Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Vet Comp Oncol ; 18(4): 664-674, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32187827

ABSTRACT

We previously reported that eBAT, an EGF-targeted angiotoxin, was safe and it improved the overall survival for dogs with splenic haemangiosarcoma when added to the standard of care in a single cycle of three administrations in the minimal residual disease setting. Our objective for the SRCBST-2 trial was to assess whether increased dosing through multiple cycles of eBAT would be well tolerated and would further enhance the benefits of eBAT. Eligibility was expanded to dogs with stage 3 haemangiosarcoma, provided that gross lesions could be surgically excised. The interval between eBAT and the start of chemotherapy was reduced, and the experimental therapy was expanded to three cycles, each administered at the biologically active dose (50 µg/kg) on a Monday/Wednesday/Friday schedule following splenectomy, and scheduled 1 week prior to the first, second and fifth doxorubicin chemotherapy. Twenty-five dogs were enrolled; six experienced acute hypotension with two requiring hospitalization. Self-limiting elevation of ALT was observed in one dog. A statistically significant survival benefit was not seen in this study in eBAT-treated dogs compared with a Contemporary comparison group of dogs with stages 1-3 haemangiosarcoma treated with standard of care alone. Our results indicate that repeated dosing cycles of eBAT starting 1 week prior to doxorubicin chemotherapy led to greater toxicity and reduced efficacy compared with a single cycle given between surgery and a delayed start of chemotherapy. Further work is needed to understand the precise mechanisms of action of eBAT in order to optimize its clinical benefits in the treatment of canine haemangiosarcoma and other tumours. IACUC Protocols 1110A06186 and 1507-32804A.


Subject(s)
Chemotherapy, Adjuvant/veterinary , Dog Diseases/drug therapy , Epidermal Growth Factor/pharmacology , Hemangiosarcoma/veterinary , Splenic Neoplasms/veterinary , Animals , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Chemotherapy, Adjuvant/methods , Dog Diseases/pathology , Dogs , Doxorubicin/therapeutic use , Female , Hemangiosarcoma/drug therapy , Male , Splenic Neoplasms/drug therapy , Splenic Neoplasms/pathology , Treatment Outcome
2.
Clin Pharmacol ; 10: 113-121, 2018.
Article in English | MEDLINE | ID: mdl-30288129

ABSTRACT

PURPOSE: Human sarcomas are rare and difficult to treat cancerous tumors typically arising from soft tissue or bone. Conversely, carcinomas are the most common cancer subtype in humans and the primary cause of mortality across all cancer patients. While conventional therapeutic modalities can prolong disease-free intervals and survival in some cases, treatment of refractory or recurrent solid tumors is challenging, and tumor-related mortality remains unacceptably high. The identification of overexpressed cell surface receptors on sarcoma and carcinoma cells has provided a valuable tool to develop targeted toxins as an alternative anticancer strategy. Recent investigation of recombinant protein-linked toxins that specifically target these cancer receptors has led to the development of highly specific, cytotoxic, and deimmunized drugs that can kill cancer cells. METHODS: This study investigated a recombinant protein called epidermal growth factor bispecific angiotoxin (eBAT), which is designed to target the epidermal growth factor receptor (EGFR) on cancer cells and the urokinase plasminogen activator receptor (uPAR) on cancer cells and associated tumor vasculature. Both receptors are expressed by a variety of human sarcomas and carcinomas. Flow cytometry techniques were used to determine binding affinity of eBAT to cancer cells, and proliferation assays were performed to calculate tumor killing ability based on half-maximal inhibitory concentrations. RESULTS: eBAT demonstrated cytotoxicity against a variety of sarcoma and carcinoma cells that overexpress EGFR and uPAR in vitro and showed greater cell killing ability and binding affinity to cancer cells compared with its monospecific counterparts. CONCLUSION: The results of our study are promising, and further studies will be necessary to confirm the applicability of eBAT as a supplementary therapy for a variety of sarcomas, carcinomas, and possibly other refractory malignancies that express EGFR and uPAR.

3.
Oncotarget ; 9(15): 11938-11947, 2018 Feb 23.
Article in English | MEDLINE | ID: mdl-29552283

ABSTRACT

Children with high risk sarcoma have a poor prognosis despite surgical resection, irradiation and chemotherapy. Alternative therapies are urgently needed. Urokinase-type plasminogen activator receptor (uPAR) and epidermal growth factor receptor (EGFR) are surface proteins expressed by some pediatric sarcomas. We show for the first time that a de-immunized bispecific ligand toxin, EGFATFKDEL, directed against EGFR and uPAR, successfully targets pediatric sarcoma. Using flow cytometry, we identified a rhabdomyosarcoma (RMS) cell line, RH30, that expresses both uPAR and EGFR, and a Ewing sarcoma (EWS) cell line, TC-71, that expresses only uPAR. We tested the differential sensitivity of these two sarcoma cell lines to toxin-induced killing, using both in vitro assays and an in vivo murine model. We show that pediatric sarcomas are highly sensitive to EGFATFKDEL (at subnanomolar concentrations) in vitro. In vivo, tumor growth was significantly attenuated after treatment with EGFTFKDEL, compared to untreated controls, in both RH30 and TC-71 tumor bearing mice. In addition, we found that simultaneously targeting both receptors in a dual positive cell line was more effective than targeting a single receptor or antigen, resulting in a greater tumor response, including complete tumor regression in an animal model of bulky disease. Our findings provide support for further exploration of bispecific targeting of pediatric sarcomas with bispecific ligand toxins, such as EGFATFKDEL.

4.
Toxins (Basel) ; 10(1)2018 01 06.
Article in English | MEDLINE | ID: mdl-29316610

ABSTRACT

Diphtheria toxin (DT) related targeted toxins are effective in cancer treatment, but efficacy diminishes in time because of their immunogenic potential and/or former vaccinations. In order to overcome this limitation for DT2219, a promising bispecific targeted toxin which targets CD19 and CD22, we deimmunized the DT moiety, and thereby developed an exciting improved drug (dDT2219) which still has the potential to sufficiently target B-cell malignancies but also limits clearance because of its reduced immunogenicity. The DT moiety was modified by inducing point mutations in prominent positions on the molecular surface. The new engineered dDT2219 was tested for activity, efficacy, and specificity using functional assays, proliferation assays, and flow cytometry. Furthermore, 12 samples of Chronic Lymphatic Leukemia (CLL) patients were used to assess binding. Immunogenicity was determined using a BALB/c mouse model. dDT2219 was efficient and specific against B-cell malignancies such as Bukitt-Lymphoma cell lines Daudi and Raji. dDT2219 showed specific binding on targets and on CLL samples. Intraperitoneal vaccination of immune competent mice showed that even after multiple administrations with increasing doses, induction of neutralizing antibodies was significantly lower in the dDT2219 treated animal group. The new dDT2219 combines potent anti-tumor cell activity with a reduced immunogenicity. With regard to the frequent development of neutralizing antibodies after multiple administrations with immunotoxins, dDT2219 shows promise to overcome this limitation and thus might maintain effectiveness even after multiple treatment cycles.


Subject(s)
Antineoplastic Agents/pharmacology , Diphtheria Toxin/pharmacology , Immunotoxins/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Lymphoma, B-Cell/drug therapy , Animals , Antibodies, Neutralizing/immunology , Antigens, CD19/immunology , Cell Line, Tumor , Diphtheria Toxin/genetics , Humans , Immunotoxins/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymphoma, B-Cell/immunology , Mice, Inbred BALB C , Sialic Acid Binding Ig-like Lectin 2/immunology
5.
Oncotarget ; 7(45): 73830-73844, 2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27650544

ABSTRACT

BACKGROUND: The design of a highly effective anti-cancer immune-engager would include targeting of highly drug refractory cancer stem cells (CSC). The design would promote effective antibody-dependent cell-mediated cytotoxicity (ADCC) and simultaneously promote costimulation to expand and self-sustain the effector NK cell population. Based on our bispecific NK cell engager platform we constructed a tetraspecific killer engager (TetraKE) comprising single-chain variable fragments (scFvs) binding FcγRIII (CD16) on NK cells, EpCAM on carcinoma cells and CD133 on cancer stem cells in order to promote ADCC. Furthermore, an Interleukin (IL)-15-crosslinker enhanced NK cell related proliferation resulting in a highly active drug termed 1615EpCAM133. RESULTS: Proliferation assays showed TetraKE promoted proliferation and enhanced NK cell survival. Drug-target binding, NK cell related degranulation, and IFN-γ production was specific for both tumor related antigens in EpCAM and CD133 bearing cancer cell lines. The TetraKE showed higher killing activity and superior dose dependent degranulation. Cytokine profiling showed a moderately enhanced IFN-γ production, enhanced GM-CSF production, but no evidence of induction of excessive cytokine release. METHODS: Assembly and synthesis of hybrid genes encoding the TetraKE were performed using DNA shuffling and ligation. The TetraKE was tested for efficacy, specificity, proliferation, survival, and cytokine production using carcinoma cell lines and functional assays measuring NK cell activity. CONCLUSION: 1615EpCAM133 combines improved induction of ADCC with enhanced proliferation, limited cytokine response, and prolonged survival and proliferation of NK cells. By linking scFv-related targeting of carcinoma and CSCs with a sustaining IL-15 signal, our new construct shows great promise to target cancer and CSCs.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Interleukin-15/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , AC133 Antigen/genetics , AC133 Antigen/metabolism , Biomarkers , Cell Degranulation/immunology , Cell Line, Tumor , Cytokines/biosynthesis , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , Humans , Interferon-gamma/biosynthesis , Interleukin-15/genetics , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Protein Binding/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism
6.
Mol Ther ; 24(7): 1312-22, 2016 08.
Article in English | MEDLINE | ID: mdl-27157665

ABSTRACT

Previously, we constructed a bispecific NK-cell-engager (BiKE) bearing single-chain variable fragments (scFv) against CD16 on NK cells and EpCAM on tumor cells. This BiKE facilitated antigen-specific antibody-dependent cell-mediated cytotoxicity (ADCC) but did not induce NK cell expansion. We incorporated a modified interleukin-15 cross-linker to create a trispecific construct (TriKE) in order to improve activation, proliferation, and survival of NK cells. Synthesis and assembly of hybrid genes encoding the TriKE was accomplished using DNA-shuffling and DNA-ligation techniques. The TriKE was tested for specificity, efficacy, proliferative capability, and cytokine profile using functional assays. The molecular modifications improved yield without compromising binding to EpCAM(+) HT-29 colorectal carcinoma cells. (51)Chromium-release and degranulation assays showed better killing rates with TriKE compared to BiKE. TriKE was more active in a variety of different carcinoma cell lines. TriKE showed the ability to stimulate expansion of CD56(+)CD3(-) NK cells. BiKE and TriKE showed enhanced but not supraphysiologic levels of cytokine secretion. 1615EpCAM TriKE drives enhanced ADCC while significantly improving proliferation, activation, and survival of NK cell effectors. The TriKE provides a selectively delivered self-sustaining signal at the NK/tumor cell synapse. Targeted cytokine stimulation, rather than systemic cytokine administration, may impact toxicity in patients rendering the TriKE a promising new off-the-shelf carcinoma therapy.


Subject(s)
Antibodies, Bispecific/genetics , Antibody-Dependent Cell Cytotoxicity/genetics , Carcinoma/immunology , Interleukin-15/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation , Mutagenesis, Insertional , Carcinoma/genetics , Cell Degranulation/immunology , Cell Line, Tumor , Cell Proliferation , Cytokines/biosynthesis , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , Gene Expression , Gene Order , Genetic Vectors/genetics , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Protein Binding , Recombinant Fusion Proteins
7.
Gynecol Oncol ; 130(3): 579-87, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23721800

ABSTRACT

OBJECTIVES: While most women with ovarian cancer will achieve complete remission after treatment, the majority will relapse within two years, highlighting the need for novel therapies. Cancer stem cells (CSC) have been identified in ovarian cancer and most other carcinomas as a small population of cells that can self-renew. CSC are more chemoresistant and radio-resistant than the bulk tumor cells; it is likely that CSC are responsible for relapse, the major problem in cancer treatment. CD133 has emerged as one of the most promising markers for CSC in ovarian cancer. The hypothesis driving this study is that despite their low numbers in ovarian cancer tumors, CSC can be eradicated using CD133 targeted therapy and tumor growth can be inhibited. METHODS: Ovarian cancer cell lines were evaluated using flow cytometry for expression of CD133. In vitro viability studies with an anti-CD133 targeted toxin were performed on one of the cell lines, NIH:OVCAR5. The drug was tested in vivo using a stably transfected luciferase-expressing NIH:OVCAR5 subline in nude mice, so that tumor growth could be monitored by digital imaging in real time. RESULTS: Ovarian cancer cell lines showed 5.6% to 16.0% CD133 expression. dCD133KDEL inhibited the in vitro growth of NIH:OVCAR5 cells. Despite low numbers of CD133-expressing cells in the tumor population, intraperitoneal drug therapy caused a selective decrease in tumor progression in intraperitoneal NIH:OVCAR5-luc tumors. CONCLUSIONS: Directly targeting CSC that are a major cause of drug resistant tumor relapse with an anti-CD133 targeted toxin shows promise for ovarian cancer therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD/metabolism , Biomarkers, Tumor/metabolism , Cytotoxins/therapeutic use , Glycoproteins/metabolism , Neoplastic Stem Cells/drug effects , Ovarian Neoplasms/drug therapy , Peptides/metabolism , AC133 Antigen , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cytotoxins/pharmacology , Disease Models, Animal , Disease Progression , Female , Glycoproteins/immunology , Humans , Mice , Mice, Nude , Molecular Targeted Therapy , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/metabolism , Peptides/immunology
8.
Leuk Res ; 33(9): 1233-42, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19327829

ABSTRACT

A bispecific ligand-directed toxin (BLT) called DT2219ARL consisting of two scFv ligands recognizing CD19 and CD22 and catalytic DT390 was genetically enhanced for superior in vivo anti-leukemia activity. Genetic alterations included reverse orienting VH-VL domains and adding aggregation reducing/stabilizing linkers. In vivo, these improvements resulted in previously unseen long-term tumor-free survivors measured in a bioluminescent xenograft imaging model in which the progression of human Raji Burkitt's lymphoma could be tracked in real time and in a Daudi model as well. Studies showed DT2219ARL was potent (IC50s 0.06-0.2 nM range) and selectively blockable. Imaging studies indicated the highly invasive nature of this B cell malignancy model and showed it likely induced pre-terminal hind limb paralysis because of metastasis to spinal regions prevented by DT2219ARL. DT2219ARL represents a new class of bispecific biological that can be continually improved by genetic mutation.


Subject(s)
Antigens, CD19/drug effects , Burkitt Lymphoma/pathology , Immunotoxins/pharmacology , Sialic Acid Binding Ig-like Lectin 2/drug effects , Amino Acid Sequence , Animals , Cell Line, Tumor , Drug Delivery Systems , Female , Flow Cytometry , Humans , Ligands , Mice , Mice, SCID
9.
Cancer Biother Radiopharm ; 22(4): 488-500, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17803443

ABSTRACT

Studies were performed to determine the suitability of using the polyethylene glycol (PEG)-labeled AHN-12 anti-CD45 monoclonal antibody to deliver the high-energy beta-particle-emitting isotope 90Y to a CD45+ B-cell Daudi lymphoma grown as flank tumors in athymic nude mice. The PEGylated radiolabeled antibody displayed a significantly better antitumor effect in the mouse tumor flank model (p<0.03) and significantly better blood pharmacokinetics in normal rats (p<0.05) than the non-PEGylated radiolabeled antibody. Studies of two different sizes of PEG showed that rats given 43 kDa of PEGylated AHN-12, but not 5 kDa of PEGylated AHN-12, had significantly higher radiolabeled antibody blood levels and, therefore, improved pharmacokinetics, as compared to rodents given non-PEGylated radiolabeled AHN-12 (p<0.05). Surviving mice revealed no signs of kidney, liver, or gastrointestinal damage by histology study. Notably, in vitro studies indicated that PEGylation did not have a major effect on labeling efficiency and the binding of labeled antibody. These findings indicate that PEGylation of radiolabeled anti-CD45 antibody may be a useful and desirable means of extending blood half-life and enhancing efficacy. Also, the final outcome may be impacted by the size of the PEG molecule used for the modification of the blood half-life.


Subject(s)
Antibodies/therapeutic use , Leukocyte Common Antigens/immunology , Neoplasms/immunology , Neoplasms/radiotherapy , Polyethylene Glycols/chemistry , Yttrium Radioisotopes/chemistry , Yttrium Radioisotopes/therapeutic use , Animals , Antibodies/immunology , Antibodies/pharmacology , Body Weight/drug effects , Cell Line, Tumor , Chromatography, Ion Exchange , Humans , Immunotherapy , Leukocyte Common Antigens/metabolism , Mice , Mice, Nude , Molecular Weight , Neoplasms/metabolism , Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
Blood ; 103(9): 3590-8, 2004 May 01.
Article in English | MEDLINE | ID: mdl-14715632

ABSTRACT

We have developed a mouse system by which to track the migration and homing of cells in a setting of bone marrow transplantation (BMT)-induced graft-versus-host disease (GVHD) after systemic infusion using enhanced green fluorescence protein (eGFP) transgenic (Tg) cells and a simple application of a fluorescence stereomicroscope outfitted with a color charge-coupled device (CCD) camera. Whole body images of anesthetized mice taken at various time points after cell infusion revealed the early migration of allogeneic cells to peripheral lymphoid organs, with later infiltration of GVHD target organs. Localization of eGFP Tg cells could be seen through the skin of shaved mice, and internal organs were easily discernible. After allogeneic or syngeneic eGFP Tg cell infusion, representative mice were dissected to better visualize deeper internal organs and tissues. Infusion of different cell populations revealed distinct homing patterns, and this method also provided a simple way to identify the critical time points for expansion of the transplanted cells in various organs. This simple application of the fluorescence stereomicroscope will be valuable for GVHD and graft-versus-tumor studies in which visualization of cellular migration, expansion, and cell-cell interactions will be more informative when analyzed by such an intravital method.


Subject(s)
Bone Marrow Transplantation/adverse effects , Graft vs Host Disease/pathology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Movement , Diagnostic Imaging/methods , Graft Survival , Green Fluorescent Proteins , Kinetics , Luminescent Proteins , Lung , Lymph Nodes , Mice , Microscopy, Fluorescence , Organ Specificity , Transplantation, Homologous , Transplantation, Isogeneic
11.
Am J Physiol Lung Cell Mol Physiol ; 286(2): L284-92, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14527928

ABSTRACT

Idiopathic pneumonia syndrome (IPS) is a significant cause of mortality post-bone marrow transplant (BMT) in humans. In our murine model, lethal pre-BMT conditioning and allogeneic T cells result in the recruitment of host antigen-presenting cells (APC) and donor T cells into the lung post-BMT concomitant with development of severe lung dysfunction. CCL2 induction is found in bronchoalveolar lavage fluid (BALF) before host monocyte influx. The major receptor for CCL2 is CCR2 present on monocytes; this interaction can play a crucial role in monocyte recruitment in inflammation. To determine whether blockade of the CCL2/CCR2 pathway could hinder host monocyte influx, lethally conditioned wild-type (WT), CCL2(-/-), or CCR2(-/-) mice were transplanted with allogeneic marrow and spleen cells. WT and (-/-) recipients exhibited equivalent lung dysfunction post-BMT. The frequencies of host macrophages as well as donor CD4(+) and CD8(+) T cells in lungs post-BMT did not differ between WT and (-/-) recipients. However, the T cell dependency of the host CD11b(+) major histocompatibility complex class II(+) cell influx was lost in CCR2(-/-) recipients. In CCR2(-/-) mice, this influx was accompanied by elevated levels of CCL20. Post-BMT BALF and sera of (-/-) mice did not reveal any decrease in cytokines or chemokines compared with WT mice. CCL2(-/-) mice had a deficiency of CCL2 in their BALF and sera post-BMT, confirming our hypothesis that CCL2 is predominantly host derived. Therefore, IPS can occur independently of host expression of CCL2 or CCR2, and compensatory mechanisms exist for regulating APC recruitment into the lung during the early post-BMT period.


Subject(s)
Bone Marrow Transplantation/adverse effects , Chemokine CCL2/genetics , Pneumonia/physiopathology , Receptors, Chemokine/genetics , Animals , Disease Models, Animal , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Graft vs Host Disease/physiopathology , Major Histocompatibility Complex/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Monocytes/immunology , Pneumonia/immunology , Pneumonia/mortality , Receptors, CCR2 , Severity of Illness Index , Survival Rate
12.
Blood ; 101(9): 3714-21, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12511416

ABSTRACT

Idiopathic pneumonia syndrome (IPS) is a significant cause of morbidity and mortality after bone marrow transplantation (BMT) in humans. We developed a murine IPS model in which lethal pre-BMT conditioning and allogeneic T cells results in the recruitment of host monocytes and then donor T cells into the lung by day 7 after BMT, concomitant with development of severe lung dysfunction. We reported the T cell-dependent production of the T cell-attracting chemokine macrophage inflammatory protein-1 alpha (MIP-1 alpha) in the lungs of such recipient mice. We reasoned that MIP-1 alpha might be a critical mediator of IPS. Lethally conditioned mice received transplants of major histocompatibility complex-disparate marrow and either wild-type (MIP-1 alpha(+/+)) or knockout (MIP-1 alpha(-/-)) spleen cells. Recipients of MIP-1 alpha(-/-) cells exhibited accelerated mortality and a decrease in specific compliance that appeared earlier than in recipients of MIP-1 alpha(+/+) cells. Donor CD4(+) and CD8(+) T cell expansion was increased in the spleens of recipients of MIP-1 alpha(-/-) cells. Lungs of recipients of MIP-1 alpha(-/-) cells had earlier recruitment of both T-cell subsets by day 3 after BMT, concomitant with the influx of cells expressing the cytolysins granzymes A and B. Monocyte recruitment was not altered. Levels of inflammatory cytokines were not increased and levels of T cell-attracting chemokines were decreased. The level of the anti-inflammatory cytokine interleukin 13 (IL-13) was lower in the serum and lungs of recipients of MIP-1 alpha(-/-) cells, indicating a skewing toward a more inflammatory T helper cell type 1 (Th1) cytokine milieu. Donor-derived MIP-1 alpha may play a role in allogeneic-induced IPS by limiting aggressive expansion of CD4(+) and CD8(+) T cells.


Subject(s)
Bone Marrow Transplantation/adverse effects , CD4-Positive T-Lymphocytes , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Macrophage Inflammatory Proteins/physiology , Pneumonia/etiology , Transplantation, Homologous/adverse effects , Animals , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/transplantation , Chemokine CCL3 , Chemokine CCL4 , Chemokines/analysis , Chemotaxis, Leukocyte , Cytokines/analysis , Graft vs Host Disease/etiology , Graft vs Host Disease/pathology , Granzymes , Lung/chemistry , Lung/immunology , Lung/physiopathology , Lung Compliance , Macrophage Inflammatory Proteins/deficiency , Macrophage Inflammatory Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/immunology , Pneumonia/pathology , Radiation Chimera , Serine Endopeptidases/analysis , Th1 Cells/immunology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...