Network


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

Hotspot


Dive into the research topics where Mark J. Dobrzanski is active.

Publication


Featured researches published by Mark J. Dobrzanski.


Journal of Immunology | 2000

Type 1 and type 2 CD8+ effector T cell subpopulations promote long-term tumor immunity and protection to progressively growing tumor.

Mark J. Dobrzanski; Joyce B. Reome; Richard W. Dutton

Cytolytic CD8+ effector cells fall into two subpopulations based on cytokine secretion. Type 1 CD8+ T cells (Tc1) secrete IFN-γ, whereas type 2 CD8+ T cells (Tc2) secrete IL-4, IL-5, and IL-10. Using an OVA-transfected B16 lung metastases model, we assessed the therapeutic effects of adoptively transferred OVA-specific Tc1 and Tc2 subpopulations in mice bearing established pulmonary malignancy. Effector cell-treated mice exhibiting high (5 × 105) tumor burdens experienced significant (p < 0.05) delays in mortality compared with those of untreated control mice, whereas high proportions (70–90%) of mice receiving therapy with low (1 × 105) tumor burdens survived indefinitely. Long-term tumor immunity was evident by resistance to lethal tumor rechallenge, heightened levels of systemic OVA Ag-specific CTL responses ex vivo, and detection of long-lived TCR transgene-positive donor cells accompanied by an elevation in the total numbers of CD8+ CD44high activated and/or memory T cells at sites of tumor growth. Long-lasting protection by Tc2 and Tc1 effector cells were dependent, in part, on both the level of tumor burden and effector cell-derived IL-4, IL-5, and IFN-γ, respectively. We conclude that Tc1 and Tc2 effector cells provide immunity by different mechanisms that subsequently potentiate host-derived antitumor responses.


Journal of Immunology | 2004

Tc1 and Tc2 Effector Cell Therapy Elicit Long-Term Tumor Immunity by Contrasting Mechanisms That Result in Complementary Endogenous Type 1 Antitumor Responses

Mark J. Dobrzanski; Joyce B. Reome; Joseph A. Hollenbaugh; Richard W. Dutton

Cytolytic CD8+ effector cells fall into two subpopulations based on cytokine secretion. Type 1 CD8+ T cells (Tc1) secrete IFN-γ, whereas type 2 CD8+ T cells (Tc2) secrete IL-4 and IL-5. Both effector cell subpopulations display predominantly perforin-dependent cytolysis in vitro. Using an OVA-transfected B16 lung metastases model, we show that adoptively transferred OVA-specific Tc1 and Tc2 cells induce considerable suppression, but not cure, of pulmonary metastases. However, long-term tumor immunity prolonged survival times indefinitely and was evident by resistance to lethal tumor rechallenge. At early stages after therapy, protection by Tc2 and Tc1 effector cells were dependent in part on effector cell-derived IL-4, IL-5, and IFN-γ, respectively. Whereas effector cell-derived perforin was not necessary. Over time the numbers of both donor cells diminished to low, yet still detectable, levels. Concomitantly, Tc1 and Tc2 effector cell therapies potentiated endogenous recipient-derived antitumor responses by inducing 1) local T cell-derived chemokines associated with type 1-like immune responses; 2) elevated levels of recipient-derived OVA tetramer-positive CD8 memory T cells that were CD44high, CD122+, and Ly6Chigh that predominantly produced IFN-γ and TNF-α; and 3) heightened numbers of activated recipient-derived Th1 and Tc1 T cell subpopulations expressing CD25+, CD69+, and CD95+ cell surface activation markers. Moreover, both Tc2 and Tc1 effector cell therapies were dependent in part on recipient-derived IFN-γ and TNF-α for long-term survival and protection. Collectively, Tc1 and Tc2 effector cell immunotherapy mediate long-term tumor immunity by different mechanisms that subsequently potentiate endogenous recipient-derived type 1 antitumor responses.


Journal of Immunology | 2001

Role of Effector Cell-Derived IL-4, IL-5, and Perforin in Early and Late Stages of Type 2 CD8 Effector Cell-Mediated Tumor Rejection

Mark J. Dobrzanski; Joyce B. Reome; Richard W. Dutton

Type 2 CD8 T cells (Tc2) secrete IL-4 and IL-5 and display perforin-dependent cytolysis in vitro. Using an OVA-transfected B16-melanoma model, we show that tumor-reactive Tc2 effector cells accumulated at the tumor site and induced tumor regression that enhanced survival in mice with pulmonary tumors. Transfer of perforin-deficient Tc2 cells generated from perforin gene knockout mice showed no differences in therapeutic efficiency when compared with wild-type Tc2 cells. In contrast, Tc2 cells derived from select cytokine gene-deficient mice showed that therapeutic effects were dependent on effector cell-derived IL-4 and IL-5 that led to a local elevation in lung-derived chemoattractants and accumulation of activated host-derived CD8/CD44high, CD4/CD44high, and OVA-specific tetramer-positive CD8 cells in vivo. Host-derived T and non-T immune cells increased in the lung over time and correlated with an elevated production of type 1-related chemokines. Conversely, donor Tc2 cell numbers markedly diminished at later times, suggesting that prolonged therapeutic responses were due to host-derived mechanisms. Moreover, type 1 host responses were detectable with increased levels of IFN-γ production by lung-derived CD4 and CD8 T cells from surviving Tc2-treated mice. Transfer of Tc2 cells into IFN-γ-deficient tumor-bearing mice was markedly less effective then into wild-type mice, suggesting that host-derived IFN-γ-dependent mechanisms play a role in Tc2-mediated antitumor responses.


Journal of Immunology | 2004

The Rate of the CD8-Dependent Initial Reduction in Tumor Volume Is Not Limited by Contact-Dependent Perforin, Fas Ligand, or TNF-Mediated Cytolysis

Joseph A. Hollenbaugh; Joyce B. Reome; Mark J. Dobrzanski; Richard W. Dutton

Established EG7 tumors expressing OVA and growing at an intradermal site become rapidly reduced in size following adoptive therapy with in vitro-generated type I CD8 T cell (Tc1) effectors generated from naive CD8 T cells from transgenic TCR OVA-specific mice. Tc1 effectors kill EG7 target cells in vitro by a perforin-dependent mechanism. However, we show that there is no quantitative diminution of the initial phase of antitumor activity in vivo, whether the Tc1 effectors are derived from perforin-, Fas ligand-, or TNF-deficient transgenic TCR mice or whether the recipients are perforin deficient. Tumors are also equally well controlled whether the Tc1 effectors come from mice deficient in perforin plus Fas ligand or perforin plus TNF. Control of tumor growth is diminished when Tc1 effectors generated from IFN-γ-deficient mice are used. We conclude that control of tumor growth is not in any way affected by loss of contact-mediated lytic mechanisms, and conclude that the CD8 effectors must act by recruiting host effector mechanisms to control tumor growth.


Cancer Research | 2004

Effector Cell-Derived Lymphotoxin α and Fas Ligand, but not Perforin, Promote Tc1 and Tc2 Effector Cell-Mediated Tumor Therapy in Established Pulmonary Metastases

Mark J. Dobrzanski; Joyce B. Reome; Joseph A. Hollenbaugh; James C. Hylind; Richard W. Dutton

Cytolytic CD8+ effector cells fall into two subpopulations based on cytokine secretion. Type 1 CD8+ T cells (Tc1) secrete IFN-γ, whereas type 2 CD8+ T cells (Tc2) secrete interleukin (IL)-4 and IL-5. Although both effector cell subpopulations display Fas ligand (FasL) and tumor necrosis factor (TNF), tumor lysis is predominantly perforin dependent in vitro. Using an ovalbumin-transfected B16 lung metastasis model, we show that heightened numbers of adoptively transferred ovalbumin-specific Tc1 and Tc2 cells accumulated at the tumor site by day 2 after therapy and induced tumor regression that enhanced survival in mice with pulmonary metastases. Transfer of either TNF-α- or perforin-deficient Tc1 or Tc2 effector cells generated from specified gene-deficient mice showed no differences in therapeutic efficiency when compared with corresponding wild-type cells. In contrast, both Tc1 and Tc2 cells, derived from either FasL or TNF-α/lymphotoxin (LT) α double knockout mice, showed that therapeutic effects were dependent, in part, on effector cell-derived FasL or LTα. Six days after effector cell therapy, elevated levels of activated endogenous CD8/CD44High and CD4/CD44High T cells localized and persisted at sites of tumor growth, whereas donor cell numbers concomitantly decreased. Both Tc1 and Tc2 effector cell subpopulations induced endogenous antitumor responses that were dependent, in part, on recipient-derived IFN-γ and TNF-α. However, neither effector cell-mediated therapy was dependent on recipient-derived perforin, IL-4, IL-5, or nitric oxide. Collectively, tumor antigen-specific Tc1 and Tc2 effector cell-mediated therapy is initially dependent, in part, on effector cell-derived FasL or LTα that may subsequently potentiate endogenous recipient-derived type 1 antitumor responses dependent on TNF-α and IFN-γ.


Immunological Investigations | 2008

Ag-specific type 1 CD8 effector cells enhance methotrexate-mediated antitumor responses by modulating endogenous CD49b-expressing CD4 and CD8 T effector cell subpopulations producing IL-10.

Mark J. Dobrzanski; Joyce B. Reome; James C. Hylind; Kathleen A. Rewers-Felkins; Khaliquzzaman Abdulsamad; Shawna L. Adams

The chemotherapeutic agent methotrexate is widely used in the treatment of breast cancer. Although its mechanism-of-action has been defined, less is known about its interaction with Ag-specific T cell-mediated antitumor responses. Type 1 CD8 T cell-mediated immune responses (Tc1) are cytolytic, produce IFN-gamma and are associated with effective antitumor responses. Using a murine transgenic TCR tumor model, we show that single-dose-treatment with methotrexate enhanced CD8-mediated type 1 antitumor responses when administered three days prior to Tc1 effector cell transfer. Co-treatment with methotrexate not only enhanced donor Tc1 cell accumulation and persistence at sites of primary tumor growth, but also promoted elevated levels of activated CD25+ expressing donor TIL cells. This correlated with a marked decrease in the appearance of endogenous differentiated (CD44High) CD3/CD8/CD49b and CD3/CD4/CD49b tumor-infiltrating effector T cells at both early (Days 1–8) and late (Days 12–20) stages following treatment when compared to that of corresponding groups receiving either MTX or Tc1 cell transfer alone. Moreover, such cellular response kinetics appeared to further correlate with the down-regulation of endogenous CD4/CD44High/CD49b effector T cells producing IL-10 and delays in tumor growth in vivo. This suggested that Ag-specific Tc1 cell transfer, in combination with chemotherapy, can enhance antitumor responses by modulating select CD49b-expressing T effector/memory cell subpopulations involved in homeostasis and immune tolerance within the tumor environment. These studies offer insight into mechanisms that enhance T cell-based immunotherapy in cancer. Supplementary materials are available for this article. Go to the publishers online edition of Immunological Investigations for the following free supplemental resource(s): Addendum 1.


Journal of Immunology | 1999

Therapeutic Effects of Tumor-Reactive Type 1 and Type 2 CD8+ T Cell Subpopulations in Established Pulmonary Metastases

Mark J. Dobrzanski; Joyce B. Reome; Richard W. Dutton


Clinical Immunology | 2001

Immunopotentiating Role of IFN-γ in Early and Late Stages of Type 1 CD8 Effector Cell-Mediated Tumor Rejection

Mark J. Dobrzanski; Joyce B. Reome; Richard W. Dutton


Clinical Immunology | 2004

Type 1 and type 2 tumor infiltrating effector cell subpopulations in progressive breast cancer

Joyce B. Reome; James C. Hylind; Richard W. Dutton; Mark J. Dobrzanski


Journal of Immunology | 2012

Enhanced levels of endogenous common {gamma} chain receptor cytokines influence CD8 T cell subpopulation numbers and phenotype in cancer patients receiving CD4 T cell immunotherapy: Observations In Four Cases Of Patients With Ovarian Cancer

Mark J. Dobrzanski; Kathleen A. Rewers-Felkins; Khaliquzzaman Samad; Imelda S. Quinlin; Catherine A. Phillips; William H. Robinson; David J. Dobrzanski; Stephen E. Wright

Collaboration


Dive into the Mark J. Dobrzanski's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kathleen A. Rewers-Felkins

Texas Tech University Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Khaliquzzaman Samad

Texas Tech University Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Stephen E. Wright

Texas Tech University Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Catherine A. Phillips

Texas Tech University Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Imelda S. Quinlin

Texas Tech University Health Sciences Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge