Network


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

Hotspot


Dive into the research topics where Rasha Abu Eid is active.

Publication


Featured researches published by Rasha Abu Eid.


Journal for ImmunoTherapy of Cancer | 2013

Anti-PD-1 antibody significantly increases therapeutic efficacy of Listeria monocytogenes (Lm)- LLO immunotherapy

Mikayel Mkrtichyan; Namju Chong; Rasha Abu Eid; Anu Wallecha; Reshma Singh; John Rothman; Samir N. Khleif

BackgroundOne of the significant tumor immune escape mechanisms and substantial barrier for successful immunotherapy is tumor-mediated inhibition of immune response through cell-to-cell or receptor/ligand interactions. Programmed death receptor-1 (PD-1) interaction with its ligands, PD-L1 and PD-L2, is one of the important strategies that many tumors employ to escape immune surveillance. Upon PD-Ls binding to PD-1, T cell receptor (TCR) signaling is dampened, causing inhibition of proliferation, decreased cytokine production, anergy and/or apoptosis. Thus PD-Ls expression by tumor cells serves as a protective mechanism, leading to suppression of tumor-infiltrating lymphocytes in the tumor microenvironment. Lm-LLO immunotherapies have been shown to be therapeutically effective due to their ability to induce potent antigen-specific immune responses. However, it has been demonstrated that infection with Lm leads to up-regulation of PD-L1 on mouse immune cells that can inhibit effector T cells through PD-1/PD-L1 pathway.MethodsTherapeutic and immune efficacy of Listeria-based vaccine (Lm-LLO-E7) in combination with anti-PD-1 antibody was tested in E7 antigen expressing TC-1 mouse tumor model. Tumor growth, survival, as well as peripheral and tumor-infiltrating immune cell profiles after immunotherapy were assessed.ResultsHere we demonstrate that the combination of an Lm-LLO immunotherapy with anti-PD-1 antibody that blocks PD-1/PD-L1 interaction, significantly improves immune and therapeutic efficacy of treatment in TC-1 mouse tumor model. Importantly, we show that in addition to significant reduction of regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSC) in both spleen and tumor microenvironment that are mediated solely by the Lm-LLO immunotherapy, the addition of anti-PD-1 antibody to the treatment results in significant increase of antigen-specific immune responses in periphery and CD8 T cell infiltration into the tumor. As a result, this combinational treatment leads to significant inhibition of tumor growth and prolonged survival/complete regression of tumors in treated animals.We also demonstrate that in vitro infection with Lm results in significant upregulation of surface PD-L1 expression on human monocyte-derived dendritic cells suggesting the translational capacity of this finding.ConclusionsOur findings demonstrate that combination of Lm-LLO-based vaccine with blocking of PD-1/PD-L1 interaction is a feasible approach with clinical translation potential that can lead to overall enhancement of the efficacy of anti-tumor immunotherapy.


Cancer immunology research | 2016

Mutant KRAS Conversion of Conventional T Cells into Regulatory T Cells

Stephanie Zdanov; Magis Mandapathil; Rasha Abu Eid; Saudat Adamson-Fadeyi; Willie Wilson; Jiahua Qian; Andrea Carnie; Nadya Tarasova; Mikayel Mkrtichyan; Jay A. Berzofsky; Theresa L. Whiteside; Samir N. Khleif

Mutant KRAS uses cell-intrinsic mechanisms to promote aggressive tumor growth. Now, mutated KRAS has been found to use cell-extrinsic mechanisms early in tumorigenesis that induce Tregs and immune evasion through activating the MEK–ERK–AP1 pathway, producing IL10 and TGFβ Constitutive activation of the KRAS oncogene in human malignancies is associated with aggressive tumor growth and poor prognosis. Similar to other oncogenes, KRAS acts in a cell-intrinsic manner to affect tumor growth or survival. However, we describe here a different, cell-extrinsic mechanism through which mutant KRAS contributes to tumor development. Tumor cells carrying mutated KRAS induced highly suppressive T cells, and silencing KRAS reversed this effect. Overexpression of the mutant KRASG12V gene in wild-type KRAS tumor cells led to regulatory T-cell (Treg) induction. We also demonstrate that mutant KRAS induces the secretion of IL10 and transforming growth factor-β1 (both required for Treg induction) by tumor cells through the activation of the MEK–ERK–AP1 pathway. Finally, we report that inhibition of KRAS reduces the infiltration of Tregs in KRAS-driven lung tumorigenesis even before tumor formation. This cell-extrinsic mechanism allows tumor cells harboring a mutant KRAS oncogene to escape immune recognition. Thus, an oncogene can promote tumor progression independent of its transforming activity by increasing the number and function of Tregs. This has a significant clinical potential, in which targeting KRAS and its downstream signaling pathways could be used as powerful immune modulators in cancer immunotherapy. Cancer Immunol Res; 4(4); 354–65. ©2016 AACR.


OncoImmunology | 2015

Akt1 and -2 inhibition diminishes terminal differentiation and enhances central memory CD8+ T-cell proliferation and survival

Rasha Abu Eid; Kevin Friedman; Mikayel Mkrtichyan; Andrea Walens; William King; John Janik; Samir N. Khleif

The CD8+ T-cell response comprises terminally differentiated effector cells and antigen-experienced memory T cells. The latter encompass central (TCM) and effector (TEM) memory cells. TCM cells are superior in their protection against viral and bacterial challenges and mediation of antitumor immunity due to their higher proliferative ability upon antigen re-encounter. Defining a mechanism to enhance TCM cells and delay terminal differentiation of CD8+ T cells is crucial for cancer immune therapy, as it can promote a better tumor immune response. The differentiation of CD8+ memory T cells is thought to be coordinated by the phosphoinositide 3-kinase (PI3K)/Akt pathway. We, therefore, investigated the role of Akt isoforms in the differentiation and proliferation of memory CD8+ T cells. We found that Akt1 and Akt2, but not Akt3, drive the terminal differentiation of CD8+ T cells, and their inhibition enhances the therapeutically superior TCM phenotype. Furthermore, the inhibition of Akt1 and Akt2, but not Akt 3, delays CD8+ T-cell exhaustion and preserves naïve and TCM CD8+ T cells, thus enhancing their proliferative ability and survival and prolonging their cytokine and Granzyme B production ability. Here, we define a mechanism in which proliferative potential, function, and survival of CD8+ T cells are enhanced by maintaining a reservoir of TCM and naïve cells using only Akt1 and Akt2 inhibition. Therefore, our findings strongly suggest the utility of using Akt1 and Akt2 inhibitors to modulate CD8+ T cells, both for adoptive cell transfer and vaccine-based cancer immune therapies.


Cancer immunology research | 2016

Old-School Chemotherapy in Immunotherapeutic Combination in Cancer, A Low-cost Drug Repurposed

Rasha Abu Eid; Ghazaleh Shoja E. Razavi; Mikayel Mkrtichyan; John Janik; Samir N. Khleif

Cancer immunotherapy has proven to be a potent treatment modality. Although often successful in generating antitumor immune responses, cancer immunotherapy is frequently hindered by tumor immune-escape mechanisms. Among immunosuppressive strategies within the tumor microenvironment, suppressive immune regulatory cells play a key role in promoting tumor progression through inhibiting the effector arm of the immune response. Targeting these suppressive cells can greatly enhance antitumor immune therapies, hence augmenting a highly effective therapeutic antitumor response. Several approaches are being tested to enhance the effector arm of the immune system while simultaneously inhibiting the suppressor arm. Some of these approaches are none other than traditional drugs repurposed as immune modulators. Cyclophosphamide, an old-school chemotherapeutic agent used across a wide range of malignancies, was found to be a potent immune modulator that targets suppressive regulatory immune cells within the tumor microenvironment while enhancing effector cells. Preclinical and clinical findings have confirmed the ability of low doses of cyclophosphamide to selectively deplete regulatory T cells while enhancing effector and memory cytotoxic T cells within the tumor microenvironment. These immune effects translate to suppressed tumor growth and enhanced survival, evidence of antitumor therapeutic efficacy. This article discusses the reincarnation of cyclophosphamide as an immune modulator that augments novel immunotherapeutic approaches. Cancer Immunol Res; 4(5); 377–82. ©2016 AACR.


Journal for ImmunoTherapy of Cancer | 2014

Functional redundancy of PI3K isoforms in conventional T cells provides a selective Treg-targeting strategy through inhibition of PI3K-delta isoform

Shamim Ahmad; Mikayel Mkrtichyan; Rasha Abu Eid; Rajeev Shrimali; Atbin Doroodchi; Samir N. Khleif

Increased regulatory T cell (Treg) numbers within tumors and circulation of cancer patients, observed in early studies, implied their involvement in pathogenesis and disease progression. Also, Treg increase in cancer patients have been associated with reduced survival and inhibition of anti-tumor immune responses. Therefore, decreasing the numbers and/or function of Tregs is needed to facilitate better outcomes for cancer patients. The phosphoinositide 3-kinase (PI3K-Akt) pathway plays important roles in cell growth, survival, and proliferation of T cells. However, little is known about the role of different isoforms of PI3K in different T subsets activation. Here, we explore the role of PI3K isoforms in Tregs and conventional T cell activation with the intention of identifying potential differences to selectively inhibit Tregs.


Journal for ImmunoTherapy of Cancer | 2014

Immune combinational therapy targeting OX40 and IDO synergistically enhances efficacy of a cancer vaccine

Zuzana Berrong; Shamim Ahmad; Rasha Abu Eid; Abdeljabar El Andaloussi; Tanusree Sen; Ross Stewart; Scott A. Hammond; Rajeev Shrimali; Mikayel Mkrtichyan; Samir N. Khleif

Enhancing an effector immune response by cancer vaccines has not been clinically successful so far. Although the necessary immune response may be elicited using vaccine-based therapies, this has not been sufficient for a positive clinical outcome; since most cancers can escape immune surveillance via multiple immune suppressive mechanisms induced in the tumor environment. A relatively recent strategy to increase the therapeutic efficacy of tumor vaccination is to combine it with alternative immunotherapeutic approaches. Here we tested if simultaneous targeting of the effector arm of the immune system with an agonist anti-OX40 mAb together with targeting of suppressive mechanisms in the tumor, via inhibition of indoleamine-(2,3)-dioxygenase (IDO), could enhance the anti-tumor activity of vaccine in mouse models of cancer. The OX40 molecule is a co-stimulatory receptor expressed on T-cells that can lead to proliferation and enhancement of T-cell effector function when triggered with an agonistic antibody. However, the tumor secretes a number of suppressive signals as a protective mechanism against its destruction, one of which is IDO. We demonstrate that treatment of TC-1 tumor bearing mice with an anti-OX40 agonist antibody in combination with vaccine leads to the enhancement of antigen-specific T cell responses. The dose of 1 mg/kg anti-OX40 antibody stimulated the effector arm of T cells, which ultimately led to a significant increase in the CD8+/regulatory T-cell (Treg) ratio within tumors. Further, this combination of vaccine and anti-OX40 antibody treatment led to a significant inhibition of tumor growth and to a prolonged survival of tumor bearing mice compared to control, and resulted in complete tumor regression in 20% of treated mice. We found, however, that this anti-tumor activity could be further enhanced through the combination of vaccine, anti-OX40 and 1-methyltryptophan (1-MT), an IDO activity inhibitor. IDO has been shown to be secreted by tumor cells, suppressive dendritic cells and macrophages in tumor environment, and is known to be responsible for suppressing effector cells and inducing Tregs. Our data demonstrate that vaccine/anti-OX40 antibody/1-MT combination leads to a more profound inhibition of tumor growth in mice and complete regression of established tumors in 60% of treated mice. In conclusion, we provide evidence that simultaneous targeting of the effector arm of the immune system, via anti-OX40 antibody, and suppressive mechanisms within the tumor, via 1-MT, in combination with cancer vaccine has a synergistic effect for tumor eradication and is a promising strategy that could potentially enhance the overall efficacy of cancer treatment in patients.


Jordan Medical Journal | 2015

Students’ Perceptions of Dental Anatomy Course at The University of Jordan

Yara Oweis; Nadia sami Eriefej; Rasha Abu Eid

Purpose: The aim of this study was to assess the students’ perception of the Dental Anatomy practical module course teaching modalities to evaluate their usefulness and the need for future changes or improvement of the course with the goal of developing the course to meet the needs of the students. Methods: Students’ opinions were sought upon completion of the Tooth Morphology sessions using a questionnaire that probed into their views about different aspects of the course. Results: The replies to the questionnaire suggest that students were generally satisfied with the course and instructors; they had positive attitudes towards the carving excercise though they had some comments about the quality of material used for tooth carving and the demonstration given in addition to the overall grading Conclusion: It was concluded that new methods for teaching dental anatomy need to be implemented keeping, at the same time, the old techniques which are important for the development of manual skills. Innovative changes have already been introduced to the course that is hoped to help meet the students’ learning needs.


Journal for ImmunoTherapy of Cancer | 2014

AKT inhibition mitigates terminal differentiation and preserves central memory phenotype of CD8 T cells

Rasha Abu Eid; Kevin Friedman; Mikayel Mkrtichyan; Samir N. Khleif

CD8 T cell response comprises effector and memory T cells. Effector CD8 T cells become terminally differentiated and are eliminated by apoptosis. Memory CD8 T cells encompass central (TCM) and effector memory T cells (TEM). TCM display a higher proliferative ability, express a higher level of CD62L and are superior in their protection against viral and bacterial challenges and mediation of anti-tumor immunity when compared to TEM. The differentiation of CD8 T cell is thought to be coordinated by the PI3K/Akt pathway.


Journal of Translational Medicine | 2014

Pre-immature dendritic cells (PIDC) pulsed with HPV16 E6 or E7 peptide are capable of eliciting specific immune response in patients with advanced cervical cancer.

Osama E. Rahma; V. E. Herrin; Rami A Ibrahim; Anton Toubaji; Sarah Bernstein; Omar Dakheel; Seth M. Steinberg; Rasha Abu Eid; Mikayel Mkrtichyan; Jay A. Berzofsky; Samir N. Khleif


Archive | 2012

Association between Graduate Entry Dental Students’ Performance in Objective Structured Practical Examination and their First Degrees

Jaya Jayasinghe; Rasha Abu Eid

Collaboration


Dive into the Rasha Abu Eid's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Samir N. Khleif

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Jay A. Berzofsky

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

John Janik

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Kevin Friedman

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Rajeev Shrimali

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Shamim Ahmad

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge