Network


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

Hotspot


Dive into the research topics where Agnieszka A. Rucki is active.

Publication


Featured researches published by Agnieszka A. Rucki.


Science | 2017

Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade

Dung T. Le; Jennifer N. Durham; Kellie Nicole Smith; Hao Wang; Bjarne Bartlett; Laveet K. Aulakh; Steve Lu; Holly Kemberling; Cara Wilt; Brandon Luber; Fay Wong; Nilofer Saba Azad; Agnieszka A. Rucki; Daniel A. Laheru; Ross C. Donehower; Atif Zaheer; George A. Fisher; Todd S. Crocenzi; James J. Lee; Tim F. Greten; Austin Duffy; Kristen K. Ciombor; Aleksandra Eyring; Bao H. Lam; Andrew K. Joe; S. Peter Kang; Matthias Holdhoff; Ludmila Danilova; Leslie Cope; Christian Meyer

Predicting responses to immunotherapy Colon cancers with loss-of-function mutations in the mismatch repair (MMR) pathway have favorable responses to PD-1 blockade immunotherapy. In a phase 2 clinical trial, Le et al. showed that treatment success is not just limited to colon cancer (see the Perspective by Goswami and Sharma). They found that a wide range of different cancer types with MMR deficiency also responded to PD-1 blockade. The trial included some patients with pancreatic cancer, which is one of the deadliest forms of cancer. The clinical trial is still ongoing, and around 20% of patients have so far achieved a complete response. MMR deficiency appears to be a biomarker for predicting successful treatment outcomes for several solid tumors and indicates a new therapeutic option for patients harboring MMR-deficient cancers. Science, this issue p. 409; see also p. 358 A pan-cancer biomarker is identified that can predict successful response to cancer immunotherapy in human patients. The genomes of cancers deficient in mismatch repair contain exceptionally high numbers of somatic mutations. In a proof-of-concept study, we previously showed that colorectal cancers with mismatch repair deficiency were sensitive to immune checkpoint blockade with antibodies to programmed death receptor–1 (PD-1). We have now expanded this study to evaluate the efficacy of PD-1 blockade in patients with advanced mismatch repair–deficient cancers across 12 different tumor types. Objective radiographic responses were observed in 53% of patients, and complete responses were achieved in 21% of patients. Responses were durable, with median progression-free survival and overall survival still not reached. Functional analysis in a responding patient demonstrated rapid in vivo expansion of neoantigen-specific T cell clones that were reactive to mutant neopeptides found in the tumor. These data support the hypothesis that the large proportion of mutant neoantigens in mismatch repair–deficient cancers make them sensitive to immune checkpoint blockade, regardless of the cancers’ tissue of origin.


Journal of Immunotherapy | 2015

PD-1/PD-L1 blockade together with vaccine therapy facilitates effector T-cell infiltration into pancreatic tumors

Kevin C. Soares; Agnieszka A. Rucki; Annie A. Wu; Kelly Olino; Qian Xiao; Yi Chai; Anthony Wamwea; Elaine Bigelow; Eric R. Lutz; Linda Liu; Sheng Yao; Robert A. Anders; Daniel A. Laheru; Christopher L. Wolfgang; Barish H. Edil; Richard D. Schulick; Elizabeth M. Jaffee; Lei Zheng

Pancreatic ductal adenocarcinoma (PDA) has a poor prognosis due to late detection and resistance to conventional therapies. Published studies show that the PDA tumor microenvironment is predominantly infiltrated with immune suppressive cells and signals that if altered, would allow effective immunotherapy. However, single-agent checkpoint inhibitors including agents that alter immune suppressive signals in other human cancers such as cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed death 1 (PD-1), and its ligand PD-L1, have failed to demonstrate objective responses when given as single agents to PDA patients. We recently reported that inhibition of the CTLA-4 pathway when given together with a T cell inducing vaccine gives objective responses in metastatic PDA patients. In this study, we evaluated blockade of the PD-1/PD-L1 pathway. We found that PD-L1 is weakly expressed at a low frequency in untreated human and murine PDAs but treatment with a granulocyte macrophage colony-stimulating factor secreting PDA vaccine (GVAX) significantly upregulates PD-L1 membranous expression after treatment of tumor-bearing mice. In addition, combination therapy with vaccine and PD-1 antibody blockade improved murine survival compared with PD-1 antibody monotherapy or GVAX therapy alone. Furthermore, PD-1 blockade increased effector CD8+ T lymphocytes and tumor-specific interferon-&ggr; production of CD8+ T cells in the tumor microenvironment. Immunosuppressive pathways, including regulatory T cells and CTLA-4 expression on T cells were overcome by the addition of vaccine and low-dose cyclophosphamide to PD-1 blockade. Collectively, our study supports combining PD-1 or PD-L1 antibody therapy with a T cell inducing agent for PDA treatment.


World Journal of Gastroenterology | 2014

Pancreatic cancer stroma: Understanding biology leads to new therapeutic strategies

Agnieszka A. Rucki; Lei Zheng

Pancreatic ductal adenocarcinoma (PDA) is among the deadliest cancers in the United States and in the world. Late diagnosis, early metastasis and lack of effective therapy are among the reasons why only 6% of patients diagnosed with PDA survive past 5 years. Despite development of targeted therapy against other cancers, little progression has been made in the treatment of PDA. Therefore, there is an urgent need for the development of new treatments. However, in order to proceed with treatments, the complicated biology of PDA needs to be understood first. Interestingly, majority of the tumor volume is not made of malignant epithelial cells but of stroma. In recent years, it has become evident that there is an important interaction between the stromal compartment and the less prevalent malignant cells, leading to cancer progression. The stroma not only serves as a growth promoting source of signals but it is also a physical barrier to drug delivery. Understanding the tumor-stroma signaling leading to development of desmoplastic reaction and tumor progression can lead to the development of therapies to decrease stromal activity and improve drug delivery. In this review, we focus on how the current understanding of biology of the pancreatic tumor microenvironment can be translated into the development of targeted therapy.


Human Molecular Genetics | 2013

The DcpS inhibitor RG3039 improves motor function in SMA mice

James P. Van Meerbeke; Rebecca M. Gibbs; Heather L. Plasterer; Wenyan Miao; Zhihua Feng; Ming Yi Lin; Agnieszka A. Rucki; Claribel D. Wee; Bing Xia; Shefali Sharma; Vincent Jacques; Darrick K. Li; Livio Pellizzoni; James R. Rusche; Chien-Ping Ko; Charlotte J. Sumner

Spinal muscular atrophy (SMA) is caused by mutations of the survival motor neuron 1 (SMN1) gene, retention of the survival motor neuron 2 (SMN2) gene and insufficient expression of full-length survival motor neuron (SMN) protein. Quinazolines increase SMN2 promoter activity and inhibit the ribonucleic acid scavenger enzyme DcpS. The quinazoline derivative RG3039 has advanced to early phase clinical trials. In preparation for efficacy studies in SMA patients, we investigated the effects of RG3039 in severe SMA mice. Here, we show that RG3039 distributed to central nervous system tissues where it robustly inhibited DcpS enzyme activity, but minimally activated SMN expression or the assembly of small nuclear ribonucleoproteins. Nonetheless, treated SMA mice showed a dose-dependent increase in survival, weight and motor function. This was associated with improved motor neuron somal and neuromuscular junction synaptic innervation and function and increased muscle size. RG3039 also enhanced survival of conditional SMA mice in which SMN had been genetically restored to motor neurons. As this systemically delivered drug may have therapeutic benefits that extend beyond motor neurons, it could act additively with SMN-restoring therapies delivered directly to the central nervous system such as antisense oligonucleotides or gene therapy.


Science Signaling | 2015

Semaphorin 3D autocrine signaling mediates the metastatic role of annexin A2 in pancreatic cancer

Kelly Foley; Agnieszka A. Rucki; Qian Xiao; Donger Zhou; Ashley Leubner; Guanglan Mo; Jennifer Kleponis; Annie A. Wu; Rajni Sharma; Qingguang Jiang; Robert A. Anders; Christine A. Iacobuzio-Donahue; Katherine A. Hajjar; Anirban Maitra; Elizabeth M. Jaffee; Lei Zheng

Annexin A2–dependent secretion of semaphorin 3D induces metastasis of pancreatic cancer cells. Semaphorin signals guide metastases Semaphorin signaling directs the migration of developing neurons, as well as new blood vessels. The abundance of semaphorin 3D (Sema3D) is increased in the tumors of patients with metastatic pancreatic ductal adenocarcinoma (PDA), and antibodies against the protein annexin A2 are present in patient sera. Using cells from a genetic mouse model of PDA, Foley et al. found that annexin A2 increased the release of Sema3D from PDA cells and promoted migratory behavior in cultured PDA cells. Secreted Sema3D activated its co-receptor plexin D1 in an autocrine manner on the surface of PDA cells. Knocking down annexin A2 decreased the colonization of injected PDA cells in the liver and lungs of mice. These prometastatic effects of annexin A2 did not involve changes in tumor growth or vascularization. Targeting annexin A2 may prevent metastatic PDA. Most patients with pancreatic ductal adenocarcinoma (PDA) present with metastatic disease at the time of diagnosis or will recur with metastases after surgical treatment. Semaphorin–plexin signaling mediates the migration of neuronal axons during development and of blood vessels during angiogenesis. The expression of the gene encoding semaphorin 3D (Sema3D) is increased in PDA tumors, and the presence of antibodies against the pleiotropic protein annexin A2 (AnxA2) in the sera of some patients after surgical resection of PDA is associated with longer recurrence-free survival. By knocking out AnxA2 in a transgenic mouse model of PDA (KPC) that recapitulates the progression of human PDA from premalignancy to metastatic disease, we found that AnxA2 promoted metastases in vivo. The expression of AnxA2 promoted the secretion of Sema3D from PDA cells, which coimmunoprecipitated with the co-receptor plexin D1 (PlxnD1) on PDA cells. Mouse PDA cells in which SEMA3D was knocked down or ANXA2-null PDA cells exhibited decreased invasive and metastatic potential in culture and in mice. However, restoring Sema3D in AnxA2-null cells did not entirely rescue metastatic behavior in culture and in vivo, suggesting that AnxA2 mediates additional prometastatic mechanisms. Patients with primary PDA tumors that have abundant Sema3D have widely metastatic disease and decreased survival compared to patients with tumors that have relatively low Sema3D abundance. Thus, AnxA2 and Sema3D may be new therapeutic targets and prognostic markers of metastatic PDA.


Oncotarget | 2015

TGF-β blockade depletes T regulatory cells from metastatic pancreatic tumors in a vaccine dependent manner.

Kevin C. Soares; Agnieszka A. Rucki; Victoria Kim; Kelly Foley; Sara Solt; Christopher L. Wolfgang; Elizabeth M. Jaffee; Lei Zheng

Our neoadjuvant clinical trial of a GM-CSF secreting allogeneic pancreas tumor vaccine (GVAX) revealed the development of tertiary lymphoid aggregates (TLAs) within the pancreatic ductal adenocarcinoma (PDA) tumor microenvironment 2 weeks after GVAX treatment. Microarray studies revealed that multiple components of the TGF-β pathway were suppressed in TLAs from patients who survived greater than 3 years and who demonstrated vaccine-enhanced mesothelin-specific T cell responses. We tested the hypothesis that combining GVAX with TGF-β inhibitors will improve the anti-tumor immune response of vaccine therapy. In a metastatic murine model of pancreatic cancer, combination therapy with GVAX vaccine and a TGF-β blocking antibody improved the cure rate of PDA-bearing mice. TGF-β blockade in combination with GVAX significantly increased the infiltration of effector CD8+ T lymphocytes, specifically anti-tumor-specific IFN-γ producing CD8+ T cells, when compared to monotherapy controls (all p < 0.05). TGF-β blockade alone did not deplete T regulatory cells (Tregs), but when give in combination with GVAX, GVAX induced intratumoral Tregs were depleted. Therefore, our PDA preclinical model demonstrates a survival advantage in mice treated with an anti-TGF-β antibody combined with GVAX therapy and provides strong rational for testing this combinational therapy in clinical trials.


Journal of gastrointestinal oncology | 2017

Shifting paradigm of developing biologics for the treatment of pancreatic adenocarcinoma

Ying Zeng; Agnieszka A. Rucki; Xu Che; Lei Zheng

Pancreatic adenocarcinoma is still widely considered as a deadly disease even though there are substantial therapeutic developments in the past decade. Using combinational chemotherapy regimens, represented by gemcitabine plus nab-paclitaxel and FOLFIRINOX, was able to improve overall survival in patients with advanced disease to a limited extent. It has been a challenge to develop targeted therapies that are focused on the neoplasm cells of pancreatic adenocarcinoma. Recently, targeting the stroma and immune compartments of pancreatic adenocarcinoma has shown promising results. The paradigm of biologics drug development therefore has been shifted by extending to these exciting areas. Although some of the preclinical and clinical researches in targeting the tumor microenvironment of pancreatic adenocarcinoma have shown promising results, others have resulted in controversial findings. Both comprehensive and in-depth researches on the basic science of the tumor microenvironment of pancreatic adenocarcinoma are thus warranted for the development of effective biologics that target the tumor microenvironment. Moreover, an ideal treatment for pancreatic adenocarcinoma shall be a combination of targeting both neoplastic cells and the tumor microenvironment.


Oncotarget | 2017

Stromal Annexin A2 expression is predictive of decreased survival in pancreatic cancer

Adrian Murphy; Kelly Foley; Agnieszka A. Rucki; Tao Xia; Elizabeth M. Jaffee; Chiung Yu Huang; Lei Zheng

Pancreatic ductal adenocarcinoma (PDA) is renowned for high rates of metastasis and poor survival. Its notoriously dense fibrotic stroma contributes to chemoresistance. Stromal signaling in PDA is recognized for its multiple roles in regulating tumor invasion and metastasis. However, no stromal biomarker which can predict survival in PDA exists. Annexin A2 (AnxA2) was formerly identified as a metastasis-associated protein in PDA and tumoral overexpression is associated with poor survival. In this study, we examined AnxA2 expression in the tumor microenvironment in a preclinical model of PDA which suggests its role in tumor colonization. We injected wild-type (KPC) and AnxA2 knockout (KPCA) pancreatic cells into C57BL/GJ (B6) and AnxA2 knockout (KO) mice using the hemi-spleen model and observed their survival. We performed quantitative immunohistochemistry examining stromal AnxA2 expression in 56 patients who had surgically resected PDA and correlated expression with clinical outcomes. B6 mice injected with KPC cells demonstrated decreased median survival compared to those injected with KPCA cells (90 days vs. not reached, p < 0.0001) whereas there was no survival difference in the AnxA2 KO mice (p = 0.63). In patient specimens, we found that high stromal AnxA2 expression (≥80th percentile) was associated with significantly reduced disease-free survival (p = 0.002) and overall survival (p < 0.001). Using multivariate Cox models, there were no significant associations between other clinical covariates apart from high stromal AnxA2 expression. This study highlights the role that stromal AnxA2 expression plays as a prognostic marker in PDA and its potential as a predictive biomarker for survival outcomes in PDA.


Cancer Research | 2013

Abstract 3935: AnnexinA2 regulates invasion and metastasis of pancreatic ductal adenocarcinoma through Semaphorin3d.

Kelly Foley; Donger Zhou; Qian Xiao; Agnieszka A. Rucki; Guanglan Mo; Lanqing Huang; Anirban Maitra; Elizabeth M. Jaffee; Lei Zheng

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Pancreatic ductal adenocarcinoma (PDA), a devastating disease with a median survival of less than six months, is characterized by its high metastatic ability and a lack of effective therapies. The mechanisms involved in promoting metastasis of PDA have yet to be elucidated. We recently identified AnnexinA2 (AnxA2) as a PDA-associated antigen. Tyr23 phosphorylated AnxA2 promotes metastasis of PDA cells, while shRNA knockdown, knockout or therapeutic inhibition of AnxA2 prevents metastasis to the liver in a mouse model. To identify downstream mediators of AnxA2 signaling, we evaluated gene expression differences in cell lines derived from the primary tumors of mice that express AnxA2 (KPC) and are metastatic or lack AnxA2 expression (KPCA) and are unable to metastasize. Using microarray analysis, we observed that expression of the ligand-receptor pair Semaphorin3d (Sema3d) and PlexinD1 (PlxnD1) was significantly down regulated in the non-metastatic KPCA cells. Sema3d and PlxnD1 were first identified as axon guidance genes. However, recently, axon guidance genes, including Sema3d and PlxnD1, were recently found by the International Cancer Genome Consortium to be among the cellular pathways that are most frequently altered by genetic mutations and amplification in human PDA (Biankin 2012). Therefore, we hypothesized that AnxA2 interacts with Sema3d and PlxnD1 in PDA to induce metastasis. We confirmed by RT-PCR and immunofluorescence analysis in multiple independent tumors from KPC and KPCA mice that Sema3d and PlxnD1 expression is down regulated in the absence of AnxA2 expression. AnxA2, Sema3d and PlxnD1 colocalize at the cell surface during pancreatic tumor development when AnxA2 is expressed. Interestingly, co-immunoprecipitation analysis demonstrated a direct interaction between Sema3d and AnxA2 in PDA cells supporting our hypothesis that AnxA2 regulates the secretion of Sema3d from PDA cells to enable its paracrine interaction with PlxnD1 resulting in invasion of PDA cells. Clinically, we have found that Sema3d expression correlates with increased lymphovascular invasion, increased perineural invasion, poor survival and increased metastatic potential in human PDA patients. Therefore, understanding the pathways that lead to metastasis of PDA will enable the development of more effective therapeutics and better clinical outcomes. Citation Format: Kelly Foley, Donger Zhou, Qian Xiao, Agnieszka Rucki, Guanglan Mo, Lanqing Huang, Anirban Maitra, Elizabeth Jaffee, Lei Zheng. AnnexinA2 regulates invasion and metastasis of pancreatic ductal adenocarcinoma through Semaphorin3d. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 3935. doi:10.1158/1538-7445.AM2013-3935


Gastroenterology | 2014

A Listeria vaccine and depletion of T-regulatory cells activate immunity against early stage pancreatic intraepithelial neoplasms and prolong survival of mice.

Bridget P. Keenan; Yvonne Saenger; Michel Kafrouni; Ashley Leubner; Peter Lauer; Anirban Maitra; Agnieszka A. Rucki; Andrew J. Gunderson; Lisa M. Coussens; Dirk G. Brockstedt; Thomas W. Dubensky; Raffit Hassan; Todd D. Armstrong; Elizabeth M. Jaffee

Collaboration


Dive into the Agnieszka A. Rucki's collaboration.

Top Co-Authors

Avatar

Lei Zheng

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kelly Foley

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qian Xiao

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Adrian Murphy

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Anirban Maitra

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Annie A. Wu

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Ashley Leubner

Johns Hopkins University

View shared research outputs
Researchain Logo
Decentralizing Knowledge