Network


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

Hotspot


Dive into the research topics where David Noyes is active.

Publication


Featured researches published by David Noyes.


International Journal of Radiation Oncology Biology Physics | 2012

Combination of External Beam Radiotherapy (EBRT) With Intratumoral Injection of Dendritic Cells as Neo-Adjuvant Treatment of High-Risk Soft Tissue Sarcoma Patients

Steven E. Finkelstein; Cristina Iclozan; Marilyn M. Bui; Matthew J. Cotter; Rupal Ramakrishnan; Jamil Ahmed; David Noyes; David Cheong; Ricardo J. Gonzalez; Randy V. Heysek; Claudia Berman; Brianna Lenox; William Janssen; Jonathan S. Zager; Vernon K. Sondak; G. Douglas Letson; Scott Antonia; Dmitry I. Gabrilovich

PURPOSE The goal of this study was to determine the effect of combination of intratumoral administration of dendritic cells (DC) and fractionated external beam radiation (EBRT) on tumor-specific immune responses in patients with soft-tissue sarcoma (STS). METHODS AND MATERIAL Seventeen patients with large (>5 cm) high-grade STS were enrolled in the study. They were treated in the neoadjuvant setting with 5,040 cGy of EBRT, split into 28 fractions and delivered 5 days per week, combined with intratumoral injection of 10(7) DCs followed by complete resection. DCs were injected on the second, third, and fourth Friday of the treatment cycle. Clinical evaluation and immunological assessments were performed. RESULTS The treatment was well tolerated. No patient had tumor-specific immune responses before combined EBRT/DC therapy; 9 patients (52.9%) developed tumor-specific immune responses, which lasted from 11 to 42 weeks. Twelve of 17 patients (70.6%) were progression free after 1 year. Treatment caused a dramatic accumulation of T cells in the tumor. The presence of CD4(+) T cells in the tumor positively correlated with tumor-specific immune responses that developed following combined therapy. Accumulation of myeloid-derived suppressor cells but not regulatory T cells negatively correlated with the development of tumor-specific immune responses. Experiments with (111)In labeled DCs demonstrated that these antigen presenting cells need at least 48 h to start migrating from tumor site. CONCLUSIONS Combination of intratumoral DC administration with EBRT was safe and resulted in induction of antitumor immune responses. This suggests that this therapy is promising and needs further testing in clinical trials design to assess clinical efficacy.


Clinical Cancer Research | 2016

HDAC inhibitors enhance T-cell chemokine expression and augment response to PD-1 immunotherapy in lung adenocarcinoma

Hong Zheng; Weipeng Zhao; Cihui Yan; Crystina Watson; Michael Massengill; Mengyu Xie; Chris Massengill; David Noyes; Gary V. Martinez; Roha Afzal; Zhihua Chen; Xiubao Ren; Scott Antonia; Eric B. Haura; Brian Ruffell; Amer A. Beg

Purpose: A significant limitation of checkpoint blockade immunotherapy is the relatively low response rate (e.g., ∼20% with PD-1 blockade in lung cancer). In this study, we tested whether strategies that increase T-cell infiltration to tumors can be efficacious in enhancing immunotherapy response. Experimental Design: We performed an unbiased screen to identify FDA-approved oncology agents with an ability to enhance T-cell chemokine expression with the goal of identifying agents capable of augmenting immunotherapy response. Identified agents were tested in multiple lung tumor models as single agents and in combination with PD-1 blockade. Additional molecular and cellular analysis of tumors was used to define underlying mechanisms. Results: We found that histone deacetylase (HDAC) inhibitors (HDACi) increased expression of multiple T-cell chemokines in cancer cells, macrophages, and T cells. Using the HDACi romidepsin in vivo, we observed increased chemokine expression, enhanced T-cell infiltration, and T-cell–dependent tumor regression. Importantly, romidepsin significantly enhanced the response to PD-1 blockade immunotherapy in multiple lung tumor models, including nearly complete rejection in two models. Combined romidepsin and PD-1 blockade also significantly enhanced activation of tumor-infiltrating T cells. Conclusions: These results provide evidence for a novel role of HDACs in modulating T-cell chemokine expression in multiple cell types. In addition, our findings indicate that pharmacologic induction of T-cell chemokine expression represents a conceptually novel approach for enhancing immunotherapy response. Finally, these results suggest that combination of HDAC inhibitors with PD-1 blockade represents a promising strategy for lung cancer treatment. Clin Cancer Res; 22(16); 4119–32. ©2016 AACR.


Journal of Immunotherapy | 2008

Phase II trial of B7-1 (CD-86) transduced, cultured autologous tumor cell vaccine plus subcutaneous interleukin-2 for treatment of stage IV renal cell carcinoma.

Mayer Fishman; Terri B. Hunter; Hatem Soliman; Patricia Thompson; Mary Dunn; Renee Smilee; Mary Jane Farmelo; David Noyes; John J. Mahany; Ji-Hyun Lee; Alan Cantor; Jane L. Messina; John D. Seigne; Julio M. Pow-Sang; William Janssen; Scott Antonia

We report a single center phase II trial of sequential vaccination followed with vaccine plus interleukin-2 (IL-2). Vaccination consisted of autologous cells cultured from primary tumor or resected metastasis, transduced to express B7.1 surface molecule and then irradiated. The vaccine would hypothetically costimulate tumor-reactive T cells before IL-2 exposure. Treatment plan was 3 subcutaneous vaccine injections at 4-week intervals and subcutaneous IL-2 treatment for 6 weeks starting at week 7. Sixty-six patients enrolled, of whom 39 received at least 1 vaccine; most observed toxicity was attributable to IL-2 not vaccine; best responses were 3% pathologic complete response, 5% partial response, 64% stable disease, and 28% disease progression. Median survival was 21.8 months (95% confidence interval 17.8 to 29.6). Significant postvaccination increases in IFN-γ responses to autologous tumor were observed in 2/26 cases. Eighty-one percent of posttreatment subdermal delayed-type hypersensitivity tests (using nontransduced, irradiated autologous tumor cells) had biopsies demonstrating injection site lymphocytic infiltration. Post hoc comparison of the median survival of subjects whose biopsies had lymphocytic infiltration appears longer than in the 19% noninfiltrated (28.4 vs. 17.8 mo, P=0.045, two-sided log-rank test). The single arm design precludes conclusive comparison of objective response rates (not different here) or median survival (longer here) versus those of historical series using similar IL-2 schedules alone. Better outcomes could be logically associated to vaccine response (detectable lymphocytic infiltrates) or to random events that a single arm study design cannot address. This vaccine approach may merit further clinical development.


Oncotarget | 2016

A phase I study of indoximod in patients with advanced malignancies

Hatem Soliman; Susan Minton; Hyo S. Han; Roohi Ismail-Khan; Anthony Neuger; Fatema Khambati; David Noyes; Richard M. Lush; Alberto Chiappori; John D. Roberts; Charles J. Link; Nicholas N. Vahanian; Mario Mautino; Howard Streicher; Daniel M. Sullivan; Scott Antonia

Purpose Indoximod is an oral inhibitor of the indoleamine 2,3-dioxygenase pathway, which causes tumor-mediated immunosuppression. Primary endpoints were maximum tolerated dose (MTD) and toxicity for indoximod in patients with advanced solid tumors. Secondary endpoints included response rates, pharmacokinetics, and immune correlates. Experimental Design Our 3+3 phase I trial comprised 10 dose levels (200, 300, 400, 600, and 800 mg once/day; 600, 800, 1200, 1600, and 2000 mg twice/day). Inclusion criteria were measurable metastatic solid malignancy, age ≥18 years, and adequate organ/marrow function. Exclusion criteria were chemotherapy ≤ 3 weeks prior, untreated brain metastases, autoimmune disease, or malabsorption. Results In 48 patients, MTD was not reached at 2000 mg twice/day. At 200 mg once/day, 3 patients previously treated with checkpoint inhibitors developed hypophysitis. Five patients showed stable disease >6 months. Indoximod plasma AUC and Cmax plateaued above 1200mg. Cmax (∼12 μM at 2000 mg twice/day) occurred at 2.9 hours, and half-life was 10.5 hours. C reactive protein (CRP) levels increased across multiple dose levels. Conclusions Indoximod was safe at doses up to 2000 mg orally twice/day. Best response was stable disease >6 months in 5 patients. Induction of hypophysitis, increased tumor antigen autoantibodies and CRP levels were observed.


Cancer Biology & Therapy | 2013

Antagonism of adenosine A2A receptor expressed by lung adenocarcinoma tumor cells and cancer associated fibroblasts inhibits their growth

Melanie Mediavilla-Varela; Kimberly Luddy; David Noyes; Farah Khalil; Anthony Neuger; Hatem Soliman; Scott Antonia

Recently it has become clear that the cost associated with the Warburg effect, which is inefficient production of ATP, is offset by selective advantages that are produced by resultant intracellular metabolic alterations. In fact tumors may be addicted to the Warburg effect. In addition these alterations result in changes in the extracellular tumor microenvironment that can also produce selective advantages for tumor cell growth and survival. One such extracellular alteration is increased adenosine concentrations that have been shown to impair T cell mediated rejection and support angiogenesis. The expression of the A2A receptor in non-small cell cancer (NSCLC) tissues, cell lines and cancer associated fibroblasts (CAF) was determined by performing immunohistrochemistry and immunoblot analysis. The efficacy of the A2A receptor antagonists in vivo was evaluated in a PC9 xenograft model. To determine the mode of cell death induced by A2A receptor antagonists flow cytometry, immunoblot, and cytotoxic analysis were performed. We found that a significant number of lung adenocarcinomas express adenosine A2A receptors. Antagonism of these receptors impaired CAF and tumor cell growth in vitro and inhibited human tumor xenograft growth in mice. These observations add to the rationale for testing adenosine A2A receptor antagonists as anticancer therapeutics. Not only could there be prevention of negative signaling in T cells within the tumor microenvironment and inhibition of angiogenesis, but also an inhibitory effect on tumor-promoting, immunosuppressive CAFs and a direct inhibitory effect on the tumor cells themselves.


Journal of Immunotherapy | 2013

Phase II trial of a GM-CSF-producing and CD40L-expressing bystander cell line combined with an allogeneic tumor cell-based vaccine for refractory lung adenocarcinoma

Ben C. Creelan; Scott Antonia; David Noyes; Terri B. Hunter; George R. Simon; Gerold Bepler; Charles Williams; Tawee Tanvetyanon; Eric B. Haura; Michael J. Schell; Alberto Chiappori

We created a vaccine in which irradiated allogeneic lung adenocarcinoma cells are combined with a bystander K562 cell line transfected with hCD40L and hGM-CSF. By recruiting and activating dendritic cells, we hypothesized that the vaccine would induce tumor regression in metastatic lung adenocarcinoma. Intradermal vaccine was given q14 days×3, followed by monthly ×3. Cyclophosphamide (300 mg/m2 IV) was administered before the first and fourth vaccines to deplete regulatory T cells. All-trans retinoic acid was given (150/mg/m2/d) after the first and fourth vaccines to enhance dendritic cell differentiation. Twenty-four participants were accrued at a single institution from October 2006 to June 2008, with a median age 64 years and median of 4 previous lines of systemic therapy. A total of 101 vaccines were administered. Common toxicities were headache (54%) and site reaction (38%). No radiologic responses were observed. Median overall survival was 7.9 months and median progression-free survival was 1.7 months. Of 14 patients evaluable for immunological study, 5 had peptide-induced CD8+ T-cell activation after vaccination. Overall, vaccine administration was feasible in an extensively pretreated population of metastatic lung cancer. Despite a suggestion of clinical activity in the subset with immune response, the trial did not meet the primary endpoint of inducing radiologic tumor regression.


Neoplasia | 2017

A Novel Antagonist of the Immune Checkpoint Protein Adenosine A2a Receptor Restores Tumor-Infiltrating Lymphocyte Activity in the Context of the Tumor Microenvironment

Melanie Mediavilla-Varela; Julio Castro; Alberto Chiappori; David Noyes; Dalia C. Hernandez; Bertrand Allard; John Stagg; Scott Antonia

BACKGROUND: Therapeutic strategies targeting immune checkpoint proteins have led to significant responses in patients with various tumor types. The success of these studies has led to the development of various antibodies/inhibitors for the different checkpoint proteins involved in immune evasion of the tumor. Adenosine present in high concentrations in the tumor microenvironment activates the immune checkpoint adenosine A2a receptor (A2aR), leading to the suppression of antitumor responses. Inhibition of this checkpoint has the potential to enhance antitumor T-cell responsiveness. METHODS: We developed a novel A2aR antagonist (PBF-509) and tested its antitumor response in vitro, in a mouse model, and in non-small cell lung cancer patient samples. RESULTS: Our studies showed that PBF-509 is highly specific to the A2aR as well as inhibitory of A2aR function in an in vitro model. In a mouse model, we found that lung metastasis was decreased after treatment with PBF-509 compared with its control. Furthermore, freshly resected tumor-infiltrating lymphocytes from lung cancer patients showed increased A2aR expression in CD4+ cells and variable expression in CD8+ cells. Ex vivo studies showed an increased responsiveness of human tumor-infiltrating lymphocytes when PBF-509 was combined with anti-PD-1 or anti-PD-L1. CONCLUSIONS: Our studies demonstrate that inhibition of the A2aR using the novel inhibitor PBF-509 could lead to novel immunotherapeutic strategies in non-small cell lung cancer.


Cancer Research | 2013

Abstract 4948: Pirfenidone as a potential therapeutic treatment for CAFs and cancer cells in NSCLC.

Melanie Mediavilla-Varela; Kimberly Luddy; David Noyes; Kingsley Boateng; Scott Antonia

Lung cancer is the leading cause of cancer death in the United States. Due to limited efficacy of current conventional chemotherapy, the majority of patients face poor prognosis and eventually succumb the disease. Until recently, the main focus in cancer research has been targeting the neoplastic cell. However, the importance of the tumor microenvironment, including cancer associated fibroblasts (CAFs), in cancer progression is now clear and targeting both, the neoplastic cell as well as the CAF, is being recognized as an important therapeutic approach. We have recently discovered that pirfenidone might be the drug to target these cells. Pirfenidone is a small molecule that has been shown to decrease fibrosis in a variety of fibrotic diseases. The exact mechanism responsible for its anti-fibrotic effects remains to be elucidated. Our hypothesis is that pirfenidone (by targeting the CAFs) in combination with standard chemotherapy (by targeting the cancer cells) will act synergistically and proffer a more potent therapeutic strategy for non-small cell lung cancer (NSCLC). Our studies show that in the presence of pirfenidone NSCLC associated CAF viability decreases in vitro. Furthermore, pirfenidone was able to induce apoptotic cell death as well as cell cycle arrest in NSCLC associated CAFs. In addition, we were able to observe that CAFs treated with pirfenidone decreased active TGFβ when compared to the control. Moreover, in a spheroid model, pirfenidone was able to affect the crosstalk between the cancer cells and CAFs leading to increased apoptosis and decreased viability. Currently, pirfenidone is being used in Phase II and III clinical trials for idiopathic pulmonary fibrosis. Our lab is the first one to recognize the potential of pirfenidone as a potential treatment of NSCLC. Citation Format: Melanie Mediavilla-Varela, Kimberly Luddy, David Noyes, Kingsley Boateng, Scott Antonia. Pirfenidone as a potential therapeutic treatment for CAFs and cancer cells in NSCLC. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4948. doi:10.1158/1538-7445.AM2013-4948


Journal of Clinical Oncology | 2018

Phase I/II study of nivolumab and ipilimumab combined with nintedanib in advanced NSCLC.

Sonam Puri; Sandrine Niyongere; Monica Sheila Chatwal; Theresa A. Boyle; Dung-Tsa Chen; David Noyes; Scott Antonia; Jhanelle E. Gray

TPS9112Background: Combination immunotherapy with nivolumab and ipilimumab has proven clinical activity in NSCLC. There is growing evidence to suggest that the tumor microenvironment (TME) may inte...


Cancer Immunology, Immunotherapy | 2018

A phase I/randomized phase II study of GM.CD40L vaccine in combination with CCL21 in patients with advanced lung adenocarcinoma

Jhanelle E. Gray; Alberto Chiappori; Charlie C. Williams; Tawee Tanvetyanon; Eric B. Haura; Ben C. Creelan; Jongphil Kim; Theresa A. Boyle; Mary Pinder-Schenck; Farah Khalil; Soner Altiok; Rebecca Devane; David Noyes; Melanie Mediavilla-Varela; Renee Smilee; Emily L. Hopewell; Linda Kelley; Scott Antonia

The GM.CD40L vaccine, which recruits and activates dendritic cells, migrates to lymph nodes, activating T cells and leading to systemic tumor cell killing. When combined with the CCL21 chemokine, which recruits T cells and enhances T-cell responses, additive effects have been demonstrated in non-small cell lung cancer mouse models. Here, we compared GM.CD40L versus GM.CD40L plus CCL21 (GM.CD40L.CCL21) in lung adenocarcinoma patients with ≥ 1 line of treatment. In this phase I/II randomized trial (NCT01433172), patients received intradermal vaccines every 14 days (3 doses) and then monthly (3 doses). A two-stage minimax design was used. During phase I, no dose-limiting toxicities were shown in three patients who received GM.CD40L.CCL21. During phase II, of evaluable patients, 5/33 patients (15.2%) randomized for GM.DCD40L (p = .023) and 3/32 patients (9.4%) randomized for GM.DCD40L.CCL21 (p = .20) showed 6-month progression-free survival. Median overall survival was 9.3 versus 9.5 months with GM.DCD40L versus GM.DCD40L.CCL21 (95% CI 0.70–2.25; p = .44). For GM.CD40L versus GM.CD40L.CCL21, the most common treatment-related adverse events (TRAEs) were grade 1/2 injection site reaction (51.4% versus 61.1%) and grade 1/2 fatigue (35.1% versus 47.2%). Grade 1 immune-mediated TRAEs were isolated to skin. No patients showed evidence of pseudo-progression or immune-related TRAEs of grade 1 or greater of pneumonitis, endocrinopathy, or colitis, and none discontinued treatment due to toxicity. Although we found no significant associations between vaccine immunogenicity and outcomes, in limited biopsies, one patient treated with GMCD40L.CCL21 displayed abundant tumor-infiltrating lymphocytes. This possible effectiveness warrants further investigation of GM.CD40L in combination approaches.

Collaboration


Dive into the David Noyes's collaboration.

Top Co-Authors

Avatar

Scott Antonia

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Alberto Chiappori

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric B. Haura

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Hatem Soliman

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Jhanelle E. Gray

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Anthony Neuger

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Ben C. Creelan

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Charles Williams

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

David Cheong

University of South Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge