Network


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

Hotspot


Dive into the research topics where Dorota Moroziewicz is active.

Publication


Featured researches published by Dorota Moroziewicz.


Journal of Clinical Oncology | 2006

Characterization of CD4+CD25+ Regulatory T Cells in Patients Treated With High-Dose Interleukin-2 for Metastatic Melanoma or Renal Cell Carcinoma

Giovanni Carlo Cesana; Gail DeRaffele; Seth M. Cohen; Dorota Moroziewicz; Josephine Mitcham; John Stoutenburg; Ken Cheung; Charles S. Hesdorffer; Seunghee Kim-Schulze; Howard L. Kaufman

PURPOSE To characterize the number and functional status of CD4+CD25+ regulatory T cells (Tregs) in patients with metastatic melanoma (MM) and renal cell carcinoma (RCC) treated with high-dose bolus interleukin-2 (IL-2). PATIENTS AND METHODS Patients with MM or RCC treated with high-dose bolus IL-2 (600,000 IU/kg every 8 hours) at a single center provided pre- and post-treatment whole blood specimens. Peripheral blood mononuclear cells were isolated by Ficoll density gradient centrifugation, separated into cellular subsets, and analyzed by flow cytometry or used for in vitro proliferation assays. RESULTS Between September 2003 and July 2005 57 patients were enrolled in the study with 48 patients available for analysis (45 MM, 12 RCC). Tregs were defined as CD4+CD25(hi) T cells, and this subset was significantly elevated in the cancer patients compared with normal donors (7.75% v 2.24%). The CD4(+)CD25(hi) T-cell pool in the patients constitutively expressed intracellular FoxP3, CTLA-4, and produced high amounts of IL-10. The Tregs were CCR7+ with 50% representing naïve and 50% central-memory T cells. The cells were functionally suppressive in mixed in vitro proliferation assays. Following IL-2 administration, the number and frequency of Tregs increased in patients with progressive disease but returned to normal levels in patients with objective clinical responses. CONCLUSION The number of Tregs, defined as CD4+CD25(hi) T cells is increased in patients with MM and RCC. High-dose IL-2 resulted in a significant decrease of Tregs in those patients achieving an objective clinical response to IL-2 therapy.


Journal of Clinical Investigation | 2005

Targeting the local tumor microenvironment with vaccinia virus expressing B7.1 for the treatment of melanoma

Howard L. Kaufman; Gail DeRaffele; Josephine Mitcham; Dorota Moroziewicz; Seth M. Cohen; Karl S. Hurst-Wicker; Ken Cheung; David S. Lee; Joseph Divito; Magalese Voulo; Julie Donovan; Kate Dolan; Kelledy Manson; Dennis Panicali; Ena Wang; Heidi Hörig; Francesco M. Marincola

Immunotherapy for the treatment of metastatic melanoma remains a major clinical challenge. The melanoma microenvironment may lead to local T cell tolerance in part through downregulation of costimulatory molecules, such as B7.1 (CD80). We report the results from the first clinical trial, to our knowledge, using a recombinant vaccinia virus expressing B7.1 (rV-B7.1) for monthly intralesional vaccination of accessible melanoma lesions. A standard 2-dose-escalation phase I clinical trial was conducted with 12 patients. The approach was well tolerated with only low-grade fever, myalgias, and fatigue reported and 2 patients experiencing vitiligo. An objective partial response was observed in 1 patient and disease stabilization in 2 patients, 1 of whom is alive without disease 59 months following vaccination. All patients demonstrated an increase in postvaccination antibody and T cell responses against vaccinia virus. Systemic immunity was tested in HLA-A*0201 patients who demonstrated an increased frequency of gp100 and T cells specific to melanoma antigen recognized by T cells 1 (MART-1), also known as Melan-A, by ELISPOT assay following local rV-B7.1 vaccination. Local immunity was evaluated by quantitative real-time RT-PCR, which suggested that tumor regression was associated with increased expression of CD8 and IFN-gamma. The local delivery of vaccinia virus expressing B7.1 was well tolerated and represents an innovative strategy for altering the local tumor microenvironment in patients with melanoma.


Journal of Translational Medicine | 2009

Phase II trial of Modified Vaccinia Ankara (MVA) virus expressing 5T4 and high dose Interleukin-2 (IL-2) in patients with metastatic renal cell carcinoma

Howard L. Kaufman; Bret Taback; William H. Sherman; Dae Won Kim; William Shingler; Dorota Moroziewicz; Gail DeRaffele; Josephine Mitcham; Miles William Carroll; Richard Harrop; Stuart Naylor; Seunghee Kim-Schulze

BackgroundInterleukin-2 (IL-2) induces durable objective responses in a small cohort of patients with metastatic renal cell carcinoma (RCC) but the antigen(s) responsible for tumor rejection are not known. 5T4 is a non-secreted membrane glycoprotein expressed on clear cell and papillary RCCs. A modified vaccinia virus Ankara (MVA) encoding 5T4 was tested in combination with high-dose IL-2 to determine the safety, objective response rate and effect on humoral and cell-mediated immunity.Methods25 patients with metastatic RCC who qualified for IL-2 were eligible and received three immunizations every three weeks followed by IL-2 (600,000 IU/kg) after the second and third vaccinations. Blood was collected for analysis of humoral, effector and regulatory T cell responses.ResultsThere were no serious vaccine-related adverse events. While no objective responses were observed, three patients (12%) were rendered disease-free after nephrectomy or resection of residual metastatic disease. Twelve patients (48%) had stable disease which was associated with improved median overall survival compared to patients with progressive disease (not reached vs. 28 months, p = 0.0261). All patients developed 5T4-specific antibody responses and 13 patients had an increase in 5T4-specific T cell responses. Although the baseline frequency of Tregs was elevated in all patients, those with stable disease showed a trend toward increased effector CD8+ T cells and a decrease in Tregs.ConclusionV accination with MVA-5T4 did not improve objective response rates of IL-2 therapy but did result in stable disease associated with an increase in the ratio of 5T4-specific effector to regulatory T cells in selected patients.Trial registration numberISRCTN83977250


Journal of Gastrointestinal Surgery | 2010

RAGE Signaling Significantly Impacts Tumorigenesis and Hepatic Tumor Growth in Murine Models of Colorectal Carcinoma

Joseph DiNorcia; Dorota Moroziewicz; Nikalesh Ippagunta; Minna K. Lee; Mark Foster; Heidrun Rotterdam; Fei Bao; Yu Shan Zhou; Shi Fang Yan; Jean C. Emond; Ann Marie Schmidt; John D. Allendorf

BackgroundThe receptor for advanced glycation end-products (RAGE) is a cell surface receptor implicated in tumor cell proliferation and migration. We hypothesized that RAGE signaling impacts tumorigenesis and metastatic tumor growth in murine models of colorectal carcinoma.Materials and MethodsTumorigenesis: Apc1638N/+ mice were crossed with Rage−/− mice in the C57BL/6 background to generate Apc1638N/+/Rage−/− mice. Metastasis: BALB/c mice underwent portal vein injection with CT26 cells (syngeneic) and received daily soluble (s)RAGE or vehicle. Rage−/− mice and Rage+/+ controls underwent portal vein injection with MC38 cells (syngeneic). Rage+/+ mice underwent portal vein injection with MC38 cells after stable transfection with full-length RAGE or mock transfection control.ResultsTumorigenesis: Apc1638N/+/Rage−/− mice had reduced tumor incidence, size, and histopathologic grade. Metastasis: Pharmacological blockade of RAGE with sRAGE or genetic deletion of Rage reduced hepatic tumor incidence, nodules, and burden. Gain of function by transfection with full-length RAGE increased hepatic tumor burden compared to vector control MC38 cells.ConclusionRAGE signaling plays an important role in tumorigenesis and hepatic tumor growth in murine models of colorectal carcinoma. Further work is needed to target the ligand–RAGE axis for possible prophylaxis and treatment of primary and metastatic colorectal carcinoma.


Journal of Immunology | 2008

Intrarectal Vaccination with Recombinant Vaccinia Virus Expressing Carcinoembronic Antigen Induces Mucosal and Systemic Immunity and Prevents Progression of Colorectal Cancer

Seunghee Kim-Schulze; Hong Sung Kim; Alberto Wainstein; Dae Won Kim; Wein Cui Yang; Dorota Moroziewicz; Phyllus Y. Mong; Michal Bereta; Bret Taback; Qin Wang; Howard L. Kaufman

The gastrointestinal mucosa contains an intact immune system that protects the host from pathogens and communicates with the systemic immune system. Absorptive epithelial cells in the mucosa give rise to malignant tumors although the interaction between tumor cells and the mucosal immune system is not well defined. The pathophysiology of colorectal cancer has been elucidated through studies of hereditary syndromes, such as familial adenomatous polyposis, a cancer predisposition syndrome caused by germline mutations in the adenomatous polyposis coli tumor suppressor gene. Patients with FAP develop adenomas and inevitably progress to invasive carcinomas by the age of 40. To better delineate the role of mucosal immunity in colorectal cancer, we evaluated the efficacy of intrarectal recombinant vaccinia virus expressing the human carcinoembryonic Ag (CEA) in a murine FAP model in which mice are predisposed to colorectal cancer and also express human CEA in the gut. Mucosal vaccination reduced the incidence of spontaneous adenomas and completely prevented progression to invasive carcinoma. The therapeutic effects were associated with induction of mucosal CEA-specific IgA Ab titers and CD8+ CTLs. Mucosal vaccination was also associated with an increase in systemic CEA-specific IgG Ab titers, CD4+ and CD8+ T cell responses and resulted in growth inhibition of s.c. implanted CEA-expressing tumors suggesting communication between mucosal and systemic immune compartments. Thus, intrarectal vaccination induces mucosal and systemic antitumor immunity and prevents progression of spontaneous colorectal cancer. These results have implications for the prevention of colorectal cancer in high-risk individuals.


Cellular Immunology | 2004

The lymphoid chemokine CCL21 costimulates naïve T cell expansion and Th1 polarization of non-regulatory CD4+ T cells

Kenneth Flanagan; Dorota Moroziewicz; Heesun Kwak; Heidi Hörig; Howard L. Kaufman


Human Gene Therapy | 2006

Local Delivery of Vaccinia Virus Expressing Multiple Costimulatory Molecules for the Treatment of Established Tumors

Howard L. Kaufman; Seth M. Cohen; Ken Cheung; Gail DeRaffele; Josephine Mitcham; Dorota Moroziewicz; Jeffrey Schlom; Charles S. Hesdorffer


Journal of Gastrointestinal Surgery | 2012

RAGE gene deletion inhibits the development and progression of ductal neoplasia and prolongs survival in a murine model of pancreatic cancer.

Joseph DiNorcia; Minna K. Lee; Dorota Moroziewicz; Megan Winner; Paritosh Suman; Fei Bao; Helen Remotti; Yu Shan Zou; Shi Fang Yan; Wanglong Qiu; Gloria H. Su; Ann Marie Schmidt; John D. Allendorf


Journal of Surgical Research | 2007

P238: Mucosal vaccination prevents spontaneous colorectal carcinoma and induces long-term immunity in mice

Seunghee Kim-Schulze; Dorota Moroziewicz; A. Wainstein; B. Elliott; Howard L. Kaufman


Journal of Clinical Oncology | 2016

A phase I clinical trial of MVA expressing 5T4 and high-dose interleukin-2 (IL-2) for metastatic renal cell carcinoma

Howard L. Kaufman; Gail DeRaffele; Josephine Mitcham; Dorota Moroziewicz; Seunghee Kim-Schulze; C. A. Petrulio; Richard Harrop; Stuart Naylor

Collaboration


Dive into the Dorota Moroziewicz's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Josephine Mitcham

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph DiNorcia

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Dae Won Kim

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Minna K. Lee

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge