UB Vice President for Research and Economic Development markup
Richard B. Bankert UB Profile
Microbiology and Immunology
Professor of Microbiology
217 Biomedical Research Building
Buffalo NY 14214
716-829-2701
Export
Publication Types
Co-Authors
  • More Coauthors +
 
Authors: Fiona Y. Glassman, Jennifer L. Schneider, Radha Ramakrishnan, Robert K. Dingman, Murali Ramanathan, Richard B. Bankert, Sathy V. Balu-Iyer
Journal-article: Journal of Pharmaceutical Sciences, 2018
Authors: Shin La Shu, Cheryl L. Allen, Yunchen Yang, Orla Maguire, Hans Minderman, Arindam Sen, Michael J. Ciesielski, Katherine A. Collins, Peter J. Bush, Martin Morgan, Yun Wu, Richard B. Bankert, Marc S. Ernstoff
Proceedings-article: Tumor Biology, 2018
Authors: Gautam N. Shenoy, Jenni Loyall, Orla Maguire, Vandana Iyer, Raymond J. Kelleher, Hans Minderman, Paul K. Wallace, Kunle Odunsi, Sathy V. Balu-Iyer, Richard B. Bankert
Journal-article: Cancer Immunology Research, vol. 6, pp. 236-247, 2018
Authors: W. Richard Burack, Janice M. Spence, John P. Spence, Stephen A. Spence, Philip J. Rock, Gautam N. Shenoy, Leonard D. Shultz, Richard B. Bankert, Steven H. Bernstein
Abstract: To discern features of non-Hodgkin lymphomas (NHL) that are autonomous from those that are shaped by the tumor environment (TE), we used patient-derived xenografts (PDX) to probe the effects on neoplastic cells of manipulating the TE. Properties of neoplastic cells that are often considered to be autonomous include their relative independence from stromal support, their relative survival and/or proliferation advantages compared with nonneoplastic cells, and their state of differentiation. Prior approaches to creation of PDX models likely select for neoplasms, which are the most capable of engraftment, potentially masking the effects of the TE. To overcome this bias, we developed a robust protocol that rapidly produced xenografts with more than 85% of unselected, cryo-preserved, B-cell NHL specimens, including low-grade tumors such as follicular and marginal zone lymphoma. To discern features that are shaped by the TE, we extensively studied 4 low-grade lymphoma specimens. B-cell engraftment required components of the native TE; specifically, CD4+ cells. The relative survival of neoplastic compared with nonneoplastic B cells was not autonomous in 2 specimens; specifically, neoplastic B cells from 2 specimens showed a greater dependence on the TE than normal B cells for engraftment. Furthermore, the differentiation of neoplastic B cells was dependent on the TE; mature B-cell neoplasms converted to plasmacytoma-like lesions in the grafts. These results highlight the central and patient-specific roles of the TE in maintaining the relative survival of neoplastic cells compared with normal cells and in controlling the differentiation of neoplastic cells.
PubMedId: 29296768
Journal-article: Blood Advances, vol. 1, pp. 1263-1273, 2017
Authors: Raymond J Kelleher, Sathy Balu-Iyer, Jenni Loyall, Anthony J Sacca, Gautam N Shenoy, Peng Peng, Vandana Iyer, Anas M Fathallah, Charles S Berenson, Paul K Wallace, Joseph Tario, Kunle Odunsi, Richard B Bankert
Abstract: The identification of immunosuppressive factors within human tumor microenvironments, and the ability to block these factors, would be expected to enhance patients' antitumor immune responses. We previously established that an unidentified factor, or factors, present in ovarian tumor ascites fluids reversibly inhibited the activation of T cells by arresting the T-cell signaling cascade. Ultracentrifugation of the tumor ascites fluid has now revealed a pellet that contains small extracellular vesicles (EV) with an average diameter of 80 nm. The T-cell arrest was determined to be causally linked to phosphatidylserine (PS) that is present on the outer leaflet of the vesicle bilayer, as a depletion of PS-expressing EV or a blockade of PS with anti-PS antibody significantly inhibits the vesicle-induced signaling arrest. The inhibitory EV were also isolated from solid tumor tissues. The presence of immunosuppressive vesicles in the microenvironments of ovarian tumors and our ability to block their inhibition of T-cell function represent a potential therapeutic target for patients with ovarian cancer.
PubMedId: 26112921
Journal-article: Cancer immunology research, vol. 3, 2015
Authors: Anas M Fathallah, Richard B Bankert, Sathy V Balu-Iyer
Abstract: The administration of therapeutic proteins via the subcutaneous route (sc) is desired for compliance and convenience, but could be challenging due to perceived immunogenic potential or unwanted immune responses. There are clinical and preclinical data supporting as well as refuting the generalized notion that sc is more immunogenic. We provide a mechanistic perspective of immunogenicity of therapeutic proteins administered via the sc route and discuss strategies and opportunities for novel therapeutic approaches to mitigate immunogenicity.
PubMedId: 23856740
Journal-article: The AAPS journal, vol. 15, 2013
Authors: Puneet Gaitonde, Radha Ramakrishnan, Jamie Chin, Raymond J Kelleher, Richard B Bankert, Sathy V Balu-Iyer
Keywords: Biotechnology Blood Coagulation Factors Dendritic Cells Factor VIII GFP-FoxP3 Immunological Tolerance Immunosuppression Phosphatidylserine Regulatory T-cell Reverse/Inverse Vaccination
Abstract: Administration of recombinant factor VIII (FVIII), an important co-factor in blood clotting cascade, elicits unwanted anti-FVIII antibodies in hemophilia A (HA) patients. Previously, FVIII associated with phosphatidylserine (PS) showed significant reduction in the anti-FVIII antibody response in HA mice. The reduction in the immune response to FVIII-PS could be due either to a failure of the immune system to recognize the antigen (i.e. immunological ignorance) or to an active induction of an antigen-specific nonresponsiveness (i.e. immunological tolerance). If it were a result of tolerance, one would predict that pre-exposure to FVIII-PS would render the mice hypo-responsive to a subsequent FVIII challenge. Here, we have demonstrated that naive HA mice that were pretreated with FVIII-PS showed a significantly reduced FVIII immune response to further challenge with native FVIII and that this decreased responsiveness could be adoptively transferred to other mice. An increase in number of FoxP3-expressing CD4(+) regulatory T-cells (Treg) was observed for the FVIII-PS-immunized group as compared with animals that received FVIII alone, suggesting the involvement of Treg in PS-mediated hypo-responsiveness. The PS-mediated reduction in antibody response was reversed by the co-administration of function-blocking anti-TGF- antibody with FVIII-PS. The decreased response to FVIII induced by FVIII-PS was determined to be antigen-specific because the immune response to another non-cross-reactive antigen (ovalbumin) was not altered. These results are consistent with the notion that FVIII-PS is tolerogenic and suggest that immunization with this tolerogenic form of the protein could be a useful treatment option to minimize immunogenicity of FVIII and other protein-based therapeutics.
PubMedId: 23649621
Journal-article: The Journal of biological chemistry, vol. 288, 2013
Authors: Joel M Bernstein, Heather Lehman, Maciej Lis, Amy Sands, Gregory E Wilding, Leonard Shultz, Richard Bankert, Libuse Bobek
PubMedId: 22724276
Journal-article: The Annals of otology, rhinology, and laryngology, vol. 121, 2012
Authors: Puneet Gaitonde, Aaron Peng, Robert M. Straubinger, Richard B. Bankert, Sathy V. Balu-iyer
Journal-article: Journal of Pharmaceutical Sciences, vol. 101, pp. 48-55, 2012
Authors: Heather K Lehman, Michelle R Simpson-Abelson, Thomas F Conway, Raymond J Kelleher, Joel M Bernstein, Richard B Bankert
Abstract: A majority of T cells from chronic inflammatory tissues derived from patients with nasal polyposis were found to express an effector memory phenotype. We report here that these memory T cells failed to activate NF-B in response to TCR stimulation but responded normally when the proximal TCR signaling molecules were bypassed with PMA and ionomycin. The dysfunction of these cells was associated with a decrease in the phosphorylation of several TCR proximal signaling molecules including ZAP70, Lck and SLP-76. In addition to the disruption in the TCR signaling pathway, the nasal polyp-associated T cells were shown to have a defect in their ability to translocate LAMP-1 to the cell surface. The results presented here establish that the phenotype and anergy of the T cells in the nasal polyp are similar to those which is seen in memory T cells derived from human tumors and other sites of chronic inflammation.
PubMedId: 22310933
Journal-article: Journal of the Association for Research in Otolaryngology : JARO, vol. 13, 2011
Authors: Dipak S Pisal, Matthew P Kosloski, C Russell Middaugh, Richard B Bankert, Sathy V Balu-Iyer
Abstract: The administration of recombinant factor VIII (FVIII) is the first-line therapy for hemophilia A (HA), but 25%-35% of patients develop an inhibitory antibody response. In general, the presence of aggregates contributes to unwanted immunogenic responses against therapeutic proteins. FVIII has been shown to form both native-like and nonnative aggregates. Previously, we showed that nonnative aggregates of FVIII are less immunogenic than the native protein. Here, we investigated the effect of native-like aggregates of FVIII on immunogenicity in HA and von Willebrand factor knockout (vWF(-/-)) mice. Mice immunized with native-like aggregates showed significantly higher inhibitory antibody titers than animals that received native FVIII. Following restimulation in vitro with native FVIII, the activation of CD4+ T-cells isolated from mice immunized with native-like aggregates is approximately fourfold higher than mice immunized with the native protein. Furthermore, this is associated with increases in the secretion of proinflammatory cytokines IL-6 and IL-17 in the native-like aggregate treatment group. The results indicate that the native-like aggregates of FVIII are more immunogenic than native FVIII for both the B-cell and the T-cell responses.
PubMedId: 22388918
Journal-article: Journal of pharmaceutical sciences, vol. 101, 2011
Authors: Puneet Gaitonde, Aaron Peng, Robert M. Straubinger, Richard B. Bankert, Sathy V. Balu-Iyer
Abstract: A major clinical complication in the treatment of Hemophilia A using exogenously administered recombinant Factor VIII (FVIII) is the development of neutralizing antibodies. It has been shown previously that FVIII complexed with phosphatidylserine (PS) reduces the development of total and neutralizing antibody titers in hemophilic mice. The effect of complexation of FVIII with PS upon dendritic cell (DC) uptake, maturation and processing, T-cell proliferation and cytokine secretion profiles was investigated. Flow cytometric studies of DC showed that PS inhibited the up-regulation of cell surface co-stimulatory markers (CD86 and CD40). PS reduced T-cell proliferation and significantly increased levels of TGF- and IL-10 but reduced secretion of IL-6 and IL-17 compared to controls. The data suggest that PS reduces immunogenicity of FVIII by regulating dendritic cell maturation and subsequent T-lymphocyte activity through modulation of cytokine secretion. A possible mechanism for PS-mediated induction of FVIII tolerance is discussed.
PubMedId: 21094627
Journal-article: Clinical Immunology, vol. 138, pp. 135-145, 2011
Authors: Richard B. Bankert, Sathy V. Balu-Iyer, Kunle Odunsi, Leonard D. Shultz, Raymond J. Kelleher, Jennifer L. Barnas, Michelle Simpson-Abelson, Robert Parsons, Sandra J. Yokota
Abstract: Ovarian cancer is the most common cause of death from gynecological cancer. Understanding the biology of this disease, particularly how tumor-associated lymphocytes and fibroblasts contribute to the progression and metastasis of the tumor, has been impeded by the lack of a suitable tumor xenograft model. We report a simple and reproducible system in which the tumor and tumor stroma are successfully engrafted into NOD-scid IL2R(null) (NSG) mice. This is achieved by injecting tumor cell aggregates derived from fresh ovarian tumor biopsy tissues (including tumor cells, and tumor-associated lymphocytes and fibroblasts) i.p. into NSG mice. Tumor progression in these mice closely parallels many of the events that are observed in ovarian cancer patients. Tumors establish in the omentum, ovaries, liver, spleen, uterus, and pancreas. Tumor growth is initially very slow and progressive within the peritoneal cavity with an ultimate development of tumor ascites, spontaneous metastasis to the lung, increasing serum and ascites levels of CA125, and the retention of tumor-associated human fibroblasts and lymphocytes that remain functional and responsive to cytokines for prolonged periods. With this model one will be able to determine how fibroblasts and lymphocytes within the tumor microenvironment may contribute to tumor growth and metastasis, and will make it possible to evaluate the efficacy of therapies that are designed to target these cells in the tumor stroma.
PubMedId: 21935406
Journal-article: PLoS ONE, vol. 6, pp. e24420, 2011
Authors: Jennifer L Barnas, Michelle R Simpson-Abelson, Stephen P Brooks, Raymond J Kelleher, Richard B Bankert
Abstract: Fibroblasts are a dominant cell type in most human solid tumors. The possibility that fibroblasts have the capacity to interact with and modulate the function of tumor-associated T lymphocytes makes them a potential therapeutic target. To address this question, primary cultures of fibroblasts derived from human lung tumors were established and cultured with T cells derived from the same tumor. The tumor fibroblasts significantly enhance the production of IFN-gamma and IL-17A by the tumor-associated T cells following a CD3/CD28-induced activation of the T cells. This enhancement was fibroblast cell dose-dependent and did not require direct contact between the two cell types. Tumor-associated fibroblast-conditioned media similarly enhanced both IFN-gamma and IL-17A in activated T cells, and this enhancement was significantly reduced by Abs to IL-6. Conditioned media derived from activated lymphocyte cultures significantly enhanced IL-6 production by tumor fibroblasts. A similar enhancement of IFN-gamma and IL-17A was observed when activated T cells from a normal donor were cultivated with skin fibroblasts derived from the same donor. These results establish that fibroblasts and autologous lymphocytes, whether derived from the tumor microenvironment or from nonmalignant tissues, have the capacity to reciprocally interact and modulate function. In contrast to other reports, fibroblasts are shown to have an immunostimulatory effect upon activated T lymphocytes. The ability of fibroblasts to enhance two T cell cytokines known to have an impact upon tumor progression suggests that fibroblasts play an important role in tumor pathogenesis that could be exploited therapeutically.
PubMedId: 20686130
Journal-article: Journal of immunology (Baltimore, Md. : 1950), vol. 185, 2010
Authors: Joel M Bernstein, Stephen P Brooks, Heather K Lehman, Liza Pope, Amy Sands, Leonard D Shultz, Richard B Bankert
Abstract: The objective was to develop a model with which to study the cellular and molecular events associated with nasal polyp progression. To accomplish this, we undertook to develop a system in which nondisrupted human nasal polyp tissue could be successfully implanted into severely immunocompromised mice, in which the histopathology of the original nasal polyp tissue, including inflammatory lymphocytes, epithelial and goblet cell hyperplasia, and subepithelial fibrosis, could be preserved for prolonged periods. Small, non-disrupted pieces of human nasal polyp tissues were subcutaneously implanted into NOD-scid IL2rgamma(null) mice. Xenografts at 8 to 12 weeks after implantation were examined histologically and immunohistochemically to identify human inflammatory leukocytes and to determine whether the characteristic histopathologic characteristics of the nasal polyps were maintained for a prolonged period. The xenografts, spleen, lung, liver, and kidneys were examined histologically and immunohistochemically and were evaluated for changes in volume. The sera of these mice were assayed for human cytokines and immunoglobulin. Xenografts of human nasal polyp tissues were established after their subcutaneous implantation into NOD-scid IL2rgamma(null) mice. The xenografts were maintained in a viable and functional state for up to 3 months, and retained a histopathologic appearance similar to that of the original tissue, with a noticeable increase in goblet cell hyperplasia and marked mucus accumulation in the submucosal glands compared to the original nasal polyp tissue. Inflammatory lymphocytes present in the polyp microenvironment were predominantly human CD8+ T cells with an effector memory phenotype. Human CD4+ T cells, CD138+ plasma cells, and CD68+ macrophages were also observed in the xenografts. Human immunoglobulin and interferon-gamma were detected in the sera of xenograft-bearing mice. The polyp-associated lymphocytes proliferated and were found to migrate from the xenografts to the spleens of the recipient mice, resulting in a significant splenomegaly. A progressive increase in the volume of the xenografts was observed with little or no evidence of mouse cell infiltration into the human leukocyte antigen-positive human tissue. An average twofold increase in polyp volume was found at 3 months after engraftment. The use of innate and adaptive immunodeficient NOD-scid mice homozygous for targeted mutations in the interleukin-2 receptor gamma-chain locus NOD-scid IL2rgamma(null) for establishing xenografts of nondisrupted pieces of human nasal polyp tissues represents a significant improvement over the previously reported xenograft model that used partially immunoincompetent CB17-scid mice as tissue recipients. The absence of the interleukin-2 receptor gamma-chain results in complete elimination of natural killer cell development, as well as severe impairments in T and B cell development. These mice, lacking both innate and adaptive immune responses, significantly improve upon the long-term engraftment of human nasal polyp tissues and provide a model with which to study how nasal polyp-associated lymphocytes and their secreted biologically active products contribute to the histopathology and progression of this chronic inflammatory disease.
PubMedId: 20112521
Journal-article: The Annals of otology, rhinology, and laryngology, vol. 118, 2010
Authors: Jennifer L. Barnas, Michelle R. Simpson-Abelson, Sandra J. Yokota, Raymond J. Kelleher, Richard B. Bankert
Keywords: Cancer Fibroblast Immunotherapy Stromal cell T lymphocyte TCR signal transduction Tumor microenvironment Xenograft model
Abstract: The immune system of cancer patients recognizes tumor-associated antigens expressed on solid tumors and these antigens are able to induce tumor-specific humoral and cellular immune responses. Diverse immunotherapeutic strategies have been used in an attempt to enhance both antibody and T cell responses to tumors. While several tumor vaccination strategies significantly increase the number of tumor-specific lymphocytes in the blood of cancer patients, most vaccinated patients ultimately experience tumor progression. CD4+ and CD8+ T cells with an effector memory phenotype infiltrate human tumor microenvironments, but most are hyporesponsive to stimulation via the T cell receptor (TCR) and CD28 under conditions that activate memory T cells derived from the peripheral blood of the cancer patients or normal donors. Attempts to identify cells and molecules responsible for the TCR signaling arrest of tumor-infiltrating T cells have focused largely upon the immunosuppressive effects of tumor cells, tolerogenic dendritic cells and regulatory T cells. Here we review potential mechanisms by which human T cell function is arrested in the tumor microenvironment with a focus on the immunomodulatory effects of stromal fibroblasts. Determining in vivo which cells and molecules are responsible for the TCR arrest in human tumor-infiltrating T cells will be necessary to formulate and test strategies to prevent or reverse the signaling arrest of the human T cells in situ for a more effective design of tumor vaccines. These questions are now addressable using novel human xenograft models of tumor microenvironments.
PubMedId: 21209773
Journal-article: Cancer Microenvironment, vol. 3, pp. 29-47, 2010
Authors: H.K. Lehman, J.M. Bernstein, R.B. Bankert
Journal-article: Journal of Allergy and Clinical Immunology, vol. 125, pp. AB62, 2010
Authors: Shannon P Hilchey, Ollivier Hyrien, Tim R Mosmann, Alexandra M Livingstone, Jonathan W Friedberg, Faith Young, Richard I Fisher, Raymond J Kelleher, Richard B Bankert, Steven H Bernstein
Abstract: The incorporation of rituximab, a chimeric anti-CD20 monoclonal antibody, into the therapeutic armamentarium for patients with follicular lymphoma (FL) has significantly improved treatment outcome for such patients. Despite the almost universal application of this therapy, however, its exact mechanism of action has not been completely defined. One proposed mechanism is that of a "vaccinal" effect, whereby FL cell kill by rituximab results in the elicitation of an FL-specific T-cell response. The demonstration that rituximab can even elicit such a response in patients has, to our knowledge, never been shown. We analyzed the response against the immunoglobulin expressed by the FL before and after rituximab monotherapy in 5 FL patients and found an increase in FL idiotype-specific T cells after rituximab in 4 of 5 patients. Our data thus provide "proof of principle" for the ability of passive immunotherapy with rituximab to elicit an active FL-specific cellular response.
PubMedId: 19196657
Journal-article: Blood, vol. 113, 2009
Authors: Michelle R Simpson-Abelson, Vivek S Purohit, Wing Man Pang, Vandana Iyer, Kunle Odunsi, Todd L Demmy, Sandra J Yokota, Jenni L Loyall, Raymond J Kelleher, Sathy Balu-Iyer, Richard B Bankert
Abstract: Using a novel loading technique, IL-12 is reported here to be efficiently encapsulated within large multilamellar liposomes. The preclinical efficacy of the cytokine loaded liposomes to deliver IL-12 into human tumors and to reactive tumor-associated T cells in situ is tested using a human tumor xenograft model. IL-12 is released in vivo from these liposomes in a biologically active form when injected into tumor xenografts that are established by the subcutaneous implantation of non-disrupted pieces of human lung, breast or ovarian tumors into immunodeficient mice. The histological architecture of the original tumor tissue, including tumor-associated leukocytes, tumor cells and stromal cells is preserved anatomically and the cells remain functionally responsive to cytokines in these xenografts. The local and sustained release of IL-12 into the tumor microenvironment reactivates tumor-associated quiescent effector memory T cells to proliferate, produce and release IFN-gamma resulting in the killing of tumor cells in situ. Very little IL-12 is detected in the serum of mice for up to 5 days after an intratumoral injection of the IL-12 liposomes. We conclude that IL-12 loaded large multilamellar liposomes provide a safe method for the local and sustained delivery of IL-12 to tumors and a therapeutically effective way of reactivating existing tumor-associated T cells in human solid tumor microenvironments. The potential of this local in situ T cell re-stimulation to induce a systemic anti-tumor immunity is discussed.
PubMedId: 19395317
Journal-article: Clinical immunology (Orlando, Fla.), vol. 132, 2008
Authors: Michelle Simpson-Abelson, Richard B Bankert
Abstract: One of the major conundrums in cancer immunotherapy is why human tumors are not rejected, and progress despite the presence of inflammatory leukocytes in the tumor microenvironment. While studies addressing the mechanisms responsible for the failure of immunocompetent cells to control tumor progression have shed considerable light upon this issue, not enough is known about the mechanisms contributing to the regulation of tumor-associated T cells in the microenvironment of human tumors. A persistent and robust response is likely to be required for the complete eradication of tumors. Such a durable immune response will require the development and persistence of functional tumor-reactive memory T cells. Various studies have investigated the mechanisms of suppression in the tumor microenvironment. However, very few studies have investigated the hyporesponsiveness of tumor-associated T cells at the molecular level. This review focuses on the hyporesponsiveness of tumor-associated T cells and how this relates to the T cell receptor signaling cascade. We postulate that this hyporesponsiveness results from a normal regulatory mechanism or TCR signaling checkpoint that is initiated by the persistence of antigen. If the TCR checkpoint is defined, it will be possible to design therapeutic strategies that reverse the TCR arrest. Essentially, this will reactivate the T cells in situ, leading to the killing and lysis of tumors locally, the release of tumor antigens and the generation of a systemic antitumor immunity.
PubMedId: 18348683
Journal-article: Expert Opinion on Therapeutic Targets, vol. 12, pp. 477-490, 2008
Authors: M. R. Simpson-Abelson, G. F. Sonnenberg, H. Takita, S. J. Yokota, T. F. Conway, R. J. Kelleher, L. D. Shultz, M. Barcos, R. B. Bankert
Journal-article: The Journal of Immunology, vol. 180, pp. 7009-7018, 2008
Authors: Michelle R Simpson-Abelson, Gregory F Sonnenberg, Hiroshi Takita, Sandra J Yokota, Thomas F Conway, Raymond J Kelleher, Leonard D Shultz, Maurice Barcos, Richard B Bankert
PubMedId: 18453623
Journal Article: Journal of immunology (Baltimore, Md. : 1950), vol. 180, 2008
Authors: M. R. Nazareth, L. Broderick, M. R. Simpson-Abelson, R. J. Kelleher, S. J. Yokota, R. B. Bankert
Journal-article: The Journal of Immunology, vol. 179, pp. 733-733, 2007
Authors: S. P. Hilchey, A. De, L. M. Rimsza, R. B. Bankert, S. H. Bernstein
Journal-article: The Journal of Immunology, vol. 178, pp. 4051-4061, 2007
Authors: L. Broderick, R.B. Bankert
Journal-article: Journal of Allergy and Clinical Immunology, vol. 119, pp. S92, 2007
Authors: M. R. Nazareth, L. Broderick, M. R. Simpson-Abelson, R. J. Kelleher, S. J. Yokota, R. B. Bankert
Journal-article: The Journal of Immunology, vol. 178, pp. 5552-5562, 2007
Authors: Michael R Nazareth, Lori Broderick, Michelle R Simpson-Abelson, Raymond J Kelleher, Sandra J Yokota, Richard B Bankert
PubMedId: 17442937
Journal Article: Journal of immunology (Baltimore, Md. : 1950), vol. 178, 2007
Authors: Lori Broderick, Stephen P. Brooks, Hiroshi Takita, Alan N. Baer, Joel M. Bernstein, Richard B. Bankert
Abstract: Memory T cells in human non-small cell lung cancer are unresponsive to progressing tumors. T cells were evaluated at the single cell level by imaging the nuclear translocation of NF-kappaB and NFAT via immunofluorescence confocal microscopy as an early measure of responsiveness to T cell receptor triggering. Little translocation of NF-kappaB or NFAT occurred in tumor-associated T cells in response to CD3+CD28 cross-linking under conditions which led to maximal translocation in normal donor peripheral blood T cells. TNF-alpha induced maximal NF-kappaB translocation in these T cells, indicating that they remain receptive to alternative signaling pathways, and pulsing with IL-12 prior to TCR triggering reversed their apparent anergy. T cells from additional chronic inflammatory microenvironments demonstrated a similar refractoriness to TCR activation, suggesting either that a common regulatory mechanism present within the microenvironment controls these cells or that with continuous antigen exposure, they remain refractory to activation via the TCR.
PubMedId: 16271513
Journal-article: Clinical Immunology, vol. 118, pp. 159-169, 2006
Authors: L. Broderick, R. B. Bankert
Journal-article: The Journal of Immunology, vol. 177, pp. 3082-3088, 2006
Authors: Lori Broderick, Richard B. Bankert
Abstract: Human tumors often progress and spread in spite of the presence of large numbers of CD4+ and CD8+ T cells with activated or memory cell phenotypes. The T cells in the microenvironment of human lung tumors fail to be activated in response to stimulation via the T cell receptor and CD28 under conditions that fully activate T cells derived from the peripheral blood of the cancer patients. A combination of regulatory mechanisms which are also observed in a variety of different chronic inflammatory conditions may contribute to the T cell unresponsiveness, and to their inability to respond to and kill tumor cells. The non-responsiveness of memory T cells isolated from human lung tumors and non-malignant chronic inflammatory tissues can be reversed in vitro by a brief pulse with IL-12, and the local and sustained release of exogenous IL-12 into the microenvironment of human tumor xenografts in SCID mice re-activates the tumor-associated T cells in situ. In the later case, the T cells proliferate, secrete interferon-gamma and initiate a cascade of events that culminate in the eradication of tumor cells from the xenograft. In transplantable and spontaneously developing tumors of mice the injection of a single tumor nodule with IL-12 loaded biodegradable microspheres activates tumor-associated T cells to kill tumor cells in situ, and provokes a systemic anti-tumor response that results in the eradication of distant metastatic tumor nodules that are not treated with the cytokine. These mice exhibit a systemic tumor specific immunity as they resist a second challenge with the same (but not a different) tumor. These findings suggest that it will be possible to provoke a systemic anti-tumor immunity in cancer patients by the direct injection of IL-12 loaded biodegradable microspheres or liposomes to locally deliver very low but sustained doses of IL-12 into a single tumor site. This strategy which is based upon the ability of IL-12 to re-activate tumor-associated T cells is termed in situ tumor vaccination.
PubMedId: 16916760
Journal-article: Immunological Investigations, vol. 35, pp. 419-436, 2006
Authors: Joel M. Bernstein, Lori Broderick, Robert R. Parsons, Richard B. Bankert
Abstract: We undertook to maintain human nasal polyp tissue in a viable and functional state in SCID (severe combined immunodeficiency) mice. Small, nondisrupted pieces of human nasal polyp tissues were subcutaneously implanted into SCID mice depleted of natural killer cells. The resulting xenografts were examined histologically, and the sera were evaluated for the presence of human protein. The original histologic architecture of the polyp was maintained in the xenografts. The tissues, including pseudostratified columnar epithelial-lined polyps and subepithelial stroma, remained viable, and goblet cells continued to produce mucin for up to 26 weeks after engraftment. Human inflammatory leukocytes, including CD3+ T cells, CD20+ B cells, CD138+ plasma cells, and CD68+ monocytes and/or macrophages, were present. Identification of human immunoglobulin and human interferon-gamma in the sera of xenograft-bearing mice indicated that the B cells or plasma cells and T cells within the xenografts remained functional for 2 weeks after engraftment. The ability to engraft and maintain nasal polyps provides an in vivo human/mouse chimeric model with which to investigate the role of inflammatory leukocytes and stromal cells in the maintenance and progression of polyposis and to determine how exogenous cytokines may alter the interaction of inflammatory cells, stromal cells, and epithelial cells in the polyp.
PubMedId: 16466102
Journal-article: Annals of Otology, Rhinology & Laryngology, vol. 115, pp. 65-73, 2006
Authors: Qiang Lou, Thomas F Conway, Nejat K Egilmez, Jenni L Loyall, Steven H Bernstein, Raymond J Kelleher, Richard B Bankert
Abstract: Protein antigens have been covalently linked randomly to surface proteins on immature dendritic cells (DC). This has been achieved under physiological conditions using a heterobifunctional reagent that couples antigens to free thiol groups expressed on DC surface proteins. This results in a significant increase in the amount of antigen that is bound to DC, and the antigen/membrane protein complexes that are formed are rapidly internalized. DC, loaded covalently with either beta-galactosidase (beta-gal) or a tumor-associated immunoglobulin (Ig) when injected into mice, induce a beta-gal- or Ig-specific T cell response, and a protective anti-tumor immunity for tumors expressing either beta-gal or the targeted Ig. This response is shown here to be significantly greater than that which is induced by DC that are loaded with these antigens via the conventional antigen pulse protocol. These results establish a novel, safe, and viable approach of enhancing the effectiveness of DC-based vaccination strategies for B cell lymphoma.
PubMedId: 16185929
Journal-article: Clinical immunology (Orlando, Fla.), vol. 118, 2005
Authors: L. Broderick, S. J. Yokota, J. Reineke, E. Mathiowitz, C. C. Stewart, M. Barcos, R. J. Kelleher, R. B. Bankert
Journal-article: The Journal of Immunology, vol. 174, pp. 898-906, 2005
Authors: Lori Broderick, Sandra J Yokota, Joshua Reineke, Edith Mathiowitz, Carleton C Stewart, Maurice Barcos, Raymond J Kelleher, Richard B Bankert
PubMedId: 15634912
Journal Article: Journal of immunology (Baltimore, Md. : 1950), vol. 174, 2005
Authors: Michael S Sabel, Stephen D Hess, Nejat K Egilmez, Thomas F Conway, Fang-An Chen, Richard B Bankert
Abstract: Previous studies by others using transplantable murine tumor models have demonstrated that the administration of antibodies that block CTLA-4 interaction with B7 can provoke the elimination of established tumors, and that the tumor suppression is mediated by T-cells and/or cells expressing NK1.1. Studies from our lab have established in a human/severe combined immunodeficient (SCID) mouse chimeric model that autologous peripheral blood leukocytes (PBL) can suppress the growth of tumor xenografts in a PBL dose-dependent fashion, and that this suppression is dependent upon the patient's T and NK cells. Using this human/mouse chimeric model, we sought to determine whether an antibody blockade of CTLA-4 would enhance the anti-tumor response of a patient's PBL. It was first important to determine whether the tumor suppression observed in the SCID model was dependent upon CD28/B7 co-stimulation. Blockade of B7 with a human CTLA-4-Ig fusion protein completely abrogated the lymphocyte-mediated tumor suppression, confirming in this model that tumor suppression is dependent upon a CD28/B7 co-stimulation. Using two different CTLA-4 specific monoclonal antibodies, we observed that CTLA-4 blockade significantly enhanced the human lymphocyte-mediated tumor suppression in mice co-engrafted with PBL and tumor cells. This enhancement was observed in both an allogeneic setting (in which the PBL were allogeneic with respect to the tumor) and an autologous setting (in which the PBL and tumor were from the same patient). These results sustain the notion that human anti-tumor immune response can be augmented (in vivo) by blocking the interaction between CTLA-4 and B7.
PubMedId: 15846492
Journal-article: Cancer immunology, immunotherapy : CII, vol. 54, 2004
Authors: Qiang Lou, Raymond J Kelleher, Alessandro Sette, Jenni Loyall, Scott Southwood, Richard B Bankert, Steven H Bernstein
Abstract: Previous studies have suggested that murine T cells are tolerant to epitopes derived from germ line variable regions of immunoglobulin (Ig) heavy (VH) or light chains. This has lead to the prediction that germ line VH-region epitopes found in neoplastic B cells cannot be used to provoke an antitumor immune response. To test these assumptions and address the question of how such a vaccine may alter the normal B-cell response, an antibody-forming B-cell hybridoma (1H6) expressing a conserved germ line VH gene with specificity for dextran was generated and used as a tumor model. Using algorithms for predicting major histocompatibility complex (MHC) binding, potential MHC class I and II binding peptides were identified within the 1H6 VH region, synthesized, and tested for MHC binding and immunogenicity. We show that germ line VH peptides, when presented by dendritic cells, are immunogenic in vitro and provoke a tumor-specific protective immune response in vivo. We conclude that (1) it is possible to induce a T-cell response to germ line VH peptides; (2) such peptides can be used to generate a B-cell tumor-specific vaccine; and (3) a vaccine targeting VH peptides expressed by the dominant dextran-specific B-cell clonotype had no effect upon the magnitude of the normal B-cell response to dextran.
PubMedId: 15054043
Journal-article: Blood, vol. 104, 2004
Authors: T M Anderson, S D Hess, N K Egilmez, C E Nwogu, J M Lenox, R B Bankert
Abstract: Leukemic cell growth in SCID mice has been reported as a predictor of disease relapse. However, there is a paucity of literature regarding xenograft growth and clinical outcomes in non-small cell lung cancer (NSCLC). Seventy-nine specimens from patients with NSCLC were either subcutaneously implanted into SCID mice and/or placed in tissue culture. Retrospective chart review was correlated with stage, histology, necrosis, disease-free interval, and survival. Tumor xenografts were successfully established with 17 of 37 (46%) tumor biopsy tissues. Thirteen of 59 (22%) specimens grew in cell culture. Patients whose tumors grew in SCID mice had no difference in survival compared to those with no growth ( n=20, p=0.42). Median survival was 36 months in 13 patients whose tumors grew in cell culture compared to 39 months in 46 patients without growth. Eight of 12 (67%) patients with metastasis showed SCID/human xenograft growth, whereas nine of 25 (36%) without metastases did so ( p=0.08). Growth of tumor cells in vitro occurred in 11 of 31 (35%) adenocarcinomas, one of 25 (4%) squamous cell carcinomas, and one of three (33%) large cell carcinomas ( p=0.02). Well or moderately differentiated tumors grew in cell culture in only two of 22 (9%), whereas poorly or undifferentiated tumors grew in 11 of 32 (34%) cases ( p=0.03). We conclude that neither the ability of a tumor to engraft and grow in SCID mice nor its ability to grow in vitro in cell culture is a reliable predictor of disease outcome or survival in patients with NSCLC. The ability to propagate tumors in vitro appears to be more dependent upon the histological type of tumor and its degree of differentiation.
PubMedId: 12923636
Journal-article: Journal of cancer research and clinical oncology, vol. 129, 2003
Authors: Nejat K E??ilmez, Richard B Bankert
Book-chapter: Liposomes, Part C, pp. 529-533, 2003
Authors: S. D. Hess, N. K. Egilmez, N. Bailey, T. M. Anderson, E. Mathiowitz, S. H. Bernstein, R. B. Bankert
Journal-article: The Journal of Immunology, vol. 170, pp. 400-412, 2003
Authors: R M Abra, R B Bankert, F Chen, N K Egilmez, K Huang, R Saville, J L Slater, M Sugano, S J Yokota
PubMedId: 12604032
Journal Article: Journal of liposome research, vol. 12, 2003
Authors: Stephen D Hess, Nejat K Egilmez, Nicola Bailey, Timothy M Anderson, Edith Mathiowitz, Steven H Bernstein, Richard B Bankert
PubMedId: 12496425
Journal Article: Journal of immunology (Baltimore, Md. : 1950), vol. 170, 2002
Authors: Nejat K Egilmez, Yong S Jong, Edith Mathiowitz, Richard B Bankert
PubMedId: 12407772
Journal Article: Methods in molecular medicine, vol. 75, 2002
Authors: Hank C Hill, Thomas F Conway, Michael S Sabel, Yong S Jong, Edith Mathiowitz, Richard B Bankert, Nejat K Egilmez
PubMedId: 12499267
Journal Article: Cancer research, vol. 62, 2002
Authors: Nejat K Egilmez, Stephen D Hess, Fang-An Chen, Hiroshi Takita, Thomas F Conway, Richard B Bankert
PubMedId: 11980657
Journal Article: Cancer research, vol. 62, 2002
Authors: Richard B Bankert
Journal-article: Frontiers in Bioscience, vol. 7, pp. c44-62, 2002
Authors: Richard B Bankert, Stephen D Hess, Nejat K Egilmez
PubMedId: 11915860
Journal Article: Frontiers in bioscience : a journal and virtual library, vol. 7, 2002
Authors: M S Sabel, H Hill, Y S Jong, E Mathiowitz, R B Bankert, N K Egilmez
Abstract: We previously demonstrated that the intratumoral injection of biodegradable polylactic acid microspheres that were loaded with interleukin (IL)-12 can induce a systemic antitumor immunity. We sought to investigate the clinical potential as neoadjuvant therapy. Mice were inoculated with 5 x 10(7) Line-1 cells subcutaneously. Six days later, a single intratumoral injection of IL-12- or BSA-loaded microspheres were given; 14 days later, autopsy was performed to document metastases. Mice were inoculated with 5 x 10(7) Line-1 cells and 10 days later either treated with IL-12- or BSA-loaded microspheres or resected. Treated tumors were resected 6 days after treatment. Mice were observed 45 days for local recurrence before autopsy. Intratumoral injection of IL-12 microspheres resulted in significant suppression of tumor growth compared with controls (599 +/- 255 mm(3) vs 1591 +/- 372 mm(3); P =.001) and pulmonary metastases (0.4 vs 3.8 nodules per mouse; P =.003). Given before the operation, IL-12-loaded microspheres both decreased the local recurrence rate (100% to 40%) and pulmonary metastases (5.2 vs 0.6 nodules per mouse; P =.06). Earlier resection did not improve local recurrence or distant metastases. Intratumoral injection of IL-12-loaded polylactic acid microspheres promotes the development of systemic antitumor immunity that can eradicate micrometastases. As a neoadjuvant therapy, this can result in decreased local and distant recurrence.
PubMedId: 11562672
Journal-article: Surgery, vol. 130, 2001
Authors: S D Hess, N K Egilmez, J Shiroko, R B Bankert
Abstract: Genes encoding the p35 and p40 subunits of human interleukin-12 (IL-12) and the bacterial aminoglycoside phosphotransferase were cloned into a mammalian expression plasmid. The resultant plasmid, pCMVIL-12neo, was used to transfect human lung tumor cell lines in vitro. Stably transfected subclones were generated and found to secrete human IL-12 for at least 10 days following a lethal dose of gamma-radiation. The ability of the IL-12--producing tumor cells to promote an antitumor response in vivo was evaluated in SCID mice co-engrafted subcutaneously with human peripheral blood lymphocytes (PBLs) and viable human lung tumor cells (SCID-Winn assay). Using this model system, it was established that IL-12 released locally into tumors by irradiated IL-12--transfected cells activated the human PBL and promoted their ability to suppress tumor development in a dose-dependent fashion. PBL subset depletion studies revealed that the antitumor effect promoted by the IL-12--modified cells was dependent on the presence of human CD8(+) T cells and, to a lesser extent, human CD56(+) natural killer cells within the xenograft. We conclude that (a) irradiated human lung tumor cells genetically modified with pCMVIL-12neo secrete bioactive human IL-12 at concentrations sufficient to promote a human lymphocyte-mediated antitumor response in the microenvironment of the xenograft, and (b) that the SCID-Winn assay provides a useful model for the preclinical evaluation of cytokine-based human immunotherapy protocols.
PubMedId: 11477457
Journal-article: Cancer gene therapy, vol. 8, 2001
Authors: Richard B Bankert, Nejat K Egilmez, Stephen D Hess
Journal-article: Trends in Immunology, vol. 22, pp. 386-393, 2001
Authors: Yasuyuki Sugiyama, Motohisa Kato, Fang-An Chen, Scott S. Williams, Yoshihiro Kawaguchi, Kiichi Miya, Yong S. Jong, Edith Mathiowitz, Nejat K. Egilmez, Richard B. Bankert
Journal-article: Journal of Immunotherapy, vol. 24, pp. 37-45, 2001
Authors: Makoto Yamada, Takashi Shiroko, Yoshihiro Kawaguchi, Yasuyuki Sugiyama, Nejat K. Egilmez, Fang-An Chen, Richard B. Bankert
Journal-article: International Journal of Cancer, vol. 92, pp. 589-599, 2001
Authors: M Sugano, T F Conway, R J Kelleher, Y Sugiyama, F A Chen, R B Bankert, S H Bernstein
PubMedId: 11484971
Journal Article: Journal of experimental & clinical cancer research : CR, vol. 20, 2001
Authors: M Yamada, T Shiroko, Y Kawaguchi, Y Sugiyama, N K Egilmez, F A Chen, R B Bankert
PubMedId: 11304696
Journal Article: International journal of cancer, vol. 92, 2001
Authors: Y Sugiyama, M Kato, F A Chen, S S Williams, Y Kawaguchi, K Miya, Y S Jong, E Mathiowitz, N K Egilmez, R B Bankert
PubMedId: 11211147
Journal Article: Journal of immunotherapy (Hagerstown, Md. : 1997), vol. 24, 2001
Authors: M Sugano, N K Egilmez, S J Yokota, F A Chen, J Harding, S K Huang, R B Bankert
PubMedId: 11156394
Journal Article: Cancer research, vol. 60, 2001
Authors: Michael S. Sabel, Thomas F. Conway, Fang-An Chen, Richard B. Bankert
Journal-article: Journal of Immunotherapy, vol. 23, pp. 362-368, 2000
Authors: Nejat K. Egilmez, Yong S. Jong, Stephen D. Hess, Jules S. Jacob, Edith Mathiowitz, Richard B. Bankert
Journal-article: Journal of Immunotherapy, vol. 23, pp. 190-195, 2000
Authors: M. Abraham Kuriakose, F-A Chen, Nejat K. Egilmez, Y.S. Jong, Edith Mathiowitz, Mark D. DeLacure, Wesley L. Hicks, Thom L. Loree, Richard B. Bankert
Journal-article: Head & Neck, vol. 22, pp. 57-63, 2000
Authors: M. Abraham Kuriakose, F???A Chen, Nejat K. Egilmez, Y.S. Jong, Edith Mathiowitz, Mark D. DeLacure, Wesley L. Hicks, Thom L. Loree, Richard B. Bankert
Journal-article: Head & Neck, vol. 22, pp. 57-63, 2000
Authors: Hank C Hill, Michael Sabel, Nejat Egilmez, Richard B Bankert
Journal-article: Journal of the American College of Surgeons, vol. 191, pp. S19-S20, 2000
Authors: Thomas F Conway, Michael S Sabel, Masahiko Sugano, John G Frelinger, Nejat K Egilmez, Fang-An Chen, Richard B Bankert
Journal-article: Journal of Immunological Methods, vol. 233, pp. 57-65, 2000
Authors: Michael S. Sabel, Makoto Yamada, Yoshihiro Kawaguchi, Fang-An Chen, Hiroshi Takita, Richard B. Bankert
Journal-article: Cancer Immunology, Immunotherapy, vol. 49, pp. 101-108, 2000
Authors: R. B. Bankert, F-A. Chen, Y. Sugiyama, N. Egilmez
Journal-article: Immunological Investigations, vol. 29, pp. 171-176, 2000
Authors: N K Egilmez, Y S Jong, M S Sabel, J S Jacob, E Mathiowitz, R B Bankert
PubMedId: 10919657
Journal Article: Cancer research, vol. 60, 2000
Authors: N K Egilmez, Y S Jong, S D Hess, J S Jacob, E Mathiowitz, R B Bankert
PubMedId: 10746545
Journal Article: Journal of immunotherapy (Hagerstown, Md. : 1997), vol. 23, 2000
Authors: T F Conway, M S Sabel, M Sugano, J G Frelinger, N K Egilmez, F Chen, R B Bankert
PubMedId: 10648856
Journal Article: Journal of immunological methods, vol. 233, 2000
Authors: M A Kuriakose, F A Chen, N K Egilmez, Y S Jong, E Mathiowitz, M D DeLacure, W L Hicks, T L Loree, R B Bankert
PubMedId: 10585606
Comparative Study: Head & neck, vol. 22, 1999
Authors: Y.S. Jong, N.K. Egilmez, F-A Chen, J.S. Jacob, L.P. Smith, T.S. Mottl, R.B. Bankert, E. Mathiowitz
Journal-article: MRS Proceedings, vol. 550, 1998
Authors: N. K. Egilmez, Yong S. Jong, Yoshimi Iwanuma, Jules S. Jacob, Camilla A. Santos, Fang-An Chen, Edith Mathiowitz, Richard B. Bankert
Journal-article: Cancer Immunology, Immunotherapy, vol. 46, pp. 21-24, 1998
Authors: N K Egilmez, Y S Jong, Y Iwanuma, J S Jacob, C A Santos, F A Chen, E Mathiowitz, R B Bankert
PubMedId: 9520288
Comparative Study: Cancer immunology, immunotherapy : CII, vol. 46, 1998
Authors: Y Iwanuma, F A Chen, N K Egilmez, H Takita, R B Bankert
PubMedId: 9230205
Journal Article: Cancer research, vol. 57, 1997
Authors: N K Egilmez, Y Iwanuma, R B Bankert
Abstract: Five commonly used cationic liposome formulations were tested for their ability to deliver DNA to established subcutaneous human tumor xenografts in SCID mice. Liposomes were complexed with a mammalian expression plasmid containing the bacterial beta-galactosidase gene and delivered to tumors by direct injection. The optimal lipid to DNA ratios in vivo were markedly different than those observed in vitro for each liposome formulation. Tumor size at the time of inoculation also effected transfection efficiency significantly. Of the five liposome formulations tested, DC-Cholesterol was found to be superior to all others in vivo. Even under optimal conditions however, the efficiency of in vivo transfection was low in our system (approximately 0.3%). Implications of these results for in vivo gene therapy of tumors are discussed.
PubMedId: 8660330
Journal-article: Biochemical and biophysical research communications, vol. 221, 1996
Authors: R.E. Cuenca, H. Takita, R. Bankert
Journal-article: Surgical Oncology, vol. 5, pp. 85-91, 1996
Authors: M. Diamond, S. S. Williams, H. Kida, S. Yokota, R. B. Bankert
Journal-article: Immunological Investigations, vol. 25, pp. 291-305, 1996
Authors: N K Egilmez, R Cuenca, S J Yokota, F Sorgi, R B Bankert
PubMedId: 8818648
Journal Article: Gene therapy, vol. 3, 1996
Authors: Harvey L. Bumpers, Thomas R. Alosco, Hui-Qun Wang, Nicholas J. Petrelli, Eddie L. Hoover, Richard B. Bankert
Journal-article: Journal of Surgical Research, vol. 61, pp. 282-288, 1996
Authors: Sabine Falkenberg, David Winter, Richard B. Bankert
Journal-article: Cellular Immunology, vol. 160, pp. 123-131, 1995
Authors: Fang-An Chen, Thomas Alosco, B. Anne Croy, K. Narumi, Dean H. Percy, Richard B. Bankert
Journal-article: Cell Adhesion and Communication, vol. 2, pp. 345-357, 1994
Authors: Thomas Alosco, Bernd Gansbacher, Richard Bankert, Hiroshi Takita, Nicholas Petrelli
Journal-article: Annals of Surgical Oncology, vol. 1, pp. 229-235, 1994
Authors: H L Bumpers, T R Alosco, H Q Wang, N J Petrelli, E L Hoover, R B Bankert
Journal-article: Journal of Clinical Investigation, vol. 94, pp. 2153-2157, 1994
Authors: Thomas Alosco, B. Anne Croy, Bernd Gansbacher, Hui-Qun Wang, Uma Rao, Richard Bankert
Journal-article: Cancer Immunology Immunotherapy, vol. 36, pp. 364-372, 1993
Authors: C. C. Gregorio, R. T. Kubo, R. B. Bankert, E. A. Repasky
Journal-article: Proceedings of the National Academy of Sciences, vol. 89, pp. 4947-4951, 1992
Authors: R. B. Bankert, T. Umemoto, Y. Sugiyama, F. A. Chen, E. Repasky, S. Yokota
Book-chapter: Current Topics in Microbiology and Immunology, pp. 201-210, 1989
Authors: John K. Lee, Jennifer D. Black, Elizabeth A. Repasky, Ralph T. Kubo, Richard B. Bankert
Journal-article: Cell, vol. 55, pp. 807-816, 1988
Authors: M. M. Abu-Hadid, R. B. Bankert, G. L. Mayers
Journal-article: Proceedings of the National Academy of Sciences, vol. 85, pp. 3990-3994, 1988
Authors: Christine S. Hughes, Elizabeth A. Repasky, Richard B. Bankert, Richard J. R. Johnson, John R. Subjeck
Journal-article: Radiation Research, vol. 112, pp. 116, 1987
Authors: M. M. Abu-hadid, R. B. Bankert, G. L. Mayers
Journal-article: Proceedings of the National Academy of Sciences, vol. 84, pp. 7232-7236, 1987
Authors: Sheng-Chang Luo, Richard B. Bankert
Book-chapter: H-2 Antigens, pp. 801-814, 1987
Authors: SHENG-CHANG LUO, RICHARD B. BANKERT
Journal-article: Hybridoma, vol. 4, pp. 319-327, 1985
Authors: SHIGETOYO SAJI, SAMUEL ZYLSTRA, BRIAN S. SCHEPART, SWAPAN K. GHOSH, YI-HER JOU, HIROSHI TAKITA, RICHARD B. BANKERT
Journal-article: Hybridoma, vol. 3, pp. 119-129, 1984
Authors: RICHARD B. BANKERT
Journal-article: Cancer Drug Delivery, vol. 1, pp. 251-267, 1984
Authors: Yi-Her Jou, Sheng-Chang Luo, Richard B. Bankert
Journal-article: Journal of Immunological Methods, vol. 65, pp. 285-292, 1983
Authors: Yi-Her Jou, Paul K. Mazzaferro, George L. Mayers, Richard B. Bankert
Book-chapter: Methods in Enzymology, pp. 257-276, 1983
Authors: Richard B. Bankert
Publication: Methods in Enzymology, pp. 182-195, 1983
Authors: R.B. Bankert, A.G. Bloor, Y-H. Jou
Journal-article: Veterinary Immunology and Immunopathology, vol. 3, pp. 147-184, 1982
Authors: Yi-Her Jou, Brian S. Schepart, Richard B. Bankert
Journal-article: Immunological Communications, vol. 11, pp. 357-375, 1982
Authors: Y. H. Jou, R. B. Bankert
Journal-article: Proceedings of the National Academy of Sciences, vol. 78, pp. 2493-2496, 1981
Authors: RICHARD B. BANKERT, DENISE MAZZAFERRO, GEORGE L. MAYERS
Journal-article: Hybridoma, vol. 1, pp. 47-58, 1981
Authors: Richard B. Bankert, George L. Mayers
Journal-article: Immunological Communications, vol. 10, pp. 457-481, 1981
Authors: Richard B. Bankert, Denise DesSoye, Laura Powers
Journal-article: Journal of Immunological Methods, vol. 35, pp. 23-32, 1980
Authors: David Pressman, Richard Bankert
Journal-article: Journal of Surgical Oncology, vol. 15, pp. 393-393, 1980
Authors: R.B. Bankert, A.K. Abbas
Book-chapter: Progress in Myeloma, pp. 109-128, 1980
Authors: R. B. Bankert, G. L. Mayers, D. Pressman
Journal-article: European Journal of Immunology, vol. 8, pp. 512-519, 1978
Authors: R.B. Bankert, B. Wolf, D. Pressman
Journal-article: Cellular Immunology, vol. 27, pp. 111-120, 1976
Authors: Nejat K. Egilmez, Yong S. Jong, Edith Mathiowitz, Richard B. Bankert
Book-chapter: Lung Cancer, pp. 687-696,
Presented by CUBS © 2024