Christina Pham
University of Florida
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Christina Pham.
Clinical Cancer Research | 2016
Christina Pham; Catherine Flores; Changlin Yang; Elaine M. Pinheiro; Jennifer H. Yearley; Elias Sayour; Yanxin Pei; Colin Moore; Roger E. McLendon; Jianping Huang; John H. Sampson; Robert J. Wechsler-Reya; Duane Mitchell
Purpose: Despite significant strides in the identification and characterization of potential therapeutic targets for medulloblastoma, the role of the immune system and its interplay with the tumor microenvironment within these tumors are poorly understood. To address this, we adapted two syngeneic animal models of human Sonic Hedgehog (SHH)-driven and group 3 medulloblastoma for preclinical evaluation in immunocompetent C57BL/6 mice. Experimental Design and Results: Multicolor flow cytometric analyses were used to phenotype and characterize immune infiltrating cells within established cerebellar tumors. We observed significantly higher percentages of dendritic cells, infiltrating lymphocytes, myeloid-derived suppressor cells, and tumor-associated macrophages in murine SHH model tumors compared with group 3 tumors. However, murine group 3 tumors had higher percentages of CD8+ PD-1+ T cells within the CD3 population. PD-1 blockade conferred superior antitumor efficacy in animals bearing intracranial group 3 tumors compared with SHH group tumors, indicating that immunologic differences within the tumor microenvironment can be leveraged as potential targets to mediate antitumor efficacy. Further analysis of anti-PD-1 monoclonal antibody localization revealed binding to PD-1+ peripheral T cells, but not tumor infiltrating lymphocytes within the brain tumor microenvironment. Peripheral PD-1 blockade additionally resulted in a marked increase in CD3+ T cells within the tumor microenvironment. Conclusions: This is the first immunologic characterization of preclinical models of molecular subtypes of medulloblastoma and demonstration that response to immune checkpoint blockade differs across subtype classification. Our findings also suggest that effective anti-PD-1 blockade does not require that systemically administered antibodies penetrate the brain tumor microenvironment. Clin Cancer Res; 22(3); 582–95. ©2015 AACR.
OncoImmunology | 2015
Catherine Flores; Christina Pham; David Snyder; Shicheng Yang; Luis Sanchez-Perez; Elias Sayour; Xiuyu Cui; Hanna Kemeny; Henry S. Friedman; Darell D. Bigner; John H. Sampson; Duane Mitchell
Adoptive cellular therapy (ACT) after lymphodepletive conditioning can induce dramatic clinical responses, but this approach has been largely limited to melanoma due to a lack of reliable methods for expanding tumor-specific lymphocytes from the majority of other solid cancers. We have employed tumor RNA-pulsed dendritic cells (DCs) to reliably expand CD4+ and CD8+ tumor-reactive T lymphocytes for curative ACT in a highly-invasive, chemotherapy- and radiation-resistant malignant glioma model. Curative treatment of established intracranial tumors involved a synergistic interaction between myeloablative (MA) conditioning, adoptively transferred tumor-specific T cells, and tumor RNA-pulsed DC vaccines. Hematopoietic stem cells (HSCs), administered for salvage from MA conditioning, rapidly migrated to areas of intracranial tumor growth and facilitated the recruitment of tumor-specific lymphocytes through HSC-elaborated chemokines and enhanced immunologic rejection of intracranial tumors during ACT. Furthermore, HSC transplant under non-myeloablative (NMA) conditions also enhanced immunologic tumor rejection, indicating a novel role for the use of HSCs in the immunologic treatment of malignant gliomas and possibly other solid tumors.
Immunotherapy | 2012
Christina Pham; Duane Mitchell
Many attempts to use genetically modified T cells to halt tumor progression have been met with disappointment and significant challenges in the successful application within human patients. Porter et al., however, describe the use of genetically modified lymphocytes bearing a chimeric antigen receptor that bypasses many of the common limitations of adoptive lymphocyte therapy. Through incorporation of a costimulatory domain within the chimeric antigen receptor, the investigators engineered lymphocytes with significantly higher tumor rejection activity and demonstrated significant expansion and prolonged survival after in vivo transfer to a single patient who showed a complete regression of refractory chronic lymphoid leukemia. This recent success in using genetically modified T cells to kill chronic lymphoid leukemia tumor cells is an encouraging advancement in the development of specific and targeted immune-based therapies against cancer.
OncoImmunology | 2017
Elias Sayour; Gabriel De Leon; Christina Pham; Adam Grippin; Hanna Kemeny; Joshua Chua; Jianping Huang; John H. Sampson; Luis Sanchez-Perez; Catherine Flores; Duane Mitchell
ABSTRACT While RNA-pulsed dendritic cell (DC) vaccines have shown promise, the advancement of cellular therapeutics is fraught with developmental challenges. To circumvent the challenges of cellular immunotherapeutics, we developed clinically translatable nanoliposomes that can be combined with tumor-derived RNA to generate personalized tumor RNA-nanoparticles (NPs) with considerable scale-up capacity. RNA-NPs bypass MHC restriction, are amenable to central distribution, and can provide near immediate immune induction. We screened commercially available nanoliposomal preparations and identified the cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) as an efficient mRNA courier to antigen-presenting cells (APCs). When administered intravenously, RNA-NPs mediate systemic activation of APCs in reticuloendothelial organs such as the spleen, liver, and bone marrow. RNA-NPs increase percent expression of MHC class I/II, B7 co-stimulatory molecules, and maturation markers on APCs (all vital for T-cell activation). RNA-NPs also increase activation markers on tumor APCs and elicit potent expansion of antigen-specific T-cells superior to peptide vaccines formulated in complete Freunds adjuvant. We demonstrate that both model antigen-encoding and physiologically-relevant tumor-derived RNA-NPs expand potent antitumor T-cell immunity. RNA-NPs were shown to induce antitumor efficacy in a vaccine model and functioned as a suitable alternative to DCs in a stringent cellular immunotherapy model for a radiation/temozolomide resistant invasive murine high-grade glioma. Although cancer vaccines have suffered from weak immunogenicity, we have advanced a RNA-NP formulation that systemically activates host APCs precipitating activated T-cell frequencies necessary to engender antitumor efficacy. RNA-NPs can thus be harnessed as a more feasible and effective immunotherapy to re-program host-immunity.
OncoImmunology | 2016
Christina Pham; Duane Mitchell
ABSTRACT Medulloblastoma (MB) is the most common pediatric brain tumor with few reports of successful immunologic targeting. We have recently demonstrated the immune tumor microenvironment as well as response to immune checkpoint blockade differ across subtypes of murine MB.
Neuro-oncology | 2018
Elias Sayour; Hector R. Mendez-Gomez; Adam Grippin; Gabriel De Leon; Brian Stover; Catherine Flores; Christina Pham; Duane Mitchell
Neuro-oncology | 2017
Catherine Flores; Tyler Wildes; Christina Pham; Rebecca Abraham; Ginger Mooree; Duane Mitchell
Neuro-oncology | 2017
Catherine Flores; Jorge Gil; Rebecca Abraham; Christina Pham; Tyler Wildes; Ginger Moore; Jeffrey Drake; Kyle Dyson; Duane Mitchell
Neuro-oncology | 2017
Elias Sayour; Gabriel De Leon; Adam Grippin; Catherine Flores; Christina Pham; Duane Mitchell
Neuro-oncology | 2016
Elias Sayour; Gabriel De Leon; Christina Pham; Adam Grippin; Catherine Flores; Duane Mitchell