Network


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

Hotspot


Dive into the research topics where Lily Shao is active.

Publication


Featured researches published by Lily Shao.


Science Translational Medicine | 2013

Blocking NRG1 and Other Ligand-Mediated Her4 Signaling Enhances the Magnitude and Duration of the Chemotherapeutic Response of Non–Small Cell Lung Cancer

Ganapati V. Hegde; Cecile de la Cruz; Cecilia Chiu; Navneet Alag; Gabriele Schaefer; Lisa Crocker; Sarajane Ross; David M. Goldenberg; Mark Merchant; Janet Tien; Lily Shao; Leslie Roth; Siao-Ping Tsai; Scott Stawicki; Zhaoyu Jin; Shelby K. Wyatt; Richard A. D. Carano; Yanyan Zheng; E. Alejandro Sweet-Cordero; Yan Wu; Erica Jackson

Inhibition of Her4 signaling enhances the response to chemotherapy and delays tumor regrowth after cessation of treatment. Regaining the Yellow Jersey Professional sports—from cycling to football and even baseball—are now cracking down on doping. The use of performance-enhancing drugs is thought to give an unfair advantage, and regulatory agencies are trying to return everyone to even ground. But whereas athletes who dope become pariahs, in some fights it’s better not to play fair. Now, Hegde et al. suggest a way to enhance chemotherapy in the fight against non–small cell lung cancer (NSCLC). Chemotherapy is a first-line treatment for NSCLC but, in some cases, cannot either adequately remove the tumor or prevent recurrence. The authors use multiple models of NSCLC and find that residual tumor cells after chemotherapy express high levels of neuregulin 1 (NRG1), which is a ligand for human epidermal growth factor receptor 3 and 4 (HER3/4). Inhibited NRG1 signaling had only variable effects on primary tumor growth, but significantly enhanced the magnitude and duration of tumor response to chemotherapy. NRG1 inhibition in combination with chemotherapy greatly impeded relapse. Although this combination remains to be tested in the clinic, this study suggests that when it comes to a competition between NSCLC and chemotherapy, all’s fair. Although standard chemotherapies are commonly used to treat most types of solid tumors, such treatment often results in inadequate response to, or relapse after, therapy. This is particularly relevant for lung cancer because most patients are diagnosed with advanced-stage disease and are treated with frontline chemotherapy. By studying the residual tumor cells that remain after chemotherapy in several in vivo non–small cell lung cancer models, we found that these cells have increased levels of human epidermal growth factor receptor (HER) signaling due, in part, to the enrichment of a preexisting NRG1HI subpopulation. Neuregulin 1 (NRG1) signaling in these models can be mediated by either the HER3 or HER4 receptor, resulting in the differential activation of downstream effectors. Inhibition of NRG1 signaling inhibits primary tumor growth and enhances the magnitude and duration of the response to chemotherapy. Moreover, we show that inhibition of ligand-mediated Her4 signaling impedes disease relapse in cases where NRG1 inhibition is insufficient. These findings demonstrate that ligand-dependent Her4 signaling plays an important role in disease relapse.


Journal of Molecular Biology | 2010

Structure of the complex between HER2 and an antibody paratope formed by side chains from tryptophan and serine.

Robert D. Fisher; Mark Ultsch; Andreas Lingel; Gabriele Schaefer; Lily Shao; Sara C. Birtalan; Sachdev S. Sidhu; Charles Eigenbrot

Engineered antibody paratopes with limited sequence diversity permit assessment of the roles played by different amino acid side chains in creating the high-affinity, high-specificity interactions characteristic of antibodies. We describe a paratope raised against the human ErbB family member HER2, using a binary diversity tryptophan/serine library displayed on phage. Fab37 binds to the extracellular domain of HER2 with sub-nanomolar affinity. An X-ray structure at 3.2 A resolution reveals a contact paratope composed almost entirely of tryptophan and serine residues. Mutagenesis experiments reveal which of these side chains are more important for direct antigen interactions and which are more important for conformational flexibility. The crystal lattice contains an unprecedented trimeric arrangement of HER2 closely related to previously observed homodimers of the related epidermal growth factor receptor.


PLOS ONE | 2012

Reorienting the Fab domains of trastuzumab results in potent HER2 activators.

Justin Scheer; Wendy Sandoval; J. Michael Elliott; Lily Shao; Elizabeth Luis; Sock-Cheng Lewin-Koh; Gabriele Schaefer; Richard Vandlen

The structure of the Fab region of antibodies is critical to their function. By introducing single cysteine substitutions into various positions of the heavy and light chains of the Fab region of trastuzumab, a potent antagonist of HER2, and using thiol chemistry to link the different Fabs together, we produced a variety of monospecific F(ab′)2-like molecules with activities spanning from activation to inhibition of breast tumor cell growth. These isomers (or bis-Fabs) of trastuzumab, with varying relative spatial arrangements between the Fv-regions, were able to either promote or inhibit cell-signaling activities through the PI3K/AKT and MAPK pathways. A quantitative phosphorylation mapping of HER2 indicated that the agonistic isomers produced a distinct phosphorylation pattern associated with activation. This study suggests that antibody geometric isomers, found both in nature and during synthetic antibody development, can have profoundly different biological activities independent of their affinities for their target molecules.


Journal of Medicinal Chemistry | 2015

Noncovalent Mutant Selective Epidermal Growth Factor Receptor Inhibitors: A Lead Optimization Case Study.

Robert Heald; Krista K. Bowman; Marian C. Bryan; Daniel J. Burdick; Bryan K. Chan; Emily Chan; Yuan Chen; Saundra Clausen; Belen Dominguez-Fernandez; Charles Eigenbrot; Richard L. Elliott; Emily J. Hanan; Philip Stephen Jackson; Hank La; Michael Lainchbury; Shiva Malek; Sam Mann; Mark Merchant; Kyle Mortara; Hans E. Purkey; Gabriele Schaefer; Stephen Schmidt; Eileen Mary Seward; Steve Sideris; Lily Shao; Shumei Wang; Kuen Yeap; Ivana Yen; Christine Yu; Timothy P. Heffron

Because of their increased activity against activating mutants, first-generation epidermal growth factor receptor (EGFR) kinase inhibitors have had remarkable success in treating non-small-cell lung cancer (NSCLC) patients, but acquired resistance, through a secondary mutation of the gatekeeper residue, means that clinical responses only last for 8-14 months. Addressing this unmet medical need requires agents that can target both of the most common double mutants: T790M/L858R (TMLR) and T790M/del(746-750) (TMdel). Herein we describe how a noncovalent double mutant selective lead compound was optimized using a strategy focused on the structure-guided increase in potency without added lipophilicity or reduction of three-dimensional character. Following successive rounds of design and synthesis it was discovered that cis-fluoro substitution on 4-hydroxy- and 4-methoxypiperidinyl groups provided synergistic, substantial, and specific potency gain through direct interaction with the enzyme and/or effects on the proximal ligand oxygen atom. Further development of the fluorohydroxypiperidine series resulted in the identification of a pair of diastereomers that showed 50-fold enzyme and cell based selectivity for T790M mutants over wild-type EGFR (wtEGFR) in vitro and pathway knock-down in an in vivo xenograft model.


Nature Communications | 2016

High cell-surface density of HER2 deforms cell membranes.

Inhee Chung; Mike Reichelt; Lily Shao; Robert W. Akita; Hartmut Koeppen; Linda Rangell; Gabriele Schaefer; Ira Mellman; Mark X. Sliwkowski

Breast cancers (BC) with HER2 overexpression (referred to as HER2 positive) progress more aggressively than those with normal expression. Targeted therapies against HER2 can successfully delay the progression of HER2-positive BC, but details of how this overexpression drives the disease are not fully understood. Using single-molecule biophysical approaches, we discovered a new effect of HER2 overexpression on disease-relevant cell biological changes in these BC. We found HER2 overexpression causes deformation of the cell membranes, and this in turn disrupts epithelial features by perturbing cell–substrate and cell–cell contacts. This membrane deformation does not require receptor signalling activities, but results from the high levels of HER2 on the cell surface. Our finding suggests that early-stage morphological alterations of HER2-positive BC cells during cancer progression can occur in a physical and signalling-independent manner.


PLOS ONE | 2017

Combined MEK and ERK inhibition overcomes therapy-mediated pathway reactivation in RAS mutant tumors

Mark Merchant; John Moffat; Gabriele Schaefer; Jocelyn Chan; Xi Wang; Christine Orr; Jason H. Cheng; Thomas Hunsaker; Lily Shao; Stephanie J. Wang; Marie-Claire Wagle; Eva Lin; Peter M. Haverty; Sheerin Shahidi-Latham; Hai Ngu; Margaret Solon; Jeffrey Eastham-Anderson; Hartmut Koeppen; Shih-Min A. Huang; Jacob B. Schwarz; Marcia Belvin; Daniel C. Kirouac; Melissa R. Junttila

Mitogen-activated protein kinase (MAPK) pathway dysregulation is implicated in >30% of all cancers, rationalizing the development of RAF, MEK and ERK inhibitors. While BRAF and MEK inhibitors improve BRAF mutant melanoma patient outcomes, these inhibitors had limited success in other MAPK dysregulated tumors, with insufficient pathway suppression and likely pathway reactivation. In this study we show that inhibition of either MEK or ERK alone only transiently inhibits the MAPK pathway due to feedback reactivation. Simultaneous targeting of both MEK and ERK nodes results in deeper and more durable suppression of MAPK signaling that is not achievable with any dose of single agent, in tumors where feedback reactivation occurs. Strikingly, combined MEK and ERK inhibition is synergistic in RAS mutant models but only additive in BRAF mutant models where the RAF complex is dissociated from RAS and thus feedback productivity is disabled. We discovered that pathway reactivation in RAS mutant models occurs at the level of CRAF with combination treatment resulting in a markedly more active pool of CRAF. However, distinct from single node targeting, combining MEK and ERK inhibitor treatment effectively blocks the downstream signaling as assessed by transcriptional signatures and phospho-p90RSK. Importantly, these data reveal that MAPK pathway inhibitors whose activity is attenuated due to feedback reactivation can be rescued with sufficient inhibition by using a combination of MEK and ERK inhibitors. The MEK and ERK combination significantly suppresses MAPK pathway output and tumor growth in vivo to a greater extent than the maximum tolerated doses of single agents, and results in improved anti-tumor activity in multiple xenografts as well as in two Kras mutant genetically engineered mouse (GEM) models. Collectively, these data demonstrate that combined MEK and ERK inhibition is functionally unique, yielding greater than additive anti-tumor effects and elucidates a highly effective combination strategy in MAPK-dependent cancer, such as KRAS mutant tumors.


Journal of Medicinal Chemistry | 2016

Correction to Noncovalent Mutant Selective Epidermal Growth Factor Receptor Inhibitors: A Lead Optimization Case Study

Robert Heald; Krista K. Bowman; Marian C. Bryan; Daniel J. Burdick; Bryan K. Chan; Emily Chan; Yuan Chen; Saundra Clausen; Belen Dominguez-Fernandez; Charles Eigenbrot; Richard L. Elliott; Emily J. Hanan; Philip Stephen Jackson; Hank La; Michael Lainchbury; Shiva Malek; Sam Mann; Mark Merchant; Kyle Mortara; Hans E. Purkey; Gabriele Schaefer; Stephen Schmidt; Eileen Mary Seward; Steve Sideris; Lily Shao; Shumei Wang; Kuen Yeap; Ivana Yen; Christine Yu; Timothy P. Heffron

Robert Heald,* Krista K. Bowman, Marian C. Bryan, Daniel Burdick, Bryan Chan, Emily Chan, Yuan Chen, Saundra Clausen, Belen Dominguez-Fernandez, Charles Eigenbrot, Richard Elliott, Emily J. Hanan, Philip Jackson, Jamie Knight, Hank La, Michael Lainchbury, Shiva Malek, Sam Mann, Mark Merchant, Kyle Mortara, Hans Purkey, Gabriele Schaefer, Stephen Schmidt, Eileen Seward, Steve Sideris, Lily Shao, Shumei Wang, Kuen Yeap, Ivana Yen, Christine Yu, and Timothy P. Heffron


Cancer Research | 2014

Abstract 1735: Discovery of novel and selective reversible inhibitors of EGFR containing the T790M drug resistance mutation with activity in vitro and in vivo

Gabriele Schaefer; Emily Hanan; Emily Chan; Lily Shao; Yuan Chen; Robert L. Yauch; Stephen Schmidt; Steven Sideris; Shiva Malek; Timothy P. Heffron

Activating mutations in the epidermal growth factor receptor (EGFR) characterize a subset of non-small cell lung cancers (NSCLC) with exquisite sensitivity to tyrosine kinase inhibitors like erlotinib or gefitinib. However, after an initial dramatic response, patients harboring these mutations progress within 8-14 months on therapy. In approximately 60% of patients, the resistance to TKIs is associated with the acquisition of a secondary mutation within the EGFR kinase domain. The mutation in the kinase domain leads to the substitution of a bulky methionine for threonine at position 790 (T790M) also referred to as the gatekeeper residue. Using structure based-design we identified and optimized a series of non-covalent, and highly selective pyridinyl-aminopyrimidine inhibitors of EGFR containing the T790M drug resistant mutation that spared the wild-type EGFR enzyme. These pyridinyl-aminopyrimidine compounds potently inhibited phosphorylation of T790M containing EGFR in enzymatic assays and cell based assays. Strong inhibition of enzyme activity resulted in anti-proliferative activity in H1975 and PC9 erlotinib resistant cells but treatment with these molecules did not affect cell lines containing wild-type EGFR. Moreover, oral dosing of mice bearing either H1975 or PC9_erlotinib resistant xenograft tumors resulted in robust inhibition of tumor growth that was accompanied by profound and dose-dependent inhibition of EGFR activity as well as downstream signaling molecules such as ERK1/2 and AKT. These results suggest that these pyridinyl-aminopyrimidine inhibitors have the potential to benefit NSCLC patients who have developed resistance to first-generation EGFR inhibitors due to the acquisition of the T790M mutation, without the skin rash and gastrointestinal toxicity associated with wild-type EGFR inhibition. *EJH and GS contributed equally to this work. Citation Format: Gabriele Schaefer, Emily J. Hanan, Emily Chan, Lily Shao, Yuan Chen, Jamie Knight, Robert L. Yauch, Stephen Schmidt, Steven Sideris, Shiva Malek, Timothy P. Heffron. Discovery of novel and selective reversible inhibitors of EGFR containing the T790M drug resistance mutation with activity in vitro and in vivo. [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 1735. doi:10.1158/1538-7445.AM2014-1735


Cancer Research | 2012

Abstract 1212: The dual action antibody MEHD7945A targeting EGFR and HER3 enhances chemotherapy induced cytotoxicity in vitro and in vivo

Lisa Crocker; Carter Fields; Lily Shao; Mark X. Sliwkowski; Gail Lewis Phillips; Gabriele Schaefer

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Dysregulation of the epidermal growth factor receptor family (EGFR, HER2, HER3, HER4) by mutation and/or overexpression plays an important role in tumorigenesis, and targeted agents directed against two members of the HER/ErbB family, epidermal growth factor receptor (EGFR/HER1), and HER2/ErbB2, are used in the treatment of cancer. Extensive crosstalk seen among these receptors implies that blocking signaling of more than one receptor may be more effective in inhibiting tumor growth and circumventing resistance mechanisms than targeting individual receptors. In particular, HER3 is considered a key mediator of resistance to many targeted agents. We generated a “two-in-one” antibody, MEHD7945A, that binds to EGFR and HER3 with high affinity, inhibits receptor function and is more broadly efficacious in various tumor types when compared to monospecific anti-EGFR or anti-HER3 antibodies. Given the ability of HER3 to potently activate the PI3K survival pathway, we investigated if antagonizing ligand-dependent HER signaling with MEHD7945A in the presence of chemotherapy augments cytotoxicity. We calculated combination index values the effects of combining MEHD7945A with commonly used chemotherapeutic agents in NSCLC and colorectal cell lines in vitro and in vivo. The NSCLC lines NCI-H292, NCI-H1666, NCI-H358 and HCC827 were treated with MEHD7945A plus gemcitabine over a wide range of drug concentrations. Cell proliferation data were analyzed using CalcuSyn software and all combination index values were <1, demonstrating that the combination of MEHD7945A and gemcitabine inhibited proliferation synergistically in these cells. To further explore the enhanced cytotoxic effect we evaluated the combination of MEHD7945A and gemcitabine, versus each single agent, in the NCI-H1975 NSCLC xenograft model, and observed enhanced tumor regression. Similar results were observed when MEHD7945A was combined with a range of chemotherapeutic agents, including pemetrexed, docetaxel and irinotecan in H1975, H441 or SW948 xenograft models. In summary, these in vitro and in vivo results demonstrate that MEHD7945A potentiates various cytotoxic agents in a variety of tumor types. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 1212. doi:1538-7445.AM2012-1212


Cancer Cell | 2011

A Two-in-One Antibody against HER3 and EGFR Has Superior Inhibitory Activity Compared with Monospecific Antibodies

Gabriele Schaefer; Lauric Haber; Lisa Crocker; Steven Shia; Lily Shao; Donald Dowbenko; Klara Totpal; Anne Wong; Chingwei V. Lee; Scott Stawicki; Robyn Clark; Carter Fields; Gail Lewis Phillips; Rodney A. Prell; Dimitry M. Danilenko; Yvonne Franke; Jean-Philippe Stephan; Jiyoung Hwang; Yan Wu; Jenny Bostrom; Mark X. Sliwkowski; Germaine Fuh; Charles Eigenbrot

Collaboration


Dive into the Lily Shao's collaboration.

Researchain Logo
Decentralizing Knowledge