Network


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

Hotspot


Dive into the research topics where Grit S. Herter-Sprie is active.

Publication


Featured researches published by Grit S. Herter-Sprie.


Nature Communications | 2016

Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints

Shohei Koyama; Esra A. Akbay; Yvonne Y. Li; Grit S. Herter-Sprie; Kevin A. Buczkowski; William G. Richards; Leena Gandhi; Amanda J. Redig; Scott J. Rodig; Hajime Asahina; Robert E. Jones; Meghana M. Kulkarni; Mari Kuraguchi; Sangeetha Palakurthi; Peter E. Fecci; Bruce E. Johnson; Pasi A. Jänne; Jeffrey A. Engelman; Sidharta P. Gangadharan; Daniel B. Costa; Gordon J. Freeman; Raphael Bueno; F. Stephen Hodi; Glenn Dranoff; Kwok-Kin Wong; Peter S. Hammerman

Despite compelling antitumour activity of antibodies targeting the programmed death 1 (PD-1): programmed death ligand 1 (PD-L1) immune checkpoint in lung cancer, resistance to these therapies has increasingly been observed. In this study, to elucidate mechanisms of adaptive resistance, we analyse the tumour immune microenvironment in the context of anti-PD-1 therapy in two fully immunocompetent mouse models of lung adenocarcinoma. In tumours progressing following response to anti-PD-1 therapy, we observe upregulation of alternative immune checkpoints, notably T-cell immunoglobulin mucin-3 (TIM-3), in PD-1 antibody bound T cells and demonstrate a survival advantage with addition of a TIM-3 blocking antibody following failure of PD-1 blockade. Two patients who developed adaptive resistance to anti-PD-1 treatment also show a similar TIM-3 upregulation in blocking antibody-bound T cells at treatment failure. These data suggest that upregulation of TIM-3 and other immune checkpoints may be targetable biomarkers associated with adaptive resistance to PD-1 blockade.


Cancer Cell | 2014

Loss of Lkb1 and Pten Leads to Lung Squamous Cell Carcinoma with Elevated PD-L1 Expression

Chunxiao Xu; Christine M. Fillmore; Shohei Koyama; Hongbo Wu; Yanqiu Zhao; Zhao Chen; Grit S. Herter-Sprie; Esra A. Akbay; Jeremy H. Tchaicha; Abigail Altabef; Jacob B. Reibel; Zandra E. Walton; Hongbin Ji; Hideo Watanabe; Pasi A. Jänne; Diego H. Castrillon; Anil K. Rustgi; Adam J. Bass; Gordon J. Freeman; Robert F. Padera; Glenn Dranoff; Peter S. Hammerman; Carla F. Kim; Kwok-Kin Wong

Lung squamous cell carcinoma (SCC) is a deadly disease for which current treatments are inadequate. We demonstrate that biallelic inactivation of Lkb1 and Pten in the mouse lung leads to SCC that recapitulates the histology, gene expression, and microenvironment found in human disease. Lkb1;Pten null (LP) tumors expressed the squamous markers KRT5, p63 and SOX2, and transcriptionally resembled the basal subtype of human SCC. In contrast to mouse adenocarcinomas, the LP tumors contained immune populations enriched for tumor-associated neutrophils. SCA1(+)NGFR(+) fractions were enriched for tumor-propagating cells (TPCs) that could serially transplant the disease in orthotopic assays. TPCs in the LP model and NGFR(+) cells in human SCCs highly expressed Pd-ligand-1 (PD-L1), suggesting a mechanism of immune escape for TPCs.


Cancer Research | 2016

STK11/LKB1 Deficiency Promotes Neutrophil Recruitment and Proinflammatory Cytokine Production to Suppress T-cell Activity in the Lung Tumor Microenvironment

Shohei Koyama; Esra A. Akbay; Yvonne Y. Li; Amir R. Aref; Ferdinandos Skoulidis; Grit S. Herter-Sprie; Kevin A. Buczkowski; Yan Liu; Mark M. Awad; Warren Denning; Lixia Diao; Jing Wang; Edwin R. Parra-Cuentas; Ignacio I. Wistuba; Margaret Soucheray; Tran C. Thai; Hajime Asahina; Shunsuke Kitajima; Abigail Altabef; Jillian D. Cavanaugh; Kevin Rhee; Peng Gao; Haikuo Zhang; Peter E. Fecci; Takeshi Shimamura; Matthew D. Hellmann; John V. Heymach; F. Stephen Hodi; Gordon J. Freeman; David A. Barbie

STK11/LKB1 is among the most commonly inactivated tumor suppressors in non-small cell lung cancer (NSCLC), especially in tumors harboring KRAS mutations. Many oncogenes promote immune escape, undermining the effectiveness of immunotherapies, but it is unclear whether the inactivation of tumor suppressor genes, such as STK11/LKB1, exerts similar effects. In this study, we investigated the consequences of STK11/LKB1 loss on the immune microenvironment in a mouse model of KRAS-driven NSCLC. Genetic ablation of STK11/LKB1 resulted in accumulation of neutrophils with T-cell-suppressive effects, along with a corresponding increase in the expression of T-cell exhaustion markers and tumor-promoting cytokines. The number of tumor-infiltrating lymphocytes was also reduced in LKB1-deficient mouse and human tumors. Furthermore, STK11/LKB1-inactivating mutations were associated with reduced expression of PD-1 ligand PD-L1 in mouse and patient tumors as well as in tumor-derived cell lines. Consistent with these results, PD-1-targeting antibodies were ineffective against Lkb1-deficient tumors. In contrast, treating Lkb1-deficient mice with an IL6-neutralizing antibody or a neutrophil-depleting antibody yielded therapeutic benefits associated with reduced neutrophil accumulation and proinflammatory cytokine expression. Our findings illustrate how tumor suppressor mutations can modulate the immune milieu of the tumor microenvironment, and they offer specific implications for addressing STK11/LKB1-mutated tumors with PD-1-targeting antibody therapies.


Frontiers in Oncology | 2013

Activating Mutations in ERBB2 and Their Impact on Diagnostics and Treatment

Grit S. Herter-Sprie; Heidi Greulich; Kwok-Kin Wong

Despite the ongoing “war on cancer,” cancer remains one of the major causes of human morbidity and mortality. A new paradigm of targeted therapies holds the most promise for the future, making identification of tumor-specific therapeutic targets of prime importance. ERBB2/HER2, best known for its role in breast cancer tumorigenesis, can be targeted by two types of pharmacological manipulation: antibody therapy against the extracellular receptor domain and small molecule compounds against the intracellular tyrosine kinase domain. Aberrant activation of ERBB2 by gene amplification has been shown to participate in the pathophysiology of breast, ovarian, gastric, colorectal, lung, brain, and head and neck tumors. However, the advent of next-generation sequencing technologies has enabled efficient identification of activating molecular alterations of ERBB2. In this review, we will focus on the functional role of these somatic mutations that cause ERBB2 receptor activation. We will additionally discuss the current preclinical and clinical therapeutic strategies for targeting mutationally activated ERBB2.


Journal of Clinical Investigation | 2013

New cast for a new era: preclinical cancer drug development revisited.

Grit S. Herter-Sprie; Andrew L. Kung; Kwok-Kin Wong

Molecularly targeted agents promise to revolutionize therapeutics by reducing morbidity and mortality in patients with cancer. However, despite an urgent need for more effective anticancer compounds, current preclinical drug evaluations largely fail to satisfy the demand. New preclinical strategies, including the improvement of sophisticated mouse models and co-clinical study designs, are being used to augment the predictive value of animal-based translational cancer research. Here, we review the development of successful preclinical antineoplastic agents, their associated limitations, and alternative methods to predict clinical outcomes.


Journal of Medicinal Chemistry | 2015

Development of Selective Covalent Janus Kinase 3 Inhibitors

Li Tan; Koshi Akahane; Randall McNally; Kathleen M. S. E. Reyskens; Scott B. Ficarro; Suhu Liu; Grit S. Herter-Sprie; Shohei Koyama; Michael J. Pattison; Katherine Labella; Liv Johannessen; Esra A. Akbay; Kwok-Kin Wong; David A. Frank; Jarrod A. Marto; Thomas Look; J. Simon C. Arthur; Michael J. Eck; Nathanael S. Gray

The Janus kinases (JAKs) and their downstream effectors, signal transducer and activator of transcription proteins (STATs), form a critical immune cell signaling circuit, which is of fundamental importance in innate immunity, inflammation, and hematopoiesis, and dysregulation is frequently observed in immune disease and cancer. The high degree of structural conservation of the JAK ATP binding pockets has posed a considerable challenge to medicinal chemists seeking to develop highly selective inhibitors as pharmacological probes and as clinical drugs. Here we report the discovery and optimization of 2,4-substituted pyrimidines as covalent JAK3 inhibitors that exploit a unique cysteine (Cys909) residue in JAK3. Investigation of structure-activity relationship (SAR) utilizing biochemical and transformed Ba/F3 cellular assays resulted in identification of potent and selective inhibitors such as compounds 9 and 45. A 2.9 Å cocrystal structure of JAK3 in complex with 9 confirms the covalent interaction. Compound 9 exhibited decent pharmacokinetic properties and is suitable for use in vivo. These inhibitors provide a set of useful tools to pharmacologically interrogate JAK3-dependent biology.


Cancer Discovery | 2016

Oncogenic deregulation of EZH2 as an opportunity for targeted therapy in lung cancer

Hailei Zhang; Jun Qi; Jaime Reyes; Liren Li; Prakash Rao; Fugen Li; Charles Y. Lin; Jennifer A. Perry; Matthew A. Lawlor; Alexander J. Federation; De Raedt T; Yvonne Y. Li; Yin Liu; Melissa Duarte; Yunyu Zhang; Grit S. Herter-Sprie; Eiki Kikuchi; Julian Carretero; Charles M. Perou; Jakob B. Reibel; Joshiawa Paulk; Roderick T. Bronson; Hideo Watanabe; Christine Fillmore Brainson; Carla F. Kim; Peter S. Hammerman; Myles Brown; Karen Cichowski; Henry W. Long; James E. Bradner

UNLABELLED As a master regulator of chromatin function, the lysine methyltransferase EZH2 orchestrates transcriptional silencing of developmental gene networks. Overexpression of EZH2 is commonly observed in human epithelial cancers, such as non-small cell lung carcinoma (NSCLC), yet definitive demonstration of malignant transformation by deregulated EZH2 remains elusive. Here, we demonstrate the causal role of EZH2 overexpression in NSCLC with new genetically engineered mouse models of lung adenocarcinoma. Deregulated EZH2 silences normal developmental pathways, leading to epigenetic transformation independent of canonical growth factor pathway activation. As such, tumors feature a transcriptional program distinct from KRAS- and EGFR-mutant mouse lung cancers, but shared with human lung adenocarcinomas exhibiting high EZH2 expression. To target EZH2-dependent cancers, we developed a potent open-source EZH2 inhibitor, JQEZ5, that promoted the regression of EZH2-driven tumors in vivo, confirming oncogenic addiction to EZH2 in established tumors and providing the rationale for epigenetic therapy in a subset of lung cancer. SIGNIFICANCE EZH2 overexpression induces murine lung cancers that are similar to human NSCLC with high EZH2 expression and low levels of phosphorylated AKT and ERK, implicating biomarkers for EZH2 inhibitor sensitivity. Our EZH2 inhibitor, JQEZ5, promotes regression of these tumors, revealing a potential role for anti-EZH2 therapy in lung cancer. Cancer Discov; 6(9); 1006-21. ©2016 AACR.See related commentary by Frankel et al., p. 949This article is highlighted in the In This Issue feature, p. 932.


JCI insight | 2016

Synergy of radiotherapy and PD-1 blockade in Kras-mutant lung cancer

Grit S. Herter-Sprie; Shohei Koyama; Houari Korideck; Josephine Hai; Jiehui Deng; Yvonne Y. Li; Kevin A. Buczkowski; Aaron K. Grant; Soumya Ullas; Kevin Rhee; Jillian D. Cavanaugh; Neermala Poudel Neupane; Camilla L. Christensen; Jan M. Herter; G. Mike Makrigiorgos; F. Stephen Hodi; Gordon J. Freeman; Glenn Dranoff; Peter S. Hammerman; Alec C. Kimmelman; Kwok-Kin Wong

Radiation therapy (RT), a critical modality in the treatment of lung cancer, induces direct tumor cell death and augments tumor-specific immunity. However, despite initial tumor control, most patients suffer from locoregional relapse and/or metastatic disease following RT. The use of immunotherapy in non-small-cell lung cancer (NSCLC) could potentially change this outcome by enhancing the effects of RT. Here, we report significant (up to 70% volume reduction of the target lesion) and durable (up to 12 weeks) tumor regressions in conditional Kras-driven genetically engineered mouse models (GEMMs) of NSCLC treated with radiotherapy and a programmed cell death 1 antibody (αPD-1). However, while αPD-1 therapy was beneficial when combined with RT in radiation-naive tumors, αPD-1 therapy had no antineoplastic efficacy in RT-relapsed tumors and further induced T cell inhibitory markers in this setting. Furthermore, there was differential efficacy of αPD-1 plus RT among Kras-driven GEMMs, with additional loss of the tumor suppressor serine/threonine kinase 11/liver kinase B1 (Stk11/Lkb1) resulting in no synergistic efficacy. Taken together, our data provide evidence for a close interaction among RT, T cells, and the PD-1/PD-L1 axis and underscore the rationale for clinical combinatorial therapy with immune modulators and radiotherapy.


Nature Communications | 2014

Image-guided radiotherapy platform using single nodule conditional lung cancer mouse models

Grit S. Herter-Sprie; Houari Korideck; Camilla L. Christensen; Jan M. Herter; Kevin Rhee; R Berbeco; David G. Bennett; Esra A. Akbay; David Kozono; Raymond H. Mak; G. Mike Makrigiorgos; Alec C. Kimmelman; Kwok-Kin Wong

Close resemblance of murine and human trials is essential to achieve the best predictive value of animal-based translational cancer research. Kras-driven genetically engineered mouse models of non-small-cell lung cancer faithfully predict the response of human lung cancers to systemic chemotherapy. Owing to development of multifocal disease, however, these models have not been usable in studies of outcomes following focal radiotherapy (RT). We report the development of a preclinical platform to deliver state-of-the-art image-guided RT in these models. Presence of a single tumour as usually diagnosed in patients is modelled by confined injection of adenoviral Cre recombinase. Furthermore, three-dimensional conformal planning and state-of-the-art image-guided dose delivery are performed as in humans. We evaluate treatment efficacies of two different radiation regimens and find that Kras-driven tumours can temporarily be stabilized upon RT, whereas additional loss of either Lkb1 or p53 renders these lesions less responsive to RT. Current genetic mouse models of lung cancer develop multifocal tumours in all lobes, which limits their applicability to model radiotherapy of human disease. Here Herter-Sprie et aldevelop a method to induce single lung tumours in these models, allowing precise evaluation of radiation regiment efficacy.


Cancer immunology research | 2016

Cytotoxic T Cells in PD-L1–Positive Malignant Pleural Mesotheliomas Are Counterbalanced by Distinct Immunosuppressive Factors

Mark M. Awad; Robert E. Jones; Hongye Liu; Patrick H. Lizotte; Elena Ivanova; Meghana M. Kulkarni; Grit S. Herter-Sprie; Xiaoyun Liao; Abigail Santos; Mark Bittinger; Lauren Keogh; Shohei Koyama; Christina G. Almonte; Jessie M. English; Julianne C Barlow; William G. Richards; David A. Barbie; Adam J. Bass; Scott J. Rodig; F.S. Hodi; Kai W. Wucherpfennig; Pasi A. Jänne; Lynette M. Sholl; Peter S. Hammerman; Kwok-Kin Wong; Raphael Bueno

In malignant pleural mesothelioma, immunohistochemical expression of PD-L1 does not accurately predict whether patients respond to treatment with PD-1 pathway inhibitors. Comprehensive immunoprofiling by flow cytometry uncovered immunophenotypes that improve our understanding of response and resistance to checkpoint blockade. PD-L1 immunohistochemical staining does not always predict whether a cancer will respond to treatment with PD-1 inhibitors. We sought to characterize immune cell infiltrates and the expression of T-cell inhibitor markers in PD-L1–positive and PD-L1–negative malignant pleural mesothelioma samples. We developed a method for immune cell phenotyping using flow cytometry on solid tumors that have been dissociated into single-cell suspensions and applied this technique to analyze 43 resected malignant pleural mesothelioma specimens. Compared with PD-L1–negative tumors, PD-L1–positive tumors had significantly more infiltrating CD45+ immune cells, a significantly higher proportion of infiltrating CD3+ T cells, and a significantly higher percentage of CD3+ cells displaying the activated HLA-DR+/CD38+ phenotype. PD-L1–positive tumors also had a significantly higher proportion of proliferating CD8+ T cells, a higher fraction of FOXP3+/CD4+ Tregs, and increased expression of PD-1 and TIM-3 on CD4+ and CD8+ T cells. Double-positive PD-1+/TIM-3+ CD8+ T cells were more commonly found on PD-L1–positive tumors. Compared with epithelioid tumors, sarcomatoid and biphasic mesothelioma samples were significantly more likely to be PD-L1 positive and showed more infiltration with CD3+ T cells and PD-1+/TIM-3+ CD8+ T cells. Immunologic phenotypes in mesothelioma differ based on PD-L1 status and histologic subtype. Successful incorporation of comprehensive immune profiling by flow cytometry into prospective clinical trials could refine our ability to predict which patients will respond to specific immune checkpoint blockade strategies. Cancer Immunol Res; 4(12); 1038–48. ©2016 AACR.

Collaboration


Dive into the Grit S. Herter-Sprie's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge