate cancer after radical prostatectomy. The primary hypothesis of this study is that clinical outcomes will be improved by the addition of enzalutamide compared to standard-of-care SRT alone and pave the path for phase III evaluation of this combination. Trial registrations ClinicaltTrials.gov Identifier: NCT02203695 Date of Registration: 06/16/2014. Date of First Participant Enrollment: 04/16/2015.
- Published
- 2019
- Full Text
- View/download PDF
10. Tumor Treating Fields: At the Crossroads Between Physics and Biology for Cancer Treatment
- Author
-
Francesca A. Carrieri, Caleb Smack, Ismaeel Siddiqui, Lawrence R. Kleinberg, and Phuoc T. Tran
- Subjects
Tumor Treating Fields ,alternating electric fields ,mitosis ,cancer treatment ,TTFields ,centrosome ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Despite extraordinary advances that have been achieved in the last few decades, cancer continues to represent a leading cause of mortality worldwide. Lethal cancer types ultimately become refractory to standard of care approaches; thus, novel effective treatment options are desperately needed. Tumor Treating Fields (TTFields) are an innovative non-invasive regional anti-mitotic treatment modality with minimal systemic toxicity. TTFields are low intensity (1–3 V/cm), intermediate frequency (100–300 kHz) alternating electric fields delivered to cancer cells. In patients, TTFields are applied using FDA-approved transducer arrays, orthogonally positioned on the area surrounding the tumor region, with side effects mostly limited to the skin. The precise molecular mechanism of the anti-tumor effects of TTFields is not well-understood, but preclinical research on TTFields suggests it may act during two phases of mitosis: at metaphase, by disrupting the formation of the mitotic spindle, and at cytokinesis, by dielectrophoretic dislocation of intracellular organelles leading to cell death. This review describes the mechanism of action of TTFields and provides an overview of the most important in vitro studies that investigate the disruptive effects of TTFields in different cancer cells, focusing mainly on anti-mitotic roles. Lastly, we summarize completed and ongoing TTFields clinical trials on a variety of solid tumors.
- Published
- 2020
- Full Text
- View/download PDF
11. Incorporating Radiation Oncology into Immunotherapy: proceedings from the ASTRO-SITC-NCI immunotherapy workshop
- Author
-
Ariel E. Marciscano, Joshua M. Walker, Heather M. McGee, Michelle M. Kim, Charles A. Kunos, Arta M. Monjazeb, Stephen L. Shiao, Phuoc T. Tran, and Mansoor M. Ahmed
- Subjects
Radiation oncology ,Radiation therapy ,Radiotherapy ,Immunotherapy ,Immune checkpoint blockade ,Immune checkpoint inhibitors ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Radiotherapy (RT) has been a fundamental component of the anti-cancer armamentarium for over a century. Approximately half of all cancer patients are treated with radiotherapy during their disease course. Over the two past decades, there has been a growing body of preclinical evidence supporting the immunomodulatory effects of radiotherapy, particularly when combined with immunotherapy, but only anecdotal clinical examples existed until recently. The renaissance of immunotherapy and the recent U.S. Food and Drug Administration (FDA) approval of several immune checkpoint inhibitors (ICIs) and other immuno-oncology (IO) agents in multiple cancers provides the opportunity to investigate how localized radiotherapy can induce systemic immune responses. Early clinical experiences have demonstrated feasibility of this approach but additional preclinical and clinical investigation is needed to understand how RT and immunotherapy can be optimally combined. To address questions that are critical to successful incorporation of radiation oncology into immunotherapy, the American Society for Radiation Oncology (ASTRO), the Society for Immunotherapy of Cancer (SITC) and the National Cancer Institute (NCI) organized a collaborative scientific workshop, Incorporating Radiation Oncology into Immunotherapy, that convened on June 15 and 16 of 2017 at the Natcher Building, NIH Campus in Bethesda, Maryland. This report summarizes key data and highlights from each session.
- Published
- 2018
- Full Text
- View/download PDF
12. A phase II randomized trial of Observation versus stereotactic ablative RadiatIon for OLigometastatic prostate CancEr (ORIOLE)
- Author
-
Noura Radwan, Ryan Phillips, Ashley Ross, Steven P. Rowe, Michael A. Gorin, Emmanuel S. Antonarakis, Curtiland Deville, Stephen Greco, Samuel Denmeade, Channing Paller, Daniel Y. Song, Maximilian Diehn, Hao Wang, Michael Carducci, Kenneth J. Pienta, Martin G. Pomper, Theodore L. DeWeese, Adam Dicker, Mario Eisenberger, and Phuoc T. Tran
- Subjects
Prostate cancer ,Stereotactic body radiation therapy ,Stereotactic ablative radiotherapy ,Oligometastasis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background We describe a randomized, non-blinded Phase II interventional study to assess the safety and efficacy of stereotactic ablative radiotherapy (SABR) for hormone-sensitive oligometastatic prostate adenocarcinoma, and to describe the biology of the oligometastatic state using immunologic, cellular, molecular, and functional imaging correlates. 54 men with oligometastatic prostate adenocarcinoma will be accrued. The primary clinical endpoint will be progression at 6 months from randomization with the hypothesis that SABR to all metastases will forestall progression by disrupting the metastatic process. Secondary clinical endpoints will include local control at 6 months post-SABR, toxicity and quality of life, and androgen deprivation therapy (ADT)-free survival (ADT-FS). Further fundamental analysis of the oligometastatic state with be achieved through correlation with investigational 18F–DCFPyL PET/CT imaging and measurement of circulating tumor cells, circulating tumor DNA, and circulating T-cell receptor repertoires, facilitating an unprecedented opportunity to characterize, in isolation, the effects of SABR on the dynamics of and immunologic response to oligometastatic disease. Methods/design Patients will be randomized 2:1 to SABR or observation with minimization to balance assignment by primary intervention, prior hormonal therapy, and PSA doubling time. Progression after 6 months will be compared using Fisher’s exact test. Hazard ratios and Kaplan-Meier estimates of progression free survival (PFS), ADT free survival (ADT-FS), time to locoregional progression (TTLP) and time to distant progression (TTDP) will be calculated based on an intention-to-treat. Local control will be assessed using Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 criteria. Withdrawal from the study prior to 6 months will be counted as progression. Adverse events will be summarized by type and grade. Quality of life pre- and post- SABR will be measured by Brief Pain Inventory. Discussion The ORIOLE trial is the first randomized, non-blinded Phase II interventional study in the North America evaluating the safety and efficacy of SABR in oligometastatic hormone-sensitive prostate cancer. Leading-edge laboratory and imaging correlates will provide unique insight into the effects of SABR on the oligometastatic state. Trial registrations ClinicalTrials.gov Identifier: NCT02680587. URL of Registry: https://clinicaltrials.gov/show/NCT02680587 Date of Registration: 02/08/2016. Date of First Participant Enrollment: 05/23/2016.
- Published
- 2017
- Full Text
- View/download PDF
13. Complete biochemical response after stereotactic ablative radiotherapy of an isolated prostate cancer pelvic soft tissue recurrence detected by 18F-DCFPyL PET/CT
- Author
-
Ryan M. Phillips, Michael A. Gorin, Steven P. Rowe, Martin G. Pomper, Kenneth J. Pienta, Ashley E. Ross, and Phuoc T. Tran
- Subjects
Prostate cancer ,Local recurrence ,PSMA ,Molecular imaging ,Radiotherapy ,Diseases of the genitourinary system. Urology ,RC870-923 - Published
- 2018
- Full Text
- View/download PDF
14. Author’s view: epithelial plasticity metabolically reprograms normal cells towards a neoplastic-prone state
- Author
-
Hailun Wang and Phuoc T. Tran
- Subjects
epithelial plasticity ,hexosamine biosynthetic pathway ,o-glcnacylation ,epithelial-mesenchymal transition ,oncogene-induced senescence ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
We have uncovered that epithelial plasticity programs metabolically reprogram epithelial lung cells by increasing expression of genes (e.g., glutamine-fructose-6-phosphate transaminase 2 – GFPT2 and UDP-N-acetylglucosamine pyrophosphorylase 1 – UAP1) critical for the hexosamine biosynthetic pathway (HBP) and elevating global protein O-GlcNAcylation – a specific type of glycosylation. We found that increased O-GlcNAcylation could suppress oncogene-induced senescence tumor suppressor pathways that ultimately led to accelerated KrasG12D-driven lung tumorigenesis.
- Published
- 2019
- Full Text
- View/download PDF
15. Contrasting impact of corticosteroids on anti-PD-1 immunotherapy efficacy for tumor histologies located within or outside the central nervous system
- Author
-
Russell Maxwell, Andrew S. Luksik, Tomas Garzon-Muvdi, Alice L. Hung, Eileen S. Kim, Adela Wu, Yuanxuan Xia, Zineb Belcaid, Noah Gorelick, John Choi, Debebe Theodros, Christopher M. Jackson, Dimitrios Mathios, Xiaobu Ye, Phuoc T. Tran, Kristin J. Redmond, Henry Brem, Drew M. Pardoll, Lawrence R. Kleinberg, and Michael Lim
- Subjects
pd-1 ,immunotherapy ,corticosteroid ,dexamethasone ,colon adenocarcinoma ,brain tumor ,glioma ,central nervous system ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Immune checkpoint blockade targeting programmed cell death protein 1 (PD-1) is emerging as an important treatment strategy in a growing list of cancers, yet its clinical benefits are limited to a subset of patients. Further investigation of tumor-intrinsic predictors of response and how extrinsic factors, such as iatrogenic immunosuppression caused by conventional therapies, impact the efficacy of anti-PD-1 therapy are paramount. Given the widespread use of corticosteroids in cancer management and their immunosuppressive nature, this study sought to determine how corticosteroids influence anti-PD-1 responses and whether their effects were dependent on tumor location within the periphery versus central nervous system (CNS), which may have a more limiting immune environment. In well-established anti-PD-1-responsive murine tumor models, corticosteroid therapy resulted in systemic immune effects, including severe and persistent reductions in peripheral CD4+ and CD8 + T cells. Corticosteroid treatment was found to diminish the efficacy of anti-PD-1 therapy in mice bearing peripheral tumors with responses correlating with peripheral CD8/Treg ratio changes. In contrast, in mice bearing intracranial tumors, corticosteroids did not abrogate the benefits conferred by anti-PD-1 therapy. Despite systemic immune changes, anti-PD-1-mediated antitumor immune responses remained intact during corticosteroid treatment in mice bearing intracranial tumors. These findings suggest that anti-PD-1 responses may be differentially impacted by concomitant corticosteroid use depending on tumor location within or outside the CNS. As an immune-specialized site, the CNS may potentially play a protective role against the immunosuppressive effects of corticosteroids, thus sustaining antitumor immune responses mediated by PD-1 blockade.
- Published
- 2018
- Full Text
- View/download PDF
16. Structure-Function Studies of the bHLH Phosphorylation Domain of TWIST1 in Prostate Cancer Cells
- Author
-
Rajendra P. Gajula, Sivarajan T. Chettiar, Russell D. Williams, Katriana Nugent, Yoshinori Kato, Hailun Wang, Reem Malek, Kekoa Taparra, Jessica Cades, Anvesh Annadanam, A-Rum Yoon, Elana Fertig, Beth A. Firulli, Lucia Mazzacurati, Timothy F. Burns, Anthony B. Firulli, Steven S. An, and Phuoc T. Tran
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The TWIST1 gene has diverse roles in development and pathologic diseases such as cancer. TWIST1 is a dimeric basic helix-loop-helix (bHLH) transcription factor existing as TWIST1-TWIST1 or TWIST1-E12/47. TWIST1 partner choice and DNA binding can be influenced during development by phosphorylation of Thr125 and Ser127 of the Thr-Gln-Ser (TQS) motif within the bHLH of TWIST1. The significance of these TWIST1 phosphorylation sites for metastasis is unknown. We created stable isogenic prostate cancer cell lines overexpressing TWIST1 wild-type, phospho-mutants, and tethered versions. We assessed these isogenic lines using assays that mimic stages of cancer metastasis. In vitro assays suggested the phospho-mimetic Twist1-DQD mutation could confer cellular properties associated with pro-metastatic behavior. The hypo-phosphorylation mimic Twist1-AQA mutation displayed reduced pro-metastatic activity compared to wild-type TWIST1 in vitro, suggesting that phosphorylation of the TWIST1 TQS motif was necessary for pro-metastatic functions. In vivo analysis demonstrates that the Twist1-AQA mutation exhibits reduced capacity to contribute to metastasis, whereas the expression of the Twist1-DQD mutation exhibits proficient metastatic potential. Tethered TWIST1-E12 heterodimers phenocopied the Twist1-DQD mutation for many in vitro assays, suggesting that TWIST1 phosphorylation may result in heterodimerization in prostate cancer cells. Lastly, the dual phosphatidylinositide 3-kinase (PI3K)-mammalian target of rapamycin (mTOR) inhibitor BEZ235 strongly attenuated TWIST1-induced migration that was dependent on the TQS motif. TWIST1 TQS phosphorylation state determines the intensity of TWIST1-induced pro-metastatic ability in prostate cancer cells, which may be partly explained mechanistically by TWIST1 dimeric partner choice.
- Published
- 2015
- Full Text
- View/download PDF
17. Erratum to: Agonist anti-GITR monoclonal antibody and stereotactic radiation induce immune-mediated survival advantage in murine intracranial glioma
- Author
-
Christina Jackson, Ada Tam, Sarah Harris-Bookman, Brian Francica, Charles G. Drake, Drew M. Pardoll, Debebe Theodros, Esteban Velarde, Betty Tyler, Xiaobu Ye, Henry Brem, Mark Selby, Dimitrios Mathios, Mira A. Patel, Jennifer E. Kim, Christina M. Kochel, Thomas R. Nirschl, Ali Ghasemzadeh, Christopher C. Jackson, Phuoc T. Tran, Vladimir Coric, and Michael Lim
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2016
- Full Text
- View/download PDF
18. Agonist anti-GITR monoclonal antibody and stereotactic radiation induce immune-mediated survival advantage in murine intracranial glioma
- Author
-
Christina Jackson, Ada Tam, Sarah Harris-Bookman, Brian Francica, Charles G. Drake, Drew M. Pardoll, Debebe Theodros, Esteban Velarde, Betty Tyler, Xiaobu Ye, Henry Brem, Mark Selby, Dimitrios Mathios, Mira A. Patel, Jennifer E. Kim, Christina M. Kochel, Thomas R. Nirschl, Ali Ghasemzadeh, Christopher C. Jackson, Phuoc T. Tran, Vladimir Coric, and Michael Lim
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Glioblastoma (GBM) is a poorly immunogenic neoplasm treated with focused radiation. Immunotherapy has demonstrated synergistic survival effects with stereotactic radiosurgery (SRS) in murine GBM. GITR is a co-stimulatory molecule expressed constitutively on regulatory T-cells and by effector T-cells upon activation. We tested the hypothesis that anti-GITR monoclonal antibody (mAb) and SRS together would confer an immune-mediated survival benefit in glioma using the orthotopic GL261 glioma model.Methods Mice received SRS and anti-GITR 10 days after implantation. The anti-GITR mAbs tested were formatted as mouse IgG1 D265A (anti-GITR (1)) and IgG2a (anti-GITR (2a)) isotypes. Mice were randomized to four treatment groups: (1) control; (2) SRS; (3) anti-GITR; (4) anti-GITR/SRS. SRS was delivered to the tumor in one fraction, and mice were treated with mAb thrice. Mice were euthanized on day 21 to analyze the immunologic profile of tumor, spleen, and tumor draining lymph nodes.Results Anti-GITR (1)/SRS significantly improved survival over either treatment alone (p
- Published
- 2016
- Full Text
- View/download PDF
19. A Phase 1 Trial of Durvalumab in Combination with Bacillus Calmette-Guerin (BCG) or External Beam Radiation Therapy in Patients with BCG-unresponsive Non-muscle-Invasive Bladder Cancer: The Hoosier Cancer Research Network GU16-243 ADAPT-BLADDER Study
- Author
-
Noah M. Hahn, Michael A. O'Donnell, Jason A. Efstathiou, Marianna Zahurak, Gary L. Rosner, Jeff Smith, Max R. Kates, Trinity J. Bivalacqua, Phuoc T. Tran, Daniel Y. Song, Alex S. Baras, Andres Matoso, Woonyoung Choi, Kellie N. Smith, Drew M. Pardoll, Luigi Marchionni, Bridget McGuire, Mary Grace Phelan, Burles A. Johnson, Tanya O'Neal, David J. McConkey, Tracy L. Rose, Marc Bjurlin, Emerson A. Lim, Charles G. Drake, James M. McKiernan, Israel Deutsch, Christopher B. Anderson, Donald L. Lamm, Daniel M. Geynisman, Elizabeth R. Plimack, Mark A. Hallman, Eric M. Horwitz, Essel Al-Saleem, David Y.T. Chen, Richard E. Greenberg, Alexander Kutikov, Gordon Guo, Timothy A. Masterson, Nabil Adra, and Hristos Z. Kaimakliotis
- Subjects
Urology - Published
- 2023
- Full Text
- View/download PDF
20. WNT Pathway Mutations in Metachronous Oligometastatic Castration-Sensitive Prostate Cancer
- Author
-
Philip Sutera, Matthew P. Deek, Kim Van der Eecken, Amol C. Shetty, Jin Hee Chang, Theresa Hodges, Yang Song, Sofie Verbeke, Jo Van Dorpe, Valérie Fonteyne, Bram De Laere, Mark Mishra, Zaker Rana, Jason Molitoris, Matthew Ferris, Ashley Ross, Edward Schaeffer, Nicholas Roberts, Daniel Y. Song, Theodore DeWeese, Kenneth J. Pienta, Emmanuel S. Antonarakis, Piet Ost, and Phuoc T. Tran
- Subjects
Cancer Research ,Radiation ,Oncology ,Radiology, Nuclear Medicine and imaging - Published
- 2023
- Full Text
- View/download PDF
21. External Beam Radiation Therapy With or Without Brachytherapy Boost in Men With Very-High-Risk Prostate Cancer: A Large Multicenter International Consortium Analysis
- Author
-
Sagar A. Patel, Ting Martin Ma, Jessica K. Wong, Bradley J. Stish, Robert T. Dess, Avinash Pilar, Chandana Reddy, Trude B. Wedde, Wolfgang A. Lilleby, Ryan Fiano, Gregory S. Merrick, Richard G. Stock, D. Jeffrey Demanes, Brian J. Moran, Phuoc T. Tran, Daniel J. Krauss, Eyad I. Abu-Isa, Thomas M. Pisansky, C. Richard Choo, Daniel Y. Song, Stephen Greco, Curtiland Deville, Theodore L. DeWeese, Derya Tilki, Jay P. Ciezki, R. Jeffrey Karnes, Nicholas G. Nickols, Matthew B. Rettig, Felix Y. Feng, Alejandro Berlin, Jonathan D. Tward, Brian J. Davis, Robert E. Reiter, Paul C. Boutros, Tahmineh Romero, Eric M. Horwitz, Rahul D. Tendulkar, Michael L. Steinberg, Daniel E. Spratt, Michael Xiang, and Amar U. Kishan
- Subjects
Cancer Research ,Radiation ,Oncology ,Radiology, Nuclear Medicine and imaging - Abstract
Very-high-risk (VHR) prostate cancer (PC) is an aggressive subgroup with high risk of distant disease progression. Systemic treatment intensification with abiraterone or docetaxel reduces PC-specific mortality (PCSM) and distant metastasis (DM) in men receiving external beam radiation therapy (EBRT) with androgen deprivation therapy (ADT). Whether prostate-directed treatment intensification with the addition of brachytherapy (BT) boost to EBRT with ADT improves outcomes in this group is unclear.This cohort study from 16 centers across 4 countries included men with VHR PC treated with either dose-escalated EBRT with ≥24 months of ADT or EBRT + BT boost with ≥12 months of ADT. VHR was defined by National Comprehensive Cancer Network (NCCN) criteria (clinical T3b-4, primary Gleason pattern 5, or ≥2 NCCN high-risk features), and results were corroborated in a subgroup of men who met Systemic Therapy in Advancing or Metastatic Prostate Cancer: Evaluation of Drug Efficacy (STAMPEDE) trials inclusion criteria (≥2 of the following: clinical T3-4, Gleason 8-10, or PSA ≥40 ng/mL). PCSM and DM between EBRT and EBRT + BT were compared using inverse probability of treatment weight-adjusted Fine-Gray competing risk regression.Among the entire cohort, 270 underwent EBRT and 101 EBRT + BT. After a median follow-up of 7.8 years, 6.7% and 5.9% of men died of PC and 16.3% and 9.9% had DM after EBRT and EBRT + BT, respectively. There was no significant difference in PCSM (sHR, 1.47 [95% CI, 0.57-3.75]; P = .42) or DM (sHR, 0.72, [95% CI, 0.30-1.71]; P = .45) between EBRT + BT and EBRT. Results were similar within the STAMPEDE-defined VHR subgroup (PCSM: sHR, 1.67 [95% CI, 0.48-5.81]; P = .42; DM: sHR, 0.56 [95% CI, 0.15-2.04]; P = .38).In this VHR PC cohort, no difference in clinically meaningful outcomes was observed between EBRT alone with ≥24 months of ADT compared with EBRT + BT with ≥12 months of ADT. Comparative analyses in men treated with intensified systemic therapy are warranted.
- Published
- 2023
- Full Text
- View/download PDF
22. Phase II Randomized Study of Salvage Radiation Therapy Plus Enzalutamide or Placebo for High-Risk Prostate-Specific Antigen Recurrent Prostate Cancer After Radical Prostatectomy: The SALV-ENZA Trial
- Author
-
Phuoc T. Tran, Kathryn Lowe, Hua-Ling Tsai, Daniel Y. Song, Arthur Y. Hung, Jason W.D. Hearn, Steven Miller, James A. Proudfoot, Matthew P. Deek, Ryan Phillips, Tamara Lotan, Channing J. Paller, Catherine H. Marshall, Mark Markowski, Shirl Dipasquale, Samuel Denmeade, Michael Carducci, Mario Eisenberger, Theodore L. DeWeese, Matthew Orton, Curtiland Deville, Elai Davicioni, Stanley L. Liauw, Elisabeth I. Heath, Stephen Greco, Neil B. Desai, Daniel E. Spratt, Felix Feng, Hao Wang, Tomasz M. Beer, and Emmanuel S. Antonarakis
- Subjects
Cancer Research ,Oncology - Abstract
PURPOSE We sought to investigate whether enzalutamide (ENZA), without concurrent androgen deprivation therapy, increases freedom from prostate-specific antigen (PSA) progression (FFPP) when combined with salvage radiation therapy (SRT) in men with recurrent prostate cancer after radical prostatectomy (RP). PATIENTS AND METHODS Men with biochemically recurrent prostate cancer after RP were enrolled into a randomized, double‐blind, phase II, placebo-controlled, multicenter study of SRT plus ENZA or placebo (ClinicalTrials.gov identifier: NCT02203695 ). Random assignment (1:1) was stratified by center, surgical margin status (R0 v R1), PSA before salvage treatment (PSA ≥ 0.5 v < 0.5 ng/mL), and pathologic Gleason sum (7 v 8‐10). Patients were assigned to receive either ENZA 160 mg once daily or matching placebo for 6 months. After 2 months of study drug therapy, external-beam radiation (66.6‐70.2 Gy) was administered to the prostate bed (no pelvic nodes). The primary end point was FFPP in the intention-to-treat population. Secondary end points were time to local recurrence within the radiation field, metastasis‐free survival, and safety as determined by frequency and severity of adverse events. RESULTS Eighty-six (86) patients were randomly assigned, with a median follow-up of 34 (range, 0-52) months. Trial arms were well balanced. The median pre-SRT PSA was 0.3 (range, 0.06-4.6) ng/mL, 56 of 86 patients (65%) had extraprostatic disease (pT3), 39 of 86 (45%) had a Gleason sum of 8-10, and 43 of 86 (50%) had positive surgical margins (R1). FFPP was significantly improved with ENZA versus placebo (hazard ratio [HR], 0.42; 95% CI, 0.19 to 0.92; P = .031), and 2-year FFPP was 84% versus 66%, respectively. Subgroup analyses demonstrated differential benefit of ENZA in men with pT3 (HR, 0.22; 95% CI, 0.07 to 0.69) versus pT2 disease (HR, 1.54; 95% CI, 0.43 to 5.47; Pinteraction = .019) and R1 (HR, 0.14; 95% CI, 0.03 to 0.64) versus R0 disease (HR, 1.00; 95% CI, 0.36 to 2.76; Pinteraction = .023). There were insufficient secondary end point events for analysis. The most common adverse events were grade 1-2 fatigue (65% ENZA v 53% placebo) and urinary frequency (40% ENZA v 49% placebo). CONCLUSION SRT plus ENZA monotherapy for 6 months in men with PSA-recurrent high-risk prostate cancer after RP is safe and delays PSA progression relative to SRT alone. The impact of ENZA on distant metastasis or survival is unknown at this time.
- Published
- 2023
- Full Text
- View/download PDF
23. Griseofulvin Radiosensitizes Non–Small Cell Lung Cancer Cells and Activates cGAS
- Author
-
Xing Wang, Natasha Raman, Ghali Lemtiri-Chlieh, Jinhee Chang, Shreya Jagtap, Dipanwita Dutta Chowdhury, Matthew Ballew, Francesca Anna Carrieri, Triet Nguyen, Katriana Nugent, Travis Peck, Michelle S. Levine, Aaron Chan, Christine Lam, Reem Malek, Tung Hoang, Ryan Phillips, ZhuoAn Cheng, Kekoa Taparra, Nick Connis, Christine L. Hann, Andrew Holland, Phuoc T. Tran, Audrey Lafargue, and Hailun Wang
- Subjects
Cancer Research ,Oncology - Abstract
Extra copies of centrosomes are frequently observed in cancer cells. To survive and proliferate, cancer cells have developed strategies to cluster extra-centrosomes to form bipolar mitotic spindles. The aim of this study was to investigate whether centrosome clustering (CC) inhibition (CCi) would preferentially radiosensitize non–small cell lung cancer (NSCLC). Griseofulvin (GF; FDA-approved treatment) inhibits CC, and combined with radiation treatment (RT), resulted in a significant increase in the number of NSCLC cells with multipolar spindles, and decreased cell viability and colony formation ability in vitro. In vivo, GF treatment was well tolerated by mice, and the combined therapy of GF and radiation treatment resulted in a significant tumor growth delay. Both GF and radiation treatment also induced the generation of micronuclei (MN) in vitro and in vivo and activated cyclic GMP-AMP synthase (cGAS) in NSCLC cells. A significant increase in downstream cGAS-STING pathway activation was seen after combination treatment in A549 radioresistant cells that was dependent on cGAS. In conclusion, GF increased radiation treatment efficacy in lung cancer preclinical models in vitro and in vivo. This effect may be associated with the generation of MN and the activation of cGAS. These data suggest that the combination therapy of CCi, radiation treatment, and immunotherapy could be a promising strategy to treat NSCLC.
- Published
- 2023
- Full Text
- View/download PDF
24. MYC and Twist1 cooperate to drive metastasis by eliciting crosstalk between cancer and innate immunity
- Author
-
Renumathy Dhanasekaran, Virginie Baylot, Minsoon Kim, Sibu Kuruvilla, David I Bellovin, Nia Adeniji, Anand Rajan KD, Ian Lai, Meital Gabay, Ling Tong, Maya Krishnan, Jangho Park, Theodore Hu, Mustafa A Barbhuiya, Andrew J Gentles, Kasthuri Kannan, Phuoc T Tran, and Dean W Felsher
- Subjects
metastasis ,macrophages ,cytokines ,liver cancer ,MYC ,Twist1 ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Metastasis is a major cause of cancer mortality. We generated an autochthonous transgenic mouse model whereby conditional expression of MYC and Twist1 enables hepatocellular carcinoma (HCC) to metastasize in >90% of mice. MYC and Twist1 cooperate and their sustained expression is required to elicit a transcriptional program associated with the activation of innate immunity, through secretion of a cytokinome that elicits recruitment and polarization of tumor associated macrophages (TAMs). Systemic treatment with Ccl2 and Il13 induced MYC-HCCs to metastasize; whereas, blockade of Ccl2 and Il13 abrogated MYC/Twist1-HCC metastasis. Further, in 33 human cancers (n = 9502) MYC and TWIST1 predict poor survival (p=4.3×10−10), CCL2/IL13 expression (p
- Published
- 2020
- Full Text
- View/download PDF
25. Evaluating potential overuse of surveillance care in cancer survivors
- Author
-
Jennifer Y. Sheng, Claire F. Snyder, Katherine C. Smith, Jennifer DeSanto, Nancy Mayonado, Susan Rall, Sharon White, Amanda L. Blackford, Fabian M. Johnston, Robert L. Joyner, Joan Mischtschuk, Kimberly S. Peairs, Elissa Thorner, Phuoc T. Tran, Antonio C. Wolff, and Youngjee Choi
- Subjects
Cancer Research ,Oncology ,Radiology, Nuclear Medicine and imaging - Abstract
Survivorship care plans (SCPs) communicate cancer-related information from oncology providers to patients and primary care providers. SCPs may limit overuse testing by specifying necessary follow-up care. From a randomized, controlled trial of SCP delivery, we examined whether cancer-related tests not specified in SCPs, but conducted after SCP receipt, were appropriate or consistent with overuse.Survivors of breast, colorectal, or prostate cancer treated at urban-academic or rural-community health systems were randomized to one of three SCP delivery arms. Tests during 18 months after SCP receipt were classified as consistent with overuse if they were (1) not included in SCPs and (2) on a guideline-based predetermined list of "not recommended surveillance." After chart abstraction, physicians performed review and adjudication of potential overuse. Descriptive analyses were conducted of tests consistent with overuse. Negative binomial regression models determined if testing consistent with overuse differed across study arms.Among 316 patients (137 breast, 67 colorectal, 112 prostate), 140 individual tests were identified as potential overuse. Upon review, 98 were deemed to be consistent with overuse: 78 tumor markers and 20 imaging tests. The majority of overuse testing was breast cancer-related (95%). Across sites, 27 patients (9%) received ≥1 test consistent with overuse; most were breast cancer patients (22/27). Exploratory analyses of overuse test frequency by study arm showed no significant difference.This analysis identified practice patterns consistent with overuse of surveillance testing and can inform efforts to improve guideline-concordant care. Future interventions may include individual practice patterns and provider education.
- Published
- 2022
- Full Text
- View/download PDF
26. Clinical and Genomic Differences Between Advanced Molecular Imaging-detected and Conventional Imaging-detected Metachronous Oligometastatic Castration-sensitive Prostate Cancer
- Author
-
Philip Sutera, Yang Song, Kim Van der Eecken, Amol C. Shetty, Keara English, Theresa Hodges, Jinhee Chang, Valérie Fonteyne, Zaker Rana, Lei Ren, Adrianna A. Mendes, Nicolaas Lumen, Louke Delrue, Sofie Verbeke, Kathia De Man, Daniel Y. Song, Kenneth Pienta, Felix Y. Feng, Steven Joniau, Tamara Lotan, Barton Lane, Ana Kiess, Steven Rowe, Martin Pomper, Theodore DeWeese, Matthew Deek, Christopher Sweeney, Piet Ost, and Phuoc T. Tran
- Subjects
Urology - Published
- 2023
- Full Text
- View/download PDF
27. Genomic classifier performance in intermediate-risk prostate cancer: Results from NRG Oncology/RTOG 0126 randomized phase III trial
- Author
-
Daniel E. Spratt, Vinnie Y.T. Liu, Jeff Michalski, Elai Davicioni, Alejandro Berlin, Jeff M. Simko, Jason A. Efstathiou, Phuoc T. Tran, Howard M. Sandler, William A. Hall, Darby JS Thompson, Matthew B. Parliament, Ian S. Dayes, Rohann Jonathan Mark Correa, John M. Robertson, Elizabeth M. Gore, Desiree E. Doncals, Eric Vigneault, Luis Souhami, Theodore G. Karrison, and Felix Y. Feng
- Subjects
Cancer Research ,Radiation ,Oncology ,Radiology, Nuclear Medicine and imaging - Published
- 2023
- Full Text
- View/download PDF
28. Artificial Intelligence Predictive Model for Hormone Therapy Use in Prostate Cancer
- Author
-
Daniel E Spratt, Siyi Tang, Yilun Sun, Huei-Chung Huang, Emmalyn Chen, Osama Mohamad, Andrew J Armstrong, Jonathan D Tward, Paul L Nguyen, Joshua M Lang, Jingbin Zhang, Akinori Mitani, Jeffry P Simko, Sandy DeVries, Douwe van der Wal, Hans Pinckaers, Jedidiah M Monson, Holly A Campbell, James Wallace, Michelle J Ferguson, Jean-Paul Bahary, Edward M Schaeffer, NRG Prostate Cancer AI Consortium, Howard M Sandler, Phuoc T Tran, Joseph P Rodgers, Andre Esteva, Rikiya Yamashita, and Felix Y Feng
- Abstract
Background Androgen deprivation therapy (ADT) with radiotherapy can benefit patients with localized prostate cancer. However, ADT can negatively impact quality of life and there remain no validated predictive models to guide its use. Methods Digital pathology image and clinical data from pre-treatment prostate tissue from 5,727 patients enrolled on five phase III randomized trials treated with radiotherapy +/- ADT were used to develop and validate an artificial intelligence (AI)-derived predictive model to assess ADT benefit with the primary endpoint of distant metastasis. After the model was locked, validation was performed on NRG/RTOG 9408 (n = 1,594) that randomized men to radiotherapy +/- 4 months of ADT. Fine-Gray regression and restricted mean survival times were used to assess the interaction between treatment and predictive model and within predictive model positive and negative subgroup treatment effects. Results In the NRG/RTOG 9408 validation cohort (14.9 years of median follow-up), ADT significantly improved time to distant metastasis (subdistribution hazard ratio [sHR] = 0.64, 95%CI [0.45–0.90], p = 0.01). The predictive model-treatment interaction was significant (p-interaction = 0.01). In predictive model positive patients (n = 543, 34%), ADT significantly reduced the risk of distant metastasis compared to radiotherapy alone (sHR = 0.34, 95%CI [0.19–0.63], p
- Published
- 2023
- Full Text
- View/download PDF
29. A novel prostate cancer subtyping classifier based on luminal and basal phenotypes
- Author
-
Adam B. Weiner, Yang Liu, Alex Hakansson, Xin Zhao, James A. Proudfoot, Julian Ho, JJ H. Zhang, Eric V. Li, R. Jeffrey Karnes, Robert B. Den, Amar U. Kishan, Robert E. Reiter, Anis A. Hamid, Ashely E. Ross, Phuoc T. Tran, Elai Davicioni, Daniel E. Spratt, Gerhardt Attard, Tamara L. Lotan, Melvin Lee Kiang Chua, Christopher J. Sweeney, and Edward M. Schaeffer
- Subjects
Cancer Research ,Oncology - Published
- 2023
- Full Text
- View/download PDF
30. Supplementary Figures from Griseofulvin Radiosensitizes Non–Small Cell Lung Cancer Cells and Activates cGAS
- Author
-
Hailun Wang, Audrey Lafargue, Phuoc T. Tran, Andrew Holland, Christine L. Hann, Nick Connis, Kekoa Taparra, ZhuoAn Cheng, Ryan Phillips, Tung Hoang, Reem Malek, Christine Lam, Aaron Chan, Michelle S. Levine, Travis Peck, Katriana Nugent, Triet Nguyen, Francesca Anna Carrieri, Matthew Ballew, Dipanwita Dutta Chowdhury, Shreya Jagtap, Jinhee Chang, Ghali Lemtiri-Chlieh, Natasha Raman, and Xing Wang
- Abstract
Figure S1 shows basal centrosome amplification in cell lines. Figure S2 shows the effect of GF on cell viability. Figure S3 shows HSET expression in cell lines. Figure S4 shows the effect of GF on clonogenic potential after RT. Figure S5 shows the induction of micronuclei after GF and/or RT. Figure S6 shows the expression of IFN-β after GF and/or RT.
- Published
- 2023
- Full Text
- View/download PDF
31. Data from Griseofulvin Radiosensitizes Non–Small Cell Lung Cancer Cells and Activates cGAS
- Author
-
Hailun Wang, Audrey Lafargue, Phuoc T. Tran, Andrew Holland, Christine L. Hann, Nick Connis, Kekoa Taparra, ZhuoAn Cheng, Ryan Phillips, Tung Hoang, Reem Malek, Christine Lam, Aaron Chan, Michelle S. Levine, Travis Peck, Katriana Nugent, Triet Nguyen, Francesca Anna Carrieri, Matthew Ballew, Dipanwita Dutta Chowdhury, Shreya Jagtap, Jinhee Chang, Ghali Lemtiri-Chlieh, Natasha Raman, and Xing Wang
- Abstract
Extra copies of centrosomes are frequently observed in cancer cells. To survive and proliferate, cancer cells have developed strategies to cluster extra-centrosomes to form bipolar mitotic spindles. The aim of this study was to investigate whether centrosome clustering (CC) inhibition (CCi) would preferentially radiosensitize non–small cell lung cancer (NSCLC). Griseofulvin (GF; FDA-approved treatment) inhibits CC, and combined with radiation treatment (RT), resulted in a significant increase in the number of NSCLC cells with multipolar spindles, and decreased cell viability and colony formation ability in vitro. In vivo, GF treatment was well tolerated by mice, and the combined therapy of GF and radiation treatment resulted in a significant tumor growth delay. Both GF and radiation treatment also induced the generation of micronuclei (MN) in vitro and in vivo and activated cyclic GMP-AMP synthase (cGAS) in NSCLC cells. A significant increase in downstream cGAS-STING pathway activation was seen after combination treatment in A549 radioresistant cells that was dependent on cGAS. In conclusion, GF increased radiation treatment efficacy in lung cancer preclinical models in vitro and in vivo. This effect may be associated with the generation of MN and the activation of cGAS. These data suggest that the combination therapy of CCi, radiation treatment, and immunotherapy could be a promising strategy to treat NSCLC.
- Published
- 2023
- Full Text
- View/download PDF
32. Supplementary Data from Inhibition of TWIST1 Leads to Activation of Oncogene-Induced Senescence in Oncogene-Driven Non–Small Cell Lung Cancer
- Author
-
Charles M. Rudin, Phuoc T. Tran, Yoon-Jae Cho, Khaled Aziz, Sarah N.H. Chatley, Saravanan Thiyagarajan, Rajendra P. Gajula, Sara C. Murphy, Irina Dobromilskaya, and Timothy F. Burns
- Abstract
Supplementary Data PDF file - 114K, Supplemental Table 1 Mutational status of cell lines studied
- Published
- 2023
- Full Text
- View/download PDF
33. Supplementary Figures from Inhibition of TWIST1 Leads to Activation of Oncogene-Induced Senescence in Oncogene-Driven Non–Small Cell Lung Cancer
- Author
-
Charles M. Rudin, Phuoc T. Tran, Yoon-Jae Cho, Khaled Aziz, Sarah N.H. Chatley, Saravanan Thiyagarajan, Rajendra P. Gajula, Sara C. Murphy, Irina Dobromilskaya, and Timothy F. Burns
- Abstract
Supplementary Figures PDF file - 3634K, S1 TWIST1 specific shRNAs suppress TWIST1 transcript and protein levels. S2 Mouse Twist1 can rescue the anti-proliferative effects of knockdown of human TWIST1 in A549 cells. S3 Growth inhibition by silencing TWIST1 is not p53-dependent in KRAS mutant NSCLC. S4 Growth inhibition by silencing TWIST1 is not p21-dependent in KRAS mutant NSCLC. S5 Growth inhibition by silencing TWIST1 is not dependent on induction of p27 in KRAS mutant NSCLC S6 Overexpression of SKP2 does not rescue loss of TWIST1 in KRAS mutant NSCLC. S7 Microarray analysis of three KRAS mutant NSCLC cell lines (H460, H727 and H358) six days after silencing of TWIST1 reveals a striking cell cycle arrest gene signature. S8 Enrichment plots after Gene Set Enrichment Analysis for A. ROSTY_CERVICAL_CANCER_PROLIFERATION_CLUSTER (left) CROONQUIST_IL6_DEPRIVATION_DN (middle) CHANG_CYCLING_GENES (right) B. KANG_DOXORUBICIN_RESISTANCE_UP C. BLUM_RESPONSE_TO_SALIRASIB DN (left) CROONQUIST_NRAS_SIGNALING_DN (right) D. FUJII_YBX1_TARGETS_DN following GSEA performed on shScram and shTWIST1 samples from three KRAS mutant cell lines (H460, H727 and H358) six days after TWIST1 knockdown samples (NOM p-values, FDR qvalues, and FWER p-values were all
- Published
- 2023
- Full Text
- View/download PDF
34. Supplementary Table 1 from A First-in-Class TWIST1 Inhibitor with Activity in Oncogene-Driven Lung Cancer
- Author
-
Timothy F. Burns, Phuoc T. Tran, Charles M. Rudin, Yoon-Jae Cho, Andrew J. Ewald, Johnathan A. Engh, Ashwin Somasundaram, Katriana Nugent, Sarah N. Chatley, Eric H.-B. Huang, Myriam A. Attar, Hailun Wang, Suman Chatterjee, Susheel K. Khetarpal, Neil M. Neumann, Lucia Mazzacurati, Jessica Cades, and Zachary A. Yochum
- Abstract
Supplementary Table 1
- Published
- 2023
- Full Text
- View/download PDF
35. Supplementary Figures 1 - 11 from The Twist Box Domain Is Required for Twist1-induced Prostate Cancer Metastasis
- Author
-
Phuoc T. Tran, Steven S. An, Venu Raman, Russell K. Hales, Joseph M. Herman, Charles M. Rudin, Christine H. Chung, Timothy F. Burns, Shyam Biswal, Elana Fertig, Jessica Cades, Tarek Salih, Jinfang Ma, Farhad Vesuna, Aaron T. Wild, Nishant Gandhi, Ruoqi Wang, Khaled Aziz, Yoshinori Kato, Saravanan Thiyagarajan, Russell D. Williams, Sivarajan T. Chettiar, and Rajendra P. Gajula
- Abstract
PDF file - 1004K, S1. The Twist box domain is required for full Twist1 transcriptional activity. S2. Quantification of immunofluorescence from isogenic prostate cancer cell lines stably expressing Twist1 and Twist1-F191G. S3. The Twist box domain is required for full Twist1-induced EMT marker phenotypes of prostate cancer cells. S4. Twist1 overexpression induces temporal changes in the material properties of prostate cancer cells during their migration in a wound healing assay. S5. The overexpression of Twist1 or Twist1-F191G does not increase cellular proliferation of prostate cancer cells in vitro. S6. The Twist box domain is required for full Twist1-induced cellular migration in PC3 cells. S7. Twist1 overexpression increases cell traction forces of individual androgenindependent PC3 prostate cancer cells. S8. Twist1 overexpression confers radioresistance to prostate cancer cells which is attenuated by mutation of the Twist box domain. S9. The Twist box domain is required for Twist1-induced soft agar anchorageindependent growth of 22Rv1 prostate cancer cells. S10. Twist1 overexpression does not confer prostate cancer cells increased primary tumorigenicity and slows primary tumor cell growth in vivo. S11. The Twist box domain is required for full Twist1-induced expression of Hoxa9/HOXA9.
- Published
- 2023
- Full Text
- View/download PDF
36. Supplementary Figures from A First-in-Class TWIST1 Inhibitor with Activity in Oncogene-Driven Lung Cancer
- Author
-
Timothy F. Burns, Phuoc T. Tran, Charles M. Rudin, Yoon-Jae Cho, Andrew J. Ewald, Johnathan A. Engh, Ashwin Somasundaram, Katriana Nugent, Sarah N. Chatley, Eric H.-B. Huang, Myriam A. Attar, Hailun Wang, Suman Chatterjee, Susheel K. Khetarpal, Neil M. Neumann, Lucia Mazzacurati, Jessica Cades, and Zachary A. Yochum
- Abstract
Supplementary Figure 1: FGF2 treatment induces branching morphogenesis of primary epithelial cells in 3D culture. Supplementary Figure 2: Basal Expression Levels of TWIST1 in KRAS mutant and MET Amplified/Mutant NSCLC Cell Lines. Supplementary Figure 3: Harmine treatment induces Oncogene-Induced Senescence (OIS) in EGFR and MET-mutant NSCLC cell lines. Supplementary Figure 4: Harmine induces apoptosis in oncogene-driven NSCLC cell lines. Supplementary Figure 5: Harmine treatment promotes TWIST1 degradation and decreases TWIST1 protein stability. Supplementary Figure 6: Silencing of E2A induces apoptosis and phenocopies silencing of TWIST1. Supplementary Figure 7: Overexpression of TWIST1 or its binding partner, E2A, rescues harmine induced growth inhibition. Supplementary Figure 8: Treatment with harmine decreases tumor growth in a KRAS mutant Patient-Derived Xenograph (PDX) model and degrades Twist1 and induces apoptosis in transgenic mouse model of Kras mutant lung cancer. Supplementary Table 1: Rank list of compounds from Connectivity mapping (CMAP) analysis Supplementary Table 2: List of primers used for qRT-PCR Supplementary Table 3: List of primers used for Taqman qRT-PCR Supplementary Table 4: List of antibodies used in current study Supplementary Table 5: Sequences for TWIST1/TCF3 shRNA (5’ â€" 3’) in pKLO.1 Supplementary Table 6: ORFs obtained from Johns Hopkins University HiT Center Supplementary Table 7: Source of Plasmids utilized
- Published
- 2023
- Full Text
- View/download PDF
37. Supplementary Table 2 from The Twist Box Domain Is Required for Twist1-induced Prostate Cancer Metastasis
- Author
-
Phuoc T. Tran, Steven S. An, Venu Raman, Russell K. Hales, Joseph M. Herman, Charles M. Rudin, Christine H. Chung, Timothy F. Burns, Shyam Biswal, Elana Fertig, Jessica Cades, Tarek Salih, Jinfang Ma, Farhad Vesuna, Aaron T. Wild, Nishant Gandhi, Ruoqi Wang, Khaled Aziz, Yoshinori Kato, Saravanan Thiyagarajan, Russell D. Williams, Sivarajan T. Chettiar, and Rajendra P. Gajula
- Abstract
PDF file - 116K, Supplemental Table S2 - Myc-CaP + Twist1-F191G versus Myc-CaP + Vector Differentially.
- Published
- 2023
- Full Text
- View/download PDF
38. Supplementary Table 1 from The Twist Box Domain Is Required for Twist1-induced Prostate Cancer Metastasis
- Author
-
Phuoc T. Tran, Steven S. An, Venu Raman, Russell K. Hales, Joseph M. Herman, Charles M. Rudin, Christine H. Chung, Timothy F. Burns, Shyam Biswal, Elana Fertig, Jessica Cades, Tarek Salih, Jinfang Ma, Farhad Vesuna, Aaron T. Wild, Nishant Gandhi, Ruoqi Wang, Khaled Aziz, Yoshinori Kato, Saravanan Thiyagarajan, Russell D. Williams, Sivarajan T. Chettiar, and Rajendra P. Gajula
- Abstract
PDF file - 384K, Supplemental Table S1 - Myc-CaP + Twist1 versus Myc-CaP + Vector Differentially.
- Published
- 2023
- Full Text
- View/download PDF
39. Supplementary Materials & Methods from Griseofulvin Radiosensitizes Non–Small Cell Lung Cancer Cells and Activates cGAS
- Author
-
Hailun Wang, Audrey Lafargue, Phuoc T. Tran, Andrew Holland, Christine L. Hann, Nick Connis, Kekoa Taparra, ZhuoAn Cheng, Ryan Phillips, Tung Hoang, Reem Malek, Christine Lam, Aaron Chan, Michelle S. Levine, Travis Peck, Katriana Nugent, Triet Nguyen, Francesca Anna Carrieri, Matthew Ballew, Dipanwita Dutta Chowdhury, Shreya Jagtap, Jinhee Chang, Ghali Lemtiri-Chlieh, Natasha Raman, and Xing Wang
- Abstract
Supplementary Materials & Methods
- Published
- 2023
- Full Text
- View/download PDF
40. Supplementary Data from A First-in-Class TWIST1 Inhibitor with Activity in Oncogene-Driven Lung Cancer
- Author
-
Timothy F. Burns, Phuoc T. Tran, Charles M. Rudin, Yoon-Jae Cho, Andrew J. Ewald, Johnathan A. Engh, Ashwin Somasundaram, Katriana Nugent, Sarah N. Chatley, Eric H.-B. Huang, Myriam A. Attar, Hailun Wang, Suman Chatterjee, Susheel K. Khetarpal, Neil M. Neumann, Lucia Mazzacurati, Jessica Cades, and Zachary A. Yochum
- Abstract
Supplementary Figure legends and methods
- Published
- 2023
- Full Text
- View/download PDF
41. Data from A First-in-Class TWIST1 Inhibitor with Activity in Oncogene-Driven Lung Cancer
- Author
-
Timothy F. Burns, Phuoc T. Tran, Charles M. Rudin, Yoon-Jae Cho, Andrew J. Ewald, Johnathan A. Engh, Ashwin Somasundaram, Katriana Nugent, Sarah N. Chatley, Eric H.-B. Huang, Myriam A. Attar, Hailun Wang, Suman Chatterjee, Susheel K. Khetarpal, Neil M. Neumann, Lucia Mazzacurati, Jessica Cades, and Zachary A. Yochum
- Abstract
TWIST1, an epithelial–mesenchymal transition (EMT) transcription factor, is critical for oncogene-driven non–small cell lung cancer (NSCLC) tumorigenesis. Given the potential of TWIST1 as a therapeutic target, a chemical–bioinformatic approach using connectivity mapping (CMAP) analysis was used to identify TWIST1 inhibitors. Characterization of the top ranked candidates from the unbiased screen revealed that harmine, a harmala alkaloid, inhibited multiple TWIST1 functions, including single-cell dissemination, suppression of normal branching in 3D epithelial culture, and proliferation of oncogene driver-defined NSCLC cells. Harmine treatment phenocopied genetic loss of TWIST1 by inducing oncogene-induced senescence or apoptosis. Mechanistic investigation revealed that harmine targeted the TWIST1 pathway through its promotion of TWIST1 protein degradation. As dimerization is critical for TWIST1 function and stability, the effect of harmine on specific TWIST1 dimers was examined. TWIST1 and its dimer partners, the E2A proteins, which were found to be required for TWIST1-mediated functions, regulated the stability of the other heterodimeric partner posttranslationally. Harmine preferentially promoted degradation of the TWIST1-E2A heterodimer compared with the TWIST-TWIST1 homodimer, and targeting the TWIST1-E2A heterodimer was required for harmine cytotoxicity. Finally, harmine had activity in both transgenic and patient-derived xenograft mouse models of KRAS-mutant NSCLC. These studies identified harmine as a first-in-class TWIST1 inhibitor with marked anti-tumor activity in oncogene-driven NSCLC including EGFR mutant, KRAS mutant and MET altered NSCLC.Implications: TWIST1 is required for oncogene-driven NSCLC tumorigenesis and EMT; thus, harmine and its analogues/derivatives represent a novel therapeutic strategy to treat oncogene-driven NSCLC as well as other solid tumor malignancies. Mol Cancer Res; 15(12); 1764–76. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
42. CCR Translation for This Article from Hypoxia in Models of Lung Cancer: Implications for Targeted Therapeutics
- Author
-
Amato J. Giaccia, Quynh-Thu Le, Alejandro Sweet-Cordero, Leanne Sayles, Dean Felsher, Phuoc T. Tran, Janine T. Erler, Ivana K. Cecic, Marta Vilalta, and Edward E. Graves
- Abstract
CCR Translation for This Article from Hypoxia in Models of Lung Cancer: Implications for Targeted Therapeutics
- Published
- 2023
- Full Text
- View/download PDF
43. Supplementary Table 1 from RK-33 Radiosensitizes Prostate Cancer Cells by Blocking the RNA Helicase DDX3
- Author
-
Venu Raman, Phuoc T. Tran, Paul J. van Diest, Kathleen Gabrielson, Reem Malek, Katriana Nugent, Marise R. Heerma van Voss, Guus M. Bol, Saritha Tantravedi, Farhad Vesuna, and Min Xie
- Abstract
Clinical characteristics of patients with low and high DDX3 expression
- Published
- 2023
- Full Text
- View/download PDF
44. Data from Hypoxia in Models of Lung Cancer: Implications for Targeted Therapeutics
- Author
-
Amato J. Giaccia, Quynh-Thu Le, Alejandro Sweet-Cordero, Leanne Sayles, Dean Felsher, Phuoc T. Tran, Janine T. Erler, Ivana K. Cecic, Marta Vilalta, and Edward E. Graves
- Abstract
Purpose: To efficiently translate experimental methods from bench to bedside, it is imperative that laboratory models of cancer mimic human disease as closely as possible. In this study, we sought to compare patterns of hypoxia in several standard and emerging mouse models of lung cancer to establish the appropriateness of each for evaluating the role of oxygen in lung cancer progression and therapeutic response.Experimental Design: Subcutaneous and orthotopic human A549 lung carcinomas growing in nude mice as well as spontaneous K-ras or Myc-induced lung tumors grown in situ or subcutaneously were studied using fluorodeoxyglucose and fluoroazomycin arabinoside positron emission tomography, and postmortem by immunohistochemical observation of the hypoxia marker pimonidazole. The response of these models to the hypoxia-activated cytotoxin PR-104 was also quantified by the formation of γH2AX foci in vitro and in vivo. Finally, our findings were compared with oxygen electrode measurements of human lung cancers.Results: Minimal fluoroazomycin arabinoside and pimonidazole accumulation was seen in tumors growing within the lungs, whereas subcutaneous tumors showed substantial trapping of both hypoxia probes. These observations correlated with the response of these tumors to PR-104, and with the reduced incidence of hypoxia in human lung cancers relative to other solid tumor types.Conclusions: These findings suggest that in situ models of lung cancer in mice may be more reflective of the human disease, and encourage judicious selection of preclinical tumor models for the study of hypoxia imaging and antihypoxic cell therapies. Clin Cancer Res; 16(19); 4843–52. ©2010 AACR.
- Published
- 2023
- Full Text
- View/download PDF
45. Supplementary Figure 5 from RK-33 Radiosensitizes Prostate Cancer Cells by Blocking the RNA Helicase DDX3
- Author
-
Venu Raman, Phuoc T. Tran, Paul J. van Diest, Kathleen Gabrielson, Reem Malek, Katriana Nugent, Marise R. Heerma van Voss, Guus M. Bol, Saritha Tantravedi, Farhad Vesuna, and Min Xie
- Abstract
Bar graph showing the percentage of positive cells stained for Ki67, cleaved Caspase 3 and gammaH2AX
- Published
- 2023
- Full Text
- View/download PDF
46. Supplemental Figure 2 from Systemic Delivery of Microencapsulated 3-Bromopyruvate for the Therapy of Pancreatic Cancer
- Author
-
Jean-François Geschwind, Bert Vogelstein, Andrew J. Ewald, Gregg L. Semenza, Joseph M. Herman, Phuoc T. Tran, Weibo Luo, MingDe Lin, Cassandra Rae Moats, Sivarajan Chettiar Thiruganasambandam, Rafael Duran, Juvenal Reyes, Shanmugasundaram Ganapathy-Kanniappan, Lynn Jeanette Savic, Surojit Sur, and Julius Chapiro
- Abstract
This figure provides the overview of the results for the viability experiments in the 3D in vitro model.
- Published
- 2023
- Full Text
- View/download PDF
47. Figure S1 from Randomized Phase II Trial of Sipuleucel-T with or without Radium-223 in Men with Bone-metastatic Castration-resistant Prostate Cancer
- Author
-
Emmanuel S. Antonarakis, Oliver Sartor, Andrew J. Armstrong, Edwin M. Posadas, Charles G. Drake, Pedro C. Barata, Nancy P. Moldawer, Patrick Cotogno, Charlotte Manogue, Julia Hurrelbrink, Michaella Afful, Serina King, Daniel Y. Song, Phuoc T. Tran, Theodore L. DeWeese, Jong Chul Park, Hao Wang, Wei Fu, and Catherine H. Marshall
- Abstract
Figure S1
- Published
- 2023
- Full Text
- View/download PDF
48. Lymphocyte gating strategy. from Combination Therapy with Anti-PD-1, Anti-TIM-3, and Focal Radiation Results in Regression of Murine Gliomas
- Author
-
Michael Lim, Drew M. Pardoll, Henry Brem, Charles G. Drake, Peter C. Burger, Janis M. Taube, Alessandro Olivi, Xiaobu Ye, Phuoc T. Tran, Betty M. Tyler, Allison M. Martin, Mary Sheu, Tomas Garzon-Muvdi, Sarah Harris-Bookman, Christopher M. Jackson, Dimitrios Mathios, Haiying Xu, Ada Tam, Eric W. Sankey, Ann Liu, Esteban Velarde, Debebe Theodros, Eileen S. Kim, Antonella Mangraviti, Mira A. Patel, and Jennifer E. Kim
- Abstract
Cells were first gated on size and singularity by forward scatter and side scatter. Nonviable cells were excluded by live/dead gating. CD3+ live cells were gated on CD4 and CD8. CD3+FoxP3+CD4+ cells were designated as Tregs. TIM-3 vs. PD-1 expression was assessed for each lymphocyte population.
- Published
- 2023
- Full Text
- View/download PDF
49. Data from Systemic Delivery of Microencapsulated 3-Bromopyruvate for the Therapy of Pancreatic Cancer
- Author
-
Jean-François Geschwind, Bert Vogelstein, Andrew J. Ewald, Gregg L. Semenza, Joseph M. Herman, Phuoc T. Tran, Weibo Luo, MingDe Lin, Cassandra Rae Moats, Sivarajan Chettiar Thiruganasambandam, Rafael Duran, Juvenal Reyes, Shanmugasundaram Ganapathy-Kanniappan, Lynn Jeanette Savic, Surojit Sur, and Julius Chapiro
- Abstract
Purpose: This study characterized the therapeutic efficacy of a systemically administered formulation of 3-bromopyruvate (3-BrPA), microencapsulated in a complex with β-cyclodextrin (β-CD), using an orthotopic xenograft mouse model of pancreatic ductal adenocarcinoma (PDAC).Experimental Design: The presence of the β-CD–3-BrPA complex was confirmed using nuclear magnetic resonance spectroscopy. Monolayer as well as three-dimensional organotypic cell culture was used to determine the half-maximal inhibitory concentrations (IC50) of β-CD–3-BrPA, free 3-BrPA, β-CD (control), and gemcitabine in MiaPaCa-2 and Suit-2 cell lines, both in normoxia and hypoxia. Phase-contrast microscopy, bioluminescence imaging (BLI), as well as zymography and Matrigel assays were used to characterize the effects of the drug in vitro. An orthotopic lucMiaPaCa-2 xenograft tumor model was used to investigate the in vivo efficacy.Results: β-CD–3-BrPA and free 3-BrPA demonstrated an almost identical IC50 profile in both PDAC cell lines with higher sensitivity in hypoxia. Using the Matrigel invasion assay as well as zymography, 3-BrPA showed anti-invasive effects in sublethal drug concentrations. In vivo, animals treated with β-CD–3-BrPA demonstrated minimal or no tumor progression as evident by the BLI signal as opposed to animals treated with gemcitabine or the β-CD (60-fold and 140-fold signal increase, respectively). In contrast to animals treated with free 3-BrPA, no lethal toxicity was observed for β-CD–3-BrPA.Conclusion: The microencapsulation of 3-BrPA represents a promising step towards achieving the goal of systemically deliverable antiglycolytic tumor therapy. The strong anticancer effects of β-CD–3-BrPA combined with its favorable toxicity profile suggest that clinical trials, particularly in patients with PDAC, should be considered. Clin Cancer Res; 20(24); 6406–17. ©2014 AACR.
- Published
- 2023
- Full Text
- View/download PDF
50. Supplemental Figures from TWIST1-WDR5-Hottip Regulates Hoxa9 Chromatin to Facilitate Prostate Cancer Metastasis
- Author
-
Phuoc T. Tran, Colm Morrissey, Paula J. Hurley, Kenneth J. Pienta, Edward M. Schaeffer, Ashley E. Ross, Theodore L. DeWeese, Steven S. An, Lawrence True, Arum R. Yoon, Ghali Lemtiri-Chlieh, Kekoa Taparra, Hailun Wang, Katriana Nugent, Brian W. Simons, Belinda Nghiem, Russell D. Williams, Rajendra P. Gajula, and Reem Malek
- Abstract
This document contains supplemental details such as antibodies and primers, list of genes overrepresented in TWIST1 and TWIST1 mutants, IHC staining for TWIST1 in mouse prostate development, IHC staining for TWIST1 and HOXA9 in various mouse models of prostate cancer, correlation of TWIST1 and HOXA9 alteration with poor survival in human prostate cancer patients, IHC for TWIST1 and HOXA9 on primary tumors and bone metastasis from prostate cancer patients, IHC for HOXA9 on cell pellets expressing HOXA9 or control and on adult mouse prostate, data showing sufficiency of HOXA9 for some pro-metastatic behaviour in prostate cancer cells, data showing requirement of WDR5 and HOTTIP for TWIST1-dependent pro-metastatic behavior, ChIP data showing binding of TWIST1 and HOXA9 at the HOXA9 promoter, data showing effect of drugs that can inhibit HOXA9 on prostate cancer cell viability and pro metastatic behaviour.
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.