Network


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

Hotspot


Dive into the research topics where Dustin A. Deming is active.

Publication


Featured researches published by Dustin A. Deming.


Journal of The National Comprehensive Cancer Network | 2017

Colon cancer, version 1.2017: Clinical practice guidelines in oncology

Al B. Benson; Alan P. Venook; Lynette Cederquist; Emily Chan; Yi Jen Chen; Harry S. Cooper; Dustin A. Deming; Paul F. Engstrom; Peter C. Enzinger; Alessandro Fichera; Jean L. Grem; Axel Grothey; Howard S. Hochster; Sarah E. Hoffe; Steven R. Hunt; Ahmed Kamel; Natalie Kirilcuk; Smitha S. Krishnamurthi; Wells A. Messersmith; Mary F. Mulcahy; James D. Murphy; Steven Nurkin; Leonard Saltz; Sunil Sharma; David Shibata; John M. Skibber; Constantinos T. Sofocleous; Elena M. Stoffel; Eden Stotsky-Himelfarb; Christopher G. Willett

This selection from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Bladder Cancer focuses on systemic therapy for muscle-invasive urothelial bladder cancer, as substantial revisions were made in the 2017 updates, such as new recommendations for nivolumab, pembrolizumab, atezolizumab, durvalumab, and avelumab. The complete version of the NCCN Guidelines for Bladder Cancer addresses additional aspects of the management of bladder cancer, including non-muscle-invasive urothelial bladder cancer and nonurothelial histologies, as well as staging, evaluation, and follow-up.


Lancet Oncology | 2017

Nivolumab for previously treated unresectable metastatic anal cancer (NCI9673): a multicentre, single-arm, phase 2 study

Van Karlyle Morris; Mohamed E. Salem; Halla Sayed Nimeiri; Syma Iqbal; Preet Paul Singh; Kristen K. Ciombor; Blase N. Polite; Dustin A. Deming; Emily Chan; James L. Wade; Lianchun Xiao; Tanios Bekaii-Saab; Luis Vence; Jorge Blando; Armeen Mahvash; Wai Chin Foo; Chimela Ohaji; Manolo Pasia; Gail Bland; Aki Ohinata; Jane E. Rogers; Amir Mehdizadeh; Kimberly C. Banks; Richard B. Lanman; Robert A. Wolff; Howard Streicher; James P. Allison; Padmanee Sharma; Cathy Eng

BACKGROUND Squamous cell carcinoma of the anal canal (SCCA) is a rare malignancy associated with infection by human papillomavirus (HPV). No consensus treatment approach exists for the treatment of metastatic disease. Because intratumoral HPV oncoproteins upregulate immune checkpoint proteins such as PD-1 to evade immune-mediated cytotoxicity, we did a trial of the anti-PD-1 antibody nivolumab for patients with metastatic SCCA. METHODS We did this single-arm, multicentre, phase 2 trial at ten academic centres in the USA. We enrolled patients with treatment-refractory metastatic SCCA, who were given nivolumab every 2 weeks (3 mg/kg). The primary endpoint was response according to Response Evaluation Criteria in Solid Tumors, version 1.1, in the intention-to-treat population. At the time of data cutoff, the study was ongoing, with patients continuing to receive treatment. The study is registered with ClinicalTrials.gov, number NCT02314169. RESULTS We screened 39 patients, of whom 37 were enrolled and received at least one dose of nivolumab. Among the 37 patients, nine (24% [95% CI 15-33]) had responses. There were two complete responses and seven partial responses. Grade 3 adverse events were anaemia (n=2), fatigue (n=1), rash (n=1), and hypothyroidism (n=1). No serious adverse events were reported. INTERPRETATION To our knowledge, this is the first completed phase 2 trial of immunotherapy for SCCA. Nivolumab is well tolerated and effective as a monotherapy for patients with metastatic SCCA. Immune checkpoint blockade appears to be a promising approach for patients with this orphan disease. FUNDING National Cancer Institute/Cancer Therapy Evaluation Program, the HPV and Anal Cancer Foundation, the E B Anal Cancer Fund, The University of Texas MD Anderson Moon Shots Program, and an anonymous philanthropic donor.


Cancer Research | 2012

Mice expressing activated PI3K rapidly develop advanced colon cancer

Alyssa A. Leystra; Dustin A. Deming; Christopher D. Zahm; Mohammed Farhoud; Terrah J. Paul Olson; Jamie N. Hadac; Laura Nettekoven; Dawn M. Albrecht; Linda Clipson; Ruth Sullivan; Mary Kay Washington; Jose Torrealba; Jamey P. Weichert; Richard B. Halberg

Aberrations in the phosphoinositide 3-kinase (PI3K) signaling pathway play a key role in the pathogenesis of numerous cancers by altering cellular growth, metabolism, proliferation, and apoptosis. Mutations in the catalytic domain of PI3K that generate a dominantly active kinase are commonly found in human colorectal cancers and have been thought to drive tumor progression but not initiation. However, the effects of constitutively activated PI3K upon the intestinal mucosa have not been previously studied in animal models. Here, we show that the expression of a dominantly active form of the PI3K protein in the mouse intestine results in hyperplasia and advanced neoplasia. Mice expressing constitutively active PI3K in the epithelial cells of the distal small bowel and colon rapidly developed invasive adenocarcinomas in the colon that spread into the mesentery and adjacent organs. The histologic characteristics of these tumors were strikingly similar to invasive mucinous colon cancers in humans. Interestingly, these tumors formed without a benign polypoid intermediary, consistent with the lack of aberrant WNT signaling observed. Together, our findings indicate a noncanonical mechanism of colon tumor initiation that is mediated through activation of PI3K. This unique model has the potential to further our understanding of human disease and facilitate the development of therapeutics through pharmacologic screening and biomarker identification.


Oncogene | 2014

PIK3CA and APC Mutations are Synergistic in the Development of Intestinal Cancers

Dustin A. Deming; Alyssa A. Leystra; Laura Nettekoven; Chelsea Sievers; Devon Miller; Malisa Middlebrooks; Linda Clipson; Dawn M. Albrecht; Jeff Bacher; Mary Kay Washington; Jamey P. Weichert; Richard B. Halberg

Human colorectal cancers are known to possess multiple mutations, though how these mutations interact in tumor development and progression has not been fully investigated. We have previously described the FCPIK3ca* murine colon cancer model, which expresses a constitutively activated phosphoinositide-3 kinase (PI3K) in the intestinal epithelium. The expression of this dominantly active form of PI3K results in hyperplasia and invasive mucinous adenocarcinomas. These cancers form via a non-canonical mechanism of tumor initiation that is mediated through activation of PI3K and not through aberrations in WNT signaling. Since the Adenomatous Polyposis Coli (APC) gene is mutated in the majority of human colon cancers and often occurs simultaneously with PIK3CA mutations, we sought to better understand the interaction between APC and PIK3CA mutations in the mammalian intestine. In this study, we have generated mice in which the expression of a constitutively active PI3K and the loss of APC occur simultaneously in the distal small intestine and colon. Here, we demonstrate that expression of a dominant active PI3K synergizes with loss of APC activity resulting in a dramatic change in tumor multiplicity, size, morphology and invasiveness. Activation of the PI3K pathway is not able to directly activate WNT signaling through the nuclear localization of CTNNB1 (β-catenin) in the absence of aberrant WNT signaling. Alterations at the transcriptional level, including increased CCND1, may be the etiology of synergy between these activated pathways.


Therapeutic Advances in Medical Oncology | 2015

Precision medicine in colorectal cancer: the molecular profile alters treatment strategies.

Nguyen H. Tran; Ludmila L. Cavalcante; Sam J. Lubner; Daniel Mulkerin; Noelle K. LoConte; Linda Clipson; Kristina A. Matkowskyj; Dustin A. Deming

When considering treatment options for patients with metastatic colorectal cancer (mCRC), molecular profiling has become a pivotal component in guiding clinical decisions. FOLFOX and FOLFIRI (fluorouracuil, leucovorin plus oxaliplatin or ininotecan, respectively) are the standard base regimens used for the treatment of mCRC. Biologic agents, such as the epidermal growth factor receptor (EGFR) targeted therapies, cetuximab and panitumumab and the vascular endothelial growth factor monoclonal antibody, bevacizumab, are safe and effective in the first-line setting. The most efficacious use of these agents in terms of timing and selection of the right patient population continues to be debated. Here we review multiple investigations into the effectiveness of treatment options as a function of the mutations present in colon cancers. Early studies have reported that KRAS mutations at exon 2 predict resistance to EGFR targeted therapies. More recently the data have expanded to include KRAS mutations at exons 3 and 4 and NRAS mutations at exons 2, 3 and 4 as well as other biomarkers including BRAF and PIK3CA, leading to the evolution of the treatment of mCRC to a more precision-based approach. As our understanding of relevant biomarkers increases, and data from both molecular profiling and treatment response become more readily available, treatment options will become more precise and their outcomes more effective.


PLOS ONE | 2013

mTOR inhibition elicits a dramatic response in PI3K-dependent colon cancers.

Dustin A. Deming; Alyssa A. Leystra; Mohammed Farhoud; Laura Nettekoven; Linda Clipson; Dawn M. Albrecht; Mary Kay Washington; Ruth Sullivan; Jamey P. Weichert; Richard B. Halberg

The phosphatidylinositide-3-kinase (PI3K) signaling pathway is critical for multiple cellular functions including metabolism, proliferation, angiogenesis, and apoptosis, and is the most commonly altered pathway in human cancers. Recently, we developed a novel mouse model of colon cancer in which tumors are initiated by a dominant active PI3K (FC PIK3ca*). The cancers in these mice are moderately differentiated invasive mucinous adenocarcinomas of the proximal colon that develop by 50 days of age. Interestingly, these cancers form without a benign intermediary or aberrant WNT signaling, indicating a non-canonical mechanism of tumorigenesis. Since these tumors are dependent upon the PI3K pathway, we investigated the potential for tumor response by the targeting of this pathway with rapamycin, an mTOR inhibitor. A cohort of FC PIK3ca* mice were treated with rapamycin at a dose of 6 mg/kg/day or placebo for 14 days. FDG dual hybrid PET/CT imaging demonstrated a dramatic tumor response in the rapamycin arm and this was confirmed on necropsy. The tumor tissue remaining after treatment with rapamycin demonstrated increased pERK1/2 or persistent phosphorylated ribosomal protein S6 (pS6), indicating potential resistance mechanisms. This unique model will further our understanding of human disease and facilitate the development of therapeutics through pharmacologic screening and biomarker identification.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Transformation of epithelial cells through recruitment leads to polyclonal intestinal tumors

Andrew T. Thliveris; Brittany Schwefel; Linda Clipson; Lauren Plesh; Christopher D. Zahm; Alyssa A. Leystra; Mary Kay Washington; Ruth Sullivan; Dustin A. Deming; Michael A. Newton; Richard B. Halberg

Intestinal tumors from mice and humans can have a polyclonal origin. Statistical analyses indicate that the best explanation for this source of intratumoral heterogeneity is the presence of interactions among multiple progenitors. We sought to better understand the nature of these interactions. An initial progenitor could recruit others by facilitating the transformation of one or more neighboring cells. Alternatively, two progenitors that are independently initiated could simply cooperate to form a single tumor. These possibilities were tested by analyzing tumors from aggregation chimeras that were generated by fusing together embryos with unequal predispositions to tumor development. Strikingly, numerous polyclonal tumors were observed even when one genetic component was highly, if not completely, resistant to spontaneous tumorigenesis in the intestine. Moreover, the observed number of polyclonal tumors could be explained by the facilitated transformation of a single neighbor within 144 μm of an initial progenitor. These findings strongly support recruitment instead of cooperation. Thus, it is conceivable that these interactions are necessary for tumors to thrive, so blocking them might be a highly effective method for preventing the formation of tumors in the intestine and other tissues.


Cancer Prevention Research | 2015

Colon Tumors with the Simultaneous Induction of Driver Mutations in APC, KRAS, and PIK3CA Still Progress through the Adenoma-to-carcinoma Sequence

Jamie N. Hadac; Alyssa A. Leystra; Terrah J. Paul Olson; Molly Maher; Susan Payne; Alexander Yueh; Alexander R. Schwartz; Dawn M. Albrecht; Linda Clipson; Cheri A. Pasch; Kristina A. Matkowskyj; Richard B. Halberg; Dustin A. Deming

Human colorectal cancers often possess multiple mutations, including three to six driver mutations per tumor. The timing of when these mutations occur during tumor development and progression continues to be debated. More advanced lesions carry a greater number of driver mutations, indicating that colon tumors might progress from adenomas to carcinomas through the stepwise accumulation of mutations following tumor initiation. However, mutations that have been implicated in tumor progression have been identified in normal-appearing epithelial cells of the colon, leaving the possibility that these mutations might be present before the initiation of tumorigenesis. We utilized mouse models of colon cancer to investigate whether tumorigenesis still occurs through the adenoma-to-carcinoma sequence when multiple mutations are present at the time of tumor initiation. To create a model in which tumors could concomitantly possess mutations in Apc, Kras, and Pik3ca, we developed a novel minimally invasive technique to administer an adenovirus expressing Cre recombinase to a focal region of the colon. Here, we demonstrate that the presence of these additional driver mutations at the time of tumor initiation results in increased tumor multiplicity and an increased rate of progression to invasive adenocarcinomas. These cancers can even metastasize to retroperitoneal lymph nodes or the liver. However, despite having as many as three concomitant driver mutations at the time of initiation, these tumors still proceed through the adenoma-to-carcinoma sequence. Cancer Prev Res; 8(10); 952–61. ©2015 AACR.


PLOS ONE | 2014

Phospholipid ether analogs for the detection of colorectal tumors.

Dustin A. Deming; Molly Maher; Alyssa A. Leystra; Joseph P. Grudzinski; Linda Clipson; Dawn M. Albrecht; Mary Kay Washington; Kristina A. Matkowskyj; Lance Hall; Sam J. Lubner; Jamey P. Weichert; Richard B. Halberg

The treatment of localized colorectal cancer (CRC) depends on resection of the primary tumor with adequate margins and sufficient lymph node sampling. A novel imaging agent that accumulates in CRCs and the associated lymph nodes is needed. Cellectar Biosciences has developed a phospholipid ether analog platform that is both diagnostic and therapeutic. CLR1502 is a near-infrared fluorescent molecule, whereas 124/131I-CLR1404 is under clinical investigation as a PET tracer/therapeutic agent imaged by SPECT. We investigated the use of CLR1502 for the detection of intestinal cancers in a murine model and 131I-CLR1404 in a patient with metastatic CRC. Mice that develop multiple intestinal tumors ranging from adenomas to locally advanced adenocarcinomas were utilized. After 96 hours post CLR1502 injection, the intestinal tumors were analyzed using a Spectrum IVIS (Perkin Elmer) and a Fluobeam (Fluoptics). The intensity of the fluorescent signal was correlated with the histological characteristics for each tumor. Colon adenocarcinomas demonstrated increased accumulation of CLR1502 compared to non-invasive lesions (total radiant efficiency: 1.76×1010 vs 3.27×109 respectively, p = 0.006). Metastatic mesenteric tumors and uninvolved lymph nodes were detected with CLR1502. In addition, SPECT imaging with 131I-CLR1404 was performed as part of a clinical trial in patients with advanced solid tumors. 131I-CLR1404 was shown to accumulate in metastatic tumors in a patient with colorectal adenocarcinoma. Together, these compounds might enhance our ability to properly resect CRCs through better localization of the primary tumor and improved lymph node identification as well as detect distant disease.


Oncogenesis | 2015

PIK3CA mutations can initiate pancreatic tumorigenesis and are targetable with PI3K inhibitors.

Susan Payne; Molly Maher; Nguyen H. Tran; D R Van De Hey; Tyler Foley; Alexander Yueh; Alyssa A. Leystra; Cheri A. Pasch; J J Jeffrey; Linda Clipson; Kristina A. Matkowskyj; Dustin A. Deming

Aberrations in the phosphoinositide 3-kinase (PI3K) signaling pathway have a key role in the pathogenesis of numerous cancers by altering cell growth, metabolism, proliferation and apoptosis. Interest in targeting the PI3K signaling cascade continues, as new agents are being clinically evaluated. PIK3CA mutations result in a constitutively active PI3K and are present in a subset of pancreatic cancers. Here we examine mutant PIK3CA-mediated pancreatic tumorigenesis and the response of PIK3CA mutant pancreatic cancers to dual PI3K/mammalian target of rapamycin (mTOR) inhibition. Two murine models were generated expressing a constitutively active PI3K within the pancreas. An increase in acinar-to-ductal metaplasia and pancreatic intraepithelial neoplasms (PanINs) was identified. In one model these lesions were detected as early as 10 days of age. Invasive pancreatic ductal adenocarcinoma developed in these mice as early as 20 days of age. These cancers were highly sensitive to treatment with dual PI3K/mTOR inhibition. In the second model, PanINs and invasive cancer develop with a greater latency owing to a lesser degree of PI3K pathway activation in this murine model. In addition to PI3K pathway activation, increased ERK1/2 signaling is common in human pancreatic cancers. Phosphorylation of ERK1/2 was also investigated in these models. Phosphorylation of ERK1/2 is demonstrated in the pre-neoplastic lesions and invasive cancers. This activation of ERK1/2 is diminished with dual PI3K/mTOR inhibition. In summary, PIK3CA mutations can initiate pancreatic tumorigenesis and these cancers are particularly sensitive to dual PI3K/mTOR inhibition. Future studies of PI3K pathway inhibitors for patients with PIK3CA mutant pancreatic cancers are warranted.

Collaboration


Dive into the Dustin A. Deming's collaboration.

Top Co-Authors

Avatar

Linda Clipson

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Kristina A. Matkowskyj

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Cheri A. Pasch

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Susan Payne

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Sam J. Lubner

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Alyssa A. Leystra

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Richard B. Halberg

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Dawn M. Albrecht

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Kyle D. Holen

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Noelle K. LoConte

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge