Network


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

Hotspot


Dive into the research topics where David J. Shields is active.

Publication


Featured researches published by David J. Shields.


PLOS ONE | 2015

Whole Exome Sequencing of Rapid Autopsy Tumors and Xenograft Models Reveals Possible Driver Mutations Underlying Tumor Progression.

Tao Xie; Monica Musteanu; Pedro P. Lopez-Casas; David J. Shields; Peter Olson; Paul A. Rejto; Manuel Hidalgo

Pancreatic Ductal Adenocarcinoma (PDAC) is a highly lethal malignancy due to its propensity to invade and rapidly metastasize and remains very difficult to manage clinically. One major hindrance towards a better understanding of PDAC is the lack of molecular data sets and models representative of end stage disease. Moreover, it remains unclear how molecularly similar patient-derived xenograft (PDX) models are to the primary tumor from which they were derived. To identify potential molecular drivers in metastatic pancreatic cancer progression, we obtained matched primary tumor, metastases and normal (peripheral blood) samples under a rapid autopsy program and performed whole exome sequencing (WES) on tumor as well as normal samples. PDX models were also generated, sequenced and compared to tumors. Across the matched data sets generated for three patients, there were on average approximately 160 single-nucleotide mutations in each sample. The majority of mutations in each patient were shared among the primary and metastatic samples and, importantly, were largely retained in the xenograft models. Based on the mutation prevalence in the primary and metastatic sites, we proposed possible clonal evolution patterns marked by functional mutations affecting cancer genes such as KRAS, TP53 and SMAD4 that may play an important role in tumor initiation, progression and metastasis. These results add to our understanding of pancreatic tumor biology, and demonstrate that PDX models derived from advanced or end-stage likely closely approximate the genetics of the disease in the clinic and thus represent a biologically and clinically relevant pre-clinical platform that may enable the development of effective targeted therapies for PDAC.


Cancer Research | 2011

Abstract 4504: PF-04449913, a small molecule inhibitor of Hedgehog signaling, is effective in inhibiting tumor growth in preclinical models

Amy Jackson-Fisher; Melissa McMahon; Justine L. Lam; Chunze Li; Lars D. Engstrom; Konstantinos Tsaparikos; David J. Shields; Douglas D. Fang; Maruja E. Lira; Zhou Zhu; Michael D. Robbins; Richard Schwab; Michael John Munchhof; Todd VanArsdale

Aberrant activation of the Hedgehog (Hh) signaling pathway has been implicated in several human cancers. Mutations in the Patched (PTCH1) gene are responsible for basal cell nevus syndrome, and are commonly found in sporadic basal cell carcinoma and in medulloblastoma. In this study we evaluated PF-04449913, an inhibitor of the Hh signaling pathway, in a Ptch1+/-p53 mouse model of medulloblastoma and in human patient derived xenograft models. Treatment of Ptch1+/-p53+/- or Ptch1+/-p53-/- medulloblastoma allografts with PF-04449913 produced potent dose-dependent inhibition of Hh pathway activity resulting in stable tumor regression. Using Gli1 transcript levels as a surrogate for Hh pathway activity, the pharmacodynamic effects of PF-04449913 were evaluated in medulloblastoma allografts following single dose and multi dose administrations of compound. PF-04449913 treated medulloblastoma allografts had reduced levels of Gli1 gene expression and down regulation of genes linked to the Hh signaling pathway. PF-04449913 was also effective when combined with a chemotherapeutic agent in a colon patient derived xenograft model and a pancreatic patient derived xenograft model, resulting in 63% and 73% tumor growth inhibition respectively. Collectively, our study demonstrates the therapeutic efficacy of a small molecule inhibitor of Hh pathway in preclinical models of multiple cancer types in either single or combination treatments. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4504. doi:10.1158/1538-7445.AM2011-4504


Cell Death & Differentiation | 2018

Therapeutic relevance of the PP2A-B55 inhibitory kinase MASTL/Greatwall in breast cancer

Mónica Álvarez-Fernández; María Sanz-Flores; Belén Sanz-Castillo; María Salazar-Roa; David Partida; Elisabet Zapatero-Solana; H. Raza Ali; Eusebio Manchado; Scott W. Lowe; Todd VanArsdale; David J. Shields; Carlos Caldas; Miguel Quintela-Fandino; Marcos Malumbres

PP2A is a major tumor suppressor whose inactivation is frequently found in a wide spectrum of human tumors. In particular, deletion or epigenetic silencing of genes encoding the B55 family of PP2A regulatory subunits is a common feature of breast cancer cells. A key player in the regulation of PP2A/B55 phosphatase complexes is the cell cycle kinase MASTL (also known as Greatwall). During cell division, inhibition of PP2A-B55 by MASTL is required to maintain the mitotic state, whereas inactivation of MASTL and PP2A reactivation is required for mitotic exit. Despite its critical role in cell cycle progression in multiple organisms, its relevance as a therapeutic target in human cancer and its dependence of PP2A activity is mostly unknown. Here we show that MASTL overexpression predicts poor survival and shows prognostic value in breast cancer patients. MASTL knockdown or knockout using RNA interference or CRISPR/Cas9 systems impairs proliferation of a subset of breast cancer cells. The proliferative function of MASTL in these tumor cells requires its kinase activity and the presence of PP2A-B55 complexes. By using a new inducible CRISPR/Cas9 system in breast cancer cells, we show that genetic ablation of MASTL displays a significant therapeutic effect in vivo. All together, these data suggest that the PP2A inhibitory kinase MASTL may have both prognostic and therapeutic value in human breast cancer.


Nature Medicine | 2018

Combinatorial inhibition of PTPN12-regulated receptors leads to a broadly effective therapeutic strategy in triple-negative breast cancer

Amritha Nair; Hsiang Ching Chung; Tingting Sun; Siddhartha Tyagi; Lacey E. Dobrolecki; Rocio Dominguez-Vidana; Sarah J. Kurley; Mayra Orellana; Alexander Renwick; David M. Henke; Panagiotis Katsonis; Earlene M. Schmitt; Doug W. Chan; Hui Li; Sufeng Mao; Ivana Petrovic; Chad J. Creighton; Carolina Gutierrez; Julien Dubrulle; Fabio Stossi; Jeffrey W. Tyner; Olivier Lichtarge; Charles Y. Lin; Bing Zhang; Kenneth L. Scott; Susan G. Hilsenbeck; Jin-Peng Sun; Xiao Yu; C. Kent Osborne; Rachel Schiff

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer diagnosed in more than 200,000 women each year and is recalcitrant to targeted therapies. Although TNBCs harbor multiple hyperactive receptor tyrosine kinases (RTKs), RTK inhibitors have been largely ineffective in TNBC patients thus far. We developed a broadly effective therapeutic strategy for TNBC that is based on combined inhibition of receptors that share the negative regulator PTPN12. Previously, we and others identified the tyrosine phosphatase PTPN12 as a tumor suppressor that is frequently inactivated in TNBC. PTPN12 restrains several RTKs, suggesting that PTPN12 deficiency leads to aberrant activation of multiple RTKs and a co-dependency on these receptors. This in turn leads to the therapeutic hypothesis that PTPN12-deficient TNBCs may be responsive to combined RTK inhibition. However, the repertoire of RTKs that are restrained by PTPN12 in human cells has not been systematically explored. By methodically identifying the suite of RTK substrates (MET, PDGFRβ, EGFR, and others) inhibited by PTPN12, we rationalized a combination RTK-inhibitor therapy that induced potent tumor regression across heterogeneous models of TNBC. Orthogonal approaches revealed that PTPN12 was recruited to and inhibited these receptors after ligand stimulation, thereby serving as a feedback mechanism to limit receptor signaling. Cancer-associated mutation of PTPN12 or reduced PTPN12 protein levels diminished this feedback mechanism, leading to aberrant activity of these receptors. Restoring PTPN12 protein levels restrained signaling from RTKs, including PDGFRβ and MET, and impaired TNBC survival. In contrast with single agents, combined inhibitors targeting the PDGFRβ and MET receptors induced the apoptosis in TNBC cells in vitro and in vivo. This therapeutic strategy resulted in tumor regressions in chemo-refractory patient-derived TNBC models. Notably, response correlated with PTPN12 deficiency, suggesting that impaired receptor feedback may establish a combined addiction to these proto-oncogenic receptors. Taken together, our data provide a rationale for combining RTK inhibitors in TNBC and other malignancies that lack receptor-activating mutations.


Embo Molecular Medicine | 2018

A versatile drug delivery system targeting senescent cells

Daniel Muñoz-Espín; Miguel Rovira; Irene Galiana; Cristina Giménez; Beatriz Lozano-Torres; Marta Paez-Ribes; Susana Llanos; Selim Chaib; Maribel Muñoz-Martin; Alvaro C Ucero; Guillermo Garaulet; Francisca Mulero; Stephen Dann; Todd VanArsdale; David J. Shields; Andrea Bernardos; José Ramón Murguía; Ramón Martínez-Máñez; Manuel Serrano

Senescent cells accumulate in multiple aging‐associated diseases, and eliminating these cells has recently emerged as a promising therapeutic approach. Here, we take advantage of the high lysosomal β‐galactosidase activity of senescent cells to design a drug delivery system based on the encapsulation of drugs with galacto‐oligosaccharides. We show that gal‐encapsulated fluorophores are preferentially released within senescent cells in mice. In a model of chemotherapy‐induced senescence, gal‐encapsulated cytotoxic drugs target senescent tumor cells and improve tumor xenograft regression in combination with palbociclib. Moreover, in a model of pulmonary fibrosis in mice, gal‐encapsulated cytotoxics target senescent cells, reducing collagen deposition and restoring pulmonary function. Finally, gal‐encapsulation reduces the toxic side effects of the cytotoxic drugs. Drug delivery into senescent cells opens new diagnostic and therapeutic applications for senescence‐associated disorders.


bioRxiv | 2018

Palbociclib and fulvestrant act in synergy to modulate central carbon metabolism in breast cancer cells

Benedikt Warth; Amelia Palermo; Nicholas J. W. Rattray; Nathan V. Lee; Zhou Zhu; Linh Hoang; Anthony Mazurek; Stephen Dann; Todd VanArsdale; Valeria R. Fantin; David J. Shields; Gary Siuzdak; Caroline H. Johnson

Palbociclib, is a selective inhibitor of cyclin-dependent kinases 4 and 6 and used as a first-line treatment for patients with estrogen receptor positive breast cancer. It has been shown that patients have improved progression-free survival when treated in combination with fulvestrant, an estrogen receptor antagonist. However, the mechanisms for this survival advantage are not known. We sought to analyze metabolic and transcriptomic changes in MCF-7 adenocarcinoma breast cancer cells following single and combined treatments to determine if selective metabolic pathways are targeted during combination therapy. Our results showed that individually, the drugs caused metabolic disruption to the same metabolic pathways, however fulvestrant additionally attenuated the pentose phosphate pathway and the production of important coenzymes. A comprehensive effect was observed when the drugs were applied together, confirming the combinatory therapy′s synergism in the cell model. This study highlights the power of merging high-dimensional datasets to unravel mechanisms involved in cancer metabolism and therapy. Highlights ○ First study employing multi-omics to investigate combined therapy on breast cancer cells ○ Fulvestrant attenuates the pentose phosphate pathway and coenzyme production ○ Synergism of palbociclib and fulvestrant was confirmed in vitro ○ Altered key pathways have been identified eTOC Blurb Johnson et al. applied an innovative multi-omics approach to decipher metabolic pathways affected by single versus combination dosing of palbociclib and fulvestrant in estrogen receptor positive breast cancer. Key metabolites and genes were correlated within metabolic pathways and shown to be involved in the drugs′ synergism.


Poster Presentation: Cancer Cell Biology | 2018

PO-028 Effectiveness and molecular basis of CDK4/6 inhibition in combination with taxanes in pancreatic cancer

B Salvador; M Álvarez; C Menéndez; P López-Casas; Todd VanArsdale; David J. Shields; Manuel Hidalgo; M Malumbres

Introduction Pancreatic Ductal Adenocarcinoma (PDAC) is among the deadliest human cancers with a 5 year survival rate of less than 5% using the standard of care gemcitabine/nab-paclitaxel. The most frequently disrupted genes in PDACs are first K-RAS and second CDKN2A, which encodes the cyclin-dependent kinase (CDK)4/6 inhibitor p16. Recently, CDK4/6 inhibitors have been approved for breast cancer treatment, and preclinical assays for PDAC are giving promising results. Material and methods PDAC Patient-Derived Xenografts (PDX) models and PDX-derived cell lines were used for in vivo and in vitro studies, respectively. Cellular studies were performed using proliferation and cell cycle assays in combination with flow cytometry, immunoblotting, fluorescence microscopy and live cell imaging techniques. Drug treatments were performed with the CDK4/6 inhibitor PD-0332991 (Palbociclib), and with Paclitaxel (Taxol) or Nab-Paclitaxel (Abraxane) for in vitro and in vivo studies, respectively. Results and discussions Treatment of different PDX-derived cell lines with the combination of taxanes and CDK4/6 inhibitors resulted in a higher anti-proliferative effect than both drugs used as single agent. Cell cycle studies showed that inhibition of CDK4/6 prevented recovery from treatment with taxol. At the molecular level we found that the combined treatment induced a clear interruption in retinoblastoma pathway, even higher than CDK4/6 inhibition in monotherapy. Gene expression profiles comparing single versus combined treatment are currently being performed to further understand the molecular basis underlying the effectiveness of this type of treatment. Moreover, to assess the efficacy of this new combined treatment in vivo, we treated nine PDAC PDX models with PD-0332991 and nab-paclitaxel, following the same schedule. Importantly, eight of them presented an increased tumour growth inhibition in the combination with respect to the monotherapies. Conclusion Although the molecular mechanism underlying the effectiveness of this treatment is not completely understood yet, our data suggest a good therapeutic value for the combination of CDK4/6 inhibitors and taxanes in PDAC treatment.


Molecular Cancer Research | 2016

Abstract A08: Mechanistic basis of Palbociclib combinatorial activity in ER+ breast cancer and non-breast indications

Stephen Dann; John Chionis; Liu Choating; Enhong Chen; Ping Wei; Koleen Eisele; David J. Shields; Paul A. Rejto; Todd VanArsdale

Phosphorylation of the retinoblastoma protein (Rb) by cyclin-dependent kinases 4 and 6 (CDK4/6) is a critical checkpoint for G1/S cell cycle progression and commitment to cellular proliferation. Human malignancies often subvert these control mechanisms through a range of genetic and biochemical adaptations. Accordingly, tumors that depend on CDK4/6 activity for proliferation and survival are particularly sensitive to inhibition of this pathway by palbociclib (Ibrance™), a highly selective inhibitor of CDK4/6 kinase activities. Treatment regimen of palbociclib with letrozole significantly improved progression-free survival in a randomized phase 2 study of women with advanced estrogen receptor-positive (ER+), HER2-negative breast cancer. Likewise, in ER+ breast cancer models palbociclib and estrogen antagonists combine for greater anti-proliferative activity, increased hallmarks of cellular senescence and prolonged durability of response following drug removal. Dual inhibition of CDK4/6 and ER signaling demonstrated robust anti-tumor activity in xenograft studies. The addition of Palbociclib to other targeted therapeutics elicits improved activity in pre-clinical models of several non-breast indications and these effects also manifest through modulation of cellular proliferation, senescence and growth arrest. Data will be presented on the molecular basis of combination benefit with Palbociclib in ER+ breast and other oncology indications. Citation Format: Stephen Dann, John Chionis, Liu Choating, Enhong Chen, Ping Wei, Koleen Eisele, David J. Shields, Paul A. Rejto, Todd VanArsdale. Mechanistic basis of Palbociclib combinatorial activity in ER+ breast cancer and non-breast indications. [abstract]. In: Proceedings of the AACR Precision Medicine Series: Cancer Cell Cycle - Tumor Progression and Therapeutic Response; Feb 28-Mar 2, 2016; Orlando, FL. Philadelphia (PA): AACR; Mol Cancer Res 2016;14(11_Suppl):Abstract nr A08.


Clinical Cancer Research | 2016

Abstract A36: Assessment of effectiveness and molecular markers of CDK4/6 inhibitor Palbociclib in Pancreatic Ductal Adenocarcinomas

Beatriz Salvador; Pedro P. Lopez-Casas; Camino Menéndez; Natalia Baños; Francesca Sarno; Yin Min-Jean; Peter Olson; Todd VanArsdale; David J. Shields; Manuel Hidalgo

Pancreatic Ductal Adenocarcinoma (PDAC) continues to be the deadliest human cancer with a 5-year survival rate of 7%. One of the current standards of care for advanced PDAC is gemcitabine plus nab-paclitaxel, a regimen our group helped develop. Finding new agents to combine with this regimen remains necessary, particularly using drug targeting strategies. At the molecular level, genetic and genomic profiling identified CDKN2A as a very frequently disrupted gene in PDAC ( In the current study, we aimed to evaluate the effectiveness of palbociclib monotherapy and in combination with nab-paclitaxel and gemcitabine/nab-paclitaxel in a cohort of PDACs patient derived xenograft (PDX) models. PDX models obtained from late stage PDAC patients were treated with palbociclib alone and in combination with gemcitabine/nab-paclitaxel. The majority of models displayed >50% tumor growth inhibition (TGI) following treatment with single agent palbociclib. Treatment with palbociclib plus nab-paclitaxel or palbociclib plus gemcitabine/nab-paclitaxel increased TGI to a greater degree than the gemcitabine/nab-paclitaxel combination and also increased the duration of response as compared to the standard of care therapy. Moving forward, the palbociclib/nab-paclitaxel combination will be evaluated in a clinical trial for PDAC patients. We are currently expanding our analyses to a larger cohort of PDX models and performing molecular characterization of these models in order to gain insights on biomarkers and mechanism of action of the drug combinations in PDAC. Citation Format: Beatriz Salvador, Pedro P. Lopez-Casas, Camino Menendez, Natalia Banos, Francesca Sarno, Yin Min-Jean, Peter Olson, Todd VanArsdale, David J. Shields, Manuel Hidalgo. Assessment of effectiveness and molecular markers of CDK4/6 inhibitor Palbociclib in Pancreatic Ductal Adenocarcinomas. [abstract]. In: Proceedings of the AACR Special Conference: Patient-Derived Cancer Models: Present and Future Applications from Basic Science to the Clinic; Feb 11-14, 2016; New Orleans, LA. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(16_Suppl):Abstract nr A36.


Cancer Research | 2016

Abstract 2740: Mechanistic basis of Palbociclib combinatorial activity in ER+ breast cancer and non-breast indications

Stephen Dann; Jing Yuan; John Chionis; Chaoting Liu; Tao Xie; Nathan V. Lee; Enhong Chen; Ping Wei; Paul A. Rejto; David J. Shields; Todd VanArsdale

Phosphorylation of the retinoblastoma protein (Rb) by cyclin-dependent kinases 4 and 6 (CDK4/6) is a critical checkpoint for G1/S cell cycle progression and commitment to cellular proliferation. Human malignancies often subvert these control mechanisms through a range of genetic and biochemical adaptations. Accordingly, tumors that depend on CDK4/6 activity for proliferation and survival are particularly sensitive to inhibition of this pathway by palbociclib (IbranceTM), a highly selective inhibitor of CDK4/6 kinase activities. Treatment regimen of palbociclib with letrozole significantly improved progression-free survival in a randomized phase 2 study of women with advanced estrogen receptor-positive (ER+), HER2-negative breast cancer. Likewise, in ER+ breast cancer models palbociclib and estrogen antagonists combine for greater anti-proliferative activity, increased hallmarks of cellular senescence and prolonged durability of response following drug removal. Dual inhibition of CDK4/6 and ER signaling demonstrated robust anti-tumor activity in xenograft studies. The addition of Palbociclib to other targeted therapeutics elicits improved activity in pre-clinical models of several non-breast indications and these effects also manifest through modulation of cellular proliferation, senescence and growth arrest. Data will be presented on the molecular basis of combination benefit with Palbociclib in ER+ breast and other oncology indications. Citation Format: Stephen Dann, Jing Yuan, John Chionis, Chaoting Liu, Tao Xie, Nathan V. Lee, Enhong Chen, Ping Wei, Paul A. Rejto, David J. Shields, Todd VanArsdale. Mechanistic basis of Palbociclib combinatorial activity in ER+ breast cancer and non-breast indications. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2740.

Collaboration


Dive into the David J. Shields's collaboration.

Top Co-Authors

Avatar

Manuel Hidalgo

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge