Network


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

Hotspot


Dive into the research topics where Neil Oronsky is active.

Publication


Featured researches published by Neil Oronsky.


Anti-cancer Agents in Medicinal Chemistry | 2014

Episensitization: Therapeutic Tumor Resensitization by Epigenetic Agents: A Review and Reassessment

Bryan Oronsky; Neil Oronsky; Susan J. Knox; Gary Fanger; Jan Scicinski

Resistance to chemotherapy, biological and targeted therapies is an important clinical problem. Resistance can arise and/or be selected for multiple mechanisms of action. Unfortunately, acquired resistance to antitumor agents or regimens is nearly inevitable in all patients with metastatic disease. Until recently, it was believed that this resistance was unalterable and irreversible, rendering retreatment with the same or similar drugs futile in most cases. However, the introduction of epigenetic therapies, including HDAC inhibitors and DNA methyltransferase inhibitors (DNMTIs), has provided oncologists with new strategies to potentially overcome this resistance. For example, if chemoresistance is the product of multiple non-genetic alterations, which develop and accumulate over time in response to treatment, then the ability to epigenetically modify the tumor to reconfigure it back to its baseline non-resistant state, holds tremendous promise for the treatment of advanced, metastatic cancer. This minireview aims (1) to explore the potential mechanisms by which a group of small molecule agents including HDACs (entinostat and vorinostat), DNA hypomethylating agents such as the DNMTIs (decitabine (DEC), 5-azacytidine (5-AZA)) and redox modulators (RRx-001) may reprogram the tumors from a refractory to non-refractory state, (2) highlight some recent findings in this area, and (3) discuss the therapeutic potential of resensitization approaches with formerly failed chemotherapies.


Translational Oncology | 2014

Rewriting the Epigenetic Code for Tumor Resensitization: A Review

Bryan Oronsky; Neil Oronsky; Jan Scicinski; Gary Fanger; Michelle Lybeck; Tony Reid

In cancer chemotherapy, one axiom, which has practically solidified into dogma, is that acquired resistance to antitumor agents or regimens, nearly inevitable in all patients with metastatic disease, remains unalterable and irreversible, rendering therapeutic rechallenge futile. However, the introduction of epigenetic therapies, including histone deacetylase inhibitors (HDACis) and DNA methyltransferase inhibitors (DNMTIs), provides oncologists, like computer programmers, with new techniques to “overwrite” the modifiable software pattern of gene expression in tumors and challenge the “one and done” treatment prescription. Taking the epigenetic code-as-software analogy a step further, if chemoresistance is the product of multiple nongenetic alterations, which develop and accumulate over time in response to treatment, then the possibility to hack or tweak the operating system and fall back on a “system restore” or “undo” feature, like the arrow icon in the Windows XP toolbar, reconfiguring the tumor to its baseline nonresistant state, holds tremendous promise for turning advanced, metastatic cancer from a fatal disease into a chronic, livable condition. This review aims 1) to explore the potential mechanisms by which a group of small molecule agents including HDACis (entinostat and vorinostat), DNMTIs (decitabine and 5-azacytidine), and redox modulators (RRx-001) may reprogram the tumor microenvironment from a refractory to a nonrefractory state, 2) highlight some recent findings, and 3) discuss whether the current “once burned forever spurned” paradigm in the treatment of metastatic disease should be revised to promote active resensitization attempts with formerly failed chemotherapies.


Frontiers in Oncology | 2015

Episensitization: Defying Time’s Arrow

Bryan Oronsky; Arnold Oronsky; Michelle Lybeck; Neil Oronsky; Jan Scicinski; Corey A. Carter; Regina M. Day; Jose F. Rodriguez Orengo; Maribel Rodriguez-Torres; Gary F. Fanger; Tony Reid

The development of cancer is driven by complex genetic and epigenetic changes that result in aberrant and uncontrolled cellular growth. Epigenetic changes, in particular, are implicated in the silencing or activation of key genes that control cellular growth and apoptosis and contribute to transformative potential. The purpose of this review is to define and assess the treatment strategy of “episensitization,” or the ability to sensitize cancer cells to subsequent therapy by resetting the epigenetic infrastructure of the tumor. One important facet is resensitization by epigenetic mechanisms, which goes against the norm, i.e., challenges the long-held doctrine in oncology that the reuse of previously tried and failed therapies is a clinically pointless endeavor. Thus, episensitization is a hybrid term, which covers recent clinically relevant observations and refers to the epigenomic mechanism of resensitization. Among the many formidable challenges in the treatment of cancer, the most inevitable is the development of acquired therapeutic resistance. Here, we present the basic principles behind episensitization and highlight the evidence suggesting that epigenetically mediated histone hypoacetylation and DNA hypermethylation events may reverse clinical drug resistance. The potential reversibility of epigenetic changes and the microenvironmental impact of epigenetic control on gene expression may mediate a return to a baseline state of treatment susceptibility. Episensitization is a novel and highly practical management strategy both to prevent the practice of permanent treatment discontinuation with the occurrence of resistance, which rapidly exhausts remaining options in the pharmaceutical armamentarium and to significantly extend patient survival. Accordingly, this review highlights several epigenetic agents including decitabine, vorinostat, entinostat, 5-azacitidine, oncolytic viruses, and RRx-001.


Translational Oncology | 2014

The implications of hyponitroxia in cancer.

Bryan Oronsky; Gary Fanger; Neil Oronsky; Susan J. Knox; Jan Scicinski

Tumors are spatially heterogeneous, with regions of relative hypoxia and normoxia. The tumor microenvironment is an important determinant of both tumor growth and response to a variety of cytotoxic and targeted therapies. In the tumor microenvironment, reactive oxygen species and nitric oxide (NO) are important mediators of the level of expression of many transcription factors and signaling cascades that affect tumor growth and responses to therapy. The primary objective of this review is to explore and discuss the seemingly dichotomous actions of NO in cancer biology as both a tumor promoter and suppressor with an emphasis on understanding the role of persistently low NO concentrations or hyponitroxia as a key mediator in tumor progression. This review will also discuss the potential role of hyponitroxia as a novel therapeutic target to treat cancer and outline an approach that provides new opportunities for pharmacological intervention.


Frontiers in Oncology | 2015

The war on cancer: a military perspective

Bryan Oronsky; Corey A. Carter; Vernon Mackie; Jan Scicinski; Arnold Oronsky; Neil Oronsky; Scott Caroen; Chris Parker; Michelle Lybeck; Tony Reid

Actually it has not quite happened yet, but almost imperceptibly, by degrees, we are learning to live with cancer. The “War on Cancer,” although generally successful in the pediatric population, has gradually been replaced with a kinder, gentler treatment paradigm that strives to contain and maintain with stalemate over checkmate, a strategy that may literally constitute the path to least resistance. The purpose of this review is (1) to critically examine the War on Cancer as a powerfully evocative metaphor that is directly responsible for a counterproductive and even potentially dangerous war-like cell-kill treatment paradigm, (2) to suggest that a reframing of this metaphor in less retaliatory and aggressive terms along with a shift in clinical practice from a maximalist to a minimalist strategy is more appropriate to the treatment of cancer, and (3) to draw on examples from the military sector as points of reference and comparison that closely parallel the three therapeutic “control and containment” strategies discussed in this review: (1) “Optimox-like” trial designs, (2) epigenetic modulation, and (3) metronomic dosing.


Case Reports in Oncology | 2016

RRx-001 in Refractory Small-Cell Lung Carcinoma: A Case Report of a Partial Response after a Third Reintroduction of Platinum Doublets

Corey A. Carter; Bryan Oronsky; Scott Caroen; Jan Scicinski; Aiste Degesys; Michelle M. Kim; Arnold Oronsky; Harry Lybeck; Pedro Cabrales; Neil Oronsky; Tony Reid; Joseph Roswarski; Christina Brzezniak

RRx-001 is a pan-active, systemically nontoxic epigenetic inhibitor under investigation in advanced non-small cell lung cancer, small-cell lung cancer and high-grade neuroendocrine tumors in a Phase II clinical trial entitled TRIPLE THREAT (NCT02489903), which reexposes patients to previously effective but refractory platinum doublets after treatment with RRx-001. The purpose of this case study is first to report a partial response to carboplatin and etoposide in a patient with small-cell lung cancer pretreated with RRx-001, indicating episensitization or resensitization by epigenetic mechanisms, and second to discuss the literature related to small-cell lung cancer and episensitization.


Neoplasia | 2015

Confirmatory Trials in the Evaluation of Anticancer Medicinal Products in Man--PFS2: A Measure of Therapeutic Action-At-A-Distance.

Bryan Oronsky; Corey A. Carter; Tony Reid; Jan Scicinski; Arnold Oronsky; Michelle Lybeck; Scott Caroen; Meaghan Stirn; Neil Oronsky; Peter Langecker

Overall survival (OS) has emerged as the definitive regulatory “be-all, end-all” for the demonstration of benefit in cancer clinical trials. The reason and the rationale for why this is so are easily appreciated: literally a “test of time,” OS is a seemingly unambiguous, agenda-free end point, independent of bias-prone variables such as the frequency and methods of assessment, clinical evaluation, and the definition of progression. However, by general consensus, OS is an imperfect end point for several reasons: First, it may often be impractical because of the length, cost, and the size of clinical trials. Second, OS captures the impact of subsequent therapies, both beneficial (i.e., active) and detrimental, on survival but it does not take into account the contribution of subsequent therapies by treatment arm; the postprogression period is treated as an unknown black box (no information about the potential influence of next-line therapies on the outcome) under the implicit assumption that the clinical trial treatment is the only clinical variable that matters: what OS explicitly measures is the destination, that is, the elapsed time between the date of randomization (or intention to treat) and the date of death, not the journey, that is, what transpires in-between. In long-term maintenance strategies, patients receive treatment in temporally separated but mutually interdependent and causally linked sequences that exert a “field of influence” akin to action-at-a-distance forces like gravity, electricity, and magnetism on both the tumor and each other. Hence, in this setting, a new end point, PFS2, is required to measure this field of influence. This article reviews the definition and use in clinical trials of PFS2 and makes the case for its potential applicability as a preferred end point to measure the mutual influence of individual regimens in long-term maintenance strategies with resensitizing agents in particular.


Journal of Cancer Science & Therapy | 2015

A Review of Two Promising Radiosensitizers in Brain Metastases: Rrx-001 and 2-Deoxyglucose

Bryan Oronsky; Neil Oronsky; Gary R. Fanger; Arnold Oronsky; Michelle Lybeck; Harry Lybeck; Scott Caroen; Christopher W. Parker; Jan Scicinski

The origin of the common phrase “your name is mud” may derive from the ordeal of 19th century physician, Dr. Samuel Mudd, who was perhaps wrongly convicted of conspiracy in the assassination of President Abraham Lincoln. Mudd’s crime may have only been bad luck: Lincoln’s assassin, John Wilkes Booth, allegedly previously unknown to the doctor, had broken his leg and happened across Mudd who, unwisely, as it turned out, set the fracture, and his own subsequent fate, which included life imprisonment with hard labor, making him a potential victim of circumstance rather than the perpetrator of a crime. Mudd’s grandson, also a physician, tried unsuccessfully to clear his grandfather’s widely reviled name, which as a result has remained, both literally and figuratively, Mudd. This historical analogy highlights the important point that radiosensitizers as a class have been ignored rather than adored due to their failed reputation. Hence, in the field of radiation oncology, the “your name is mud” expression applies to radiation sensitizers, which from hyperbaric oxygen and the nitroimidazoles, to motexafin gadolinium, tirapazamine and efaproxiral have generally overpromised and under delivered with respect to survival treatment benefits in multiple different indications. However, newer non-toxic radiosensitizers on the horizon such as the antienergetic epigenetic redox modulator, RRx-001, that will start a Phase 2 clinical trial with concurrent whole brain radiotherapy (WBRT) in subjects with brain metastases, may finally validate the underlying promise and unrealized potential of these agents. The successful treatment of brain metastases is at least a four-hurdle process involving penetration, retention, selectivity and toxicity. This article will review the mechanism of the radiosensitizers, RRx- 001, and 2-deoxyglucose, as examples or “role models” for therapies that theoretically are able to overcome these substantial in vivo obstacles to successfully treat brain metastases. It is the thesis of this review that new radiosensitizers are urgently needed and their poor reputation should be overcome.


OncoImmunology | 2017

No patient left behind: The promise of immune priming with epigenetic agents

Corey A. Carter; Bryan Oronsky; Joseph Roswarski; Arnold Oronsky; Neil Oronsky; Jan Scicinski; Harry Lybeck; Michelle M. Kim; Michelle Lybeck; Tony Reid

ABSTRACT Checkpoint inhibitors, monoclonal antibodies that inhibit PD-1 or CTLA-4, have revolutionized the treatment of multiple cancers. Despite the enthusiasm for the clinical successes of checkpoint inhibitors, and immunotherapy, in general, only a minority of patients with specific tumor types actually benefit from treatment. Emerging evidence implicates epigenetic alterations as a mechanism of clinical resistance to immunotherapy. This review presents evidence for that association, summarizes the epi-based mechanisms by which tumors evade immunogenic cell death, discusses epigenetic modulation as a component of an integrated strategy to boost anticancer T cell effector function in relation to a tumor immunosuppression cycle and, finally, makes the case that the success of this no-patient-left-behind strategy critically depends on the toxicity profile of the epigenetic agent(s).


Oncotarget | 2016

Addressing the elephant in the room, therapeutic resistance in non-small cell lung cancer, with epigenetic therapies

Corey A. Carter; Karen Zeman; Regina M. Day; Patrick Richard; Arnold Oronsky; Neil Oronsky; Michelle Lybeck; Jan Scicinski; Bryan Oronsky

Like Chinese boxes nesting inside each other, the classification of non-small cell lung cancer (NSCLC) is subdivided into smaller and smaller subtypes on the basis of histological and molecular attributes. The latter characterizes NSCLC by its molecular alterations and the identification of inhibitors that target these cancer-specific “driver” mutations. Despite the initial promise of precision-guided therapies to inhibit a finer and finer array of molecular subcategories, despite even the curative potential of immunotherapeutic checkpoint blockade, in particular, casualties still abound and true clinical success stories are few and far between; the ever-present, if sometimes unmentioned, “elephant in the room”, is the acquisition of resistance, which, sooner or later, rears its ugly head to undermine treatment success and shorten survival. Emerging data suggests that epigenetic therapies are able to reprogram the aberrant tumor-associated epigenome and ‘tame the beast of resistance’, thereby prolonging survival. This article reviews the role of epigenetic dysregulation in NSCLC, explores PFS2 as a possible surrogate endpoint, briefly mentions possible biomarkers and highlights combinatorial treatment epigenetic strategies to “prime” tumors and reverse resistance.

Collaboration


Dive into the Neil Oronsky's collaboration.

Top Co-Authors

Avatar

Bryan Oronsky

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Arnold Oronsky

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Jan Scicinski

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Corey A. Carter

Walter Reed National Military Medical Center

View shared research outputs
Top Co-Authors

Avatar

Tony Reid

University of California

View shared research outputs
Top Co-Authors

Avatar

Scott Caroen

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Pedro Cabrales

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gary Fanger

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge