Network


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

Hotspot


Dive into the research topics where Steve Burke is active.

Publication


Featured researches published by Steve Burke.


Blood | 2011

Application of dual affinity retargeting molecules to achieve optimal redirected T-cell killing of B-cell lymphoma.

Paul A. Moore; Wenjun Zhang; G. Jonah Rainey; Steve Burke; Hua Li; Ling Huang; Sergey Gorlatov; Maria Concetta Veri; Sudeepta Aggarwal; Yinhua Yang; Kalpana Shah; Linda Jin; Sunan Zhang; Leilei He; Tengfei Zhang; Valentina Ciccarone; Scott Koenig; Ezio Bonvini; Syd Johnson

We describe the application of a novel, bispecific antibody platform termed dual affinity retargeting (DART) to eradicate B-cell lymphoma through coengagement of the B cell-specific antigen CD19 and the TCR/CD3 complex on effector T cells. Comparison with a single-chain, bispecific antibody bearing identical CD19 and CD3 antibody Fv sequences revealed DART molecules to be more potent in directing B-cell lysis. The enhanced activity with the CD19xCD3 DART molecules was observed on all CD19-expressing target B cells evaluated using resting and prestimulated human PBMCs or purified effector T-cell populations. Characterization of a CD19xTCR bispecific DART molecule revealed equivalent potency with the CD19xCD3 DART molecule, demonstrating flexibility of the DART structure to support T-cell/B-cell associations for redirected T cell-killing applications. The enhanced level of killing mediated by DART molecules was not accompanied by any increase in nonspecific T-cell activation or lysis of CD19(-) cells. Cell-association studies indicated that the DART architecture is well suited for maintaining cell-to-cell contact, apparently contributing to the high level of target cell killing. Finally, the ability of the CD19xTCR DART to inhibit B-cell lymphoma in NOD/SCID mice when coadministered with human PBMCs supports further evaluation of DART molecules for the treatment of B-cell malignancies.


Immunology | 2007

Monoclonal antibodies capable of discriminating the human inhibitory Fcγ-receptor IIB (CD32B) from the activating Fcγ-receptor IIA (CD32A): biochemical, biological and functional characterization

Maria-Concetta Veri; Sergey Gorlatov; Hua Li; Steve Burke; Syd Johnson; Jeffrey B. Stavenhagen; Kathryn E. Stein; Ezio Bonvini; Scott Koenig

Human CD32B (FcγRIIB), the low‐affinity inhibitory Fcγ receptor (FcγR), is highly homologous in its extracellular domain to CD32A (FcγRIIA), an activating FcγR. Available monoclonal antibodies (mAb) against the extracellular region of CD32B recognize both receptors. Through immunization of mice transgenic for human CD32A, we generated a set of antibodies specific for the extracellular region of CD32B with no cross‐reactivity with CD32A, as determined by enzyme‐linked immunosorbent assay and surface plasmon resonance with recombinant CD32A and CD32B, and by fluorescence‐activated cell sorting analysis of CD32 transfectants. A high‐affinity mAb, 2B6, was used to explore the expression of CD32B by human peripheral blood leucocytes. While all B lymphocytes expressed CD32B, only a fraction of monocytes and almost no polymorphonuclear cells stained with 2B6. Likewise, natural killer cells, which express CD32C, a third CD32 variant, did not react with 2B6. Immune complexes co‐engage the inhibitory receptor with activating Fcγ receptors, a mechanism that limits cell responses. 2B6 competed for immune complex binding to CD32B as a monomeric Fab, suggesting that it directly recognizes the Fc‐binding region of the receptor. Furthermore, when co‐ligated with an activating receptor, 2B6 triggered CD32B‐mediated inhibitory signalling, resulting in diminished release of inflammatory mediators by FcεRI in an in vitro allergy model or decreased proliferation of human B cells induced by B‐cell receptor stimulation. These antibodies form the basis for the development of investigational tools and therapeutics with multiple potential applications, ranging from adjuvants in FcγR‐mediated responses to the treatment of allergy and autoimmunity.


Clinical Cancer Research | 2012

Development of an Fc-Enhanced Anti–B7-H3 Monoclonal Antibody with Potent Antitumor Activity

Deryk Loo; Ralph Alderson; Francine Chen; Ling Huang; Wenjun Zhang; Sergey Gorlatov; Steve Burke; Valentina Ciccarone; Hua Li; Yinhua Yang; Tom Son; Y Chen; Ann Easton; Jonathan C. Li; Jill Rillema; Monica Licea; Claudia Fieger; Tony W. Liang; Jennie P. Mather; Scott Koenig; Stanford J. Stewart; Syd Johnson; Ezio Bonvini; Paul A. Moore

Purpose: The goal of this research was to harness a monoclonal antibody (mAb) discovery platform to identify cell-surface antigens highly expressed on cancer and develop, through Fc optimization, potent mAb therapies toward these tumor-specific antigens. Experimental Design: Fifty independent mAbs targeting the cell-surface immunoregulatory B7-H3 protein were obtained through independent intact cell-based immunizations using human tissue progenitor cells, cancer cell lines, or cell lines displaying cancer stem cell properties. Binding studies revealed this natively reactive B7-H3 mAb panel to bind a range of independent B7-H3 epitopes. Immunohistochemical analyses showed that a subset displayed strong reactivity to a broad range of human cancers while exhibiting limited binding to normal human tissues. A B7-H3 mAb displaying exquisite tumor/normal differential binding was selected for humanization and incorporation of an Fc domain modified to enhance effector-mediated antitumor function via increased affinity for the activating receptor CD16A and decreased binding to the inhibitory receptor CD32B. Results: MGA271, the resulting engineered anti–B7-H3 mAb, mediates potent antibody-dependent cellular cytotoxicity against a broad range of tumor cell types. Furthermore, in human CD16A-bearing transgenic mice, MGA271 exhibited potent antitumor activity in B7-H3–expressing xenograft models of renal cell and bladder carcinoma. Toxicology studies carried out in cynomolgus monkeys revealed no significant test article-related safety findings. Conclusions: This data supports evaluation of MGA271 clinical utility in B7-H3–expressing cancer, while validating a combination of a nontarget biased approach of intact cell immunizations and immunohistochemistry to identify novel cancer antigens with Fc-based mAb engineering to enable potent antitumor activity. Clin Cancer Res; 18(14); 3834–45. ©2012 AACR.


Breast Cancer Research | 2011

Anti-tumor activity and toxicokinetics analysis of MGAH22, an anti-HER2 monoclonal antibody with enhanced Fcγ receptor binding properties

Jeffrey L. Nordstrom; Sergey Gorlatov; Wenjun Zhang; Yinhua Yang; Ling Huang; Steve Burke; Hua Li; Valentina Ciccarone; Tengfei Zhang; Jeffrey B. Stavenhagen; Scott Koenig; Stanford J Stewart; Paul A. Moore; Syd Johnson; Ezio Bonvini

IntroductionResponse to trastuzumab in metastatic breast cancer correlates with expression of the high binding variant (158V) of the activating Fcγ receptor IIIA (CD16A). We engineered MGAH22, a chimeric anti-HER2 monoclonal antibody with specificity and affinity similar to trastuzumab, with an Fc domain engineered for increased binding to both alleles of human CD16A.MethodsMGAH22 was compared to an identical anti-HER2 mAb except for a wild type Fc domain. Antibody-dependent cell cytotoxicity (ADCC) assays were performed with HER2-expressing cancer cells as targets and human PBMC or purified NK cells as effectors. Xenograft studies were conducted in mice with wild type murine FcγRs; in mice lacking murine CD16; or in mice lacking murine CD16 but transgenic for human CD16A-158F, the low-binding variant. The latter model reproduces the differential binding between wild type and the Fc-optimized mAb for human CD16A. The JIMT-1 human breast tumor line, derived from a patient that progressed on trastuzumab therapy, was used in these studies. Single and repeat dose toxicology studies with MGAH22 administered intravenously at high dose were conducted in cynomolgus monkeys.ResultsThe optimized Fc domain confers enhanced ADCC against all HER2-positive tumor cells tested, including cells resistant to trastuzumabs anti-proliferative activity or expressing low HER2 levels. The greatest improvement occurs with effector cells isolated from donors homozygous or heterozygous for CD16A-158F, the low-binding allele. MGAH22 demonstrates increased activity against HER2-expressing tumors in mice transgenic for human CD16A-158F. In single and repeat-dose toxicology studies in cynomolgus monkeys, a species with a HER2 expression pattern comparable to that in humans and Fcγ receptors that exhibit enhanced binding to the optimized Fc domain, MGAH22 was well tolerated at all doses tested (15-150 mg/kg) and exhibited pharmacokinetic parameters similar to that of other anti-HER2 antibodies. Induction of cytokine release by MGAH22 in vivo or in vitro was similar to that induced by the corresponding wild type mAb or trastuzumab.ConclusionsThe data support the clinical development of MGAH22, which may have utility in patients with low HER2 expressing tumors or carrying the CD16A low-binding allele.


Antibodies | 2016

Development of PF-06671008, a Highly Potent Anti-P-cadherin/Anti-CD3 Bispecific DART Molecule with Extended Half-Life for the Treatment of Cancer

Adam R. Root; Wei Cao; Bilian Li; Peter LaPan; Caryl Meade; Jocelyn Sanford; Macy Jin; Cliona O’Sullivan; Emma Cummins; Matthew Lambert; Alfredo Darmanin Sheehan; Weijun Ma; Scott Gatto; Kelvin M. Kerns; Khetemenee Lam; Aaron M. D’Antona; Lily Zhu; William A. Brady; Susan Benard; Amy King; Tao He; Lisa A. Racie; Maya Arai; Dianah Barrett; Wayne Stochaj; Edward R. Lavallie; James R. Apgar; Kristine Svenson; Lidia Mosyak; Yinhua Yang

Bispecific antibodies offer a promising approach for the treatment of cancer but can be challenging to engineer and manufacture. Here we report the development of PF-06671008, an extended-half-life dual-affinity re-targeting (DART®) bispecific molecule against P-cadherin and CD3 that demonstrates antibody-like properties. Using phage display, we identified anti-P-cadherin single chain Fv (scFv) that were subsequently affinity-optimized to picomolar affinity using stringent phage selection strategies, resulting in low picomolar potency in cytotoxic T lymphocyte (CTL) killing assays in the DART format. The crystal structure of this disulfide-constrained diabody shows that it forms a novel compact structure with the two antigen binding sites separated from each other by approximately 30 Å and facing approximately 90° apart. We show here that introduction of the human Fc domain in PF-06671008 has produced a molecule with an extended half-life (~4.4 days in human FcRn knock-in mice), high stability (Tm1 > 68 °C), high expression (>1 g/L), and robust purification properties (highly pure heterodimer), all with minimal impact on potency. Finally, we demonstrate in vivo anti-tumor efficacy in a human colorectal/human peripheral blood mononuclear cell (PBMC) co-mix xenograft mouse model. These results suggest PF-06671008 is a promising new bispecific for the treatment of patients with solid tumors expressing P-cadherin.


Clinical Cancer Research | 2017

MGD011, A CD19 x CD3 Dual-Affinity Retargeting Bi-specific Molecule Incorporating Extended Circulating Half-life for the Treatment of B-Cell Malignancies

Liqin Liu; Chia-Ying K. Lam; Vatana Long; Lusiana Widjaja; Yinhua Yang; Hua Li; Linda Jin; Steve Burke; Sergey Gorlatov; Jennifer R. Brown; Ralph Alderson; Margaret D. Lewis; Jeffrey L. Nordstrom; Scott Koenig; Paul A. Moore; Syd Johnson; Ezio Bonvini

Purpose: CD19, a B-cell lineage-specific marker, is highly represented in B-cell malignancies and an attractive target for therapeutic interventions. MGD011 is a CD19 x CD3 DART bispecific protein designed to redirect T lymphocytes to eliminate CD19-expressing cells. MGD011 has been engineered with a modified human Fc domain for improved pharmacokinetic (PK) properties and designed to cross-react with the corresponding antigens in cynomolgus monkeys. Here, we report on the preclinical activity, safety and PK properties of MGD011. Experimental Design: The activity of MGD011 was evaluated in several in vitro and in vivo models. PK, safety and pharmacodynamic activity was also assessed in dose-escalation and repeat-dose studies of MGD011 administered once weekly in cynomolgus monkeys. Results: MGD011 mediated killing of human B-cell lymphoma lines by human or cynomolgus monkey PBMCs as well as autologous B-cell depletion in PBMCs from both species. MGD011-mediated killing was accompanied by target-dependent T-cell activation and expansion, cytokine release and upregulation of perforin and granzyme B. MGD011 demonstrated antitumor activity against localized and disseminated lymphoma xenografts reconstituted with human PBMCs. In cynomolgus monkeys, MGD011 displayed a terminal half-life of 6.7 days; once weekly intravenous infusion of MGD011 at doses up to 100 μg/kg, the highest dose tested, was well tolerated and resulted in dose-dependent, durable decreases in circulating B cells accompanied by profound reductions of B lymphocytes in lymphoid organs. Conclusions: The preclinical activity, safety and PK profile support clinical investigation of MGD011 as a therapeutic candidate for the treatment of B-cell malignancies. Clin Cancer Res; 23(6); 1506–18. ©2016 AACR.


Cancer Research | 2014

Abstract 669: Development of MGD007, a gpA33 x CD3 bi-specific DART for T-cell immunotherapy of metastatic colorectal cancer

Paul A. Moore; Ralph Alderson; Kalpana Shah; Yinhua Yang; Steve Burke; Hua Li; Valentine Ciccarone; Ezio Bonvini; Syd Johnson

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Introduction: Encouraging clinical responses have been observed through various strategies designed to harness T-cells for their anti-tumor properties, including immune checkpoint inhibition, chimeric antigen receptor-expressing T-cells or bispecific molecules designed to co-engage T-cells and cancer cells. Furthermore, a positive correlation has been observed in colorectal cancer between the degree and type of T-cell infiltration and prognosis suggesting that T-cell recruitment strategies may be particularly advantageous in this type of cancer. We have developed MGD007, a Dual-Affinity Re-Targeting (DART®) protein designed to redirect T-cells to target gpA33 expressing colon cancer. MGD007 has enhanced pharmacokinetic properties via incorporation of a neonatal FcR-binding Fc domain. The gpA33 target was selected based on its ubiquitous expression in colorectal cancer including reactivity with putative cancer stem cell populations (Li 2013, AACR #3763). To enable preclinical toxicokinetics and dose optimization, MGD007 was designed to cross-react with non-human primates. Methods: MGD007 was stably expressed in CHO cells and purified to homogeneity via a standard antibody-purification platform; in vitro functional studies were performed with a range of colorectal cancer cell lines and primary human T-cells; tumor growth inhibition studies were performed in NOD-SCID mice co-implanted with Colo205 and human T-cells (1:1 E:T ratio) and treated IV with MGD007; pharmacokinetic analyses were performed in cynomolgus monkeys. Results: MGD007 displays the anticipated bispecific binding properties and mediates potent lysis of gpA33-positive - but not gpA33-negative cancer cell lines through recruitment of either human or cynomolgus monkey T-cells. Concomitant with CTL activity, both T-cell activation and expansion are observed in a gpA33-dependent manner. No cytokine activation was observed with human PBMC alone, consistent with the absence of gpA33 expression on peripheral blood cell populations. Both CD8 and CD4 T-cells mediated lysis of gpA33-expressing tumor cells, with activity accompanied by increases in granzyme levels. Xenograft studies showed tumor growth inhibition at doses as low as 4ug/kg. In cynomologus monkeys, 4 weekly doses of 200ug/kg were well tolerated, with prolonged PK consistent with that of an Fc-containing molecule. Conclusions: MGD007 displays potent activity against colorectal cancer cells consistent with a mechanism of action endowed in its design. Furthermore it displays favorable PK in cynomolgus monkeys supporting convenient dosing. Taken together, these data support further investigation of MGD007 as a potential novel therapeutic treatment for colorectal cancer. Citation Format: Paul A. Moore, Ralph Alderson, Kalpana Shah, Yinhua Yang, Steve Burke, Hua Li, Valentine Ciccarone, Ezio Bonvini, Syd Johnson. Development of MGD007, a gpA33 x CD3 bi-specific DART for T-cell immunotherapy of metastatic colorectal cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 669. doi:10.1158/1538-7445.AM2014-669


Molecular Cancer Therapeutics | 2018

Development of MGD007, a gpA33 x CD3 bispecific DART® protein for T-cell immunotherapy of metastatic colorectal cancer

Paul A. Moore; Kalpana Shah; Yinhua Yang; Ralph Alderson; Penny Roberts; Vatana Long; Daorong Liu; Jonathan Li; Steve Burke; Valentina Ciccarone; Hua Li; Claudia Fieger; Jeff Hooley; Ann Easton; Monica Licea; Sergey Gorlatov; Kathleen L. King; Peter Young; Arash Adami; Deryk Loo; Gurunadh Chichili; Liqin Liu; Douglas H. Smith; Jennifer G. Brown; Francine Chen; Scott Koenig; Jennie P. Mather; Ezio Bonvini; Syd Johnson

We have developed MGD007 (anti-glycoprotein A33 x anti-CD3), a DART protein designed to redirect T cells to target gpA33 expressing colon cancer. The gpA33 target was selected on the basis of an antibody-based screen to identify cancer antigens universally expressed in both primary and metastatic colorectal cancer specimens, including putative cancer stem cell populations. MGD007 displays the anticipated-bispecific binding properties and mediates potent lysis of gpA33-positive cancer cell lines, including models of colorectal cancer stem cells, through recruitment of T cells. Xenograft studies showed tumor growth inhibition at doses as low as 4 μg/kg. Both CD8 and CD4 T cells mediated lysis of gpA33-expressing tumor cells, with activity accompanied by increases in granzyme and perforin. Notably, suppressive T-cell populations could also be leveraged to mediate lysis of gpA33-expressing tumor cells. Concomitant with CTL activity, both T-cell activation and expansion are observed in a gpA33-dependent manner. No cytokine activation was observed with human PBMC alone, consistent with the absence of gpA33 expression on peripheral blood cell populations. Following prolonged exposure to MGD007 and gpA33 positive tumor cells, T cells express PD-1 and LAG-3 and acquire a memory phenotype but retain ability to support potent cell killing. In cynomolgus monkeys, 4 weekly doses of 100 μg/kg were well tolerated, with prolonged PK consistent with that of an Fc-containing molecule. Taken together, MGD007 displays potent activity against colorectal cancer cells consistent with a mechanism of action endowed in its design and support further investigation of MGD007 as a potential novel therapeutic treatment for colorectal cancer. Mol Cancer Ther; 17(8); 1761–72. ©2018 AACR.


Cancer Research | 2010

Abstract 5629: Application of a novel bispecific antibody-based scaffold for optimal redirected T-cell killing of cancer cells

Paul A. Moore; Wenjun Zhang; Jonah Rainey; Steve Burke; Ling Huang; Hua Li; Maria-Concetta Veri; Yinhua Yang; Val Ciccarone; Ralph Alderson; Scott Koenig; Ezio Bonvini; Syd Johnson

Despite significant progress, improved treatment modalities are required to eradicate cancer. Cytotoxic T lymphocytes are among the most potent cell populations capable of killing target cells and considerable effort has been dedicated to developing strategies aimed at redirecting T cells toward cancer, including the generation of bispecific molecules capable of co-engaging a tumor target together with an activating receptor on T lymphocytes. Limitations of the existing technology, however, including shortcomings in manufacturability, stability, specificity and potency, have often hampered progress in harnessing the power of T cells with bi-specific molecules. We have applied a novel antibody-based scaffold, termed DART (Dual-Affinity Re-Targeting), to redirect T-cell killing toward tumor cells in the absence of cognate interactions. DART molecules are structurally comprised of a covalently linked “diabody” generated through co-expression in mammalian cells of two separate Fv-encoding chains. To evaluate the ability of the platform to redirect T-cell killing, DARTs were designed to target hematological malignancies by comprising a B-cell lineage specificity (CD19) together with T-cell recognition (CD3 or TCR), while a DART comprising specificities for the HER2 target and the TCR was used to evaluate applicability to solid tumors. All three DART molecules assembled efficiently and were evaluated in a series of redirected T-cell killing assays to evaluate potency and mechanistic aspects of cytotoxicity. Both the CD19xCD3 and CD19xTCR DART molecules demonstrated efficient co-ligation of B-cell lymphoma cells with human T cells and mediated equivalently potent redirected T-cell killing against a panel of CD19-positive B-cell lymphoma cell lines but not CD19-ve cell lines. In similar fashion, the TCRxHER2 DART revealed potent T-cell mediated killing of a panel of HER2 expressing cell lines, including those expressing low HER2 levels (1+ by HercepTest), but was inactive against HER2-negative lines. Analyses of the mechanism of T-cell mediated killing by the DART molecules revealed a dependence on the granzyme B/perforin pathways, with potent killing observed during short-term incubation of the effector populations with target cells and at low E:T ratios. Furthermore, activation of human T cells by DART molecules proceeded strictly in a target-dependent manner. Finally, analyses in xenograft models reconstituted with human peripheral blood mononuclear cells, demonstrated efficient tumor regression by TCR-based DART molecules. In conclusion, T-cell-directed DARTs represent a robust and flexible platform to recruit T-cell populations for cancer cell killing and may offer potential application toward tumor eradication in cancer treatment. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 5629.


Cancer Research | 2016

Abstract 583: Evaluation of EphA2 as a therapeutic target for redirected T-cell killing by DART® bispecific molecules

Claudia Fieger; Kalpana Shah; Gurunadh Chichili; Jonathan J. Li; Thomas Son; Jeffrey Hooley; Francine Chen; Sergey Gorlatov; Steve Burke; Valentina Ciccarone; Ralph Alderson; Deryk Loo; Syd Johnson; Ezio Bonvini; Paul A. Moore

Introduction: EphA2 is a receptor tyrosine kinase that plays a critical role in cancer progression through both ligand-dependent and independent mechanisms. The broad overexpression in tumors but limited normal tissue expression of EphA2 makes it an attractive therapeutic target amenable for redirected T-cell killing via an EphA2 x CD3 Dual-Affinity Re-Targeting (DART®) molecules designed to co-engage cytotoxic T cells with EphA2 expressing tumor cells. Methods: Anti-EphA2 monoclonal antibodies (mAbs) were identified by target-specific screening of a library generated by whole-cell immunization with proprietary cancer cell lines, including models of cancer stem cells. The binding and signaling properties of the antibodies were characterized by ELISA, SPR, flow cytometry and phosphorylation assays. Receptor binding regions were determined by ELISA based competition assays and by utilizing human-mouse chimeric EphA2 molecules. Immunohistochemistry (IHC) was performed on frozen normal and tumor tissues. In vitro functional studies were performed with various cancer as well as transfected cell lines, and primary human T cells or PBMCs. In vivo activity was evaluated in xenograft models in immune deficient mice. Results: EphA2 mAbs encompassing diversity in binding kinetics and effects on receptor phosphorylation were classified in 5 discrete binding groups. The majority interacted with the N-terminal ligand-binding domain of EphA2 and most mAbs within that group interfered with ligand binding. The majority of mAbs displayed little IHC reactivity with normal tissue, while strong staining of cancer tissues was observed, including colon, lung, pancreas, ovary, bladder and breast cancers. Seven mAbs recognizing independent epitopes were selected for conversion into EphA2 x CD3 DART molecules that showed a range of potency in redirecting T cells to kill EphA2-expressing target cells. A lead EphA2 x CD3 DART molecule was selected based on potency and cross-reactivity with the cynomolgus monkey ortholog; this lead was engineered with a human Fc domain to confer an extended circulating half-life. The resulting Fc-bearing EphA2 x CD3 DART molecule demonstrated in vivo anti-tumor activity at doses as low as 20 mcg/kg in NOD/SCID/IL-2 gamma chain null mice co-implanted with activated human T cells and MDA-MB231 breast cancer cells. Conclusion: EphA2 is a potential cancer target for redirected T-cell killing applications independent of ligand mediated mechanisms. Further preclinical assessment of EphA2 x CD3 DART molecules as a strategy for targeting EphA2-expressing malignancies is warranted. Citation Format: Claudia B. Fieger, Kalpana Shah, Gurunadh Chichili, Jonathan Li, Thomas Son, Jeffrey Hooley, Francine Chen, Sergey Gorlatov, Steve Burke, Valentina Ciccarone, Ralph Alderson, Deryk Loo, Syd Johnson, Ezio Bonvini, Paul Moore. Evaluation of EphA2 as a therapeutic target for redirected T-cell killing by DART® bispecific molecules. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 583.

Collaboration


Dive into the Steve Burke's collaboration.

Top Co-Authors

Avatar

Ezio Bonvini

Food and Drug Administration

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Scott Koenig

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Valentina Ciccarone

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ralph Alderson

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge