Network


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

Hotspot


Dive into the research topics where Nicholas P. Morris is active.

Publication


Featured researches published by Nicholas P. Morris.


Cancer Research | 2013

OX40 Is a Potent Immune-Stimulating Target in Late-Stage Cancer Patients

Brendan D. Curti; Magdalena Kovacsovics-Bankowski; Nicholas P. Morris; Edwin B. Walker; Lana Chisholm; Kevin Floyd; Joshua M. Walker; Iliana Gonzalez; Tanisha Meeuwsen; Bernard A. Fox; Tarsem Moudgil; W.R. Miller; Daniel Haley; Todd Coffey; Brenda Fisher; Laurie Delanty-Miller; Nicole Rymarchyk; Tracy L Kelly; Todd Crocenzi; Eric D. Bernstein; Rachel Sanborn; Walter J. Urba; Andrew D. Weinberg

OX40 is a potent costimulatory receptor that can potentiate T-cell receptor signaling on the surface of T lymphocytes, leading to their activation by a specifically recognized antigen. In particular, OX40 engagement by ligands present on dendritic cells dramatically increases the proliferation, effector function, and survival of T cells. Preclinical studies have shown that OX40 agonists increase antitumor immunity and improve tumor-free survival. In this study, we performed a phase I clinical trial using a mouse monoclonal antibody (mAb) that agonizes human OX40 signaling in patients with advanced cancer. Patients treated with one course of the anti-OX40 mAb showed an acceptable toxicity profile and regression of at least one metastatic lesion in 12 of 30 patients. Mechanistically, this treatment increased T and B cell responses to reporter antigen immunizations, led to preferential upregulation of OX40 on CD4(+) FoxP3(+) regulatory T cells in tumor-infiltrating lymphocytes, and increased the antitumor reactivity of T and B cells in patients with melanoma. Our findings clinically validate OX40 as a potent immune-stimulating target for treatment in patients with cancer, providing a generalizable tool to favorably influence the antitumor properties of circulating T cells, B cells, and intratumoral regulatory T cells.


Immunological Reviews | 2011

Science gone translational: the OX40 agonist story

Andrew D. Weinberg; Nicholas P. Morris; Magdalena Kovacsovics-Bankowski; Walter J. Urba; Brendan D. Curti

Summary:  OX40 (CD134) is a tumor necrosis factor (TNF) receptor expressed primarily on activated CD4+ and CD8+ T cells and transmits a potent costimulatory signal when engaged. OX40 is transiently expressed after T‐cell receptor engagement and is upregulated on the most recently antigen‐activated T cells within inflammatory lesions (e.g. sites of autoimmune destruction and on tumor‐infiltrating lymphocytes). Hence, it is an attractive target to modulate immune responses: OX40 blocking agents to inhibit undesirable inflammation or OX40 agonists to enhance immune responses. In regards to this review, OX40 agonists enhance anti‐tumor immunity, which leads to therapeutic effects in mouse tumor models. A team of laboratory and clinical scientists at the Providence Cancer Center has collaborated to bring the preclinical observations in cancer models from the bench to the bedside. This review describes the journey from in vitro experiments through preclinical mouse models to the successful translation of the first OX40 agonist to the clinic for the treatment of patients with cancer.


Seminars in Oncology | 2010

Signaling Through OX40 Enhances Antitumor Immunity

Shawn M. Jensen; Levi D. Maston; Michael J. Gough; Carl E. Ruby; William L. Redmond; Marka Crittenden; Yuhuan Li; Sachin Puri; Christian H. Poehlein; Nicholas P. Morris; Magdalena Kovacsovics-Bankowski; Tarsem Moudgil; Chris Twitty; Edwin B. Walker; Hong-Ming Hu; Walter J. Urba; Andrew D. Weinberg; Brendan D. Curti; Bernard A. Fox

The existence of tumor-specific T cells, as well as their ability to be primed in cancer patients, confirms that the immune response can be deployed to combat cancer. However, there are obstacles that must be overcome to convert the ineffective immune response commonly found in the tumor environment to one that leads to sustained destruction of tumor. Members of the tumor necrosis factor (TNF) superfamily direct diverse immune functions. OX40 and its ligand, OX40L, are key TNF members that augment T-cell expansion, cytokine production, and survival. OX40 signaling also controls regulatory T-cell differentiation and suppressive function. Studies over the past decade have demonstrated that OX40 agonists enhance antitumor immunity in preclinical models using immunogenic tumors; however, treatment of poorly immunogenic tumors has been less successful. Combining strategies that prime tumor-specific T cells together with OX40 signaling could generate and maintain a therapeutic antitumor immune response.


Journal of Immunotherapy | 2006

Anti-OX40 (CD134) administration to nonhuman primates: immunostimulatory effects and toxicokinetic study.

Andrew D. Weinberg; Colin Thalhofer; Nicholas P. Morris; Joshua M. Walker; Donald Seiss; Scott W. Wong; Michael K. Axthelm; Louis J. Picker; Walter J. Urba

The immune-stimulatory properties of anti-CD134 (OX40) antibodies have been well documented in rodents, including their ability to enhance antitumor immunity. In this study, an anti-OX40 antibody (Ab) known to costimulate monkey T cells in vitro, was infused into rhesus macaque monkeys during immunization with the simian immunodeficiency virus protein, gp130. The draining lymph nodes from immunized monkeys treated with anti-OX40 were enlarged compared with immunized monkeys injected with mouse Ig. Anti-OX40-treated monkeys had increased gp130-specific Ab titers, and increased long-lived T-cell responses, compared with controls. There were no overt signs of toxicity in the anti-OX40-treated monkeys. The encouraging immune-stimulatory effects led to the good manufacturing practice production of an anti-OX40 Ab for clinical trials in cancer patients. A detailed toxicology study was performed with anti-OX40 in nonhuman primates. Three groups of 8 monkeys received anti-OX40 at 1 of 3 dose levels (0.4, 2.0, and 10 mg/kg) and a control group received saline. No clinical toxicity was observed, but acute splenomegaly and enlarged gut-associated lymph nodes were observed in the anti-OX40-treated animals; splenomegaly and lymphadenopathy resolved by day 28. These studies demonstrate the immune-stimulatory properties and safety of anti-OX40 in primates and provide a strong scientific rationale to pursue clinical trials in humans.


Scientific Reports | 2016

Combinational Immunotherapy with Allo-DRibble Vaccines and Anti-OX40 Co-Stimulation Leads to Generation of Cross-Reactive Effector T Cells and Tumor Regression

Guangjie Yu; Yuhuan Li; Zhihua Cui; Nicholas P. Morris; Andrew D. Weinberg; Bernard A. Fox; Walter J. Urba; Li-Xin Wang; Hong-Ming Hu

It is well-known that vaccines comprising of irradiated whole tumor cells or tumor-derived heat shock proteins can generate tumor-specific immune responses. In contrast, we showed recently that vaccines composed of autophagosomes (DRibbles) derived from syngeneic sarcomas could induce cross-reactive T-cell responses and cross-protection against the tumor. This unusual property of DRibbles was related to the selective recruitment of defective ribosomal products (DRiPs) and other short-lived proteins (SLiPs) into autophagosomes via sequestosome (SQSTM1, p62) mediated association of ubiquitinated SLiPs to the autophagy gene product LC3. Here, we extend our observations to mammary carcinomas from mice of different genetic background. We demonstrated that combined of intranodal administration of autologous or allogeneic DRibbles together with anti-OX40 antibody led to robust proliferation, expansion, and differentiation of memory and effector T cells. We also showed that SLiPs is an excellent source of antigen for cross-priming of CD8+ T-cells that recognize shared tumor antigens in the context of host MHC class I molecules. Thus, our results provide a strong basis for novel clinical trials that combine allogeneic “off-the-shelf” DRibble vaccines together with antibodies against co-stimulatory molecules.


Molecular Cancer Therapeutics | 2018

Potent Immune Modulation by MEDI6383, an Engineered Human OX40 Ligand IgG4P Fc Fusion Protein

Michael Oberst; Catherine Auge; Chad Morris; Stacy Kentner; Kathy Mulgrew; Kelly McGlinchey; James Hair; Shino Hanabuchi; Qun Du; Melissa Damschroder; Hui Feng; Steven Eck; Nicholas Buss; Lolke de Haan; Andrew Pierce; Haesun Park; Andrew W. Sylwester; Michael K. Axthelm; Louis J. Picker; Nicholas P. Morris; Andrew D. Weinberg; Scott A. Hammond

Ligation of OX40 (CD134, TNFRSF4) on activated T cells by its natural ligand (OX40L, CD252, TNFSF4) enhances cellular survival, proliferation, and effector functions such as cytokine release and cellular cytotoxicity. We engineered a recombinant human OX40L IgG4P Fc fusion protein termed MEDI6383 that assembles into a hexameric structure and exerts potent agonist activity following engagement of OX40. MEDI6383 displayed solution-phase agonist activity that was enhanced when the fusion protein was clustered by Fc gamma receptors (FcγRs) on the surface of adjacent cells. The resulting costimulation of OX40 on T cells induced NFκB promoter activity in OX40-expressing T cells and induced Th1-type cytokine production, proliferation, and resistance to regulatory T cell (Treg)-mediated suppression. MEDI6383 enhanced the cytolytic activity of tumor-reactive T cells and reduced tumor growth in the context of an alloreactive human T cell:tumor cell admix model in immunocompromised mice. Consistent with the role of OX40 costimulation in the expansion of memory T cells, MEDI6383 administered to healthy nonhuman primates elicited peripheral blood CD4 and CD8 central and effector memory T-cell proliferation as well as B-cell proliferation. Together, these results suggest that OX40 agonism has the potential to enhance antitumor immunity in human malignancies. Mol Cancer Ther; 17(5); 1024–38. ©2018 AACR.


Molecular Immunology | 2007

Development and characterization of recombinant human Fc:OX40L fusion protein linked via a coiled-coil trimerization domain

Nicholas P. Morris; Carmen Peters; Ryan Montler; Hong-Ming Hu; Brendan D. Curti; Walter J. Urba; Andrew D. Weinberg


Archive | 2006

Trimeric ox40-immunoglobulin fusion protein and methods of use

Andrew D. Weinberg; Nicholas P. Morris; Carmen Peters


Cancer Research | 2015

Abstract 4275: Agonist OX40 ligand fusion proteins induce effector T cell proliferation, block regulatory T cell function and can combine with immune checkpoint inhibitors to promote antitumor immunity in preclinical models

Kelly McGlinchey; Kathy Mulgrew; Chad Morris; Catherine Auge; Nicholas Holoweckyj; Nicholas M. Durham; Karen Coffman; James Hair; Terrance O'Day; Nicholas P. Morris; Andrew D. Weinberg; Ching Ching Leow; Michael Oberst; Scott A. Hammond


Archive | 2006

Proteína de fusión OX40-inmunoglobulina trimérica y métodos de uso

Andrew D. Weinberg; Nicholas P. Morris; Carmen Peters

Collaboration


Dive into the Nicholas P. Morris's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carmen Peters

Providence Portland Medical Center

View shared research outputs
Top Co-Authors

Avatar

Walter J. Urba

Providence Portland Medical Center

View shared research outputs
Top Co-Authors

Avatar

Brendan D. Curti

Providence Portland Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hong-Ming Hu

Providence Portland Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Edwin B. Walker

Providence Portland Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge