Network


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

Hotspot


Dive into the research topics where Peter S. Goedegebuure is active.

Publication


Featured researches published by Peter S. Goedegebuure.


Immunologic Research | 1995

THE ROLE OF CD4+ TUMOR-INFILTRATING LYMPHOCYTES IN HUMAN SOLID TUMORS

Peter S. Goedegebuure; Timothy J. Eberlein

Many, if not all, solid tumors are characterized by a T cell infiltrate, usually consisting of CD4+ and CD8+ T cells. Characterization of both subsets of tumor-infiltrating lymphocytes (TIL) have shown that each population can be divided into tumor-specific and tumor-nonspecific T cells. A small proportion of tumor-specific CD4+ TIL can directly lyse tumor cells in an HLA class I- or II-restricted fashion. The majority of tumor-specific CD4+ TIL, however, recognize tumor antigens presented on HLA class II molecules by antigen-presenting cells (APC). At the same time, APC in the tumor environment express elevated levels of heat shock antigen (Hsp) 70 (and perhaps other antigens) that can be specifically recognized by tumor-nonspecific CD4+ TIL when presented by HLA class II. Functionally, CD4+ TIL cells can be distinguished into Th0 (production of IL-2, IL-4, and IFN-γ), Th1 (IL-2 and IFN-γ), and Th2 (IL-4). In addition, stressed CD4+ TIL have the ability to produce the growth factors heparin binding epidermal growth factor and basic fibroblast growth factor that support tumor growth. Since the efficacy of an antitumor immune response is codetermined by the net effect of stimulatory and inhibitory cytokines, a detailed understanding of the developmental pathways of CD4+ TIL subsets and their interactions is critical for the design of clinical protocols.


Surgical Oncology-oxford | 1994

Reactivation of murine tumour-infiltrating lymphocytes with solid-phase anti-CD3 antibody: in vitro cytokine production is associated with in vivo efficacy

Peter S. Goedegebuure; M. Zuber; D.L. Leonard-Vidal; Ú.L. Burger; James C. Cusack; M.P. Chang; L.M. Douville; Timothy J. Eberlein

Previously we described the use of solid-phase anti-CD3 monoclonal antibody (mAb) to stimulate murine tumour-infiltrating lymphocytes (TIL) and their subsequent expansion in recombinant interleukin 2 (rIL-2). In a pulmonary metastases model using the methylcholanthrene-induced sarcoma MCA-105 anti-CD3 activated TIL were capable of eradicating disease similar to TIL cultured in rIL-2 only. Here we extend these observations by characterizing the biological effects of sequential solid-phase anti-CD3 activation. TIL from MCA-105 tumour activated with solid-phase anti-CD3 on day 1 were reactivated on day 14, or day 26, or both and compared to TIL grown in rIL-2 only or TIL activated with anti-CD3 once on day 1. Reactivation enhanced in vitro proliferation 1.8- to 4-fold compared to TIL activated once with anti-CD3 (P < 0.05). In addition, the total lytic capacity of the cultures was enhanced after reactivation without changing the phenotype of the TIL cultures. Reactivation resulted in a greater in vivo efficacy when the TIL were administered within 72 h of reactivation. In contrast, TIL activated with anti-CD3 on day 1 and day 14 were least effective of all TIL cultures (P < 0.05). This correlated with in vitro cytokine production. The most effective TIL cultures in vivo produced 4- to 100-fold higher amounts of cytokines, especially interferon gamma (IFN gamma) and granulocyte macrophage colony stimulating factor (GM-CSF), than the other cultures. On the other hand, the least effective in vivo TIL culture, TIL activated with anti-CD3 on day 1 and 14, produced little or no cytokines. These data suggest that in vitro production of cytokines is indicative of in vivo efficacy of anti-CD3 activated TIL.


Annals of Surgical Oncology | 1996

Vaccine therapy for cancer.

David C. Linehan; Peter S. Goedegebuure; Timothy J. Eberlein

AbstractBackground: Tumor-specific cytotoxic T-lymphocytes (CTLs) can be isolated from the solid tumors, draining lymph nodes, metastatic effusions, and peripheral blood of cancer patients. Despite this evidence for a cell-mediated immune response to cancer, attempts at active specific immunotherapy using cancer vaccines have met with little success in clinical trials.nMethods: We have reviewed the immunobiology of the cell-mediated immune response to cancer by focusing on what is known about the major histocompatibility complex (MHC)-restricted interaction between tumor cells and CD8+ or CD4+ T-cells. In addition, we review the recent advances in the identification of tumor-associated antigens (TAAs) that are recognized by tumor-specific CTLs in melanoma and other cancers. In discussing these antigens, we highlight the recent identification of several MHC-restricted antigenic peptides that are recognized by CTLs from patients with melanoma and those with ovarian and breast cancer. We examine the implications that the discovery of these TAAs and peptides will have on the development of new anticancer vaccines. We review the most recent vaccine trials in melanoma and other cancers and focus on current concepts aimed at improving the therapeutic efficacy of future vaccines, including genetically engineered tumor cell vaccines.nConclusions: With the recent identification of several TAAs and antigenic peptide epitopes in melanoma and other cancers, immunotherapy researchers are now focusing on new strategies for the development of anticancer vaccines. As the repertoire of known TAAs increases and our understanding of the immunobiology of cell-mediated immunity to cancer improves, immunotherapists remain cautiously optimistic in their quest for effective cancer vaccines.


Surgery | 1995

Immunoregulatory effects of CD4+ T helper subsets in human melanoma

Kyung-Yung Lee; Peter S. Goedegebuure; David C. Linehan; Timothy J. Eberlein

BACKGROUNDnThe elucidation of CD4+ T helper (Th) cell traits is important for the understanding of immunoregulatory mechanisms in patients with cancer, in particular the Th-cell effect on cytotoxic CD8+ tumor-specific lymphocytes (CTL).nnnMETHODSnSixty-six T-cell receptor alpha beta+/CD4+ clones were generated from tumor-infiltrating lymphocytes of five patients with melanoma and classified into subsets by cytokine production. Transwell experiments were performed to test how the soluble factors of each Th-clone subset affected the cytotoxicity of the tumor-specific CTL against autologous tumor.nnnRESULTSnTh0 clones enhanced cytotoxicity of the CD8+ CTL compared with control CTL cultured in cytokine-free medium. Th1-clone supernatant also enhanced cytotoxicity by CD8+ CTL. In contrast, Th2 clones decreased killing compared with control CTL. Replacement of the Th clones by exogenous interleukin (IL)-2 in concentrations similar to that produced by Th0 and Th1 clones enhanced cytotoxicity. However, suppression of cytotoxicity was observed when similar concentrations of IL-4 were added instead. The helper effect of Th0-soluble factors could be inhibited by anti-IL-2 antibody, whereas anti-IL-4 antibody did not show a significant enhancement.nnnCONCLUSIONSnThe majority of the CD4+ tumor-infiltrating lymphocytes (Th0) in patients with melanoma enhance the CTL response to autologous tumor by their soluble factors, whereas Th2 cells suppress the CTL response.


Surgery | 1997

Generation of peptide-specific cytotoxic T lymphocytes using allogeneic dendritic cells capable of lysing human pancreatic cancer cells☆☆☆

Matthias Peiper; Peter S. Goedegebuure; Timothy J. Eberlein

BACKGROUNDnDendritic cells (DCs) are potent antigen presenting cells (APCs), able to efficiently induce primary T cell-mediated responses to foreign antigens. In earlier studies we were able to identify a histocompatibility antigen (HLA)-A 2-restricted nine amino acid peptide (GP2, peptide 654-662) from the transmembrane portion of the protooncogene HER2/neu as a tumor-associated antigen (TAA) in human pancreatic cancer.nnnMETHODSnPeripheral blood mononuclear cells (PBMCs) of HLA-A2+ and HLA-A2 healthy volunteers were isolated. PBMCs were grown with initial anti-CD3, low-dose interleukin-2 (IL-2), and peptide-pulsed DC stimulation. T-cell lines were analyzed in functional studies.nnnRESULTSnAfter 4 weeks, T-cell cultures were more than 50% CD8+. All peptide-pulsed T cells significantly lysed APC pulsed with the immunizing antigen in an HLA-A2 restricted fashion. Furthermore, HLA-A2+,HER2/neu+ human pancreatic cancer cells were lysed significantly higher than HLA-A2 HER2/neu+ pancreatic cancer cells. Transfection of an HLA-A2 pancreatic cancer cell line with the HLA-A2 gene resulted in a significantly higher lysis of the transfected cell line compared to the wild type. In HLA-A2+ pancreatic cancer targets, specific lysis was HLA-A2 restricted.nnnCONCLUSIONnThe ability to use DCs for presentation of either tumor or peptide antigen in an HLA-restricted fashion to stimulate T-cell proliferation, as well as cytotoxicity, demonstrates the potential of this technology for future development of a pancreatic cancer vaccine.


Surgery | 1995

Recruitment of host CD8+ T cells by tumor-infiltrating lymphocytes and recombinant interleukin-2 during adoptive immunotherapy of cancer.

Ulrike L. Burger; Maximilian P. Chang; Peter S. Goedegebuure; Timothy J. Eberlein

BACKGROUNDnPreviously we demonstrated that optimal doses of tumor-infiltrating lymphocytes (TIL) concomitant with recombinant interleukin-2 (rIL-2) effectively mediated complete tumor regression of murine 3-day pulmonary metastases.nnnMETHODSnIn the present study we have investigated the contribution of the host immune response to the effectiveness of adoptive immunotherapy with TIL in combination with low-dose rIL-2. All experiments were performed in a murine pulmonary metastases model induced by intravenous injection of methylcholanthrene-induced sarcoma (MCA-105) cells into C57BL/6 mice. As a novel approach we used monoclonal antibody specific for CD4+ or CD8+ T cells to deplete the host of its T-cell subpopulations.nnnRESULTSnDepletion of host CD8+ T cells 24 hours after tumor injection and 48 hours before TIL+rIL-2 treatment abrogated all antitumor activity of this type of immunotherapy and resulted in significant metastatic pulmonary disease (p < 0.001). In contrast, depletion of host CD4+ T cells did not alter the efficacy of TIL+rIL-2 treatment in tumor eradication. The loss of tumoricidal activity of TIL+rIL-2 treatment in a CD8+ T cell-depleted host could be overcome by adding back normal uneducated splenocytes 2 hours after TIL therapy (p < 0.001). In contrast, adding back CD8- CD4+ splenocytes to a CD8+ T cell-depleted host 2 hours after TIL+rIL-2 treatment resulted in significant pulmonary disease comparable to untreated animals.nnnCONCLUSIONSnWe conclude that the recruitment of host CD8+ T cells by adoptively transferred TIL+rIL-2 appears to be important for effective tumor eradication in this type of immunotherapy.


Surgical Oncology-oxford | 1995

In vitro stimulation of ovarian tumour-associated lymphocytes with a peptide derived from HER2/neu induces cytotoxicity against autologous tumour.

David C. Linehan; G.E. Peoples; D.T. Hess; I.C. Summerhayes; A.S. Parikh; Peter S. Goedegebuure; Timothy J. Eberlein

The proto-oncogene HER2/neu encodes for a 185 kDa transmembrane protein with extensive homology to the epidermal growth factor (EGF) receptor. We have previously shown a correlation between HER2/neu expression and the level of in vitro cytotoxicity of tumour-associated lymphocytes (TAL) versus autologous tumour. In addition, we have recently demonstrated that tumour-associated cytotoxic T-lymphocytes (CTL) from ovarian and breast cancer patients can recognize a HER2/neu derived peptide epitope when presented in the context of HLA-A2. Since repeated tumour stimulation of CTL enhances both proliferation and cytotoxicity against autologous tumour, we hypothesized that repeated peptide antigen stimulation would have a similar effect. To be therapeutically useful, the peptide antigen must meet the following conditions: (1) the peptide must be immunogenic and cause a proliferation of CTL to adequate therapeutic numbers, and (2) the peptide-specific CTL which are generated must be cytotoxic against autologous tumour. To test our hypothesis, T-lymphocytes isolated from the ascites of four consecutive HER2/neu+ ovarian cancer patients were initially stimulated with solid phase anti-CD3 antibody and divided into three groups: (1) treatment with recombinant interleukin-2 (IL-2) alone, (2) IL-2 plus weekly stimulation with irradiated autologous tumour cells, and (3) IL-2 plus weekly stimulation with a HER2/neu derived peptide. Peptide-stimulated and tumour-stimulated CTL showed similar increases in proliferation with both groups consistently reaching therapeutic numbers. Peptide-stimulated CTL demonstrated significantly enhanced cytotoxicity against autologous tumour in 4-h chromium release assays as compared to the IL-2 alone group.(ABSTRACT TRUNCATED AT 250 WORDS)


International Journal of Cancer | 1999

Tumor-specific cytokine release by donor T cells induces an effective host anti-tumor response through recruitment of host naive antigen presenting cells

Makoto Nagoshi; Noriaki Sadanaga; Hong-Gu Joo; Peter S. Goedegebuure; Timothy J. Eberlein

We recently reported that tumor eradication induced by immunotherapy (IT) in a congenic mouse model using tumor infiltrating lymphocytes (TIL) + recombinant interleukin‐2 (rIL‐2) is dependent on recruitment of naive host immune cells at the tumor sites. The recruitment of host immune cells was induced mainly through a local secretion of interferon‐γ (IFN‐γ) produced by donor T cells. We now further investigated how a non‐specific inflammatory response progresses to a host T‐cell‐mediated tumor‐specific response. In crossover experiments using MCA‐105 and MCA‐205 sarcoma tumors, pulmonary metastatic disease was eradicated only in mice treated with tumor‐matched TIL + rIL‐2. In vitro, TIL stimulated with the tumor of origin secreted relatively high levels of IFN‐γ and granulocyte‐macrophage colony stimulating factor (GM‐CSF) compared to TIL stimulated with mismatched tumor cells. In lungs of tumor‐bearing mice treated with matched TIL + rIL‐2, significant increases in the percentages of IFN‐γ, GM‐CSF and tumor necrosis factor‐α (TNF‐α) positive cells were detected, as well as of macrophages, natural killer (NK) cells and dendritic cells. Depletion of macrophages or NK cells did not inhibit the efficacy. In contrast, depletion of dendritic cells partially inhibited the efficacy of the treatment. Combined depletion of dendritic cells and macrophages abrogated more than 80% of the efficacy. Our data suggest that successful IT may require 3 steps: (1) release of inflammatory cytokines by donor TIL after restimulation by tumor cells; (2) infiltration of host immune cells in response to local cytokine production; and (3) activation of tumor‐specific host immune cells by dendritic cells and to a lesser extent by macrophages. Int. J. Cancer 80:308–314, 1999.


Annals of Surgical Oncology | 1996

Improved in vivo efficacy of tumor-infiltrating lymphocytes after restimulation with irradiated tumor cells in vitro

Ulrike L. Burger; Maximilian P. Chang; Makoto Nagoshi; Peter S. Goedegebuure; Timothy J. Eberlein

AbstractBackground: We investigated different culture conditions for tumor-infiltrating lymphocytes (TILs) with regard to proliferation, phenotypic changes, in vitro cytotoxicity, and in vivo therapeutic efficacy.nMethods: After enzymatic digestion of the murine fibrosarcoma, MCA-105, TIL cultures were initiated as pure lymphocyte (groups 1 and 2) or mixed lymphocyte/tumor suspensions (groups 3 and 4). Group 1 TILs were grown in culture medium containing 100 IU/ml recombinant interleukin-2 (rIL-2). Group 2 TILs were stimulated with solid-phase anti-CD3 monoclonal antibody (mAb) for 48 h and cultured in rIL-2 (100 IU/ml)-containing medium. Group 3, which consisted initially of a surplus of tumor cells, received the same treatment as group 2. Group 4 was also activated with anti-CD3 mAb and rIL-2 but was additionally restimulated weekly with irradiated tumor cells (TILs to tumor, 20:1).nResults: Groups 1 and 2 showed up to twofold higher increases in TIL numbers compared with groups 3 and 4 by the end of culture week 5. Although the original lymphocyte/tumor cell suspension consisted of 12.0 ± 3.8% CD4+ T cells and 5.3 ± 3.3% CD8+ T cells, all four TIL cultures showed ∼80% CD8+ TILs and no CD4+ TILs by the end of culture week 4. In vitro cytotoxicity did not correlate with in vivo efficacy of the examined TIL cultures. By using the MCA-105 pulmonary metastases model in C57BL/6 mice, only suboptimal doses of TILs (2 × 106) from group 4, which had been restimulated weekly with irradiated tumor, showed significant tumor eradication compared with all other treatment groups (p<0.01).nConclusions: We conclude that in vitro tumor restimulation of TILs improves in vivo efficacy, most likely through the education of tumor-reactive T cells.


Surgery | 1995

Antitumor activity of three mouse mammary cancer cell lines after interferon-gamma gene transfection

Yvedt L. Matory; Man Chen; David M. Dorfman; Amanda Williams; Peter S. Goedegebuure; Timothy J. Eberlein

BACKGROUNDnThe antitumor effects of three mouse mammary tumors transfected to express interferon-gamma were evaluated.nnnMETHODSnThree immunologically different tumors were used: DA3, EMT6, and 410. All three cell lines were successfully transfected with highly efficient viral vectors. Wild type or transfected tumor cells were injected subcutaneously into Balb/c mice. Animals were observed for tumor growth and the induction of immunologic memory.nnnRESULTSnA significant decrease occurred in the size of all transfected tumors, EMT6 1.9 cm2, DA3 1.7, and 410 1.8 compared with nontransfected control tumors with a mean size of 4 cm2 on day 30. To further test the development of immunity, animals were injected with either nontransfected or transfected tumors and challenged with nontransfected tumor. Animals immunized with transfected tumor cells had significantly smaller tumors, EMT6 2.5 cm2, DA3 3.1, and 410 2.4 compared with controls with a mean size of 4 cm2. No specific splenocyte cytotoxicity was shown. Expression of major histocompatibility complex class I antigens was enhanced in the 410 and DA3 tumor lines.nnnCONCLUSIONSnSignificant antitumor effects were observed after interferon-gamma gene transfection of three mouse mammary cancer cell lines. Up-regulation of major histocompatibility complex class I antigen expression is a partial explanation of these findings. These results provide preliminary studies for gene therapy of human breast cancer.

Collaboration


Dive into the Peter S. Goedegebuure's collaboration.

Top Co-Authors

Avatar

Timothy J. Eberlein

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

David C. Linehan

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

George E. Peoples

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Yvedt L. Matory

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Ichiro Yoshino

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Man Chen

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

David M. Dorfman

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Kyung-Yung Lee

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Cara C. Douville

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge