Network


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

Hotspot


Dive into the research topics where Nathan G. Dolloff is active.

Publication


Featured researches published by Nathan G. Dolloff.


Science Translational Medicine | 2013

Dual inactivation of Akt and ERK by TIC10 signals Foxo3a nuclear translocation, TRAIL gene induction, and potent antitumor effects.

Joshua E. Allen; Gabriel S. Krigsfeld; Patrick A. Mayes; Luv Patel; David T. Dicker; Akshal S. Patel; Nathan G. Dolloff; Evangelos Messaris; Kimberly A. Scata; Wenge Wang; Jun Ying Zhou; Gen Sheng Wu; Wafik S. El-Deiry

TIC10 is a small molecule that activates Foxo3a through dual inactivation of Akt and ERK, up-regulates the expression of the TRAIL gene, an endogenous tumor suppressor, and effectively improves the therapeutic properties and utility of TRAIL as an anticancer therapy. TIC’ing Up the TRAIL TRAIL is a naturally occurring tumor suppressor: It stimulates cell death pathways in a variety of human cancers and thus has been a popular target for the development of anticancer drugs. Previous TRAIL-targeting strategies include synthesis of the recombinant protein and stimulatory antibodies. All of these agents exhibit some of the typical drawbacks of protein-based therapeutics, such as short half-lives and a need to administer the drugs directly into the bloodstream or even into the tumor. Now, Allen and colleagues have discovered a drug, TIC10, which can stimulate production of TRAIL while avoiding the shortcomings of protein-based therapies. The authors demonstrated that TIC10 can increase TRAIL and stimulate the death of multiple types of human cancer cells both in culture and in mice. The drug was equally effective when given orally or intravenously and effectively penetrated the blood-brain barrier to target glioblastoma, a difficult-to-treat brain tumor. Whereas recombinant TRAIL displayed a short half-life of ~30 min, TIC10 activity persisted in the mice for days, allowing for once-a-week dosing. Toxicity analysis in mice showed no detectable adverse effects from treatment with TIC10. The authors also showed that TIC10 boosts TRAIL function through inactivation of the Akt and MEK signaling proteins, which results in translocation of the transcription factor Foxo3a into the cell nucleus, where it stimulates TRAIL gene expression. Before TIC10 can be used to treat patients, the drug will need to be tested in clinical trials to confirm safety and efficacy results from mouse studies. In addition, further work is needed to determine the mechanism by which TIC10 causes the dephosphorylation and resulting inactivation of Akt and MEK. However, the discovery of TIC10 clears a path to versatile TRAIL-based cancer therapies. Recombinant tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) is an antitumor protein that is in clinical trials as a potential anticancer therapy but suffers from drug properties that may limit efficacy such as short serum half-life, stability, cost, and biodistribution, particularly with respect to the brain. To overcome such limitations, we identified TRAIL-inducing compound 10 (TIC10), a potent, orally active, and stable small molecule that transcriptionally induces TRAIL in a p53-independent manner and crosses the blood-brain barrier. TIC10 induces a sustained up-regulation of TRAIL in tumors and normal cells that may contribute to the demonstrable antitumor activity of TIC10. TIC10 inactivates kinases Akt and extracellular signal–regulated kinase (ERK), leading to the translocation of Foxo3a into the nucleus, where it binds to the TRAIL promoter to up-regulate gene transcription. TIC10 is an efficacious antitumor therapeutic agent that acts on tumor cells and their microenvironment to enhance the concentrations of the endogenous tumor suppressor TRAIL.


Cell Cycle | 2009

The p53 target Plk2 interacts with TSC proteins impacting mTOR signaling, tumor growth and chemosensitivity under hypoxic conditions

Elizabeth M. Matthew; Lori S. Hart; Aristotelis Astrinidis; Arunasalam Navaraj; Nathan G. Dolloff; David T. Dicker; Elizabeth P. Henske; Wafik S. El-Deiry

Tuberous sclerosis complex 1 (TSC1) inhibits mammalian target of rapamycin (mTOR), a central promotor of cell growth and proliferation. The protein product of the TSC1 gene, hamartin (referred to as TSC1) is known to interact with Polo-like kinase 1 (Plk1) in a cell cycle regulated, phosphorylation-dependent manner. We hypothesized that the p53 target gene, Plk2, is a tumor suppressor, mediating its tumor suppressor function through interactions with TSC1 that facilitate TSC1/2 restraint of mTOR under hypoxic stress. We found that human lung tumor cells deficient in Plk2 grew larger than control tumors, and that Plk2 interacts with endogenous TSC1 protein. Additionally, C-terminal Plk2-GST fusion protein bound both TSC1 and TSC2 proteins. TSC1 levels were elevated in response to Adriamycin and cells transiently over-expressing Plk2 demonstrated decreased phosphorylation of the downstream target of mTOR, ribosomal protein p70S6 kinase during hypoxia. Plk2 levels were inversely correlated with cytoplasmic p70S6K phosphorylation. Plk2 levels did not increase in response to DNA damage (Adriamycin, CPT-11) when HCT 116 and H460 cells were exposed to hypoxia. TSC1-deficient mouse embryonic fibroblasts with TSC1 added back demonstrated decreased S6K phosphorylation, which was further decreased when Plk2 was transiently over-expressed. Interestingly, under normoxia, Plk2 deficient tumor cells demonstrated increased apoptosis in response to various chemotherapeutic agents including CPT-11 but increased resistance to apoptotic death after CPT-11 treatment under hypoxia, and tumor xenografts comprised of these Plk2-deficient cells were resistant to CPT-11. Our results point to a novel Plk2-TSC1 interaction with effects on mTOR signaling during hypoxia, and tumor growth that may enable targeting Plk2 signaling in cancer therapy.


Cell Cycle | 2011

Human colon cancer stem cells are enriched by insulin-like growth factor-1 and are sensitive to figitumumab

Lori S. Hart; Nathan G. Dolloff; David T. Dicker; Constantinos Koumenis; James G. Christensen; Adda Grimberg; Wafik S. El-Deiry

Cancer stem cells (CSCs) are recognized as contributors to cancer progression and therapeutic resistance in liquid and solid malignancies. We analyzed a panel of human colon cancer cell lines for CSC populations by side population and aldehyde dehydrogenase activity. IGF-1 enriches these putative colon CSC populations in a β-catenin-dependent manner. Chemical inhibition of Akt depletes SP cells, and conversely, the overexpression of a constitutively active mutant version of Akt is sufficient to enrich CSC populations. CP-751,871, a fully human antibody with specificity to the IGF-1 receptor, is currently being tested in clinical trials for a variety of solid tumors. CP-751,871 reduces CSC populations in colon cancer cell lines in vitro and reduces tumor growth in vivo. We have identified a novel role for IGF-1 in the enrichment of chemo-resistant CSC populations. Our results suggest that CP-751,871 has preferential activity against putative CSC populations and, therefore, may complement current standard chemotherapeutic regimens that target cycling cells.


Molecular Cancer Therapeutics | 2013

Profiling Bortezomib Resistance Identifies Secondary Therapies in a Mouse Myeloma Model

Holly A.F. Stessman; Linda B. Baughn; Aaron L. Sarver; Tian Xia; Raamesh Deshpande; Aatif Mansoor; Susan A. Walsh; John Sunderland; Nathan G. Dolloff; Michael A. Linden; Fenghuang Zhan; Siegfried Janz; Chad L. Myers; Brian Van Ness

Multiple myeloma is a hematologic malignancy characterized by the proliferation of neoplastic plasma cells in the bone marrow. Although the first-to-market proteasome inhibitor bortezomib (Velcade) has been successfully used to treat patients with myeloma, drug resistance remains an emerging problem. In this study, we identify signatures of bortezomib sensitivity and resistance by gene expression profiling (GEP) using pairs of bortezomib-sensitive (BzS) and bortezomib-resistant (BzR) cell lines created from the Bcl-XL/Myc double-transgenic mouse model of multiple myeloma. Notably, these BzR cell lines show cross-resistance to the next-generation proteasome inhibitors, MLN2238 and carfilzomib (Kyprolis) but not to other antimyeloma drugs. We further characterized the response to bortezomib using the Connectivity Map database, revealing a differential response between these cell lines to histone deacetylase (HDAC) inhibitors. Furthermore, in vivo experiments using the HDAC inhibitor panobinostat confirmed that the predicted responder showed increased sensitivity to HDAC inhibitors in the BzR line. These findings show that GEP may be used to document bortezomib resistance in myeloma cells and predict individual sensitivity to other drug classes. Finally, these data reveal complex heterogeneity within multiple myeloma and suggest that resistance to one drug class reprograms resistant clones for increased sensitivity to a distinct class of drugs. This study represents an important next step in translating pharmacogenomic profiling and may be useful for understanding personalized pharmacotherapy for patients with multiple myeloma. Mol Cancer Ther; 12(6); 1140–50. ©2013 AACR.


Cancer Biology & Therapy | 2011

Quinacrine sensitizes hepatocellular carcinoma cells to TRAIL and chemotherapeutic agents.

Wenge Wang; Jean-Nicolas Gallant; Sharyn I. Katz; Nathan G. Dolloff; Charles D. Smith; Junaid Abdulghani; Joshua E. Allen; David T. Dicker; Bo Hong; Arunasalam Navaraj; Wafik S. El-Deiry

Quinacrine has been widely explored in treatment of malaria, giardiasis, and rheumatic diseases. We find that quinacrine stabilizes p53 and induces p53-dependent and independent cell death. Treatment by quinacrine alone at concentrations of 10–20 mM for 1–2 d cannot kill hepatocellular carcinoma cells, such as HepG2, Hep3B, Huh7, which are also resistant to TRAIL. However, quinacrine renders these cells sensitive to treatment by TRAIL. Co-treatment of these cells with quinacrine and TRAIL induces overwhelming cell death within 3–4 h. Levels of DR5, a pro-apoptotic death receptor of TRAIL, are increased upon treatment with quinacrine, while levels of Mcl-1, an anti-apoptotic member of the Bcl-2 family, are decreased. While the synergistic effect of quinacrine with TRAIL appears to be in part independent of p53, knockdown of p53 in HepG2 cells by siRNA results in more cell death after treatment by quinacrine and TRAIL. The mechanism by which quinacrine sensitizes hepatocellular carcinoma cells to TRAIL and chemotherapies, and the potential for clinical application currently are being further explored. Lastly, quinacrine synergizes with chemotherapeutics, such as adriamycin, 5-FU, etoposide, CPT11, sorafenib, and gemcitabine, in killing hepatocellular carcinoma cells in vitro and the drug enhances the activity of sorafenib to delay tumor growth in vivo.


Cancer Research | 2007

Human Bone Marrow Activates the Akt Pathway in Metastatic Prostate Cells through Transactivation of the α-Platelet–Derived Growth Factor Receptor

Nathan G. Dolloff; Mike R. Russell; Nick Loizos; Alessandro Fatatis

The factors regulating the bone tropism of disseminated prostate cancer cells are still vaguely defined. We report that prostate cancer cells that metastasize to the skeleton respond to human bone marrow with a robust stimulation of the phosphatidylinositol 3-kinase/Akt pathway, whereas prostate cells that lack bone-metastatic potential respond negligibly. The majority of this Akt activation is dependent on α-platelet–derived growth factor receptor (α-PDGFR) signaling, which was shown using the small-molecule inhibitor of PDGFR signaling AG1296. Low concentrations of PDGF-AA and PDGF-BB found in bone marrow aspirates, which were detected by ELISA, do not account for the high levels of α-PDGFR signaling. Additionally, neutralizing PDGF binding using a α-PDGFR–specific antibody (IMC-3G3) failed to produce a significant inhibition of bone marrow–induced Akt activation. However, the inhibitory effect of IMC-3G3 rivaled that of AG1296 when incubation was done under conditions that stimulated α-PDGFR internalization. We conclude that α-PDGFR is activated by multiple soluble factors contained within human bone marrow, in addition to its natural ligands, and this transactivation is dependent on receptor localization to the plasma membrane. Therefore, α-PDGFR expression may provide select prostate phenotypes with a growth advantage within the bone microenvironment. [Cancer Res 2007;67(2):555–62]


Cell Cycle | 2013

CDK1 stabilizes HIF-1α via direct phosphorylation of Ser668 to promote tumor growth

Noel A. Warfel; Nathan G. Dolloff; David T. Dicker; Jozef Malysz; Wafik S. El-Deiry

Hypoxia-inducible factor 1 (HIF-1) is a major mediator of tumor physiology, and its activation is correlated with tumor progression, metastasis, and therapeutic resistance. HIF-1 is activated in a broad range of solid tumors due to intratumoral hypoxia or genetic alterations that enhance its expression or inhibit its degradation. As a result, decreasing HIF-1α expression represents an attractive strategy to sensitize hypoxic tumors to anticancer therapies. Here, we show that cyclin-dependent kinase 1 (CDK1) regulates the expression of HIF-1α, independent of its known regulators. Overexpression of CDK1 and/or cyclin B1 is sufficient to stabilize HIF-1α under normoxic conditions, whereas inhibition of CDK1 enhances the proteasomal degradation of HIF-1α, reducing its half-life and steady-state levels. In vitro kinase assays reveal that CDK1 directly phosphorylates HIF-1α at a previously unidentified regulatory site, Ser668. HIF-1α is stabilized under normoxic conditions during G2/M phase via CDK1-mediated phosphorylation of Ser668. A phospho-mimetic construct of HIF-1α at Ser668 (S668E) is significantly more stable under both normoxic and hypoxic conditions, resulting in enhanced transcription of HIF-1 target genes and increased tumor cell invasion and migration. Importantly, HIF-1α (S668E) displays increased tumor angiogenesis, proliferation, and tumor growth in vivo compared with wild-type HIF-1α. Thus, we have identified a novel link between CDK1 and HIF-1α that provides a potential molecular explanation for the elevated HIF-1 activity observed in primary and metastatic tumors, independent of hypoxia, and offers a molecular rationale for the clinical translation of CDK inhibitors for use in tumors with constitutively active HIF-1.


Clinical Lymphoma, Myeloma & Leukemia | 2010

Beyond the CRAB Symptoms: A Study of Presenting Clinical Manifestations of Multiple Myeloma

Giampaolo Talamo; Umar Farooq; Maurizio Zangari; Jason Liao; Nathan G. Dolloff; Thomas P. Loughran; Elliot Epner

BACKGROUND Although the typical clinical manifestations of multiple myeloma (MM) are summarized by the CRAB symptoms (hypercalcemia, renal insufficiency, anemia, and bone lesions), a significant proportion of patients with MM present with a variety of other clinical manifestations. We conducted a study evaluating the presenting symptoms that led to the diagnosis of MM. PATIENTS AND METHODS We conducted a retrospective review of 170 consecutive patients with MM seen at the Penn State Hershey Cancer Institute. RESULTS Among patients with symptomatic MM, 74% presented with CRAB symptoms, 20% presented with non-CRAB manifestations, and 6% had both clinical features. Ten categories of non-CRAB manifestations were found, in order of decreasing frequency: neuropathy (because of spinal cord compression, nerve root compression, or peripheral neuropathy), extramedullary involvement, hyperviscosity syndrome, concomitant amyloidosis (eg, nephrotic syndrome or cardiopathy), hemorrhage/coagulopathy, systemic symptoms (eg, fever or weight loss), primary plasma cell leukemia, infections, cryoglobulinemia, and secondary gout. Kaplan-Meier estimates of survival in patients with non-CRAB manifestations did not show a significant difference from the survival of patients presenting with CRAB symptoms. CONCLUSION Presenting symptoms of MM may be grouped in a total of 14 categories, 4 for the CRAB and 10 for the less common non-CRAB features. Grouped together, non-CRAB manifestations do not appear to confer a negative effect on the prognosis of patients with MM.


Science Translational Medicine | 2011

Off-target lapatinib activity sensitizes colon cancer cells through TRAIL death receptor up-regulation.

Nathan G. Dolloff; Patrick A. Mayes; Lori S. Hart; David T. Dicker; Robin Humphreys; Wafik S. El-Deiry

The breast cancer drug lapatinib increases the sensitivity of colon cancer cells to apoptosis through a HER2- and EGFR-independent mechanism. Coaxing Colon Cancer Cells to an Easy Death In a utopian future, physicians will have a well-stocked arsenal from which to choose the ideal drug for every subtype of cancer. One essential weapon will be agents that trigger apoptosis in cancer cells, overriding their reluctance to die. These will include molecules that activate TRAIL (tumor necrosis factor–related apoptosis-inducing ligand) death receptors, which normally initiate the so-called extrinsic apoptotic pathway, causing cell death. Dolloff et al. have found a way to boost the ability of colon cancer cells to die in response to TRAIL, by pretreating with lapatinib, a drug already approved for use in breast cancer. The sensitizing effect results from actions of the drug other than its ability to inhibit HER2/EGFR (epidermal growth factor receptor family members), its targets in breast cancer. In a panel of TRAIL-resistant colon cancer cells, 2 days of pretreatment with lapatinib sensitized the cells to a subsequent 24-hour treatment with TRAIL, causing substantial caspase activation and cell death. This combined therapy could also suppress tumor growth in mice. But the effectiveness of TRAIL may be limited in the clinic because of its brief half-life. Of higher interest are the antibodies mapatumumab and lexatumumab, which target TRAIL death receptors and are currently being tested in clinical trials. Indeed, these therapeutic antibodies too induce apoptosis more readily after pretreatment of cancer cells with lapatinib. What does lapatinib do to coax these cells to die? Measurements of the levels of the death receptors themselves, DR4 and DR5, show that they are increased after lapatinib treatment via induction of the c-Jun N-terminal kinase (JNK)/c-Jun/activating protein–1 (AP-1) pathway. Although lapatinib was designed as an EGFR- and HER2-targeted agent, the authors suspected that it was not acting through these known targets to promote TRAIL receptor–induced apoptosis. The concentrations required were higher than needed for HER2/EGFR inhibition. Indeed, lapatinib up-regulated TRAIL death receptors and enhanced TRAIL sensitivity in cells that lack EGFR; moreover, the mere inhibition of EGFR and HER2 with other selective inhibitors was not sufficient to enhance TRAIL-induced cancer cell death. If confirmed by clinical validation, this newly described, non-EGFR/HER2–mediated action of lapatinib in sensitizing colon cancer cells to apoptotic stimuli will qualify this drug for a new place among anticancer munitions. Lapatinib, a dual HER2/EGFR (human epidermal growth factor receptor 2/epidermal growth factor receptor) inhibitor, is a recently approved targeted therapy for metastatic breast cancer. Because lapatinib enhances the efficacy of the chemotherapeutic agent capecitabine in breast cancer patients, we tested whether lapatinib also enhances the activity of anticancer agents in colorectal cancer. We found that lapatinib improved the proapoptotic effects of tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) and two TRAIL receptor agonists, the antibodies mapatumumab and lexatumumab. Tumors from mice treated with a combination of lapatinib and TRAIL exhibited more immunostaining for cleaved caspase-8, a marker of the extrinsic cell death pathway, than did tumors from mice treated with lapatinib or TRAIL alone. Furthermore, combination therapy suppressed tumor growth more effectively than either agent alone. Lapatinib up-regulated the proapoptotic TRAIL death receptors DR4 and DR5, leading to more efficient induction of apoptosis in the presence of TRAIL receptor agonists. This activity of lapatinib was independent of EGFR and HER2. The off-target induction of DR5 by lapatinib resulted from activation of the c-Jun amino-terminal kinase (JNK)/c-Jun signaling axis. This activity of lapatinib on TRAIL death receptor expression and signaling may confer therapeutic benefit when increased doses of lapatinib are used in combination with TRAIL receptor–activating agents.


Nature Cell Biology | 2008

ERK and MDM2 prey on FOXO3a

Wensheng Yang; Nathan G. Dolloff; Wafik S. El-Deiry

Constitutively active ERK signalling stimulates cell proliferation, thereby contributing to tumorigenesis. We now learn that ERK activity induces phosphorylation and MDM2-mediated degradation of the tumour-suppressing transcription factor FOXO3a, thus gaining new information on valuable targets for human cancer therapeutic intervention.

Collaboration


Dive into the Nathan G. Dolloff's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Giampaolo Talamo

Penn State Milton S. Hershey Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lori S. Hart

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Joshua E. Allen

Penn State Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Patrick A. Mayes

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Arunasalam Navaraj

Penn State Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Jozef Malysz

Penn State Milton S. Hershey Medical Center

View shared research outputs
Top Co-Authors

Avatar

Robin Humphreys

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Wenge Wang

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge