Network


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

Hotspot


Dive into the research topics where Amer Najjar is active.

Publication


Featured researches published by Amer Najjar.


Cancer Research | 2008

Targeting Src Family Kinases Inhibits Growth and Lymph Node Metastases of Prostate Cancer in an Orthotopic Nude Mouse Model

In Park Serk; Jing Zhang; Kacy A. Phillips; John C. Araujo; Amer Najjar; Andrei Volgin; Juri G. Gelovani; Sun Jin Kim; Zhengxin Wang; Gary E. Gallick

Aberrant expression and/or activity of members of the Src family of nonreceptor protein tyrosine kinases (SFK) are commonly observed in progressive stages of human tumors. In prostate cancer, two SFKs (Src and Lyn) have been specifically implicated in tumor growth and progression. However, there are no data in preclinical models demonstrating potential efficacy of Src inhibitors against prostate cancer growth and/or metastasis. In this study, we used the small molecule SFK/Abl kinase inhibitor dasatinib, currently in clinical trials for solid tumors, to examine in vitro and in vivo effects of inhibiting SFKs in prostate tumor cells. In vitro, dasatinib inhibits both Src and Lyn activity, resulting in decreased cellular proliferation, migration, and invasion. In orthotopic nude mouse models, dasatinib treatment effectively inhibits expression of activated SFKs, resulting in inhibition of both tumor growth and development of lymph node metastases in both androgen-sensitive and androgen-resistant tumors. In primary tumors, SFK inhibition leads to decreased cellular proliferation (determined by immunohistochemistry for proliferating cell nuclear antigen). In vitro, small interfering RNA (siRNA)-mediated inhibition of Lyn affects cellular proliferation; siRNA inhibition of Src affects primarily cellular migration. Therefore, we conclude that SFKs are promising therapeutic targets for treatment of human prostate cancer and that Src and Lyn activities affect different cellular functions required for prostate tumor growth and progression.


Cancer Research | 2015

Tuning Sensitivity of CAR to EGFR Density Limits Recognition of Normal Tissue While Maintaining Potent Antitumor Activity

Hillary G. Caruso; Lenka V. Hurton; Amer Najjar; David Rushworth; Sonny Ang; Simon Olivares; Tiejuan Mi; Kirsten Switzer; Harjeet Singh; Helen Huls; Dean A. Lee; Amy B. Heimberger; Richard E. Champlin; Laurence J.N. Cooper

Many tumors overexpress tumor-associated antigens relative to normal tissue, such as EGFR. This limits targeting by human T cells modified to express chimeric antigen receptors (CAR) due to potential for deleterious recognition of normal cells. We sought to generate CAR(+) T cells capable of distinguishing malignant from normal cells based on the disparate density of EGFR expression by generating two CARs from monoclonal antibodies that differ in affinity. T cells with low-affinity nimotuzumab-CAR selectively targeted cells overexpressing EGFR, but exhibited diminished effector function as the density of EGFR decreased. In contrast, the activation of T cells bearing high-affinity cetuximab-CAR was not affected by the density of EGFR. In summary, we describe the generation of CARs able to tune T-cell activity to the level of EGFR expression in which a CAR with reduced affinity enabled T cells to distinguish malignant from nonmalignant cells.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Molecular imaging of active mutant L858R EGF receptor (EGFR) kinase-expressing nonsmall cell lung carcinomas using PET/CT

Hsin Hsien Yeh; Kazuma Ogawa; Julius Balatoni; Asutosh Pal; Carlos Gonzalez-Lepera; Aleksandr Shavrin; Suren Soghomonyan; Leo G. Flores; Daniel Young; Andrei Volgin; Amer Najjar; Victor Krasnykh; William P. Tong; Mian M. Alauddin; Juri G. Gelovani

The importance of the EGF receptor (EGFR) signaling pathway in the development and progression of nonsmall cell lung carcinomas (NSCLC) is widely recognized. Gene sequencing studies revealed that a majority of tumors responding to EGFR kinase inhibitors harbor activating mutations in the EGFR kinase domain. This underscores the need for novel biomarkers and diagnostic imaging approaches to identify patients who may benefit from particular therapeutic agents and approaches with improved efficacy and safety profiles. To this goal, we developed 4-[(3-iodophenyl)amino]-7-{2-[2-{2-(2-[2-{2-([18F]fluoroethoxy)-ethoxy}-ethoxy]-ethoxy)-ethoxy}-ethoxy]-quinazoline-6-yl-acrylamide ([18F]F-PEG6-IPQA), a radiotracer with increased selectivity and irreversible binding to the active mutant L858R EGFR kinase. We show that PET with [18F]F-PEG6-IPQA in tumor-bearing mice discriminates H3255 NSCLC xenografts expressing L858R mutant EGFR from H441 and PC14 xenografts expressing EGFR or H1975 xenografts with L858R/T790M dual mutation in EGFR kinase domain, which confers resistance to EGFR inhibitors (i.e., gefitinib). The T790M mutation precludes the [18F]F-PEG6-IPQA from irreversible binding to EGFR. These results suggest that PET with [18F]F-PEG6-IPQA could be used for the selection of NSCLC patients for individualized therapy with small molecular inhibitors of EGFR kinase that are currently used in the clinic and have a similar structure (i.e., iressa, gefitinib, and erlotinib).


PLOS ONE | 2013

Antigen presenting cell-mediated expansion of human umbilical cord blood yields log-scale expansion of natural killer cells with anti-myeloma activity.

Nina Shah; Beatriz Martín-Antonio; Hong Yang; Stephanie Ku; Dean A. Lee; Laurence J.N. Cooper; William K. Decker; Sufang Li; Simon N. Robinson; Takuya Sekine; Simrit Parmar; John G. Gribben; Michael Wang; Katy Rezvani; Eric Yvon; Amer Najjar; Jared K. Burks; Indreshpal Kaur; Richard E. Champlin; Catherine M. Bollard; Elizabeth J. Shpall

Natural killer (NK) cells are important mediators of anti-tumor immunity and are active against several hematologic malignancies, including multiple myeloma (MM). Umbilical cord blood (CB) is a promising source of allogeneic NK cells but large scale ex vivo expansion is required for generation of clinically relevant CB-derived NK (CB-NK) cell doses. Here we describe a novel strategy for expanding NK cells from cryopreserved CB units using artificial antigen presenting feeder cells (aAPC) in a gas permeable culture system. After 14 days, mean fold expansion of CB-NK cells was 1848-fold from fresh and 2389-fold from cryopreserved CB with >95% purity for NK cells (CD56+/CD3−) and less than 1% CD3+ cells. Though surface expression of some cytotoxicity receptors was decreased, aAPC-expanded CB-NK cells exhibited a phenotype similar to CB-NK cells expanded with IL-2 alone with respect to various inhibitory receptors, NKG2C and CD94 and maintained strong expression of transcription factors Eomesodermin and T-bet. Furthermore, CB-NK cells formed functional immune synapses with and demonstrated cytotoxicity against various MM targets. Finally, aAPC-expanded CB-NK cells showed significant in vivo activity against MM in a xenogenic mouse model. Our findings introduce a clinically applicable strategy for the generation of highly functional CB-NK cells which can be used to eradicate MM.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Bioengineering T cells to target carbohydrate to treat opportunistic fungal infection

Pappanaicken R. Kumaresan; Pallavi R. Manuri; Nathaniel D. Albert; Sourindra Maiti; Harjeet Singh; Tiejuan Mi; Jason Roszik; Brian Rabinovich; Simon Olivares; Janani Krishnamurthy; Ling Zhang; Amer Najjar; M. Helen Huls; Dean A. Lee; Richard E. Champlin; Dimitrios P. Kontoyiannis; Laurence J.N. Cooper

Significance Patients with compromised T-cell function are at risk for opportunistic fungal infections. We have developed a novel approach to restore immunity by using a fungal pattern-recognition receptor Dectin-1 to redirect T-cell specificity to carbohydrate antigen in the fungal cell wall. We did so by genetically modifying T cells using the nonviral Sleeping Beauty gene-transfer system to enforce expression of a chimeric antigen receptor (CAR) that recapitulates the specificity of Dectin-1 (D-CAR). The D-CAR+ T cells can be electroporated and propagated on artificial activating and propagating cells in a manner suitable for human application, enabling this immunology to be translated into immunotherapy. This approach has implications for genetically modifying T cells to express CARs with specificity for carbohydrate and thus broadening their application in the investigational treatment of pathogens and malignancies. Clinical-grade T cells are genetically modified ex vivo to express chimeric antigen receptors (CARs) to redirect their specificity to target tumor-associated antigens in vivo. We now have developed this molecular strategy to render cytotoxic T cells specific for fungi. We adapted the pattern-recognition receptor Dectin-1 to activate T cells via chimeric CD28 and CD3-ζ (designated “D-CAR”) upon binding with carbohydrate in the cell wall of Aspergillus germlings. T cells genetically modified with the Sleeping Beauty system to express D-CAR stably were propagated selectively on artificial activating and propagating cells using an approach similar to that approved by the Food and Drug Administration for manufacturing CD19-specific CAR+ T cells for clinical trials. The D-CAR+ T cells exhibited specificity for β-glucan which led to damage and inhibition of hyphal growth of Aspergillus in vitro and in vivo. Treatment of D-CAR+ T cells with steroids did not compromise antifungal activity significantly. These data support the targeting of carbohydrate antigens by CAR+ T cells and provide a clinically appealing strategy to enhance immunity for opportunistic fungal infections using T-cell gene therapy.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Tethered IL-15 augments antitumor activity and promotes a stem-cell memory subset in tumor-specific T cells

Lenka V. Hurton; Harjeet Singh; Amer Najjar; Kirsten Switzer; Tiejuan Mi; Sourindra Maiti; Simon Olivares; Brian Rabinovich; Helen Huls; Marie Andrée Forget; Vrushali Datar; Partow Kebriaei; Dean A. Lee; Richard E. Champlin; Laurence J.N. Cooper

Significance We describe an approach based on cytokine therapeutics to enhance the persistence and effectiveness of T-cell–based immunotherapies using chimeric antigen receptors (CARs). This strategy is effective without the use of high-dose exogenous cytokines that are typically associated with toxicities. Moreover, we report that the persistence of the least differentiated memory T cell, the T-memory stem cell, was promoted by signaling induced by a membrane-bound chimeric IL-15 cytokine-fusion molecule. These findings may contribute to improving the safety and therapeutic efficacy of CAR-based immunotherapies of patients with advanced cancer. Adoptive immunotherapy retargeting T cells to CD19 via a chimeric antigen receptor (CAR) is an investigational treatment capable of inducing complete tumor regression of B-cell malignancies when there is sustained survival of infused cells. T-memory stem cells (TSCM) retain superior potential for long-lived persistence, but challenges exist in manufacturing this T-cell subset because they are rare among circulating lymphocytes. We report a clinically relevant approach to generating CAR+ T cells with preserved TSCM potential using the Sleeping Beauty platform. Because IL-15 is fundamental to T-cell memory, we incorporated its costimulatory properties by coexpressing CAR with a membrane-bound chimeric IL-15 (mbIL15). The mbIL15-CAR T cells signaled through signal transducer and activator of transcription 5 to yield improved T-cell persistence independent of CAR signaling, without apparent autonomous growth or transformation, and achieved potent rejection of CD19+ leukemia. Long-lived T cells were CD45ROnegCCR7+CD95+, phenotypically most similar to TSCM, and possessed a memory-like transcriptional profile. Overall, these results demonstrate that CAR+ T cells can develop long-term persistence with a memory stem-cell phenotype sustained by signaling through mbIL15. This observation warrants evaluation in clinical trials.


The Journal of Nuclear Medicine | 2009

Molecular–Genetic PET Imaging Using an HSV1-tk Mutant Reporter Gene with Enhanced Specificity to Acycloguanosine Nucleoside Analogs

Amer Najjar; Ryuichi Nishii; David Maxwell; Andrei Volgin; Uday Mukhopadhyay; William G. Bornmann; William P. Tong; Mian M. Alauddin; Juri G. Gelovani

Imaging 2 different molecular–genetic events in a single subject by PET is essential in a variety of in vivo applications. Using herpes simplex virus-1 thymidine kinase (HSV1-tk) mutants with narrower substrate specificities in combination with wild-type HSV1-tk (wtHSV1-tk) would enable differential imaging with corresponding radiotracers, namely 2′-deoxy-2′-18F-fluoro-5-ethyl-1-β-d-arabinofuranosyl-uracil (18F-FEAU) and the acycloguanosine derivative 9-(4-18F-fluoro-3-[hydroxymethyl]butyl)guanine (18F-FHBG). In this study, we evaluated wtHSV1-tk and the A168H mutant, which has been reported to exhibit enhanced acycloguanosine substrate catalytic activity and diminished pyrimidine phosphorylating activity, as PET reporter genes. Methods: Computational analysis was performed to assess the binding mode of FHBG and FEAU to wtHSV1-tk and the A168H variant. U87 cells were stably transduced with wtHSV1-tk or HSV1-tk(A168H) fused with green fluorescent protein and sorted to obtain equivalent transgene expression. In vitro uptake studies were performed to determine rates of substrate accumulation and retention. Nude mice bearing tumors expressing HSV1-tk variants were subsequently imaged using 18F-FHBG and 18F-FEAU. Results: Docking results indicate that binding of FHBG to the A168H variant is unaffected whereas the binding of FEAU is hindered because of a steric clash with the bulkier mutant residues. U87 cells expressing HSV1-tk(A168H) accumulated 18F-FHBG in in vitro uptake studies at a 3-fold higher rate than did cells expressing wtHSV1-tk without any detectable accumulation of 3H-FEAU. Furthermore, HSV1-tk(A168H) demonstrated no thymidine phosphorylation activity. In contrast, U87 cells expressing wtHSV1-tk preferentially accumulated 3H-FEAU at an 18-fold higher rate than they did 18F-FHBG. Tumors expressing wtHSV1-tk or HSV1-tk(A168H) were distinctly imaged with 18F-FEAU or 18F-FHBG, respectively. Hence, tumors expressing HSV1-tk(A168H) accumulated 8.4-fold more 18F-FHBG than did tumors expressing wtHSV1-tk. In addition, wtHSV1-tk tumors, compared with HSV1-tk(A168H)–expressing tumors (which retained baseline levels of the radiotracer), preferentially accumulated 18F-FEAU. Conclusion: The FEAU and FHBG substrate discrimination capacity of the wtHSV1-tk and HSV1-tk(A168H) reporter enzymes was validated in vivo by PET of mice with tumor xenografts established from U87 cells expressing these different reporters. Thus, HSV1-tk(A168H) may potentially be used as a second reporter gene in combination with wtHSV1-tk to achieve differential PET.


Cytotherapy | 2014

Fucosylation with fucosyltransferase VI or fucosyltransferase VII improves cord blood engraftment

Simon N. Robinson; Michael W. Thomas; Paul J. Simmons; Junjun Lu; Hong Yang; Simrit Parmar; Xiaoying Liu; Nina Shah; Beatriz Martín-Antonio; Catherine M. Bollard; Gianpietro Dotti; Barbara Savoldo; Laurence J.N. Cooper; Amer Najjar; Katayoun Rezvani; Indreshpaul Kaur; Ian McNiece; Richard E. Champlin; Leonard Miller; Patrick A. Zweidler-McKay; Elizabeth J. Shpall

BACKGROUND AIMS Advantages associated with the use of cord blood (CB) transplantation include the availability of cryopreserved units, ethnic diversity and lower incidence of graft-versus-host disease compared with bone marrow or mobilized peripheral blood. However, poor engraftment remains a major obstacle. We and others have found that ex vivo fucosylation can enhance engraftment in murine models, and now ex vivo treatment of CB with fucosyltransferase (FT) VI before transplantation is under clinical evaluation (NCT01471067). However, FTVII appears to be more relevant to hematopoietic cells and may alter acceptor substrate diversity. The present study compared the ability of FTVI and FTVII to improve the rapidity, magnitude, multi-lineage and multi-tissue engraftment of human CB hematopoietic stem and progenitor cells (HSPCs) in vivo. METHODS CD34-selected CB HSPCs were treated with recombinant FTVI, FTVII or mock control and then injected into immunodeficient mice and monitored for multi-lineage and multi-tissue engraftment. RESULTS Both FTVI and FTVII fucosylated CB CD34⁺ cells in vitro, and both led to enhanced rates and magnitudes of engraftment compared with untreated CB CD34⁺ cells in vivo. Engraftment after treatment with either FT was robust at multiple time points and in multiple tissues with similar multi-lineage potential. In contrast, only FTVII was able to fucosylate T and B lymphocytes. CONCLUSIONS Although FTVI and FTVII were found to be similarly able to fucosylate and enhance the engraftment of CB CD34⁺ cells, differences in their ability to fucosylate lymphocytes may modulate graft-versus-tumor or graft-versus-host effects and may allow further optimization of CB transplantation.


Cytotherapy | 2014

Third-party umbilical cord blood-derived regulatory T cells prevent xenogenic graft-versus-host disease.

Simrit Parmar; Xiaoying Liu; Shawndeep Tung; Simon N. Robinson; Gabriel Rodriguez; Laurence J.N. Cooper; Hui Yang; Nina Shah; Hong Yang; Marina Konopleva; Jeffery J Molldrem; Guillermo Garcia-Manero; Amer Najjar; Eric Yvon; Ian McNiece; Katy Rezvani; Barbara Savoldo; Catherine M. Bollard; Elizabeth J. Shpall

BACKGROUND AIMS Naturally occurring regulatory T cells (Treg) are emerging as a promising approach for prevention of graft-versus-host disease (GvHD), which remains an obstacle to the successful outcome of allogeneic hematopoietic stem cell transplantation. However, Treg only constitute 1-5% of total nucleated cells in cord blood (CB) (<3 × 10⁶ cells), and therefore novel methods of Treg expansion to generate clinically relevant numbers are needed. METHODS Several methodologies are currently being used for ex vivo Treg expansion. We report a new approach to expand Treg from CB and demonstrate their efficacy in vitro by blunting allogeneic mixed lymphocyte reactions and in vivo by preventing GvHD through the use of a xenogenic GvHD mouse model. RESULTS With the use of magnetic cell sorting, naturally occurring Treg were isolated from CB by the positive selection of CD25⁺ cells. These were expanded to clinically relevant numbers by use of CD3/28 co-expressing Dynabeads and interleukin (IL)-2. Ex vivo-expanded Treg were CD4⁺25⁺ FOXP3⁺127(lo) and expressed a polyclonal T-cell receptor, Vβ repertoire. When compared with conventional T-lymphocytes (CD4⁺25⁻ cells), Treg consistently showed demethylation of the FOXP3 TSDR promoter region and suppression of allogeneic proliferation responses in vitro. CONCLUSIONS In our NOD-SCID IL-2Rγ(null) xenogeneic model of GvHD, prophylactic injection of third-party, CB-derived, ex vivo-expanded Treg led to the prevention of GvHD that translated into improved GvHD score, decreased circulating inflammatory cytokines and significantly superior overall survival. This model of xenogenic GvHD can be used to study the mechanism of action of CB Treg as well as other therapeutic interventions.


Circulation-cardiovascular Imaging | 2012

Molecular imaging of mesenchymal stem cell: mechanistic insight into cardiac repair after experimental myocardial infarction.

Jingxiong Wang; Amer Najjar; Sui Zhang; Brian Rabinovich; James T. Willerson; Juri G. Gelovani; Edward T.H. Yeh

Background— Mesenchymal stem cells (MSCs) can differentiate into endothelial cells in vivo. However, it is unknown if the differentiated MSCs persist in vivo and if this potential persistence contributes to functional improvement after experimental myocardial infarction. Methods and Results— We generated a lentivector encoding 2 distinct reporter genes, one driven by a constitutive murine stem cell virus promoter and the other driven by an endothelial-specific Tie-2 promoter. The endothelial specificity of the lentivector was validated by its expression in endothelial cells but not in human MSCs (hMSCs). The lentivirus-transduced hMSCs were injected into peri-infarct areas of the hearts of severe combined immune-deficient mice. Persistence of injected cells was tracked by bioluminescence imaging (BLI) and verified by immunohistochemical staining. The BLI signal from the endothelial-specific reporter revealed that hMSCs differentiated into endothelial cells 48 hours after injection. However, both the constitutive and endothelial-specific BLI signals disappeared by day 50. Nonetheless, the improvement in left ventricle ejection fraction with hMSC therapy persisted for up to 6 months. Immunohistochemical staining showed that hMSC-derived endothelial cells integrated into endogenous CD31+ vessels. Furthermore, hMSC-transplanted hearts had more CD31+ vessels and a lesser degree of cardiac fibrosis compared with the controls at 6 months. Conclusions— hMSCs differentiated into endothelial cells and integrated into blood vessels after experimental myocardial infarction. The differentiated hMSCs only lasted for up to 50 days in vivo, but improvement in cardiac function persisted for up to 6 months. Increased angiogenesis and decreased fibrosis were associated with cardiac functional improvement after hMSC transplantation.Background— Mesenchymal stem cells (MSCs) can differentiate into endothelial cells in vivo. However, it is unknown if the differentiated MSCs persist in vivo and if this potential persistence contributes to functional improvement after experimental myocardial infarction. Methods and Results— We generated a lentivector encoding 2 distinct reporter genes, one driven by a constitutive murine stem cell virus promoter and the other driven by an endothelial-specific Tie-2 promoter. The endothelial specificity of the lentivector was validated by its expression in endothelial cells but not in human MSCs (hMSCs). The lentivirus-transduced hMSCs were injected into peri-infarct areas of the hearts of severe combined immune-deficient mice. Persistence of injected cells was tracked by bioluminescence imaging (BLI) and verified by immunohistochemical staining. The BLI signal from the endothelial-specific reporter revealed that hMSCs differentiated into endothelial cells 48 hours after injection. However, both the constitutive and endothelial-specific BLI signals disappeared by day 50. Nonetheless, the improvement in left ventricle ejection fraction with hMSC therapy persisted for up to 6 months. Immunohistochemical staining showed that hMSC-derived endothelial cells integrated into endogenous CD31+ vessels. Furthermore, hMSC-transplanted hearts had more CD31+ vessels and a lesser degree of cardiac fibrosis compared with the controls at 6 months. Conclusions— hMSCs differentiated into endothelial cells and integrated into blood vessels after experimental myocardial infarction. The differentiated hMSCs only lasted for up to 50 days in vivo, but improvement in cardiac function persisted for up to 6 months. Increased angiogenesis and decreased fibrosis were associated with cardiac functional improvement after hMSC transplantation.

Collaboration


Dive into the Amer Najjar's collaboration.

Top Co-Authors

Avatar

Juri G. Gelovani

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Laurence J.N. Cooper

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Elizabeth J. Shpall

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Richard E. Champlin

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Brian Rabinovich

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Harjeet Singh

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Simon N. Robinson

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Simon Olivares

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Tiejuan Mi

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hong Yang

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge