Network


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

Hotspot


Dive into the research topics where Christina Delgado is active.

Publication


Featured researches published by Christina Delgado.


Clinical Cancer Research | 2007

Antigenic Profiling of Glioma Cells to Generate Allogeneic Vaccines or Dendritic Cell–Based Therapeutics

Jian Gang Zhang; Junichi Eguchi; Carol A. Kruse; German G. Gomez; Habib Fakhrai; Stephanie Schroter; Wenxue Ma; Neil Hoa; Boris Minev; Christina Delgado; H. Terry Wepsic; Hideho Okada; Martin R. Jadus

Purpose: Allogeneic glioma cell lines that are partially matched to the patient at class I human leukocyte antigen (HLA) loci and that display tumor-associated antigens (TAA) or antigenic precursors [tumor antigen precursor proteins (TAPP)] could be used for generating whole tumor cell vaccines or, alternatively, for extraction of TAA peptides to make autologous dendritic cell vaccines. Experimental Design: Twenty human glioma cell lines were characterized by molecular phenotyping and by flow cytometry for HLA class I antigen expression. Twelve of the 20 cell lines, as well as analyses of freshly resected glioma tissues, were further characterized for protein and/or mRNA expression of 16 tumor antigen precursor proteins or TAA. Results: These 20 human glioma cell lines potentially cover 77%, 85%, and 78% of the U.S. Caucasian population at HLA-A, HLA-B, and HLA-C alleles, respectively. All cells exhibited multiple TAA expressions. Most glioma cells expressed antigen isolated from immunoselected melanoma-2 (Aim-2), B-cyclin, EphA2, GP100, β1,6-N-acetylglucosaminyltransferase V (GnT-V), IL13Rα2, Her2/neu, hTert, Mage, Mart-1, Sart-1, and survivin. Real-time PCR technology showed that glioblastoma specimens expressed most of the TAA as well. Tumor-infiltrating lymphocytes and CD8+ CTL killed T2 cells when loaded with specific HLA-A2+ restricted TAA, or gliomas that were both HLA-A2+ and also positive for specific TAA (Mart-1, GP100, Her2/neu, and tyrosinase) but not those cells negative for HLA-A2 and/or lacking the specific epitope. Conclusions: These data provide proof-in-principle for the use of allogeneic, partially HLA patient–matched glioma cells for vaccine generation or for peptide pulsing with allogeneic glioma cell extracts of autologous patient dendritic cells to induce endogenous CTL in brain tumor patients.


International Immunopharmacology | 2008

Macrophage colony stimulating factor: not just for macrophages anymore! A gateway into complex biologies.

Thomas Douglass; Lara Driggers; Jian Gang Zhang; Neil Hoa; Christina Delgado; Christopher C. Williams; Qinhong Dan; Ramon Sanchez; Edward W. B. Jeffes; H. Terry Wepsic; Michael P. Myers; Kirston Koths; Martin R. Jadus

Macrophage colony stimulating factor (M-CSF, also called colony stimulating factor-1) has traditionally been viewed as a growth/differentiation factor for monocytes, macrophages, and some female-specific tumors. As a result of alternative mRNA splicing and post-translational processing, several forms of M-CSF protein are produced: a secreted glycoprotein, a longer secreted form containing proteoglycan, and a short membrane-bound isoform. These different forms of M-CSF all initiate cell signaling in cells bearing the M-CSF receptor, called c-fms. Here we review the biology of M-CSF, which has important roles in bone physiology, the intestinal tract, cancer metastases to the bone, macrophage-mediated tumor cell killing and tumor immunity. Although this review concentrates mostly on the membrane form of human M-CSF (mM-CSF), the biology of the soluble forms and the M-CSF receptor will also be discussed for comparative purposes. The mechanisms of the biological effects of the membrane-bound M-CSF reveal that this cytokine is unexpectedly involved in many complex molecular events. Recent experiments suggest that a tumor vaccine based on membrane-bound M-CSF-transduced tumor cells, combined with anti-angiogenic therapy, should be evaluated further for use in clinical trials.


PLOS ONE | 2009

Molecular mechanisms of paraptosis induction: implications for a non-genetically modified tumor vaccine.

Neil Hoa; Michael P. Myers; Thomas Douglass; Jian Gang Zhang; Christina Delgado; Lara Driggers; Linda L. Callahan; Gerald Vandeusen; Jimmy T. H. Pham; Nirav Bhakta; Lisheng Ge; Martin R. Jadus

Paraptosis is the programmed cell death pathway that leads to cellular necrosis. Previously, rodent and human monocytes/macrophages killed glioma cells bearing the membrane macrophage colony stimulating factor (mM-CSF) through paraptosis, but the molecular mechanism of this killing process was never identified. We have demonstrated that paraptosis of rat T9 glioma cells can be initiated through a large potassium channel (BK)-dependent process initiated by reactive oxygen species. Macrophage mediated cytotoxicity upon the mM-CSF expressing T9-C2 cells was not prevented by the addition of the caspase inhibitor, zVAD-fmk. By a combination of fluorescent confocal and electron microscopy, flow cytometry, electrophysiology, pharmacology, and genetic knock-down approaches, we demonstrated that these ion channels control cellular swelling and vacuolization of rat T9 glioma cells. Cell lysis is preceded by a depletion of intracellular ATP. Six-hour exposure to BK channel activation caused T9 cells to over express heat shock proteins (Hsp 60, 70, 90 and gp96). This same treatment forced HMGB1 translocation from the nuclear region to the periphery. These last molecules are “danger signals” that can stimulate immune responses. Similar inductions of mitochondrial swelling and increased Hsp70 and 90 expressions by BK channel activation were observed with the non-immunogenic F98 glioma cells. Rats injected with T9 cells which were killed by prolonged BK channel activation developed immunity against the T9 cells, while the injection of x-irradiated apoptotic T9 cells failed to produce the vaccinating effect. These results are the first to show that glioma cellular death induced by prolonged BK channel activation improves tumor immunogenicity; this treatment reproduces the vaccinating effects of mM-CSF transduced cells. Elucidation of strategies as described in this study may prove quite valuable in the development of clinical immunotherapy against cancer.


Laboratory Investigation | 2007

Human monocytes kill M-CSF-expressing glioma cells by BK channel activation.

Neil Hoa; Jian Gang Zhang; Christina Delgado; Michael P. Myers; Linda L. Callahan; Gerald Vandeusen; Patric M. Schiltz; H. Terry Wepsic; Martin R. Jadus

In this study, human monocytes/macrophages were observed to kill human U251 glioma cells expressing membrane macrophage colony-stimulating factor (mM-CSF) via a swelling and vacuolization process called paraptosis. Human monocytes responded to the mM-CSF-transduced U251 glioma cells, but not to viral vector control U251 glioma cells (U251-VV), by producing a respiratory burst within 20 min. Using patch clamp techniques, functional big potassium (BK) channels were observed on the membrane of the U251 glioma cell. It has been previously reported that oxygen indirectly regulates BK channel function. In this study, it was demonstrated that prolonged BK channel activation in response to the respiratory burst induced by monocytes initiates paraptosis in selected glioma cells. Forced BK channel opening within the glioma cells by BK channel activators (phloretin or pimaric acid) induced U251 glioma cell swelling and vacuolization occurred within 30 min. U251 glioma cell cytotoxicity, induced by using BK channel activators, required between 8 and 12 h. Swelling and vacuolization induced by phloretin and pimaric acid was prevented by iberiotoxin, a specific BK channel inhibitor. Confocal fluorescence microscopy demonstrated BK channels co-localized with the endoplasmic reticulum and mitochondria, the two targeted organelles affected in paraptosis. Iberiotoxin prevented monocytes from producing death in mM-CSF-expressing U251glioma cells in a 24 h assay. This study demonstrates a novel mechanism whereby monocytes can induce paraptosis via the disruption of internal potassium ion homeostasis.


Journal of Immunology | 2005

Antiangiogenic Drugs Synergize with a Membrane Macrophage Colony-Stimulating Factor-Based Tumor Vaccine to Therapeutically Treat Rats with an Established Malignant Intracranial Glioma

Edward W. B. Jeffes; Jian Gang Zhang; Neil Hoa; Animesh Petkar; Christina Delgado; Samuel Chong; Andre Obenaus; Ramon Sanchez; Sakineh Khalaghizadeh; Tetyana Khomenko; Brandon Knight; Reza Alipanah; Tuong-Vi Nguyen; Chirag Shah; Seema Vohra; Jing-Li Zhuang; Jessie Liu; H. Terry Wepsic; Martin R. Jadus

Combining a T9/9L glioma vaccine, expressing the membrane form of M-CSF, with a systemic antiangiogenic drug-based therapy theoretically targeted toward growth factor receptors within the tumor’s vasculature successfully treated >90% of the rats bearing 7-day-old intracranial T9/9L gliomas. The antiangiogenic drugs included (Z)-3-[4-(dimethylamino)benzylidenyl]indolin-2-one (a platelet-derived growth factor receptor β and a fibroblast growth factor receptor 1 kinase inhibitor) and oxindole (a vascular endothelial growth factor receptor 2 kinase inhibitor). A total of 20–40% of the animals treated with the antiangiogenic drugs alone survived, while all nontreated controls and tumor vaccine-treated rats died within 40 days. In vitro, these drugs inhibited endothelial cells from proliferating in response to the angiogenic factors produced by T9/9L glioma cells and prevented endothelial cell tubulogenesis. FITC-labeled tomato lectin staining demonstrated fewer and constricted blood vessels within the intracranial tumor after drug therapy. Magnetic resonance imaging demonstrated that the intracranial T9 glioma grew much slower in the presence of these antiangiogenic drugs. These drugs did not affect in vitro glioma cell growth nor T cell mitogenesis. Histological analysis revealed that the tumor destruction occurred at the margins of the tumor, where there was a heavy lymphocytic infiltrate. Real-time PCR showed more IL-2-specific mRNA was present within the gliomas in the vaccinated rats treated with the drugs. Animals that rejected the established T9/9L glioma by the combination therapy proved immune against an intracranial rechallenge by T9/9L glioma, but showed no resistance to an unrelated MADB106 breast cancer.


PLOS ONE | 2012

Differential Glioma-Associated Tumor Antigen Expression Profiles of Human Glioma Cells Grown in Hypoxia

Lisheng Ge; Andrew N. Cornforth; Neil Hoa; Christina Delgado; Shiun Kwei Chiou; Yi-Hong Zhou; Martin R. Jadus

Human U251 and D54 glioma cells were tested for expression of 25 glioma-associated tumor antigen precursor proteins (TAPP) under hypoxic (1% O2) or normoxic (21% O2) conditions. Hypoxic glioma cell lines increased their mRNA expression for nine TAPP (Aim2, Art-4, EphA2, EZH2, Fosl1, PTH-rP, Sox 11, Whsc2 and YKL-40), as assessed by quantitative reverse transcriptase real-time/polymerase chain reaction (qRT-PCR). Increased differences with three hypoxic-induced TAPP: EZH2, Whsc2 and YKL-40 were shown at the protein levels by fluorescent antibody staining and quantitative electrophoretic analysis. Two TAPP (MRP3 and Trp1) were down-regulated by hypoxia in glioma cell lines. Growing the glioma cells under hypoxia for 13 days, followed by returning them back to normoxic conditions for 7 days, and restored the original normoxic TAPP profile. Thus, hypoxia was an environmental factor that stimulated the transient expression of these antigens. Intracranial xenografts grown in nude mice derived from U251 cells that had been cultured under neurosphere stem cell conditions showed increased expression of Whsc2 or YKL-40, demonstrating that these in vitro properties of glioma also occur in vivo. Whsc2-specific cytotoxic T lymphocytes killed the hypoxic U251 glioma cells better than normoxic glioma cells. The antigens expressed by hypoxic tumor cells may be a better source of starting tumor material for loading dendritic cells for novel immunotherapy of glioma using tumor-associated antigens.


Cellular Immunology | 2009

Cytotoxic T cell immunity against the non-immunogenic, murine, hepatocellular carcinoma Hepa1-6 is directed towards the novel alternative form of macrophage colony stimulating factor

Lisheng Ge; Jian Gang Zhang; Christina A. Samathanam; Christina Delgado; Mary Tarbiyat-Boldaji; Qinghong Dan; Neil Hoa; Tuong-Vi Nguyen; Reza Alipanah; Jimmy T. H. Pham; Ramon Sanchez; H. Terry Wepsic; Timothy R. Morgan; Martin R. Jadus

Mouse Hepa1-6 hepatocellular carcinoma (HCC) cells were transduced with the membrane form of macrophage colony stimulating factor (mM-CSF). When mM-CSF transduced Hepa1-6 cells were injected subcutaneously into mice, these cells did not form tumors. The spleens of these immunized mice contained cytotoxic CD8+ T lymphocytes (CTL) that killed the unmodified Hepa1-6 cells. We show that the alternative form of macrophage colony stimulating factor (altM-CSF) induced CTL-mediated immunity against Hepa1-6 cells. AltM-CSF is restricted to the H-2D(b) allele. CTLs killed RMA-S cells loaded with exogenous altM-CSF peptide. Vaccination of mice with dendritic cells pulsed with the altM-CSF peptide stimulated anti-Hepa1-6 CTLs. Hyper-immunization of mice with mM-CSF Hepa1-6 cells showed inflammation of the liver and kidneys. Although altM-CSF was expressed within liver and kidney cells, its intensity was lower than Hepa1-6 cells. AltM-CSF was detected within the human HepG2 cell line. These studies suggest that altM-CSF may be a tumor antigen for HCC.


Molecular Therapy | 2004

585. Anti-Angiogenic Drugs Synergize with a Membrane Macrophage Colony Stimulating Factor (mM-CSF) Based Tumor Vaccine to Therapeutically Treat Rats with an Established Intracranial Glioma

Martin R. Jadus; Edward W. B. Jeffes; Ramon Sanchez; Christina Delgado; Jian G. Zhang; Sakineh Khalaghizadeh; Tetyana Khomenko; Animesh Petkar; Chirag Shah; Brandon Knight; Seema Vohra; Reza Jahroudi; Terry Wepsic

Glioblastoma multiforme (GBM) are lethal brain tumors in humans with survival times of less than a year. We report that we successfully treated >90% of the rats bearing an established intracranial T9/9L glioma by combining a T9/9L glioma vaccine, which expresses the membrane form of macrophage colony stimulating factor (mM-CSF), and an anti-angiogenic drug-based therapy targeting growth factor receptors within the tumors vasculature. The anti-angiogenic drugs included DMBI [a platelet derived growth factor receptor β and a fibroblast growth factor receptor 1 kinase inhibitor] and oxindole [a vascular endothelial growth factor receptor 2 kinase inhibitor]. Twenty to forty percent of the animals treated with the anti-angiogenic drugs alone survived. All non-treated controls and tumor vaccine- treated rats died within 40 days. In vitro, the drugs inhibited the endothelial cell formation of tubes and sprouts; they inhibited endothelial cells from proliferating in response to the angiogenic factors produced by T9/9L glioma cells. The drugs had no effect upon glioma cell growth or T cell mitogenesis. Animals that rejected the established T9/9L glioma by means of the combination therapy proved immune to an intracranial rechallenge by T9/9L glioma, but not to an unrelated MADB106 breast cancer. Anti-angiogenic drugs such as epigallocatechin gallate and thalidomide failed to enhance the tumor vaccine. This works demonstrates that particular anti-angiogenic drugs can successfully synergize with an mM-CSF-based tumor vaccine to treat more advanced tumors.


Blood | 2002

Living T9 glioma cells expressing membrane macrophage colony-stimulating factor produce immediate tumor destruction by polymorphonuclear leukocytes and macrophages via a "paraptosis"-induced pathway that promotes systemic immunity against intracranial T9 gliomas

Yijun Chen; Thomas Douglass; Edward W. B. Jeffes; Qingcheng Xu; Christopher C. Williams; Neary Arpajirakul; Christina Delgado; Michael T. Kleinman; Ramon Sanchez; Qinghong Dan; Ronald C. Kim; H. Terry Wepsic; Martin R. Jadus


Molecular Therapy | 2001

Membrane Macrophage Colony-Stimulating Factor on MADB106 Breast Cancer Cells Does Not Activate Cytotoxic Macrophages but Immunizes Rats against Breast Cancer

Christopher C. Williams; Hao Trinh; Thanh-Van Tran; Qinghong Dan; Ramon Sanchez; Christina Delgado; Yijun Chen; Brian Sippel; Edward W. B. Jeffes; H. Terry Wepsic; Martin R. Jadus

Collaboration


Dive into the Christina Delgado's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Neil Hoa

United States Department of Veterans Affairs

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ramon Sanchez

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas Douglass

California State University

View shared research outputs
Top Co-Authors

Avatar

Linda L. Callahan

California State University

View shared research outputs
Top Co-Authors

Avatar

Lisheng Ge

United States Department of Veterans Affairs

View shared research outputs
Researchain Logo
Decentralizing Knowledge