Network


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

Hotspot


Dive into the research topics where Thomas L. Sims is active.

Publication


Featured researches published by Thomas L. Sims.


Clinical Cancer Research | 2007

Bevacizumab-induced transient remodeling of the vasculature in neuroblastoma xenografts results in improved delivery and efficacy of systemically administered chemotherapy.

Paxton V. Dickson; John B. Hamner; Thomas L. Sims; Charles H. Fraga; Catherine Y.C. Ng; Surender Rajasekeran; Nikolaus Hagedorn; M. Beth McCarville; Clinton F. Stewart; Andrew M. Davidoff

Purpose: Dysfunctional tumor vessels can be a significant barrier to effective cancer therapy. However, increasing evidence suggests that vascular endothelial growth factor (VEGF) inhibition can effect transient “normalization” of the tumor vasculature, thereby improving tumor perfusion and, consequently, delivery of systemic chemotherapy. We sought to examine temporal changes in tumor vascular function in response to the anti-VEGF antibody, bevacizumab. Experimental Design: Established orthotopic neuroblastoma xenografts treated with bevacizumab were evaluated at serial time points for treatment-associated changes in intratumoral vascular physiology, penetration of systemically administered chemotherapy, and efficacy of combination therapy. Results: After a single bevacizumab dose, a progressive decrease in tumor microvessel density to <30% of control was observed within 7 days. Assessment of the tumor microenvironment revealed a rapid, sustained decrease in both tumor vessel permeability and tumor interstitial fluid pressure, whereas intratumoral perfusion, as assessed by contrast-enhanced ultrasonography, was improved, although this latter change abated by 1 week. Intratumoral drug delivery mirrored these changes; penetration of chemotherapy was improved by as much as 81% when given 1 to 3 days after bevacizumab, compared with when both drugs were given concomitantly, or 7 days apart. Finally, administering topotecan to tumor-bearing mice 3 days after bevacizumab resulted in greater tumor growth inhibition (36% of control size) than with monotherapy (88% bevacizumab, 54% topotecan) or concomitant administration of the two drugs (44%). Conclusions: Bevacizumab-mediated VEGF blockade effects alterations in tumor vessel physiology that allow improved delivery and efficacy of chemotherapy, although careful consideration of drug scheduling is required to optimize antitumor activity.


International Journal of Radiation Oncology Biology Physics | 2010

Improved Intratumoral Oxygenation Through Vascular Normalization Increases Glioma Sensitivity to Ionizing Radiation

Mackenzie McGee; J. Blair Hamner; Regan F. Williams; Shannon F. Rosati; Thomas L. Sims; Catherine Y.C. Ng; M. Waleed Gaber; Christopher Calabrese; Jianrong Wu; Amit C. Nathwani; Christopher Duntsch; Thomas E. Merchant; Andrew M. Davidoff

PURPOSE Ionizing radiation, an important component of glioma therapy, is critically dependent on tumor oxygenation. However, gliomas are notable for areas of necrosis and hypoxia, which foster radioresistance. We hypothesized that pharmacologic manipulation of the typically dysfunctional tumor vasculature would improve intratumoral oxygenation and, thus, the antiglioma efficacy of ionizing radiation. METHODS AND MATERIALS Orthotopic U87 xenografts were treated with either continuous interferon-beta (IFN-beta) or bevacizumab, alone, or combined with cranial irradiation (RT). Tumor growth was assessed by quantitative bioluminescence imaging; the tumor vasculature using immunohistochemical staining, and tumor oxygenation using hypoxyprobe staining. RESULTS Both IFN-beta and bevaziumab profoundly affected the tumor vasculature, albeit with different cellular phenotypes. IFN-beta caused a doubling in the percentage of area of perivascular cell staining, and bevacizumab caused a rapid decrease in the percentage of area of endothelial cell staining. However, both agents increased intratumoral oxygenation, although with bevacizumab, the effect was transient, being lost by 5 days. Administration of IFN-beta or bevacizumab before RT was significantly more effective than any of the three modalities as monotherapy or when RT was administered concomitantly with IFN-beta or bevacizumab or 5 days after bevacizumab. CONCLUSION Bevacizumab and continuous delivery of IFN-beta each induced significant changes in glioma vascular physiology, improving intratumoral oxygenation and enhancing the antitumor activity of ionizing radiation. Additional investigation into the use and timing of these and other agents that modify the vascular phenotype, combined with RT, is warranted to optimize cytotoxic activity.


Surgery | 2008

Bevacizumab Suppresses Neuroblastoma Progression in the Setting of Minimal Disease

Thomas L. Sims; Regan F. Williams; Cathy Ng; Shannon F. Rosati; Yunyu Spence; Andrew M. Davidoff

BACKGROUND We hypothesized that vascular endothelial growth factor (VEGF) contributes to autocrine stimulation of neuroblastoma and that inhibition of its signaling pathway contributes to the anticancer activity of bevacizumab, an anti-VEGF monoclonal antibody. METHODS For in vitro studies, 2 neuroblastoma cell lines, CHLA-255 and NB1691, were treated with VEGF+/-bevacizumab. For in vivo studies, disseminated neuroblastoma was established by intravenous administration of luciferase-expressing tumor cells in SCID mice prior to bevacizumab treatment. RESULTS Exogenous VEGF increased cell counts after 48 h (NB1691: 58,878 +/- 8279 vs 137,500 +/- 13,108 cells, P < .001; CHLA: 1.56 x 10(6) +/- 866 vs 1.81 x 10(6) +/- 2550 cells, P <.001); the addition of bevacizumab abrogated this stimulation. In vivo, mice with disseminated disease treated twice weekly with intraperitoneal bevacizumab had a decreased tumor burden at day 14 and prolonged survival (NB1691: 50 +/- 2 vs 43 +/- 2 days, P < .001; CHLA: 53 +/- 3 vs 42 +/- 1 days, P = .006). Interestingly, VEGF and basic fibroblast growth factor expression was increased in treated NB1691 tumors, which likely occurred in response to VEGF signaling inhibition. CONCLUSION Our results suggest that VEGF has a role in neuroblastoma autocrine signaling. Maintenance therapy with bevacizumab may be useful for disease suppression after maximal cytoreductive therapy; however, upregulation of proangiogenic factors may provide resistance to this approach, which suggests that maximal antitumor efficacy may require combination therapy.


Journal of Pediatric Surgery | 2008

Avoiding misdiagnosing neuroblastoma as Wilms tumor

Paxton V. Dickson; Thomas L. Sims; Christian J. Streck; M. Beth McCarville; Victor M. Santana; Lisa M. McGregor; Wayne L. Furman; Andrew M. Davidoff

PURPOSE Although occasionally difficult, distinguishing abdominal neuroblastoma (NBL) from Wilms tumor (WT) at presentation is important, as surgical management differs significantly. We reviewed our 20-year experience (1987-2006) treating patients with NBL, focusing on those with an initial diagnosis of WT, to determine presenting features that would have suggested the correct preoperative diagnosis. METHODS Retrospective case cohort study reviewing charts and imaging of patients with NBL initially diagnosed clinically with WT. Preoperative symptoms, laboratory studies, and imaging were evaluated. Similar variables were assessed in the 20 patients with WT most recently treated at our institution. RESULTS Nine patients with NBL were identified as those who had an exploratory laparotomy with a preoperative diagnosis of WT; 8 underwent nephrectomy at exploration. Children with NBL had symptoms such as fever and weight loss at presentation (67%) more often than patients with WT (20%). Preoperative computed tomography demonstrated intratumoral calcifications, vascular encasement, or both in 78% of patients with NBL but were never seen in WT patients. Of interest, preoperative urinary catecholamines were elevated in 5 patients ultimately diagnosed with NBL. CONCLUSION Although NBL can be mistaken for WT at presentation, the presence of constitutional symptoms, or intratumoral calcification or vascular encasement on preoperative imaging should heighten suspicion for NBL. In addition, laboratory evaluation, including urinary catecholamines, should be completed before surgery when the etiology of an abdominal tumor is uncertain.


Journal of Pediatric Surgery | 2009

Neural progenitor cell–mediated delivery of osteoprotegerin limits disease progression in a preclinical model of neuroblastoma bone metastasis☆

Thomas L. Sims; J. Blair Hamner; Rebecca A. Bush; Peter E. Fischer; Seung U. Kim; Karen S. Aboody; Beth McCarville; Mary K. Danks; Andrew M. Davidoff

PURPOSE Osteoprotegerin (OPG) inhibits osteoclast activation and reduces osteolysis in bone tumors. We hypothesized that tumor-tropic neural progenitor cells (NPCs) engineered to express OPG would reduce neuroblastoma disease burden in the bone. METHODS Stable expression of green fluorescent protein (NPC-GFP) and OPG (NPC-OPG) was established in human NPCs by lentivirus-mediated transduction. Bone disease was established by intrafemoral injection of luciferase-expressing human neuroblastoma (CHLA-255) cells into 20 SCID mice. Three weeks later, mice began receiving intravenous injection of 2 x 10(6) NPC-OPG or NPC-GFP (control) every 10 days x 3 doses. Disease was monitored with quantitative bioluminescence imaging and x-ray images, which were evaluated on a scale of 0 to 4. These studies were approved by the Institutional Animal Care and Use Committee. RESULTS Osteoprotegerin treatment in vitro produced no direct toxicity to tumor cells. Coculture of tumor cells with bone marrow significantly increased activation of bone marrow-derived osteoclasts as assessed by tartrate-resistant acid phosphatase staining (156 +/- 10.8 osteoclasts per well) compared to bone marrow culture alone (91.67 +/- 4.7, P = .005). This increase was abrogated by adding OPG-containing media (68.3 +/- 2.8, P = .001). NPC-OPG slowed tumor progression (108-fold increase from pretreatment) compared to mice treated with NPC-GFP (538-fold), as judged by bioluminescence imaging. X-rays subjectively demonstrated less bone disease in NPC-OPG-treated mice (2.27 +/- 0.25) compared to NPC-GFP-treated mice (3.25 +/- 0.22, P = .04). CONCLUSIONS Neural progenitor cell-mediated delivery of OPG slowed disease progression in a preclinical model of neuroblastoma bone metastasis. The decrease in bone disease was not from direct tumor cell toxicity but likely occurred indirectly through inhibition of osteoclast-directed bone resorption. Thus, targeted delivery of OPG by NPCs may be effective in the treatment of neuroblastoma bone metastasis.


Journal of Pediatric Surgery | 2008

The efficacy of combination therapy using adeno-associated virus—interferon β and trichostatin A in vitro and in a murine model of neuroblastoma

John B. Hamner; Thomas L. Sims; Aaron Cutshaw; Paxton V. Dickson; Shannon F. Rosati; Mackenzie McGee; Catherine Y.C. Ng; Andrew M. Davidoff

PURPOSE Trichostatin A (TSA) is a potent histone deacetylase inhibitor and has demonstrated significant antitumor activity against a variety of cancer cell lines. Type I interferons have also shown significant antitumor as well as antiangiogenic activity. In this study, we examined the effectiveness of combination therapy of TSA and interferon beta (IFN-beta) on human neuroblastoma cells in vitro and in vivo using a murine model of retroperitoneal neuroblastoma. MATERIALS AND METHODS For in vitro experiments, plated human neuroblastoma cells (NB-1643 and NB-1691) were treated with vehicle or with IFN-beta, TSA, or both for 24 hours. Cytotoxicity was assessed by counting cells and expressing the results as a percentage of controls. Expression of the tumor suppressor p21(Waf1) was assessed by Western blot. For in vivo experiments, retroperitoneal neuroblastomas were established in severe combined immune deficiency (SCID) mice. Interferon beta was given using a gene therapy approach, administering 1.5 x 10(10) particles of an adeno-associated virus vector encoding human IFN-beta (AAV hIFN-beta) via tail vein as a single dose per mouse. Trichostatin A was given at a dose of 5 mg/kg every 48 hours subcutaneously. Treatment groups included controls, AAV hIFN-beta alone, TSA alone, and AAV hIFN-beta together with TSA. Tumor volume was assessed 2 weeks after the treatment began. RESULTS After 24 hours, treatment with IFN-beta, TSA, and a combination of both resulted in a 45.3%, 68.1%, and 75% reduction in cell count relative to controls in the NB-1691 cell line. In the NB-1643 line, cell counts were reduced by 23%, 58%, and 62.3% respectively. In addition, NB-1691 cells treated with TSA showed increased expression of p21(Waf1) on Western blot. For in vivo experiments, control-, AAV hIFN-beta-, TSA-, and combination-treated tumors had the following final volumes: 1577.7 +/- 264.2 mm(3) (n = 3); 128.5 +/- 74.4 mm(3) (n = 4; P = .0001); 1248.7 +/- 673.9 mm(3) (n = 4; P = .48); and 127.5 +/- 36.8 mm(3) (n = 4; P = .0007), respectively. CONCLUSION Neuroblastoma, because of its unique biology, continues to be a challenging tumor to treat, and many times these tumors are refractory to standard chemotherapeutic regimens. These data show that both TSA and IFN-beta inhibit neuroblastoma growth and that the combination may potentially provide a unique way to treat this difficult disease.


Surgery | 2011

Continuous Local Delivery of Interferon-β Stabilizes Tumor Vasculature in an Orthotopic Glioblastoma Xenograft Resection Model

Jason W. Denbo; Regan F. Williams; W. Shannon Orr; Thomas L. Sims; Catherine Y.C. Ng; Junfang Zhou; Yunyu Spence; Christopher L. Morton; Amit C. Nathwani; Christopher Duntsch; Lawrence M. Pfeffer; Andrew M. Davidoff

BACKGROUND High-grade glioblastomas have immature, leaky tumor blood vessels that impede the efficacy of adjuvant therapy. We assessed the ability of human interferon (hIFN)-β delivered locally via gene transfer to effect vascular stabilization in an orthotopic model of glioblastoma xenograft resection. METHODS Xenografts were established by injecting 3 grade IV glioblastoma cell lines (GBM6-luc, MT330-luc, and SJG2-luc) into the cerebral cortex of nude rats. Tumors underwent subtotal resection, and then had gel foam containing an adeno-associated virus vector encoding either hIFN-β or green fluorescence protein (control) placed in the resection cavity. The primary endpoint was stabilization of tumor vasculature, as evidenced by CD34, α-SMA, and CA IX staining. Overall survival was a secondary endpoint. RESULTS hIFN-β treatment altered the tumor vasculature of GBM6-luc and SJG2-luc xenografts, decreasing the density of endothelial cells, stabilizing vessels with pericytes, and decreasing tumor hypoxia. The mean survival for rats with these neoplasms was not improved, however. In rats with MT330-luc xenografts, hIFN-β resulted in tumor regression with a 6-month survival of 55% (INF-β group) and 9% (control group). CONCLUSION The use of AAV hIFN-β in our orthotopic model of glioblastoma resection stabilized tumor vasculature and improved survival in rats with MT330 xenografts.


Journal of Pediatric Surgery | 2010

Targeting multiple angiogenic pathways for the treatment of neuroblastoma

Regan F. Williams; Adrianne L. Myers; Thomas L. Sims; Catherine Y.C. Ng; Amit C. Nathwani; Andrew M. Davidoff

PURPOSE Resistance to angiogenesis inhibition can occur through the upregulation of alternative mediators of neovascularization. We used a combination of angiogenesis inhibitors with different mechanisms of action, interferon-beta (IFN-beta) and rapamycin, to target multiple angiogenic pathways to treat neuroblastoma xenografts. METHODS Subcutaneous and retroperitoneal neuroblastoma xenografts (NB-1691 and SK-N-AS) were used. Continuous delivery of IFN-beta was achieved with adeno-associated virus vector-mediated, liver-targeted gene transfer. Rapamycin was delivered intraperitoneally (5 mg/kg per day). After 2 weeks of treatment, tumor size was measured, and tumor vasculature was evaluated with intravital microscopy and immunohistochemistry. RESULTS Rapamycin and IFN-beta, alone and in combination, had little effect on tumor cell viability in vitro. In vivo, combination therapy led to fewer intratumoral vessels (69% of control), and the remaining vessels had an altered phenotype, being covered with significantly more pericytes (13x control). Final tumor size was significantly less than controls in all tumor models, with combination therapy having a greater antitumor effect than either monotherapy. CONCLUSION The combination of IFN-beta and rapamycin altered the vasculature of neuroblastoma xenografts and resulted in significant tumor inhibition. The use of combinations of antiangiogenic agents should be further evaluated for the treatment of neuroblastoma and other solid tumors.


Journal of Surgical Research | 2007

2: Bortezomib inhibits angiogenesis and reduces tumor burden in a murine model of neuroblastoma

John B. Hamner; Paxton V. Dickson; Thomas L. Sims; Junfang Zhou; Yunyu Spence; Cathy Ng; Andrew M. Davidoff

BACKGROUND Bortezomib is a proteasome inhibitor with pleiotropic antitumor activity. Here we investigate the antiangiogenic and antitumor efficacy of bortezomib against neuroblastoma both in vitro and in a murine model of localized and disseminated disease. METHODS In vitro activity of bortezomib was assessed by evaluating its effect on cell proliferation and cell cycle status. Localized tumor burden was followed with caliper measurements and total-body bioluminescence in mice with disseminated disease. The antiangiogenic activity was evaluated with immunohistochemistry and human vascular endothelial growth factor (VEGF) enzyme-linked immunosorbent assay on tumor protein extracts. RESULTS Bortezomib treatment resulted in dose and time-dependent decreases in cell proliferation and resulted in cell cycle arrest. In vivo, bortezomib restricted tumor growth in a model of localized disease and decreased bioluminescence in mice with disseminated disease. That decreased bioluminescence reflected decreased tumor burden was confirmed at necropsy by assessing disease in specific organs. In addition, treatment resulted in a decrease in intratumoral vessel counts and reduced tumor VEGF expression. CONCLUSION Bortezomib shows significant activity against neuroblastoma in vitro, and it inhibits tumor growth and angiogenesis in vivo. These results suggest that clinical studies of bortezomib are warranted for the treatment of this difficult disease.


Surgery | 2007

Bortezomib inhibits angiogenesis and reduces tumor burden in a murine model of neuroblastoma.

John B. Hamner; Paxton V. Dickson; Thomas L. Sims; Junfang Zhou; Yunyu Spence; Cathy Ng; Andrew M. Davidoff

Collaboration


Dive into the Thomas L. Sims's collaboration.

Top Co-Authors

Avatar

Andrew M. Davidoff

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Regan F. Williams

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Catherine Y.C. Ng

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Cathy Ng

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

John B. Hamner

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Paxton V. Dickson

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Junfang Zhou

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Mackenzie McGee

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Yunyu Spence

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Christopher Duntsch

University of Tennessee Health Science Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge