Network


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

Hotspot


Dive into the research topics where Nicholas N. Vahanian is active.

Publication


Featured researches published by Nicholas N. Vahanian.


OncoImmunology | 2012

IDO inhibits a tryptophan sufficiency signal that stimulates mTOR: A novel IDO effector pathway targeted by D-1-methyl-tryptophan.

Richard Metz; Sonja Rust; James B. DuHadaway; Mario R. Mautino; David H. Munn; Nicholas N. Vahanian; Charles J. Link; George C. Prendergast

Tryptophan catabolism by indoleamine 2,3-dioxygenase (IDO) alters inflammation and favors T-cell tolerance in cancer, but the underlying molecular mechanisms remain poorly understood. The integrated stress response kinase GCN2, a sensor of uncharged tRNA that is activated by amino acid deprivation, is recognized as an important effector of the IDO pathway. However, in a mouse model of inflammatory carcinogenesis, ablation of Gcn2 did not promote resistance against tumor development like the absence of IDO does, implying the existence of additional cancer-relevant pathways that operate downstream of IDO. Addressing this gap in knowledge, we report that the IDO-mediated catabolism of tryptophan also inhibits the immunoregulatory kinases mTOR and PKC-Θ, along with the induction of autophagy. These effects were relieved specifically by tryptophan but also by the experimental agent 1-methyl-D-tryptophan (D-1MT, also known as NLG8189), the latter of which reversed the inhibitory signals generated by IDO with higher potency. Taken together, our results implicate mTOR and PKC-Θ in IDO-mediated immunosuppressive signaling, and they provide timely insights into the unique mechanism of action of D-1MT as compared with traditional biochemical inhibitors of IDO. These findings are important translationally, because they suggest broader clinical uses for D-1MT against cancers that overexpress any tryptophan catabolic enzyme (IDO, IDO2 or TDO). Moreover, they define mTOR and PKC-Θ as candidate pharmacodynamic markers for D-1MT responses in patients recruited to ongoing phase IB/II cancer trials, addressing a current clinical need.


Virology | 2010

A Replication-incompetent Rift Valley Fever Vaccine: Chimeric Virus-like Particles Protect Mice and Rats Against Lethal Challenge

Robert B. Mandell; Ramesh Koukuntla; Laura J.K. Mogler; Andrea K. Carzoli; Alexander N. Freiberg; Brian K. Martin; William Staplin; Nicholas N. Vahanian; Charles J. Link; Ramon Flick

Abstract Virus-like particles (VLPs) present viral antigens in a native conformation and are effectively recognized by the immune system and therefore are considered as suitable and safe vaccine candidates against many viral diseases. Here we demonstrate that chimeric VLPs containing Rift Valley fever virus (RVFV) glycoproteins GN and GC, nucleoprotein N and the gag protein of Moloney murine leukemia virus represent an effective vaccine candidate against Rift Valley fever, a deadly disease in humans and livestock. Long-lasting humoral and cellular immune responses are demonstrated in a mouse model by the analysis of neutralizing antibody titers and cytokine secretion profiles. Vaccine efficacy studies were performed in mouse and rat lethal challenge models resulting in high protection rates. Taken together, these results demonstrate that replication-incompetent chimeric RVF VLPs are an efficient RVFV vaccine candidate.


Cancer Research | 2005

Effective Treatment of Preexisting Melanoma with Whole Cell Vaccines Expressing α(1,3)-Galactosyl Epitopes

Gabriela R. Rossi; Mario R. Mautino; Robert C. Unfer; Tatiana Seregina; Nicholas N. Vahanian; Charles J. Link

The hyperacute immune response in humans is a potent mechanism of xenograft rejection mediated by complement-fixing natural antibodies recognizing alpha(1,3)-galactosyl epitopes (alphaGal) not present on human cells. We exploited this immune mechanism to create a whole cell cancer vaccine to treat melanoma tumors. B16 melanoma vaccines genetically engineered to express alphaGal epitopes (B16alphaGal) effectively treated preexisting s.c. and pulmonary alphaGal-negative melanoma (B16Null) tumors in the alpha(1,3)-galactosyltransferase knockout mouse model. T cells from mice vaccinated with B16alphaGal recognized B16Null melanoma cells measured by detection of intracellular tumor necrosis factor-alpha. We showed successful adoptive transfer of immunity to recipient mice bearing lung melanoma metastasis. Mice receiving lymphocytes from donors previously immunized with B16alphaGal had reduced pulmonary metastases. The transfer of lymphocytes from mice vaccinated with control vaccine had no effect in the pulmonary metastasis burden. This study unequivocally establishes for the first time efficacy in the treatment of preexisting melanoma tumors using whole cell vaccines expressing alphaGal epitopes. Vaccination with B16alphagal induced strong long-lasting cell-mediated antitumor immunity extended to B16Null. These data formed the basis for the testing of this therapeutic strategy in human clinical trials currently under way.


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.


Stem Cells | 2008

Hematopoietic Mixed Chimerism Derived from Allogeneic Embryonic Stem Cells Prevents Autoimmune Diabetes Mellitus in NOD Mice

Larissa Verda; Duck-An Kim; Susumu Ikehara; Laisvyde Statkute; Delphine Bronesky; Yevgeniya Petrenko; Yu Oyama; Xiang He; Charles J. Link; Nicholas N. Vahanian; Richard K. Burt

Embryonic stem cell (ESC)‐derived hematopoietic stem cells (HSC), unlike HSC harvested from the blood or marrow, are not contaminated by lymphocytes. We therefore evaluated whether ESC‐derived HSC could produce islet cell tolerance, a phenomenon termed graft versus autoimmunity (GVA), without causing the usual allogeneic hematopoietic stem cell transplant complication, graft‐versus‐host disease (GVHD). Herein, we demonstrate that ESC‐derived HSC may be used to prevent autoimmune diabetes mellitus in NOD mice without GVHD or other adverse side effects. ESC were cultured in vitro to induce differentiation toward HSC, selected for c‐kit expression, and injected either i.v. or intra‐bone marrow (IBM) into sublethally irradiated NOD/LtJ mice. Nine of 10 mice from the IBM group and 5 of 8 from the i.v. group did not become hyperglycemic, in contrast to the control group, in which 8 of 9 mice developed end‐stage diabetes. All mice with >5% donor chimerism remained free of diabetes and insulitis, which was confirmed by histology. Splenocytes from transplanted mice were unresponsive to glutamic acid decarboxylase isoform 65, a diabetic‐specific autoantigen, but responded normally to third‐party antigens. ESC‐derived HSC can induce an islet cell tolerizing GVA effect without GVHD. This study represents the first instance, to our knowledge, of ESC‐derived HSC cells treating disease in an animal model.


Cancer Research | 2013

Abstract 491: NLG919, a novel indoleamine-2,3-dioxygenase (IDO)-pathway inhibitor drug candidate for cancer therapy.

Mario R. Mautino; Firoz Jaipuri; Jesse Waldo; Sanjeev Kumar; James Adams; Clarissa Van Allen; Agnieszka Marcinowicz-Flick; David H. Munn; Nicholas N. Vahanian; Charles J. Link

The IDO pathway mediates immunosuppressive effects through the metabolization of tryptophan (Trp) to kynurenine (Kyn), triggering downstream signaling through GCN2, mTOR and AHR that can affect differentiation and proliferation of T cells. Expression of the IDO1 gene by tumor cells or host APCs can inhibit tumor-specific effector CD8+ T cells and enhance the suppressor activity of Tregs, and high expression of IDO correlates with worse clinical prognosis in patients with a variety of malignancies. Therefore, targeting the IDO pathway via inhibition of the IDO enzyme or blocking its downstream signaling effects is a prime target for small-molecule immunomodulatory drugs in cancer. Here we describe the pharmacological and biological properties of NLG919, a novel small-molecule IDO-pathway inhibitor. NLG919 potently inhibits this pathway in vitro and in cell based assays (Ki=7 nM; EC50 =75 nM). It is orally bioavailable (F>70%); and has a favorable pharmacokinetic and toxicity profile. In mice, a single oral administration of NLG919 reduces the concentration of plasma and tissue Kyn by ∼ 50%. Using IDO-expressing human monocyte-derived DCs in allogeneic MLR reactions, NLG919 potently blocked IDO-induced T cell suppression and restored robust T cell responses with an ED50=80 nM. Similarly, using IDO-expressing mouse DCs from tumor-draining lymph nodes, NLG919 abrogated IDO-induced suppression of antigen-specific T cells (OT-I) in vitro, with ED50=120 nM. In vivo, in mice bearing large established B16F10 tumors, administration of NLG919 markedly enhanced the anti-tumor responses of naive, resting pmel-1 cells to vaccination with cognate hgp100 peptide plus CpG-1826 in IFA. In this stringent established-tumor model, NLG919 plus pmel 1/vaccine produced a dramatic collapse of tumor size within 4 days of vaccination (∼95% reduction in tumor volume compared to control animals receiving pmel-1/vaccine alone without NLG919). In conclusion, NLG919 is a potent IDO pathway inhibitor with desirable pharmacological properties, suitable for the treatment of immunosuppression associated with cancer. Citation Format: Mario R. Mautino, Firoz A. Jaipuri, Jesse Waldo, Sanjeev Kumar, James Adams, Clarissa Van Allen, Agnieszka Marcinowicz-Flick, David Munn, Nicholas Vahanian, Charles J. Link. NLG919, a novel indoleamine-2,3-dioxygenase (IDO)-pathway inhibitor drug candidate for cancer therapy. [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 491. doi:10.1158/1538-7445.AM2013-491


Journal of Virological Methods | 2010

Novel suspension cell-based vaccine production systems for Rift Valley fever virus-like particles.

Robert B. Mandell; Ramesh Koukuntla; Laura J.K. Mogler; Andrea K. Carzoli; Brian K. Martin; Nicholas N. Vahanian; Charles J. Link; Ramon Flick

Rift Valley fever virus (RVFV) is an arthropod-borne pathogen that often results in severe morbidity and mortality in both humans and livestock. As its geographic range continues to expand, it presents a real threat to naïve populations around the world by accidental introduction (e.g., the result of increased travel) or intentional release (e.g., a bioterror event). While there is a clear need for a safe and efficacious vaccine against this emerging and re-emerging pathogen, no FDA-approved vaccine is currently available. This need was addressed by the establishment of novel mammalian and insect suspension cell line systems for the efficient production of RVF virus-like particle (VLP)-based vaccine candidates. A direct comparison of the production of RVF VLPs in these systems was performed. Optimization and characterization resulted in a production platform suitable for scale-up. Furthermore, RVF VLP-based vaccines were tested in a lethal challenge model and showed full protection, demonstrating that RVF VLPs present promising RVFV vaccine candidates.


Journal of Immunotherapy | 2008

Allogeneic Melanoma Vaccine Expressing αGal Epitopes Induces Antitumor Immunity to Autologous Antigens in Mice Without Signs of Toxicity

Gabriela R. Rossi; Mario R. Mautino; Dana Z. Awwad; Katie Husske; Henry Lejukole; Marie Koenigsfeld; William Jay Ramsey; Nicholas N. Vahanian; Charles J. Link

Owing to the absence of αGal epitopes in human cells and constant stimulation of the immune system by the symbiotic bacterial flora, humans develop high titers of natural antibodies against these epitopes. It has been demonstrated that syngeneic whole cell vaccines modified to express αGal epitopes could be used to generate a potent anticancer vaccine. In this study, we tested whether allogeneic whole cell cancer vaccines modified to express αGal epitopes would be effective for the treatment of murine melanoma. The α(1,3)galactosyltransferase (αGT) knockout mice (H-2b/b) with preexisting subcutaneous and pulmonary tumors [αGal(−) B16, H-2b/b] received therapeutic vaccinations with S91M3αGal(+) (H-2d/d) whole cell allogeneic vaccines. These mice had better survival and reduced pulmonary metastasis burden compared with control mice treated with S91M3 vaccine cells. Vaccination with S91M3αGal-induced cytotoxic CD8+ T cells recognizing the syngeneic αGal(−) B16 tumors measured by adoptive transfer to recipients bearing pulmonary metastases. The presence of allo-antigens did not dominate the induction of immunity to “cryptic” tumor antigens and had helped in the generation of a more efficient vaccine to treat preexisting tumors when compared with classic autologous vaccines. Vaccination with allogeneic αGal(+) vaccines did not induce signs of toxicity including changes in weight, hematology, chemistry, and histopathology of major perfused organs or autoimmunity in long-term murine models for breast, lung, and melanoma. This study established the safety and efficacy data of allogeneic αGal(+) whole cell vaccines and constituted the basis for the initiation of human clinical trials to treat human malignancies.


Cancer Research | 2017

Abstract CT117: Interim analysis of the Phase 2 clinical trial of the IDO pathway inhibitor indoximod in combination with pembrolizumab for patients with advanced melanoma

Yousef Zakharia; Robert R. McWilliams; Monaster Shaheen; Kenneth Grossman; Joseph J. Drabick; Mohammed M. Milhem; Olivier Rixie; Samir N. Khleif; Ryan Lott; Eugene Kennedy; David H. Munn; Nicholas N. Vahanian; Charles J. Link

Background: The indoleamine 2,3-dioxygenase (IDO) pathway is a key counter-regulatory mechanism that normally inhibits immune responses when appropriate. In the setting of cancer, IDO pathway-mediated immune suppression is exploited by tumors in order to prevent and defeat anti-tumor immunity. Small-molecule inhibitors of the IDO pathway, such as indoximod, are an increasingly validated class of potential cancer therapeutics. Additionally, pre-clinical tumor models have shown complementary effects with indoximod / anti-PD1 checkpoint inhibitor treatment combinations. A clinical trial was developed based upon these data. Methods: Upon successful completion of a Phase 1b dose escalation cohort, metastatic melanoma patients were enrolled in a single arm Phase 2 trial evaluating the addition of indoximod to standard of care checkpoint inhibitors approved for melanoma. Treating physicians were allowed to administer their choice of approved checkpoint inhibitor. The large majority of patients received indoximod with pembrolizumab and this interim report is limited to those patients. Indoximod was administered continuously in 21 days cycles (1200mg po twice daily) concurrently with pembrolizumab (3mg/kg q21 days). Study endpoint is best overall response (objective response rate (ORR) = complete response rate (CR) + partial response rate (PR)) per site reported RECIST criteria. Results: At time of data cut-off, 60 patients had received indoximod /pembrolizumab and were evaluable for response, defined as having at least one follow-up imaging study performed. The ORR was 52% (31/60) with a CRR of 8% (5/60). The combination was well tolerated. The most frequently reported adverse events (regardless of attribution), occurring in ≥ 20% of subjects, were fatigue, diarrhea, nausea, arthralgia, headache, cough, rash, pruritus, and hypertension. The most frequently reported laboratory abnormalities (regardless of attribution), were anemia (17%) and hyperglycemia (17%). Conclusions: The interim analysis of the combination of indoximod and pembrolizumab demonstrates an ORR of 52% which compares favorably with the established ORR for pembrolizumab alone. Updated data to be presented. NCT02073123. Citation Format: Yousef Zakharia, Robert McWilliams, Monaster Shaheen, Kenneth Grossman, Joseph Drabick, Mohammed Milhem, Olivier Rixie, Samir Khleif, Ryan Lott, Eugene Kennedy, David Munn, Nicholas Vahanian, Charles Link. Interim analysis of the Phase 2 clinical trial of the IDO pathway inhibitor indoximod in combination with pembrolizumab for patients with advanced melanoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr CT117. doi:10.1158/1538-7445.AM2017-CT117


Journal for ImmunoTherapy of Cancer | 2014

A Phase I study of NLG919 for adult patients with recurrent advanced solid tumors

Asha Nayak; Zhonglin Hao; R. Sadek; Nicholas N. Vahanian; William Jay Ramsey; Eugene Kennedy; Mario R. Mautino; Charles J. Link; Pamela Bourbo; Robin Dobbins; Kelly Adams; Allison Diamond; Lisa Marshall; David H. Munn; John Janik; Samir N. Khleif

Meeting abstracts Clinical Trial Registration Number: [NCT02048709][1] The enzyme Indoleamine 2,3-dioxygenase (IDO1) catalyzes the cleavage of L-tryptophan, resulting in the production of kynurenine. Tryptophan depletion and kynurenine metabolites enhance the number and function of Tregs (

Collaboration


Dive into the Nicholas N. Vahanian's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gabriela R. Rossi

National Foundation for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Eugene Kennedy

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

David H. Munn

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Mario R. Mautino

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yousef Zakharia

University of Iowa Hospitals and Clinics

View shared research outputs
Top Co-Authors

Avatar

R. Sadek

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Hatem Soliman

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

John C. Morris

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Scott Antonia

University of South Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge