Network


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

Hotspot


Dive into the research topics where Curtis B. Thompson is active.

Publication


Featured researches published by Curtis B. Thompson.


Gut | 2013

Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer

Michael A. Jacobetz; Derek S. Chan; Albrecht Neesse; Tashinga E. Bapiro; Natalie Cook; Kristopher K. Frese; Christine Feig; Tomoaki Nakagawa; Meredith E. Caldwell; Heather I Zecchini; Martijn P. Lolkema; Ping Jiang; Anne Kultti; Curtis B. Thompson; Daniel C. Maneval; Duncan I. Jodrell; Gregory I. Frost; Harold Michael Shepard; Jeremy N. Skepper; David A. Tuveson

Objective Pancreatic ductal adenocarcinoma (PDA) is characterised by stromal desmoplasia and vascular dysfunction, which critically impair drug delivery. This study examines the role of an abundant extracellular matrix component, the megadalton glycosaminoglycan hyaluronan (HA), as a novel therapeutic target in PDA. Methods Using a genetically engineered mouse model of PDA, the authors enzymatically depleted HA by a clinically formulated PEGylated human recombinant PH20 hyaluronidase (PEGPH20) and examined tumour perfusion, vascular permeability and drug delivery. The preclinical utility of PEGPH20 in combination with gemcitabine was assessed by short-term and survival studies. Results PEGPH20 rapidly and sustainably depleted HA, inducing the re-expansion of PDA blood vessels and increasing the intratumoral delivery of two chemotherapeutic agents, doxorubicin and gemcitabine. Moreover, PEGPH20 triggered fenestrations and interendothelial junctional gaps in PDA tumour endothelia and promoted a tumour-specific increase in macromolecular permeability. Finally, combination therapy with PEGPH20 and gemcitabine led to inhibition of PDA tumour growth and prolonged survival over gemcitabine monotherapy, suggesting immediate clinical utility. Conclusions The authors demonstrate that HA impedes the intratumoral vasculature in PDA and propose that its enzymatic depletion be explored as a means to improve drug delivery and response in patients with pancreatic cancer.


Molecular Cancer Therapeutics | 2010

Enzymatic Depletion of Tumor Hyaluronan Induces Antitumor Responses in Preclinical Animal Models

Curtis B. Thompson; H. Michael Shepard; Patrick O'Connor; Salam Kadhim; Ping Jiang; Ryan Osgood; Louis H. Bookbinder; Xiaoming Li; Barry J. Sugarman; Robert J. Connor; Sinisa Nadjsombati; Gregory I. Frost

Hyaluronan (HA) is a glycosaminoglycan polymer that often accumulates in malignancy. Megadalton complexes of HA with proteoglycans create a hydrated connective tissue matrix, which may play an important role in tumor stroma formation. Through its colloid osmotic effects, HA complexes contribute to tumor interstitial fluid pressure, limiting the effect of therapeutic molecules on malignant cells. The therapeutic potential of enzymatic remodeling of the tumor microenvironment through HA depletion was initially investigated using a recombinant human HA-degrading enzyme, rHuPH20, which removed HA-dependent tumor cell extracellular matrices in vitro. However, rHuPH20 showed a short serum half-life (t1/2 < 3 minutes), making depletion of tumor HA in vivo impractical. A pegylated variant of rHuPH20, PEGPH20, was therefore evaluated. Pegylation improved serum half-life (t1/2 = 10.3 hours), making it feasible to probe the effects of sustained HA depletion on tumor physiology. In high-HA prostate PC3 tumors, i.v. administration of PEGPH20 depleted tumor HA, decreased tumor interstitial fluid pressure by 84%, decreased water content by 7%, decompressed tumor vessels, and increased tumor vascular area >3-fold. Following repeat PEGPH20 administration, tumor growth was significantly inhibited (tumor growth inhibition, 70%). Furthermore, PEGPH20 enhanced both docetaxel and liposomal doxorubicin activity in PC3 tumors (P < 0.05) but did not significantly improve the activity of docetaxel in low-HA prostate DU145 tumors. The ability of PEGPH20 to enhance chemotherapy efficacy is likely due to increased drug perfusion combined with other tumor structural changes. These results support enzymatic remodeling of the tumor stroma with PEGPH20 to treat tumors characterized by the accumulation of HA. Mol Cancer Ther; 9(11); 3052–64. ©2010 AACR.


BioMed Research International | 2014

Accumulation of extracellular hyaluronan by hyaluronan synthase 3 promotes tumor growth and modulates the pancreatic cancer microenvironment.

Anne Kultti; Chunmei Zhao; Netai C. Singha; Susan Zimmerman; Ryan Osgood; Rebecca Symons; Ping Jiang; Xiaoming Li; Curtis B. Thompson; Jeffrey R. Infante; Michael A. Jacobetz; David A. Tuveson; Gregory I. Frost; H. Michael Shepard; Zhongdong Huang

Extensive accumulation of the glycosaminoglycan hyaluronan is found in pancreatic cancer. The role of hyaluronan synthases 2 and 3 (HAS2, 3) was investigated in pancreatic cancer growth and the tumor microenvironment. Overexpression of HAS3 increased hyaluronan synthesis in BxPC-3 pancreatic cancer cells. In vivo, overexpression of HAS3 led to faster growing xenograft tumors with abundant extracellular hyaluronan accumulation. Treatment with pegylated human recombinant hyaluronidase (PEGPH20) removed extracellular hyaluronan and dramatically decreased the growth rate of BxPC-3 HAS3 tumors compared to parental tumors. PEGPH20 had a weaker effect on HAS2-overexpressing tumors which grew more slowly and contained both extracellular and intracellular hyaluronan. Accumulation of hyaluronan was associated with loss of plasma membrane E-cadherin and accumulation of cytoplasmic β-catenin, suggesting disruption of adherens junctions. PEGPH20 decreased the amount of nuclear hypoxia-related proteins and induced translocation of E-cadherin and β-catenin to the plasma membrane. Translocation of E-cadherin was also seen in tumors from a transgenic mouse model of pancreatic cancer and in a human non-small cell lung cancer sample from a patient treated with PEGPH20. In conclusion, hyaluronan accumulation by HAS3 favors pancreatic cancer growth, at least in part by decreasing epithelial cell adhesion, and PEGPH20 inhibits these changes and suppresses tumor growth.


Diabetes | 2013

Hyaluronan Accumulates with High Fat Feeding and Contributes to Insulin Resistance

Li Kang; Louise Lantier; Arion Kennedy; Jeffrey S. Bonner; Wesley H. Mayes; Deanna P. Bracy; Louis H. Bookbinder; Alyssa H. Hasty; Curtis B. Thompson; David H. Wasserman

Increased deposition of specific extracellular matrix (ECM) components is a characteristic of insulin-resistant skeletal muscle. Hyaluronan (HA) is a major constituent of the ECM. The hypotheses that 1) HA content is increased in the ECM of insulin-resistant skeletal muscle and 2) reduction of HA in the muscle ECM by long-acting pegylated human recombinant PH20 hyaluronidase (PEGPH20) reverses high-fat (HF) diet–induced muscle insulin resistance were tested. We show that muscle HA was increased in HF diet–induced obese (DIO) mice and that treatment of PEGPH20, which dose-dependently reduced HA in muscle ECM, decreased fat mass, adipocyte size, and hepatic and muscle insulin resistance in DIO mice at 10 mg/kg. Reduced muscle insulin resistance was associated with increased insulin signaling, muscle vascularization, and percent cardiac output to muscle rather than insulin sensitization of muscle per se. Dose-response studies revealed that PEGPH20 dose-dependently increased insulin sensitivity in DIO mice with a minimally effective dose of 0.01 mg/kg. PEGPH20 at doses of 0.1 and 1 mg/kg reduced muscle HA to levels seen in chow-fed mice, decreased fat mass, and increased muscle glucose uptake. These findings suggest that ECM HA is a target for treatment of insulin resistance.


Cancers | 2012

Therapeutic Targeting of Hyaluronan in the Tumor Stroma

Anne Kultti; Xiaoming Li; Ping Jiang; Curtis B. Thompson; Gregory I. Frost; H. Michael Shepard

The tumor stroma, consisting of non-malignant cells and the extracellular matrix, undergoes significant quantitative and qualitative changes throughout malignant transformation and tumor progression. With increasing recognition of the role of the tumor microenvironment in disease progression, stromal components of the tumor have become attractive targets for therapeutic intervention. Stromal accumulation of the glycosaminoglycan hyaluronan occurs in many tumor types and is frequently associated with a negative disease prognosis. Hyaluronan interacts with other extracellular molecules as well as cellular receptors to form a complex interaction network influencing physicochemical properties, signal transduction, and biological behavior of cancer cells. In preclinical animal models, enzymatic removal of hyaluronan is associated with remodeling of the tumor stroma, reduction of tumor interstitial fluid pressure, expansion of tumor blood vessels and facilitated delivery of chemotherapy. This leads to inhibition of tumor growth and increased survival. Current evidence shows that abnormal accumulation of hyaluronan may be an important stromal target for cancer therapy. In this review we highlight the role of hyaluronan and hyaluronan-mediated interactions in cancer, and discuss historical and recent data on hyaluronidase-based therapies and the effect of hyaluronan removal on tumor growth.


Journal of Immunology | 2014

Recombinant Human Hyaluronidase PH20 Does Not Stimulate an Acute Inflammatory Response and Inhibits Lipopolysaccharide-Induced Neutrophil Recruitment in the Air Pouch Model of Inflammation

Zhongdong Huang; Chunmei Zhao; Yanling Chen; Jessica Cowell; Ge Wei; Anne Kultti; Lei Huang; Curtis B. Thompson; Sanna Rosengren; Gregory I. Frost; H. Michael Shepard

Hyaluronidase (Hyal) and low m.w. hyaluronan (LMW HA) fragments have been widely reported to stimulate the innate immune response. However, most hyaluronidases used were purified from animal tissues (e.g., bovine testis Hyal [BTH]), and contain endotoxin and other unrelated proteins. We tested a highly purified recombinant human Hyal (rHuPH20) and endotoxin-free HA fragments from Mr 5,000 to 1,500,000 in the rodent air pouch model of inflammation to determine their potential for stimulation of the innate immune response. Exogenous LMW HA fragments (average Mr 200,000) failed to induce either cytokine/chemokine production or neutrophil infiltration into the air pouch. Challenging the air pouch with LPS or BTH stimulated production of cytokines and chemokines but rHuPH20 did not, suggesting that neither PH20 nor generation of LMW HA fragments in situ stimulates cytokine and chemokine production. LPS and BTH also induced neutrophil infiltration into the air pouch, which was not observed with rHuPH20 treatment. Endotoxin-depleted BTH had much reduced proinflammatory activity, suggesting that the difference in inflammatory responses between rHuPH20 and BTH is likely due to endotoxin contaminants in BTH. When rHuPH20 was dosed with LPS, the induction of cytokines and chemokines was the same as LPS alone, but neutrophil infiltration was inhibited, likely by interrupting HA–CD44 interaction. Our results indicate that neither rHuPH20 nor its directly generated HA catabolites have inflammatory properties in the air pouch model, and rHuPH20 can instead inhibit some aspects of inflammation, such as neutrophil infiltration into the air pouch.


Cancer Cell | 2014

Response to Chauhan et al.: Interstitial Pressure and Vascular Collapse in Pancreas Cancer—Fluids and Solids, Measurement and Meaning

Kathleen E. DelGiorno; Markus A. Carlson; Ryan Osgood; Paolo P. Provenzano; J. Scott Brockenbough; Curtis B. Thompson; H. Michael Shepard; Gregory I. Frost; John D. Potter; Sunil R. Hingorani

Chauhan et al. suggest that vascular collapse and hypoperfusion in pancreatic ductal adenocarcinoma (PDA) are caused by solid stress (SS) (Chauhan et al., 2014) instead of the elevated interstitial fluid pressure (IFP) associated with high extravascular concentrations of hyaluronan (Provenzano et al., 2012). We appreciate their attention to our work and the opportunity to clarify underlying mechanisms. Chauhan et al. make four important claims, to which we respond.


Cancer Research | 2012

Abstract 5635: Pegylated recombinant human hyaluronidase PH20 (PEGPH20) enhances Nab-Paclitaxel efficacy in BxPC-3 human pancreatic cancer xenografts

Ryan Osgood; James Skipper; Ping Jiang; Susan Zimmerman; H. Michael Shepard; Daniel C. Maneval; Curtis B. Thompson

Hyaluronan (HA) accumulates in the extracellular matrix (ECM) of many solid tumors, including those of the prostate, colon, breast, stomach, ovary, and pancreas. This accumulation is associated with tumor progression and a negative clinical outcome. Accordingly, an HA-degrading enzyme, pegylated recombinant human hyaluronidase PH20 (PEGPH20), was developed to target tumor-associated HA in the ECM. Preclinical studies demonstrated that PEGPH20-mediated removal of HA from HA-rich xenograft tumors in mice decreased tumor interstitial fluid pressure and tumor water content resulting in a decompression of tumor vasculature, increased tumor vascular perfusion, tumor growth inhibition (TGI) and enhanced chemotherapeutic activity (Thompson 2010). We further characterized HA expression across multiple human tumor types and identified pancreatic ductal adenocarcinoma (PDA) as the cancer type with the most HA (∼87% express high levels). These observations coupled with a lack of curative therapy for PDA led us to evaluate alternative treatment strategies for PDA. Specifically, using the peritibial BxPC-3 human pancreatic cancer xenograft model, we investigated whether the antitumor activity of Nab-Paclitaxel was significantly enhanced with PEGPH20 treatment. Nude mice were inoculated with human PDA BxPC-3 cells adjacent to the right tibial periosteum, and tumor growth was monitored with ultrasonography. When tumors reached ∼400 mm3 (n≥8/group), mice were staged into 8 treatment groups: (1) vehicle control; (2) PEGPH20 monotherapy, 4.5 mg/kg; (3) Nab-Paclitaxel, 3 mg/kg; (4) Nab-Paclitaxel, 10 mg/kg; (5) Nab-Paclitaxel, 30 mg/kg; (6) Nab-Paclitaxel, 3 mg/kg, plus PEGPH20; (7) Nab-Paclitaxel, 10 mg/kg, plus PEGPH20; or (8) Nab-Paclitaxel, 30 mg/kg, plus PEGPH20. Vehicle or PEGPH20 ± Nab-Paclitaxel was administered intravenously starting on study day 0, and then dosed every third day for 15 days. At study termination, the average TGIs from animals treated with either PEGPH20 alone (12.2%), low dose (3 mg/kg) Nab-Paclitaxel (20.3%), or low dose Nab-Paclitaxel (3 mg/kg) plus PEGPH20 (25%) were not significantly different from vehicle-treated animals. However, Nab-Paclitaxel alone at both 10 mg/kg (61.5%, p Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 5635. doi:1538-7445.AM2012-5635


Cancer Research | 2016

Abstract 4886: PEGylated recombinant hyaluronidase PH20 (PEGPH20) enhances checkpoint inhibitor efficacy in syngeneic mouse models of cancer

Sanna Rosengren; Renee Clift; Susan Zimmerman; Jennifer Souratha; Benjamin J. Thompson; Barbara Blouw; Xiaoming Li; Qiping Zhao; Michael Shepard; Dan C. Maneval; Christopher D. Thanos; Curtis B. Thompson

Hyaluronan (HA), a major extracellular matrix component in many solid tumors, has been proposed to contribute to tumor progression, and to play a complex role in T lymphocyte biology. Its depletion by intravenous PEGylated recombinant human hyaluronidase PH20 (PEGPH20) remodels the tumor stroma, reduces intratumoral pressure, decompresses tumor blood vessels, and facilitates tumor drug delivery. However, the impact of HA removal on intra-tumoral immune responses and the efficacy of immune checkpoint inhibitors is unknown. To evaluate checkpoint blockade efficacy with PEGPH20, two mouse tumor cell lines, CT26 (colon) and MH194 (pancreatic, derived from spontaneous tumors in KrasLSL-G12D/+Trp53LSL-R172H/+Cre mice) were transduced with hyaluronan synthase-3 (HAS3) to generate syngeneic HA-high tumor models. For anti-CTLA4 studies, parental CT26 and CT26/HAS3 cells were implanted peritibially in Balb/C mice. While treatment with anti-mouse-CTLA4 alone (clone 9D9) inhibited tumor growth in CT26 tumors (37%), PEGPH20 alone did not significantly inhibit tumor growth or increase anti-CTLA4 efficacy. In contrast, tumor growth of CT26/HAS3 tumors was inhibited to a greater extent by the combination of PEGPH20 and anti-CTLA4 (79%) (PEGPH20 treatment 24h prior to anti-CTLA4 treatment), compared to anti-CTLA4 alone (60%, p = 0.002) or PEGPH20 alone (43%, p = 0.0001). Furthermore, gene expression of markers associated with immune suppression, such as IL10 and FoxP3, was higher in CT26/HAS3 than in CT26 tumors; suggesting an association between HA content and immune suppression. To evaluate the effect of PEGPH20 on tumor growth inhibition by PD-1 blockade, MH194/HAS3 cells were implanted peritibially in C57BL/6 mice along with immortalized pancreatic stellate cells. Growth of MH194/HAS3 tumors was significantly inhibited (33%, p = 0.049) by anti-mouse-PD-L1 antibody (clone 10F.9G2), and the addition of PEGPH20 (24h prior to anti-PD-L1) to anti-PD-L1 further enhanced tumor growth inhibition (79%, p Citation Format: Sanna Rosengren, Renee Clift, Susan J. Zimmerman, Jennifer Souratha, Benjamin J. Thompson, Barbara Blouw, Xiaoming Li, Qiping Zhao, Michael Shepard, Dan C. Maneval, Christopher D. Thanos, Curtis B. Thompson. PEGylated recombinant hyaluronidase PH20 (PEGPH20) enhances checkpoint inhibitor efficacy in syngeneic mouse models of cancer. [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 4886.


Cancer Cell | 2014

Interstitial pressure and vascular collapse in pancreas cancer:fluids and solids, measurement and meaning

Kathleen E. DelGiorno; Markus A. Carlson; Ryan Osgood; Paolo P. Provenzano; J. Scott Brockenbough; Curtis B. Thompson; H. Michael Shepard; Gregory I. Frost; John D. Potter; Sunil R. Hingorani

Chauhan et al. suggest that vascular collapse and hypoperfusion in pancreatic ductal adenocarcinoma (PDA) are caused by solid stress (SS) (Chauhan et al., 2014) instead of the elevated interstitial fluid pressure (IFP) associated with high extravascular concentrations of hyaluronan (Provenzano et al., 2012). We appreciate their attention to our work and the opportunity to clarify underlying mechanisms. Chauhan et al. make four important claims, to which we respond.

Collaboration


Dive into the Curtis B. Thompson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

David A. Tuveson

Cold Spring Harbor Laboratory

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kathleen E. DelGiorno

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Markus A. Carlson

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Paolo P. Provenzano

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Sunil R. Hingorani

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Daniel D. Von Hoff

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Don J. Diamond

City of Hope National Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge