Network


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

Hotspot


Dive into the research topics where Michael Chastain is active.

Publication


Featured researches published by Michael Chastain.


Journal of Virology | 2003

Comparative Immunogenicity in Rhesus Monkeys of DNA Plasmid, Recombinant Vaccinia Virus, and Replication-Defective Adenovirus Vectors Expressing a Human Immunodeficiency Virus Type 1 gag Gene

Danilo R. Casimiro; Ling Chen; Tong-Ming Fu; Robert K. Evans; Michael J. Caulfield; Mary-Ellen Davies; Aimin Tang; Minchun Chen; Lingyi Huang; Virginia Harris; Daniel C. Freed; Keith A. Wilson; Sheri A. Dubey; De-Min Zhu; Denise K. Nawrocki; Henryk Mach; Robert Troutman; Lynne Isopi; Donna M. Williams; William Hurni; Zheng Xu; Jeffrey G. Smith; Su Wang; Xu Liu; Liming Guan; Romnie Long; Wendy L. Trigona; Gwendolyn J. Heidecker; Helen C. Perry; Natasha Persaud

ABSTRACT Cellular immune responses, particularly those associated with CD3+ CD8+ cytotoxic T lymphocytes (CTL), play a primary role in controlling viral infection, including persistent infection with human immunodeficiency virus type 1 (HIV-1). Accordingly, recent HIV-1 vaccine research efforts have focused on establishing the optimal means of eliciting such antiviral CTL immune responses. We evaluated several DNA vaccine formulations, a modified vaccinia virus Ankara vector, and a replication-defective adenovirus serotype 5 (Ad5) vector, each expressing the same codon-optimized HIV-1 gag gene for immunogenicity in rhesus monkeys. The DNA vaccines were formulated with and without one of two chemical adjuvants (aluminum phosphate and CRL1005). The Ad5-gag vector was the most effective in eliciting anti-Gag CTL. The vaccine produced both CD4+ and CD8+ T-cell responses, with the latter consistently being the dominant component. To determine the effect of existing antiadenovirus immunity on Ad5-gag-induced immune responses, monkeys were exposed to adenovirus subtype 5 that did not encode antigen prior to immunization with Ad5-gag. The resulting anti-Gag T-cell responses were attenuated but not abolished. Regimens that involved priming with different DNA vaccine formulations followed by boosting with the adenovirus vector were also compared. Of the formulations tested, the DNA-CRL1005 vaccine primed T-cell responses most effectively and provided the best overall immune responses after boosting with Ad5-gag. These results are suggestive of an immunization strategy for humans that are centered on use of the adenovirus vector and in which existing adenovirus immunity may be overcome by combined immunization with adjuvanted DNA and adenovirus vector boosting.


PLOS ONE | 2010

Characterization of notch1 antibodies that inhibit signaling of both normal and mutated notch1 receptors

Miguel Aste-Amezaga; Ningyan Zhang; Janet Lineberger; Beth Anne Arnold; Timothy J. Toner; Mingcheng Gu; Lingyi Huang; Salvatore Vitelli; Kim Vo; Peter Haytko; Jing Zhang Zhao; Frederic Baleydier; Sarah L'heureux; Hongfang Wang; Wendy R. Gordon; Elizabeth Thoryk; Marie Blanke Andrawes; Kittichoat Tiyanont; Kimberly Stegmaier; Giovanni Roti; Kenneth N. Ross; Laura L. Franlin; Hui Wang; Fubao Wang; Michael Chastain; Andrew J. Bett; Laurent P. Audoly; Stephen C. Blacklow; Hans E. Huber

Background Notch receptors normally play a key role in guiding a variety of cell fate decisions during development and differentiation of metazoan organisms. On the other hand, dysregulation of Notch1 signaling is associated with many different types of cancer as well as tumor angiogenesis, making Notch1 a potential therapeutic target. Principal Findings Here we report the in vitro activities of inhibitory Notch1 monoclonal antibodies derived from cell-based and solid-phase screening of a phage display library. Two classes of antibodies were found, one directed against the EGF-repeat region that encompasses the ligand-binding domain (LBD), and the second directed against the activation switch of the receptor, the Notch negative regulatory region (NRR). The antibodies are selective for Notch1, inhibiting Jag2-dependent signaling by Notch1 but not by Notch 2 and 3 in reporter gene assays, with EC50 values as low as 5±3 nM and 0.13±0.09 nM for the LBD and NRR antibodies, respectively, and fail to recognize Notch4. While more potent, NRR antibodies are incomplete antagonists of Notch1 signaling. The antagonistic activity of LBD, but not NRR, antibodies is strongly dependent on the activating ligand. Both LBD and NRR antibodies bind to Notch1 on human tumor cell lines and inhibit the expression of sentinel Notch target genes, including HES1, HES5, and DTX1. NRR antibodies also strongly inhibit ligand-independent signaling in heterologous cells transiently expressing Notch1 receptors with diverse NRR “class I” point mutations, the most common type of mutation found in human T-cell acute lymphoblastic leukemia (T-ALL). In contrast, NRR antibodies failed to antagonize Notch1 receptors bearing rare “class II” or “class III” mutations, in which amino acid insertions generate a duplicated or constitutively sensitive metalloprotease cleavage site. Signaling in T-ALL cell lines bearing class I mutations is partially refractory to inhibitory antibodies as compared to cell-penetrating gamma-secretase inhibitors. Conclusions/Significance Antibodies that compete with Notch1 ligand binding or that bind to the negative regulatory region can act as potent inhibitors of Notch1 signaling. These antibodies may have clinical utility for conditions in which inhibition of signaling by wild-type Notch1 is desired, but are likely to be of limited value for treatment of T-ALLs associated with aberrant Notch1 activation.


Proceedings of the National Academy of Sciences of the United States of America | 2008

An oligosaccharide-based HIV-1 2G12 mimotope vaccine induces carbohydrate-specific antibodies that fail to neutralize HIV-1 virions.

Joseph G. Joyce; Isaac J. Krauss; Hong C. Song; David Opalka; Karen M. Grimm; Deborah D. Nahas; Mark T. Esser; Renee Hrin; Meizhen Feng; Vadim Y. Dudkin; Michael Chastain; John W. Shiver; Samuel J. Danishefsky

The conserved oligomannose epitope, Man9GlcNAc2, recognized by the broadly neutralizing human mAb 2G12 is an attractive prophylactic vaccine candidate for the prevention of HIV-1 infection. We recently reported total chemical synthesis of a series of glycopeptides incorporating one to three copies of Man9GlcNAc2 coupled to a cyclic peptide scaffold. Surface plasmon resonance studies showed that divalent and trivalent, but not monovalent, compounds were capable of binding 2G12. To test the efficacy of the divalent glycopeptide as an immunogen capable of inducing a 2G12-like neutralizing antibody response, we covalently coupled the molecule to a powerful immune-stimulating protein carrier and evaluated immunogenicity of the conjugate in two animal species. We used a differential immunoassay to demonstrate induction of high levels of carbohydrate-specific antibodies; however, these antibodies showed poor recognition of recombinant gp160 and failed to neutralize a panel of viral isolates in entry-based neutralization assays. To ascertain whether antibodies produced during natural infection could recognize the mimetics, we screened a panel of HIV-1-positive and -negative sera for binding to gp120 and the synthetic antigens. We present evidence from both direct and competitive binding assays that no significant recognition of the glycopeptides was observed, although certain sera did contain antibodies that could compete with 2G12 for binding to recombinant gp120.


BMC Medical Genomics | 2010

A gene expression signature of RAS pathway dependence predicts response to PI3K and RAS pathway inhibitors and expands the population of RAS pathway activated tumors

Andrey Loboda; Michael Nebozhyn; Rich Klinghoffer; Jason Frazier; Michael Chastain; William T. Arthur; Brian Roberts; Theresa Zhang; Melissa Chenard; Brian B. Haines; Jannik N. Andersen; Kumiko Nagashima; Cloud Paweletz; Bethany Lynch; Igor Feldman; Hongyue Dai; Pearl S. Huang; James Watters

BackgroundHyperactivation of the Ras signaling pathway is a driver of many cancers, and RAS pathway activation can predict response to targeted therapies. Therefore, optimal methods for measuring Ras pathway activation are critical. The main focus of our work was to develop a gene expression signature that is predictive of RAS pathway dependence.MethodsWe used the coherent expression of RAS pathway-related genes across multiple datasets to derive a RAS pathway gene expression signature and generate RAS pathway activation scores in pre-clinical cancer models and human tumors. We then related this signature to KRAS mutation status and drug response data in pre-clinical and clinical datasets.ResultsThe RAS signature score is predictive of KRAS mutation status in lung tumors and cell lines with high (> 90%) sensitivity but relatively low (50%) specificity due to samples that have apparent RAS pathway activation in the absence of a KRAS mutation. In lung and breast cancer cell line panels, the RAS pathway signature score correlates with pMEK and pERK expression, and predicts resistance to AKT inhibition and sensitivity to MEK inhibition within both KRAS mutant and KRAS wild-type groups. The RAS pathway signature is upregulated in breast cancer cell lines that have acquired resistance to AKT inhibition, and is downregulated by inhibition of MEK. In lung cancer cell lines knockdown of KRAS using siRNA demonstrates that the RAS pathway signature is a better measure of dependence on RAS compared to KRAS mutation status. In human tumors, the RAS pathway signature is elevated in ER negative breast tumors and lung adenocarcinomas, and predicts resistance to cetuximab in metastatic colorectal cancer.ConclusionsThese data demonstrate that the RAS pathway signature is superior to KRAS mutation status for the prediction of dependence on RAS signaling, can predict response to PI3K and RAS pathway inhibitors, and is likely to have the most clinical utility in lung and breast tumors.


Journal of Virology | 2005

Universal Influenza B Vaccine Based on the Maturational Cleavage Site of the Hemagglutinin Precursor

Elisabetta Bianchi; Xiaoping Liang; Paolo Ingallinella; Marco Finotto; Michael Chastain; Jiang Fan; Tong-Ming Fu; Hong Chang Song; Melanie Horton; Daniel C. Freed; Walter Manger; Emily Wen; Li Shi; Roxana Ionescu; Colleen Price; Marc Wenger; Emilio A. Emini; Riccardo Cortese; Gennaro Ciliberto; John W. Shiver; Antonello Pessi

ABSTRACT Conventional influenza vaccines can prevent infection, but their efficacy depends on the degree of antigenic “match” between the strains used for vaccine preparation and those circulating in the population. A universal influenza vaccine based on invariant regions of the virus, able to provide broadly cross-reactive protection, without requiring continuous manufacturing update, would solve a major medical need. Since the temporal and geographical dominance of the influenza virus type and/or subtype (A/H3, A/H1, or B) cannot yet be predicted, a universal vaccine, like the vaccines currently in use, should include both type A and type B influenza virus components. However, while encouraging preclinical data are available for influenza A virus, no candidate universal vaccine is available for influenza B virus. We show here that a peptide conjugate vaccine, based on the highly conserved maturational cleavage site of the HA0 precursor of the influenza B virus hemagglutinin, can elicit a protective immune response against lethal challenge with viruses belonging to either one of the representative, non-antigenically cross-reactive influenza B virus lineages. We demonstrate that protection by the HA0 vaccine is mediated by antibodies, probably through effector mechanisms, and that a major part of the protective response targets the most conserved region of HA0, the P1 residue of the scissile bond and the fusion peptide domain. In addition, we present preliminary evidence that the approach can be extended to influenza A virus, although the equivalent HA0 conjugate is not as efficacious as for influenza B virus.


Journal of Virology | 2004

Heterologous Human Immunodeficiency Virus Type 1 Priming-Boosting Immunization Strategies Involving Replication-Defective Adenovirus and Poxvirus Vaccine Vectors

Danilo R. Casimiro; Andrew J. Bett; Tong-Ming Fu; Mary-Ellen Davies; Aimin Tang; Keith A. Wilson; Minchun Chen; Romnie Long; Troy McKelvey; Michael Chastain; Sanjay Gurunathan; Jim Tartaglia; Emilio A. Emini; John W. Shiver

ABSTRACT We compared the human immunodeficiency virus type 1 (HIV-1)-specific cellular immune responses elicited in nonhuman primates by HIV-1 gag-expressing replication-defective adenovirus serotype 5 (Ad5) or poxvirus vectors, used either alone or in combination with each other. The responses arising from a heterologous Ad5 priming-poxvirus boosting regimen were significantly greater than those elicited by homologous regimens with the individual vectors or by a heterologous poxvirus priming-Ad5 boosting regimen. The heterologous Ad5 priming-poxvirus boosting approach may have potential utility in humans as a means of inducing high levels of cellular immunity.


Journal of Virology | 2002

Vaccine-Induced Immune Responses in Rodents and Nonhuman Primates by Use of a Humanized Human Immunodeficiency Virus Type 1 pol Gene

Danilo R. Casimiro; Aimin Tang; Helen C. Perry; Romnie Long; Minchun Chen; Gwendolyn J. Heidecker; Mary-Ellen Davies; Daniel C. Freed; Natasha V. Persaud; Sheri A. Dubey; Jeffrey G. Smith; Diane V. Havlir; Douglas D. Richman; Michael Chastain; Adam J. Simon; Tong-Ming Fu; Emilio A. Emini; John W. Shiver

ABSTRACT A synthetic gene consisting of the reverse transcriptase (RT) and integrase (IN) domains of human immunodeficiency virus type 1 (HIV-1) pol was constructed using codons most frequently used in humans. The humanized pol gave dramatically improved levels of Rev-independent, in vitro protein production in mammalian cells and elicited much stronger cellular immunity in rodents than did virus-derived gene. Specifically, BALB/c mice were immunized with plasmids and/or recombinant vaccinia virus constructs expressing the synthetic gene. High frequencies of Pol-specific T lymphocytes were detected in these animals by the gamma interferon enzyme-linked immunospot assay against pools of short overlapping peptides. Characterization of the stimulatory peptides from these pools indicates that the optimized gene constructs are able to effectively activate both CD4+ and CD8+ T cells. Immunization of rhesus macaques with DNA vaccines expressing the humanized pol coupled to a human tissue plasminogen activator leader sequence led to pronounced in vitro cytotoxic T-lymphocyte killing activities and enhanced levels of circulating Pol-specific T cells, comparable to those observed in HIV-1-infected human subjects. Thus, optimizing the immunogenic properties of HIV-1 Pol at the level of the gene sequence validates it as an antigen and provides an important step toward the construction of a potent pol-based HIV-1 vaccine component.


Biologics: Targets & Therapy | 2010

Distinct expression profiles of Notch-1 protein in human solid tumors: Implications for development of targeted therapeutic monoclonal antibodies

Yuan Li; Janine Burns; Carol A. Cheney; Ningyan Zhang; Salvatore Vitelli; Fubao Wang; Andrew J. Bett; Michael Chastain; Laurent P. Audoly; Zhi Qiang Zhang

Biological therapies, such as monoclonal antibodies (mAbs) that target tumor-associated antigens have been considered an effective therapeutic approach in oncology. In considering Notch-1 receptor as a potential target, we performed immunohistochemistry on tissue microarrays to determine 1) whether the receptor is overexpressed in tumor cells as compared to their corresponding normal tissues and 2) the clinical significance of its expression levels in human breast, colorectal, lung and prostate cancers. We found that the expression of Notch-1 protein was overexpressed in primary colorectal adenocarcinoma and nonsmall cell lung carcinoma (NSCLC), but not in primary ductal breast carcinoma or prostate adenocarcinoma. Further analysis revealed that higher levels of Notch-1 protein expression were significantly associated with poorer differentiation of breast and prostate tumors. Strikingly, for NSCLC, the expression levels of Notch-1 protein were found to be inversely correlated with tumor differentiation and progression. For colorectal tumors, however, no correlation of Notch-1 protein expression was found with any tumor clinicopathological parameters, in spite of its overexpression in tumor cells. Our data demonstrated the complexity of Notch-1 protein expression in human solid tumors and further supported the notion that the roles of Notch-1 expression in tumorigenesis are highly context-dependent. The findings could provide the basis for development of distinct therapeutic strategies of Notch-1 mAbs for its applications in the treatment of suitable types of human cancers.


Archive | 2006

A Universal Influenza B Peptide Vaccine

Paolo Ingallinella; Elisabetta Bianchi; Xiaoping Liang; Marco Finotto; Michael Chastain; Jiang Fan; Tong-Ming Fu; Hong Chang Song; Melanie Horton; Daniel C. Freed; Walter Manger; Emily Wen; Li Shi; Roxana Ionescu; Colleen Price; Marc Wenger; Emilio A. Emini; Riccardo Cortese; Gennaro Ciliberto; John W. Shiver; Antonello Pessi

Paolo Ingallinella, Elisabetta Bianchi, Xiaoping Liang, Marco Finotto, Michael Chastain, Jiang Fan, Tong-Ming Fu, Hong Chang Song, Melanie Horton, Daniel Freed, Walter Manger, Emily Wen, Li Shi, Roxana Ionescu, Colleen Price, Marc Wenger, Emilio Emini, Riccardo Cortese, Gennaro Ciliberto, John Shiver and Antonello Pessi IRBM P.Angeletti, Pomezia (Rome), Italy; Merck Research Laboratories, West Point, PA, USA


Science | 2000

Control of Viremia and Prevention of Clinical AIDS in Rhesus Monkeys by Cytokine-Augmented DNA Vaccination

Dan H. Barouch; Sampa Santra; Jörn E. Schmitz; Marcelo J. Kuroda; Tong-Ming Fu; Wendeline Wagner; Miroslawa Bilska; Abie Craiu; Xin Xiao Zheng; Georgia R. Krivulka; Kristin Beaudry; Michelle A. Lifton; Christine E. Nickerson; Wendy L. Trigona; Kara Punt; Dan C. Freed; Liming Guan; Sheri A. Dubey; Danilo R. Casimiro; Adam J. Simon; Mary-Ellen Davies; Michael Chastain; Terry B. Strom; Rebecca Gelman; David C. Montefiori; Mark G. Lewis; Emilio A. Emini; John W. Shiver; Norman L. Letvin

Collaboration


Dive into the Michael Chastain's collaboration.

Top Co-Authors

Avatar

Emilio A. Emini

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Tong-Ming Fu

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Mary-Ellen Davies

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Aimin Tang

United States Military Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge