1
|
Nakamura T, Sasaki S, Sato Y, Harashima H. Cancer Immunotherapy with Lipid Nanoparticles Loaded with a Stimulator of Interferon Genes Agonist against Renal Tumor Lung Metastasis. Pharmaceutics 2023; 16:31. [PMID: 38258042 PMCID: PMC10819482 DOI: 10.3390/pharmaceutics16010031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Metastatic renal cell carcinoma (RCC) has a poor prognosis, and the major organ of metastasis is the lung. Immunotherapy with immune checkpoint inhibitors (ICIs) is the first-line therapy, but the response rates are low. Thus, the development of a more effective immunotherapy against metastatic RCC would be highly desirable. We previously demonstrated how a stimulator of an interferon gene (STING) agonist-loaded lipid nanoparticles (STING-LNPs) significantly activates natural killer (NK) cells and induces an antitumor effect against cases of melanoma lung metastasis that have shown ICI resistance. In this study, we evaluated the potential of using STING-LNPs in the treatment of lung metastatic RCC (Renca). An intravenous injection of STING-LNPs drastically decreased the amount of Renca tumor colonies. In contrast, monotherapies using ICIs showed no antitumor effect, and even a combination of ICI and STING-LNP therapies failed to enhance the antitumor effects. The main effector cells would be NK cells, and the activation of NK cells by the STING-LNPs may avoid the increased expression of immune checkpoint molecules. These findings provide useful insights into the development of an effective immunotherapy against metastatic RCC.
Collapse
Affiliation(s)
- Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | | | | | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| |
Collapse
|
2
|
Trott JF, Abu Aboud O, McLaughlin B, Anderson KL, Modiano JF, Kim K, Jen KY, Senapedis W, Chang H, Landesman Y, Baloglu E, Pili R, Weiss RH. Anti-Cancer Activity of PAK4/NAMPT Inhibitor and Programmed Cell Death Protein-1 Antibody in Kidney Cancer. KIDNEY360 2020; 1:376-388. [PMID: 35224510 PMCID: PMC8809296 DOI: 10.34067/kid.0000282019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/12/2020] [Indexed: 06/14/2023]
Abstract
BACKGROUND Kidney cancer (or renal cell carcinoma, RCC) is the sixth most common malignancy in the United States and is increasing in incidence. Despite new therapies, including targeted therapies and immunotherapies, most RCCs are resistant to treatment. Thus, several laboratories have been evaluating new approaches to therapy, both with single agents as well as combinations. Although we have previously shown efficacy of the dual PAK4/nicotinamide phosphoribosyltransferase (NAMPT) inhibitor KPT-9274, and the immune checkpoint inhibitors (CPI) have shown utility in the clinic, there has been no evaluation of this combination either clinically or in an immunocompetent animal model of kidney cancer. METHODS In this study, we use the renal cell adenocarcinoma (RENCA) model of spontaneous murine kidney cancer. Male BALB/cJ mice were injected subcutaneously with RENCA cells and, after tumors were palpable, they were treated with KPT-9274 and/or anti-programmed cell death 1 (PDCD1; PD1) antibody for 21 days. Tumors were measured and then removed at animal euthanasia for subsequent studies. RESULTS We demonstrate a significant decrease in allograft growth with the combination treatment of KPT-9274 and anti-PD1 antibody without significant weight loss by the animals. This is associated with decreased (MOUSE) Naprt expression, indicating dependence of these tumors on NAMPT in parallel to what we have observed in human RCC. Histology of the tumors showed substantial necrosis regardless of treatment condition, and flow cytometry of antibody-stained tumor cells revealed that the enhanced therapeutic effect of KPT-9274 and anti-PD1 antibody was not driven by infiltration of T cells into tumors. CONCLUSIONS This study highlights the potential of the RENCA model for evaluating immunologic responses to KPT-9274 and checkpoint inhibitor (CPI) and suggests that therapy with this combination could improve efficacy in RCC beyond what is achievable with CPI alone.
Collapse
Affiliation(s)
- Josephine F. Trott
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, California
| | - Omran Abu Aboud
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, California
| | - Bridget McLaughlin
- Comprehensive Cancer Center, University of California, Davis, California
| | - Katie L. Anderson
- Animal Cancer Care and Research Program, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota
| | - Jaime F. Modiano
- Animal Cancer Care and Research Program, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Kyoungmi Kim
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, California
| | - Kuang-Yu Jen
- Department of Pathology and Laboratory Medicine, University of California, Davis, California
| | - William Senapedis
- Research and Translational Development, Karyopharm Therapeutics Inc., Newton, Massachusetts
| | - Hua Chang
- Research and Translational Development, Karyopharm Therapeutics Inc., Newton, Massachusetts
| | - Yosef Landesman
- Research and Translational Development, Karyopharm Therapeutics Inc., Newton, Massachusetts
| | - Erkan Baloglu
- Research and Translational Development, Karyopharm Therapeutics Inc., Newton, Massachusetts
| | - Roberto Pili
- Simon Cancer Center, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Robert H. Weiss
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, California
- Comprehensive Cancer Center, University of California, Davis, California
- Medical Service, Veterans Affairs Northern California Health Care System, Sacramento, California
| |
Collapse
|
3
|
Sobczuk P, Brodziak A, Khan MI, Chhabra S, Fiedorowicz M, Wełniak-Kamińska M, Synoradzki K, Bartnik E, Cudnoch-Jędrzejewska A, Czarnecka AM. Choosing The Right Animal Model for Renal Cancer Research. Transl Oncol 2020; 13:100745. [PMID: 32092671 PMCID: PMC7036425 DOI: 10.1016/j.tranon.2020.100745] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/04/2020] [Accepted: 01/06/2020] [Indexed: 12/17/2022] Open
Abstract
The increase in the life expectancy of patients with renal cell carcinoma (RCC) in the last decade is due to changes that have occurred in the area of preclinical studies. Understanding cancer pathophysiology and the emergence of new therapeutic options, including immunotherapy, would not be possible without proper research. Before new approaches to disease treatment are developed and introduced into clinical practice they must be preceded by preclinical tests, in which animal studies play a significant role. This review describes the progress in animal model development in kidney cancer research starting from the oldest syngeneic or chemically-induced models, through genetically modified mice, finally to xenograft, especially patient-derived, avatar and humanized mouse models. As there are a number of subtypes of RCC, our aim is to help to choose the right animal model for a particular kidney cancer subtype. The data on genetic backgrounds, biochemical parameters, histology, different stages of carcinogenesis and metastasis in various animal models of RCC as well as their translational relevance are summarized. Moreover, we shed some light on imaging methods, which can help define tumor microstructure, assist in the analysis of its metabolic changes and track metastasis development.
Collapse
Affiliation(s)
- Paweł Sobczuk
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland; Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland.
| | - Anna Brodziak
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland; Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland.
| | - Mohammed Imran Khan
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada.
| | - Stuti Chhabra
- Department of Biochemistry, CSIR-Central Drug Research Institute, Lucknow, India.
| | - Michał Fiedorowicz
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| | - Marlena Wełniak-Kamińska
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| | - Kamil Synoradzki
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| | - Ewa Bartnik
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.
| | - Anna M Czarnecka
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| |
Collapse
|
4
|
Xu Q, Junttila S, Scherer A, Giri KR, Kivelä O, Skovorodkin I, Röning J, Quaggin SE, Marti HP, Shan J, Samoylenko A, Vainio SJ. Renal carcinoma/kidney progenitor cell chimera organoid as a novel tumorigenesis gene discovery model. Dis Model Mech 2017; 10:1503-1515. [PMID: 29084770 PMCID: PMC5769601 DOI: 10.1242/dmm.028332] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 10/16/2017] [Indexed: 12/13/2022] Open
Abstract
Three-dimensional (3D) organoids provide a new way to model various diseases, including cancer. We made use of recently developed kidney-organ-primordia tissue-engineering technologies to create novel renal organoids for cancer gene discovery. We then tested whether our novel assays can be used to examine kidney cancer development. First, we identified the transcriptomic profiles of quiescent embryonic mouse metanephric mesenchyme (MM) and of MM in which the nephrogenesis program had been induced ex vivo. The transcriptome profiles were then compared to the profiles of tumor biopsies from renal cell carcinoma (RCC) patients, and control samples from the same kidneys. Certain signature genes were identified that correlated in the developmentally induced MM and RCC, including components of the caveolar-mediated endocytosis signaling pathway. An efficient siRNA-mediated knockdown (KD) of Bnip3, Gsn, Lgals3, Pax8, Cav1, Egfr or Itgb2 gene expression was achieved in mouse RCC (Renca) cells. The live-cell imaging analysis revealed inhibition of cell migration and cell viability in the gene-KD Renca cells in comparison to Renca controls. Upon siRNA treatment, the transwell invasion capacity of Renca cells was also inhibited. Finally, we mixed E11.5 MM with yellow fluorescent protein (YFP)-expressing Renca cells to establish chimera organoids. Strikingly, we found that the Bnip3-, Cav1- and Gsn-KD Renca-YFP+ cells as a chimera with the MM in 3D organoid rescued, in part, the RCC-mediated inhibition of the nephrogenesis program during epithelial tubules formation. Altogether, our research indicates that comparing renal ontogenesis control genes to the genes involved in kidney cancer may provide new growth-associated gene screens and that 3D RCC-MM chimera organoids can serve as a novel model with which to investigate the behavioral roles of cancer cells within the context of emergent complex tissue structures. Editor’s Choice: Chimeras between embryonic kidney cells and renal carcinoma cells serve as a novel model to assay the roles of co-regulated genes in kidney development and renal carcinogenesis.
Collapse
Affiliation(s)
- Qi Xu
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland
| | - Sanna Junttila
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland
| | | | - Khem Raj Giri
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland
| | - Oona Kivelä
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland.,ValiFinn, FI-90220 Oulu, Finland
| | - Ilya Skovorodkin
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland
| | - Juha Röning
- Department of Computer Science and Engineering, University of Oulu, FI-90014 Oulu, Finland
| | - Susan E Quaggin
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland.,Feinberg Cardiovascular Research Institute, Division of Medicine-Nephrology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Hans-Peter Marti
- Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway
| | - Jingdong Shan
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland
| | - Anatoly Samoylenko
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland
| | - Seppo J Vainio
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland
| |
Collapse
|
5
|
Alomar SY, Alkhuriji A, Trayhyrn P, Alhetheel A, Al-Jurayyan A, Mansour L. Association of the genetic diversity of killer cell immunoglobulin-like receptor genes and HLA-C ligand in Saudi women with breast cancer. Immunogenetics 2016; 69:69-76. [PMID: 27631728 DOI: 10.1007/s00251-016-0950-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 08/31/2016] [Indexed: 01/12/2023]
Abstract
Breast cancer (BC) progression and metastases have been linked to antitumor immunity inefficiency and particularly to natural killer (NK) cells. Killer cell immunoglobulin-like receptors (KIRs) are the most polymorphic receptors of NK cells. Through their interactions with human leukocyte antigen (HLA)-C ligands, they modulate NK and T cell actions against target cells. Therefore, we studied the combinatorial effect of KIR genes and their HLA-C ligands on the susceptibility to development of BC in Saudi women. The presence of KIR genes and HLA-C1 and HLA-C2 groups was typed in 50 Saudi patients living in Riyadh and 65 healthy controls using polymerase chain reaction with sequence-specific primers. Our results indicated a protective effect by the KIR2DS2, 2DS3, and 2DL5A genes against BC (OR = 0.25, 0.21, and 0.27, respectively, and p < 0.01). The synergistic action of the three genes was observed when they occurred together, and the absence of the three genes increased BC occurrence by 6.5-fold. Distribution of the HLA-C1/C2 ligand between patients and controls showed an increase in the risk of BC occurrence for the heterozygote C1/C2 (OR = 2.33; 95 % CI = 1.08-5.02; p = 0.037) and a protective effect of the homozygote C2C2 (OR = 0.03; 95 % CI = 0.009-0.098; p < 0.001). Combinatory analyses of KIR genes and their HLA-C ligands showed protective effects of KIR2DL2 and 2DL3 in the absence of their HLA-C1 ligand. These results suggested that KIR-gene content combined with their ligand could influence the risk of BC development in women in Saudi Arabia.
Collapse
Affiliation(s)
- Suliman Y Alomar
- Zoology Department, College of Sciences, King Saud University, Post Office Box 2455, Riyadh, 11451, Saudi Arabia
| | - Afrah Alkhuriji
- Zoology Department, College of Sciences, King Saud University, Post Office Box 2455, Riyadh, 11451, Saudi Arabia
| | - Paul Trayhyrn
- Zoology Department, College of Sciences, King Saud University, Post Office Box 2455, Riyadh, 11451, Saudi Arabia
- Clore Laboratory, Buckingham Institute for Translational Medicine, University of Buckingham, Buckingham, UK
| | - Abdulkarim Alhetheel
- Department of Microbiology/Pathology, King Khalid University Hospital, College of Medicine King Saud University, Riyadh, Saudi Arabia
| | - Abdullah Al-Jurayyan
- Immunology and HLA section, Pathology and Clinical Laboratory Medicine, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Lamjed Mansour
- Zoology Department, College of Sciences, King Saud University, Post Office Box 2455, Riyadh, 11451, Saudi Arabia.
- Faculty of Sciences of Gabès, University of Gabès, Erriadh City, 6072, Zrig Gabès, Tunisia.
| |
Collapse
|
6
|
Fend L, Remy-Ziller C, Foloppe J, Kempf J, Cochin S, Barraud L, Accart N, Erbs P, Fournel S, Préville X. Oncolytic virotherapy with an armed vaccinia virus in an orthotopic model of renal carcinoma is associated with modification of the tumor microenvironment. Oncoimmunology 2015; 5:e1080414. [PMID: 27057460 PMCID: PMC4801465 DOI: 10.1080/2162402x.2015.1080414] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 07/31/2015] [Accepted: 07/31/2015] [Indexed: 12/21/2022] Open
Abstract
Oncolytic virotherapy is an emergent promising therapeutic approach for the treatment of cancer. We have constructed a vaccinia virus (WR strain) deleted for thymidine kinase (TK) and ribonucleotide reductase (RR) genes that expressed the fusion suicide gene FCU1 derived from the yeast cytosine deaminase and uracil phosphoribosyltransferase genes. We evaluated this construct (VV-FCU1) in the orthotopic model of renal carcinoma (RenCa). Systemic administration of VV-FCU1 resulted in orthotopic tumor growth inhibition, despite temporary expression of viral proteins. VV-FCU1 treatment was associated with an infiltration of tumors by CD8+ T lymphocytes and a decrease in the proportion of infiltrating Tregs, thus modifying the ratio of CD8+/CD4+ Treg in favor of CD8+cytotoxic T cells. We demonstrated that VV-FCU1 treatment prolonged survival of animals implanted with RenCa cells in kidney. Depletion of CD8+ T cells abolished the therapeutic effect of VV-FCU1 while depletion of CD4+ T cells enhanced its protective activity. Administration of the prodrug 5-fluorocytosine (5-FC) resulted in a sustained control of tumor growth but did not extend survival. This study shows the importance of CD4+ and CD8+ T cells in vaccinia virus-mediated oncolytic virotherapy and suggests that this approach may be evaluated for the treatment of human renal cell carcinoma.
Collapse
Affiliation(s)
- Laetitia Fend
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| | - Christelle Remy-Ziller
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| | - Johann Foloppe
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| | - Juliette Kempf
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| | - Sandrine Cochin
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| | - Luc Barraud
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| | - Nathalie Accart
- Novartis Institutes for Biomedical Research, Analytical Sciences and Imaging , WSJ386, Basel, Switzerland
| | - Philippe Erbs
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| | - Sylvie Fournel
- Laboratoire de Conception et Application de Molécules Bioactives, Equipe de Biovectorologie, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie , 74 Route du Rhin- BP60024 , Illkirch-Graffenstaden Cedex, France
| | - Xavier Préville
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| |
Collapse
|
7
|
Yin H, Frontini MJ, Arpino JM, Nong Z, O'Neil C, Xu Y, Balint B, Ward AD, Chakrabarti S, Ellis CG, Gros R, Pickering JG. Fibroblast Growth Factor 9 Imparts Hierarchy and Vasoreactivity to the Microcirculation of Renal Tumors and Suppresses Metastases. J Biol Chem 2015; 290:22127-42. [PMID: 26183774 DOI: 10.1074/jbc.m115.652222] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Indexed: 12/11/2022] Open
Abstract
Tumor vessel normalization has been proposed as a therapeutic paradigm. However, normal microvessels are hierarchical and vasoreactive with single file transit of red blood cells through capillaries. Such a network has not been identified in malignant tumors. We tested whether the chaotic tumor microcirculation could be reconfigured by the mesenchyme-selective growth factor, FGF9. Delivery of FGF9 to renal tumors in mice yielded microvessels that were covered by pericytes, smooth muscle cells, and a collagen-fortified basement membrane. This was associated with reduced pulmonary metastases. Intravital microvascular imaging revealed a haphazard web of channels in control tumors but a network of arterioles, bona fide capillaries, and venules in FGF9-expressing tumors. Moreover, whereas vasoreactivity was absent in control tumors, arterioles in FGF9-expressing tumors could constrict and dilate in response to adrenergic and nitric oxide releasing agents, respectively. These changes were accompanied by reduced hypoxia in the tumor core and reduced expression of the angiogenic factor VEGF-A. FGF9 was found to selectively amplify a population of PDGFRβ-positive stromal cells in the tumor and blocking PDGFRβ prevented microvascular differentiation by FGF9 and also worsened metastases. We conclude that harnessing local mesenchymal stromal cells with FGF9 can differentiate the tumor microvasculature to an extent not observed previously.
Collapse
Affiliation(s)
- Hao Yin
- From the Robarts Research Institute and
| | | | | | | | | | - Yiwen Xu
- From the Robarts Research Institute and Medical Biophysics
| | | | | | | | | | - Robert Gros
- From the Robarts Research Institute and Physiology and Pharmacology, and Medicine, University of Western Ontario
| | - J Geoffrey Pickering
- From the Robarts Research Institute and Departments of Biochemistry, Medical Biophysics, Medicine, University of Western Ontario, London Health Sciences Centre, London, Ontario N6A 5A5, Canada
| |
Collapse
|
8
|
Neal RE, Rossmeisl JH, Robertson JL, Arena CB, Davis EM, Singh RN, Stallings J, Davalos RV. Improved local and systemic anti-tumor efficacy for irreversible electroporation in immunocompetent versus immunodeficient mice. PLoS One 2013; 8:e64559. [PMID: 23717630 PMCID: PMC3663742 DOI: 10.1371/journal.pone.0064559] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 04/16/2013] [Indexed: 12/18/2022] Open
Abstract
Irreversible electroporation (IRE) is a non-thermal focal ablation technique that uses a series of brief but intense electric pulses delivered into a targeted region of tissue, killing the cells by irrecoverably disrupting cellular membrane integrity. This study investigates if there is an improved local anti-tumor response in immunocompetent (IC) BALB/c versus immunodeficient (ID) nude mice, including the potential for a systemic protective effect against rechallenge. Subcutaneous murine renal carcinoma tumors were treated with an IRE pulsing protocol that used 60% of the predicted voltage required to invoke complete regressions in the ID mice. Tumors were followed for 34 days following treatment for 11 treated mice from each strain, and 7 controls from each strain. Mouse survival based on tumor burden and the progression-free disease period was substantially longer in the treated IC mice relative to the treated ID mice and sham controls for both strains. Treated IC mice were rechallenged with the same cell line 18 days after treatment, where growth of the second tumors was shown to be significantly reduced or prevented entirely. There was robust CD3+ cell infiltration in some treated BALB/C mice, with immunocytes focused at the transition between viable and dead tumor. There was no difference in the low immunocyte presence for untreated tumors, nude mice, and matrigel-only injections in both strains. These findings suggest IRE therapy may have greater therapeutic efficacy in immunocompetent patients than what has been suggested by immunodeficient models, and that IRE may invoke a systemic response beyond the targeted ablation region.
Collapse
Affiliation(s)
- Robert E Neal
- Department of Radiology, The Alfred Hospital, Melbourne, Victoria, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Egenolf DD, Rafferty P, Brosnan K, Walker M, Jordan J, Makropoulos D, Kavalkovich K, Watson S, Johns L, Volk A, Bugelski PJ. Development of a murine model of lymph node metastases suitable for immunotoxicity studies. J Pharmacol Toxicol Methods 2011; 63:236-49. [DOI: 10.1016/j.vascn.2010.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 12/01/2010] [Indexed: 12/27/2022]
|
10
|
Liang S, Kahlenberg MS, Rousseau DL, Hornsby PJ. Neoplastic conversion of human colon smooth muscle cells: No requirement for telomerase. Mol Carcinog 2008; 47:478-84. [PMID: 18085530 DOI: 10.1002/mc.20405] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The role of telomerase as an essential requirement for the neoplastic conversion of human cells has been controversial. In the model of conversion of normal human cells to cancer cells by the combination of simian virus 40 (SV40) early region genes and oncogenic Ras (H-Ras(G12V)), telomerase (hTERT) was originally described as essential in conjunction with these other genes. Here we used primary cultures of colon smooth muscle cells isolated from surgical specimens. SV40 large T antigen (TAg) and oncogenic Ras(G12V) were introduced into the cells by retroviral transduction and cells were rapidly transplanted into the subrenal capsule space in immunodeficient mice, without selection in culture. Malignant tumors were formed from transduced cells. Extensive invasion into the kidney occurred even when tumors were small; in contrast, at the same tumor size, oncogene-expressing fibroblasts did not show much invasion. Increased invasiveness was also observed in vitro. However, cells in these cancers showed morphological evidence of crisis, consistent with their lack of telomerase. These experiments on human colon smooth muscle cells support the concept that Ras(G12V) and SV40 TAg form a minimal set of genes that can convert normal human cells to cancer cells without a requirement for hTERT.
Collapse
Affiliation(s)
- Sitai Liang
- Department of Physiology, Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, Texas 78245, USA
| | | | | | | |
Collapse
|
11
|
Liu D, Hornsby PJ. Fibroblast stimulation of blood vessel development and cancer cell invasion in a subrenal capsule xenograft model: stress-induced premature senescence does not increase effect. Neoplasia 2007; 9:418-26. [PMID: 17534447 PMCID: PMC1877979 DOI: 10.1593/neo.07205] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2007] [Revised: 03/27/2007] [Accepted: 03/29/2007] [Indexed: 11/18/2022] Open
Abstract
Fibroblast cooperation with cancer cells in xenograft development was investigated by transplanting MDA-MB-231 cells under the kidney capsule of immunodeficient mice. Control fibroblasts and fibroblasts subjected to stress-induced premature senescence by treatment with bleomycin were used. In other xenograft models, senescent fibroblasts have shown a growth-stimulatory effect greater than that of control cells. In this model, both types of fibroblasts accelerated the formation and growth of xenografts. Blood vessel development, as evidence by von Willebrand factor staining, was greatly accelerated by the presence of fibroblasts, and invasion into the kidney was also increased. Control and senescent fibroblasts had very similar effects. These actions of fibroblasts were partially recapitulated in in vitro experiments. Both control and senescent fibroblasts stimulated the tubulogenesis of endothelial cells in culture and stimulated the invasion of MDA-MB-231 cells through Matrigel in vitro. In this xenograft model, in which fibroblasts are cotransplanted with a cancer cell into an internal organ rather than subcutaneously, senescence was not an important factor in the effects of cotransplanted fibroblasts on growth, blood vessel development, and invasion. Therefore, cancer promotion by the senescence of adjacent stromal cells may be restricted to certain organ and tissue types.
Collapse
Affiliation(s)
- Dan Liu
- Department of Physiology and Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, TX, USA
| | | |
Collapse
|
12
|
Samlowski WE, McGregor JR, Jurek M, Baudys M, Zentner GM, Fowers KD. ReGel® Polymer-based Delivery of Interleukin-2 as a Cancer Treatment. J Immunother 2006; 29:524-35. [PMID: 16971808 DOI: 10.1097/01.cji.0000211306.05869.25] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
ReGel is an aqueous, filter sterilizable ABA tri-block polymer consisting of poly-(lactide-co-glycolide) and polyethylene glycol. We tested the suitability of this polymer to provide sustained interleukin-2 (IL-2) delivery for cancer immunotherapy. ReGel/IL-2 is liquid at or below room temperature, and is easily injectable through narrow gauge needles, but undergoes a reversible thermal transition into a bioerodible depot at body temperature. We demonstrated that ReGel/IL-2 releases IL-2 over 72 to 96 hours in vitro, without loss of bioactivity. Pharmacokinetic studies after peritumoral injection of 0.1 mL ReGel/IL-2 in mice demonstrated an early burst of IL-2 release, followed by more sustained release kinetics over 96 hours (T(1/2)beta 48 h). Less than 1.5% of the injected dose was detectable in blood or kidneys during the first 48 hours. A single peritumoral dose of ReGel/IL-2 [1 to 4 million international units (MIU) ReGel/IL-2, split into 4 quadrant injections] was administered to mice bearing subcutaneous RD-995 spindle cell carcinoma. Only the highest dose of ReGel/IL-2 tested (4.0 MIU) resulted in significant hypotension on day 3 after injection. Weekly treatment of Meth A fibrosarcoma and RENCA renal carcinoma with ReGel/IL-2 (2 MIU/dose) induced a significant reduction in tumor growth and improved survival. Reduction in tumor growth at implants remote from treated lesions was also observed, suggesting systemic activation of antitumor immunity. These findings establish that peritumoral injection of ReGel/IL-2 is an effective delivery system for cancer immunotherapy, while decreasing IL-2 toxicity. This polymer delivery system is likely to be broadly applicable for sustained delivery of other cytokines and peptides.
Collapse
Affiliation(s)
- Wolfram E Samlowski
- Multidisciplinary Melanoma Program, Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT 84112, USA.
| | | | | | | | | | | |
Collapse
|
13
|
Sun B, Chen M, Hawks CL, Pereira-Smith OM, Hornsby PJ. The minimal set of genetic alterations required for conversion of primary human fibroblasts to cancer cells in the subrenal capsule assay. Neoplasia 2005; 7:585-93. [PMID: 16036109 PMCID: PMC1501282 DOI: 10.1593/neo.05172] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2005] [Accepted: 03/16/2005] [Indexed: 02/04/2023] Open
Abstract
Based on previous studies, a minimal set of genetic alterations that is required to convert normal human fibroblasts into cancer cells has been defined. Essential roles for telomere maintenance and alterations in phosphatase 2A activity were inferred from experiments in which tumorigenicity was tested by injecting cells under the skin of immunodeficient mice. However, in the present experiments, the combination of SV40 large T antigen and activated Ras, without hTERT or SV40 small t antigen, was sufficient to convert nine different primary human fibroblast cell strains to a fully malignant state. The malignant behavior of the cells was demonstrated by growth of the cells into invasive tumors when the cells were injected beneath the kidney capsule of immunodeficient mice. Lung metastases and circulating tumor cells were also detected. These tumors were not immortal; cells entered crisis, from which they could be rescued by expression of hTERT. However, the same cell populations were not tumorigenic when they were injected under the skin. In this site, tumorigenicity required the expression of hTERT and SV40 small t antigen as well as SV40 large T antigen and Ras. The cellular pathways targeted by SV40 large T antigen (p53 and pRb) and those targeted by activated Ras represent a minimal set of genetic alterations required for the conversion of normal human fibroblasts into cancer cells.
Collapse
Affiliation(s)
- Beicheng Sun
- Department of Physiology, University of Texas Health Science Center, San Antonio, TX 78245, USA
| | | | | | | | | |
Collapse
|
14
|
FUJIKAWA KEITA, MATSUI YOSHIYUKI, MIURA KATSUKI, KOBAYASHI TAKASHI, OKA HIROYA, FUKUZAWA SHIGEKI, TAKEUCHI HIDEO. SERUM IMMUNOSUPPRESSIVE ACIDIC PROTEIN AND NATURAL KILLER CELL ACTIVITY IN PATIENTS WITH METASTATIC RENAL CELL CARCINOMA BEFORE AND AFTER NEPHRECTOMY. J Urol 2000. [DOI: 10.1016/s0022-5347(05)67278-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- KEITA FUJIKAWA
- From the Department of Urology, Kobe City General Hospital, Kobe City, Japan
| | - YOSHIYUKI MATSUI
- From the Department of Urology, Kobe City General Hospital, Kobe City, Japan
| | - KATSUKI MIURA
- From the Department of Urology, Kobe City General Hospital, Kobe City, Japan
| | - TAKASHI KOBAYASHI
- From the Department of Urology, Kobe City General Hospital, Kobe City, Japan
| | - HIROYA OKA
- From the Department of Urology, Kobe City General Hospital, Kobe City, Japan
| | - SHIGEKI FUKUZAWA
- From the Department of Urology, Kobe City General Hospital, Kobe City, Japan
| | - HIDEO TAKEUCHI
- From the Department of Urology, Kobe City General Hospital, Kobe City, Japan
| |
Collapse
|
15
|
Fujikawa K, Matsui Y, Miura K, Kobayashi T, Oka H, Fukuzawa S, Takeuchi H. Serum immunosuppressive acidic protein and natural killer cell activity in patients with metastatic renal cell carcinoma before and after nephrectomy. J Urol 2000; 164:673-5. [PMID: 10953123 DOI: 10.1097/00005392-200009010-00013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE To our knowledge the impact of nephrectomy on stage M1 renal cell carcinoma remains to be determined. We previously reported that nephrectomy is beneficial in patients with elevated serum C-reactive protein before treatment, and those in whom nadir C-reactive protein decreases postoperatively to within the normal range may expect longer survival when surgery is combined with postoperative immunotherapy. In this study we determine the effect of nephrectomy on the immune response in patients with metastatic renal cell carcinoma. MATERIALS AND METHODS We retrospectively reviewed the records of 40 patients with metastatic renal cell carcinoma diagnosed at our institution between 1986 and 1999. These patients underwent nephrectomy before cytokine therapy with interferon. Before and after nephrectomy we measured serum C-reactive protein, serum immunosuppressive acidic protein and peripheral blood natural killer cell activity. RESULTS In 15 patients with pretreatment serum C-reactive protein within the normal range (less than 1 ng./ml.) there was no significant difference before and after nephrectomy in the serum immunosuppressive acidic protein level or natural killer cell activity (p = 0.4587 and 0.3892, respectively). On the other hand, in 25 patients with serum C-reactive protein elevated before treatment to 1 ng./ml. or greater serum immunosuppressive acidic protein decreased significantly and natural killer cell activity increased significantly after cytoreductive surgery (p = 0.0002 and 0.0286, respectively). CONCLUSIONS Our study implies that nephrectomy may be beneficial in patients with elevated serum C-reactive protein before treatment. Further evaluation by a prospective study is needed to make a definitive conclusion.
Collapse
Affiliation(s)
- K Fujikawa
- Department of Urology, Kobe City General Hospital, Japan
| | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
There is increasing evidence that tumors express putative target molecules for a therapeutic immune reaction. Yet, tumor cells lack the prerequisites for appropriate antigen presentation and--hence--the immune system does not respond. This difficulty can probably be circumvented when tumor antigens are processed by conventional antigen presenting cells. Thus, the identification of immunogenic tumor-associated antigens may allow new modes of vaccination with the hope of adding a fourth and hopefully powerful weapon to surgery, radiation and chemotherapy in the fight against cancer.
Collapse
Affiliation(s)
- M Zöller
- Department of Tumor Progression and Immune Defense, German Cancer Research Center, Heidelberg.
| | | |
Collapse
|
17
|
Oya M, Marumo K, Murai M, Tazaki H. Pharmacokinetics and antitumor effects of an interleukin-2 immunocomplexing agent in murine renal cell carcinoma. Int J Urol 1996; 3:141-4. [PMID: 8689509 DOI: 10.1111/j.1442-2042.1996.tb00499.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Conventional therapy for renal cell carcinoma (RCC) using systemic administration of interleukin-2 (IL-2) has shown limited anti-tumor action. The purpose of this study was to investigate the anti-tumor effects of a newly developed immune complex of IL-2 (IC) against RCC. METHODS IC was prepared by mixing IL-2 and an anti-IL-2 monoclonal antibody at a molar ratio of 2:1. The pharmacokinetics and anti-tumor effects of IC were then studied in a murine RCC line, Renca. RESULTS Serum IL-2 levels were sustained longer in mice given IC than in mice given IL-2 alone after either subcutaneous or intratumoral injections. After an intratumoral injection of IC, the IL-2 concentration in the tumor nodules remained higher compared with mice given IL-2 alone. The anti-tumor effect was most pronounced in mice treated with intratumoral injections of IC. CONCLUSIONS Results obtained here indicate that an immune complex of IL-2 provides a useful tool for the treatment of RCC by altering the pharmacokinetics of IL-2 in vivo.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/therapeutic use
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/immunology
- Cytotoxicity, Immunologic/drug effects
- Female
- Immunoconjugates/administration & dosage
- Immunoconjugates/therapeutic use
- Injections, Intralesional
- Injections, Subcutaneous
- Interleukin-2/administration & dosage
- Interleukin-2/pharmacokinetics
- Interleukin-2/therapeutic use
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/immunology
- Killer Cells, Natural/immunology
- Mice
- Mice, Inbred BALB C
- Neoplasm Transplantation
- Recombinant Proteins/administration & dosage
- Recombinant Proteins/therapeutic use
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- M Oya
- Department of Urology, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | |
Collapse
|
18
|
Huang S, Xie K, Singh RK, Gutman M, Bar-Eli M. Suppression of tumor growth and metastasis of murine renal adenocarcinoma by syngeneic fibroblasts genetically engineered to secrete the JE/MCP-1 cytokine. J Interferon Cytokine Res 1995; 15:655-65. [PMID: 7553238 DOI: 10.1089/jir.1995.15.655] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The murine monocyte chemoattractant protein 1, JE/MCP-1, like its human counterpart monocyte chemotactic and activating factor (MCAF), attracts monocytes-macrophages to tumor tissues. In previous studies we reported that expression of the JE/MCP-1 gene in murine colon carcinoma cells reduced their tumorigenicity and suppressed their metastatic potential. We now demonstrate that the growth and metastasis of the renal adenocarcinoma cell line RENCA are reduced when it was admixed with syngeneic fibroblasts engineered to secrete the JE/MCP-1 cytokine before injection. Culture supernatants of JE/MCP-1-expressing cells plus lipopolysaccharide (LPS) synergistically activated tumoricidal properties in syngeneic macrophages against RENCA cells. This activity was blocked by anti-JE/MCP-1 antibody, indicating that JE/MCP-1 was involved in priming the macrophages to respond to LPS. Moreover, alveolar macrophages isolated shortly after iv injections of JE/MCP-1 transfected cells were cytotoxic to RENCA cells in vitro. Collectively, these data suggest that in addition to its chemotactic properties, JE/MCP-1 can synergize with bacterial endotoxins to activate macrophages, thus providing a rationale for the use of the JE/MCP-1 protein as a modality for treatment of metastasis.
Collapse
Affiliation(s)
- S Huang
- Department of Cell Biology, University of Texas M.D. Anderson Cancer Center, Houston 77030, USA
| | | | | | | | | |
Collapse
|
19
|
Hillman GG, Younes E, Visscher D, Ali E, Lam JS, Montecillo E, Pontes JE, Haas GP, Puri RK. Systemic treatment with interleukin-4 induces regression of pulmonary metastases in a murine renal cell carcinoma model. Cell Immunol 1995; 160:257-63. [PMID: 7720087 DOI: 10.1016/0008-8749(95)80036-i] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Advanced metastatic renal cell carcinoma has been shown to be responsive to immunotherapy but the response rate is still limited. We have investigated the therapeutic potential of systemic interleukin-4 (IL-4) administration for the treatment of pulmonary metastases in the murine Renca renal adenocarcinoma model. Renca cells were injected iv in Balb/c mice to induce multiple pulmonary tumor nodules. From Day 5, Renca-bearing mice were treated with two daily injections of recombinant murine IL-4 for 5 consecutive days. IL-4 treatment induced a significant reduction in the number of lung metastases in a dose-dependent manner and significantly augmented the survival of treated animals. Immunohistochemistry studies, performed on lung sections, showed macrophage and CD8+ T cell infiltration in the tumor nodules 1 day after the end of IL-4 treatment. The CD8 infiltration increased by Day 7 after IL-4 treatment. Granulocyte infiltration was not detectable. To clarify further the role of the immune system in IL-4 anti-tumor effect, mice were depleted of lymphocyte subpopulations by in vivo injections of specific antibodies prior to treatment with IL-4. Depletion of CD8+ T cells or AsGM1+ cells abrogated the effect of IL-4 on lung metastases, whereas depletion of CD4+ T cells had no impact. These data indicate that CD8+ T cells and AsGM1+ cells are involved in IL-4-induced regression of established renal cell carcinoma.
Collapse
Affiliation(s)
- G G Hillman
- Department of Urology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Fujioka T, Ishikura K, Hasegawa M, Ogyu K, Matsushita Y, Sato M, Sato F, Aoki H, Kubo T. Antitumor effects of oral administration of an interferon-inducing pyrimidinone, Bropirimine, on murine renal-cell carcinoma. Cancer Chemother Pharmacol 1995; 36:7-12. [PMID: 7536641 DOI: 10.1007/bf00685725] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Bropirimine [2-amino-5-bromo-6-phenyl-4-(3H)-pyrimidinone] is a low-molecular-weight compound that acts as an inducer of interferon in several animal species. Experiments were designed to explore the possibility of using this drug for the treatment of renal-cell carcinoma (RCC). Euthymic BALB/c mice were inoculated with murine RCC (Renca) cells and given graded doses of Bropirimine p.o. for 5 consecutive days beginning on day 1 following tumor inoculation. These mice were killed and tumors were excised on day 21. Bropirimine significantly (P < 0.01) inhibited the tumor growth at a daily dose of 1,000 or 2,000 mg/kg. No adverse effect or toxicity was noted at 1,000 mg/kg, and at 2,000 mg/kg there was only a marginal body-weight reduction without any other appreciable side effect. In addition to the inhibition of tumor growth, there was a small yet significant (P < 0.05) increase in the duration of survival (in days) in the Bropirimine-treated animals. When the treatment was delayed to begin on day 6 following tumor inoculation, Bropirimine did not suppress tumor growth in euthymic mice, pointing to the importance of the timing of the treatment. In athymic nude BALB/c mice lacking T-cells or T-cell function, Bropirimine also inhibited tumor growth (P < 0.01). The antitumor effect of this drug was abolished by pretreatment with anti-asialo GM1 serum, which eliminated natural killer (NK) activity in euthymic mice. In vivo treatment with Bropirimine augmented the cytotoxicity of lymphocytes isolated from the spleens or lungs of the tumor-bearing mice, which were active against Renca and YAC-1 cells in vitro. This activity was NK-cell-dependent as judged on the basis of the results of the in vitro complement-dependent cytotoxicity assay. Since Bropirimine induced interferon (IFN)-alpha/beta production, significantly (P < 0.05) elevating its serum concentration, and since this drug mimics the effects of IFN-alpha/beta, it seemed likely that the Bropirimine-induced NK cell augmentation we found was mediated by IFN-alpha/beta. These results suggest that Bropirimine, a booster of NK activity, may have potential as an adjunct to other therapeutic modalities in the treatment of human RCC.
Collapse
Affiliation(s)
- T Fujioka
- Department of Urology, Iwate Medical University School of Medicine, Morioka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lau BH, Ruckle HC, Botolazzo T, Lui PD. Chinese medicinal herbs inhibit growth of murine renal cell carcinoma. CANCER BIOTHERAPY 1994; 9:153-61. [PMID: 7812364 DOI: 10.1089/cbr.1994.9.153] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Tumors are known to produce factors suppressing immune functions. We previously showed that a murine renal cell carcinoma (Renca) suppressed macrophage function in vitro and that this suppression was abolished by co-incubation with extracts of two Chinese medicinal herbs. We now report that these phytochemicals are capable of inhibiting growth of Renca in vivo. BALB/c mice were transplanted intraperitoneally (IP) with 1-2 x 10(5) Renca cells. One day after tumor transplant, mice were randomized into two groups. One group was treated IP, daily for 10 days, with 100 microliters of phytochemicals containing 500 micrograms each of Astragalus membranaceus and Ligustrum lucidum, while the other group received saline as controls. A cure rate of 57% was obtained with these phytochemicals when the initial tumor load was 2 x 10(5), and 100% when the initial tumor load was 1 x 10(5). Additional experiments were performed to investigate the mechanisms involved in this protection. Splenic macrophages from tumor-bearing mice were shown to have depressed chemiluminescent oxidative burst activity, and this depression was restored with phytochemical treatment. Splenocytes from mice transplanted with Renca responded less favorably to interleukin-2 (IL-2) in generating lymphokine-activated killer (LAK) cells; again this depression was restored with phytochemical treatment. Our data suggest that these phytochemicals may have exerted their antitumor effects via augmentation of phagocyte and LAK cell activities.
Collapse
Affiliation(s)
- B H Lau
- Department of Microbiology, School of Medicine, Loma Linda University, California 92350
| | | | | | | |
Collapse
|
22
|
Abstract
Renal cell carcinoma is diagnosed frequently when it already has metastasized. The use of immunotherapy has provided a new source of drugs to be used in the therapy of renal cell carcinoma. Interferons were the first biologics to be evaluated demonstrating a response rate of 10-30 percent. The second cytokine to be actively developed was interleukin-2, a T-cell growth factor which demonstrated activity in patients with renal cell carcinoma of 20-30 percent alone or in combination with LAK cells and other cytokines (such as interferons and tumor necrosis factor). Biologic response modifiers such as interferons and interleukins alone and in combination with other drugs have opened a new era in the therapy of renal cell carcinoma. Future development of other cytokines may pave the way for higher response rates with durable duration for this disease.
Collapse
Affiliation(s)
- M Choudhury
- Department of Urology, New York Medical College, Valhalla
| | | | | |
Collapse
|
23
|
Dybal EJ, Haas GP, Maughan RL, Sud S, Pontes JE, Hillman GG. Synergy of radiation therapy and immunotherapy in murine renal cell carcinoma. J Urol 1992; 148:1331-7. [PMID: 1404669 DOI: 10.1016/s0022-5347(17)36903-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The treatment of metastatic renal cell carcinoma with immunotherapy has resulted in objective anti-tumor responses in 15-30% of patients. To enhance the therapeutic effects of immunotherapy, it is becoming evident that this approach should be combined with other treatment modalities. In this study, a spontaneously metastasizing murine renal adenocarcinoma (Renca), transplanted under the renal capsule, was treated with either radiation therapy, immunotherapy or a combination of both. In order to distinguish between the local and systemic effects of radiation therapy, total body irradiation was compared to irradiation of the tumor-bearing kidney only, or irradiation of the whole mouse with the tumor-bearing kidney shielded. Immunotherapy was administered with interleukin-2 (IL-2) alone or with IL-2 and lymphokine activated killer (LAK) cells. Combined radiation and immunotherapy induced a better anti-tumor response than either modality alone. The best response was obtained by local tumor irradiation and IL-2 therapy and resulted in a significant reduction in primary tumor size, elimination of lung metastases and a significant increase in survival.
Collapse
Affiliation(s)
- E J Dybal
- Department of Veterans Affairs, VA Medical Center, Allen Park, Michigan
| | | | | | | | | | | |
Collapse
|
24
|
Feruglio C, Zambello R, Trentin L, Bulian P, Franceschi T, Cetto GL, Semenzato G. Cytotoxic in vitro function in patients with metastatic renal cell carcinoma before and after alpha-2b-interferon therapy. Effects of activation with recombinant interleukin-2. Cancer 1992; 69:2525-31. [PMID: 1568175 DOI: 10.1002/1097-0142(19920515)69:10<2525::aid-cncr2820691023>3.0.co;2-d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In this study the characteristics of the cytotoxic function in a series of patients with metastatic renal cell carcinoma (RCC) were analyzed and the possibility of modulating this capacity in vitro with the use of biologic response modifiers (BMR) such as alpha-interferon (alpha-IFN) and recombinant interleukin-2 (rIL-2) was verified, with the ultimate goal of providing a rationale for a therapeutic approach to this disease with these molecules. Peripheral blood mononuclear cells (PBMC) of patients with advanced RCC were tested for natural killer (NK) and lymphokine-activated killer (LAK) activity both before and after alpha-IFN therapy. In addition, surface markers of unstimulated and stimulated cells were analyzed and in vitro assays were performed to determine the proliferative capacity in response to the stimulus with rIL-2. During an evaluation before treatment, defective NK activity was observed that could be corrected by incubating the cells with rIL-2. In these subjects, LAK cells could be consistently generated after PBMC were activated with this cytokine in vitro. No changes in NK and LAK activity were found after alpha-IFN therapy. In contrast, treatment with alpha-IFN affected the proliferative response of PBMC to rIL-2, and a significant decrease in this in vitro capacity was observed during follow-up. The ability to restore NK activity and obtain an adequate LAK cytotoxicity from the PBMC of patients with RCC supports a therapeutic approach with BRM. However, the fact that this type of treatment affects the proliferative response of PBMC to rIL-2 must be considered when clinical trials are designed for patients with RCC.
Collapse
Affiliation(s)
- C Feruglio
- Department of Clinical Medicine, Padua University School of Medicine, Padova, Italy
| | | | | | | | | | | | | |
Collapse
|
25
|
Salup RR, Sicker DC, Wolmark N, Herberman RB, Hakala TR. Chemoimmunotherapy of metastatic murine renal cell carcinoma using flavone acetic acid and interleukin 2. J Urol 1992; 147:1120-3. [PMID: 1552607 DOI: 10.1016/s0022-5347(17)37499-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Metastatic renal cell carcinoma (RCC) remains largely incurable. We used a murine RCC (Renca) in BALB/c mice to investigate the treatment possibilities with chemoimmunotherapy using in vivo boosters of natural killer (NK) activity. Diffuse pulmonary metastases were induced by intravenous (i.v.) inoculation with 100,000 Renca cells. All untreated control animals died within one month from pulmonary metastases. Chemoimmunotherapy using the NK immunostimulator flavonic-8-acetic acid (FAA) at 200 mg./kg. i.v. was given on the third day post tumor inoculation, followed by four consecutive days of twice daily intraperitoneal (i.p.) administration of 10,000 units human recombinant interleukin-2 (rIL-2). This chemoimmunotherapy regimen consistently cured 70% of tumor-bearing animals. Mice cured by this chemoimmunotherapy regimen did not reject subsequent reinoculation with Renca, indicating absence of specific antitumor immunity as a result of the treatment. While FAA and rIL-2 have no demonstrable in vitro cytotoxicity for Renca, they are excellent boosters of in vivo NK activity. These data suggest a potential alternative treatment method for metastatic RCC, a tumor type for which no efficient cytostatic drugs are available.
Collapse
Affiliation(s)
- R R Salup
- Department of Surgery, Pittsburgh Cancer Institute, Pennsylvania
| | | | | | | | | |
Collapse
|
26
|
Salup RR, Sicker DC, Ballou BT, Flack CE, Wolmark N, Hakala TR. Improved lymphocyte cytotoxicity against murine renal cell carcinoma. J Urol 1992; 147:491-5. [PMID: 1732630 DOI: 10.1016/s0022-5347(17)37286-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In this paper we describe the generation of antibody dependent cellular cytotoxicity against a murine renal cell carcinoma. Using human recombinant interleukin-2 and in vitro adherence to plastic, we generated lymphokine activated killer and adherent lymphokine activated killer cells. Adherent lymphokine activated killer cells had significant (p less than 0.05) higher unrestricted cytotoxicity than LAK cells. Using a rabbit antibody against Renca developed in our laboratory, we induced significant (p less than 0.01) antibody dependent cellular cytotoxicity using fresh spleen, lymphokine activated killer and adherent lymphokine activated killer cells. The strongest antibody dependent cellular cytotoxicity killing was mediated by adherent lymphokine activated killer cells and was restricted only to the renal cell carcinoma target. Using FACS cell surface analysis and antibody and complement depletion of selected effector cell subsets, we also demonstrate that the antibody dependent cellular cytotoxicity effector cell population consists of asialoGM1+ Lyt 2.1- natural killer cells. This first description of antibody dependent cellular cytotoxicity against renal cell carcinoma by activated natural killer cells suggests a novel method for more efficient use of cytotoxic effector cells against this type of cancer.
Collapse
Affiliation(s)
- R R Salup
- Department of Surgery, University of Pittsburgh, Pennsylvania
| | | | | | | | | | | |
Collapse
|
27
|
Golumbek PT, Lazenby AJ, Levitsky HI, Jaffee LM, Karasuyama H, Baker M, Pardoll DM. Treatment of established renal cancer by tumor cells engineered to secrete interleukin-4. Science 1991; 254:713-6. [PMID: 1948050 DOI: 10.1126/science.1948050] [Citation(s) in RCA: 502] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The generation of antigen-specific antitumor immunity is the ultimate goal in cancer immunotherapy. When cells from a spontaneously arising murine renal cell tumor were engineered to secrete large doses of interleukin-4 (IL-4) locally, they were rejected in a predominantly T cell-independent manner. However, animals that rejected the IL-4-transfected tumors developed T cell-dependent systemic immunity to the parental tumor. This systemic immunity was tumor-specific and primarily mediated by CD8+ T cells. Established parental tumors could be cured by the systemic immune response generated by injection of the genetically engineered tumors. These results provide a rationale for the use of lymphokine gene-transfected tumor cells as a modality for cancer therapy.
Collapse
Affiliation(s)
- P T Golumbek
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21205
| | | | | | | | | | | | | |
Collapse
|
28
|
Utsugi T, Dinney CP, Killion JJ, Fidler IJ. In situ activation of mouse macrophages and therapy of spontaneous renal cell cancer metastasis by liposomes containing the lipopeptide CGP 31362. Cancer Immunol Immunother 1991; 33:375-81. [PMID: 1878891 PMCID: PMC11038907 DOI: 10.1007/bf01741597] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/1991] [Accepted: 05/03/1991] [Indexed: 12/29/2022]
Abstract
We determined whether the intravenous administration of multilamellar vesicle liposomes (MLV) containing a lipopeptide analogue of a fragment from the cell wall of gram-negative bacteria (CGP 31362) can render BALB/c mouse alveolar macrophages tumoricidal in situ and reduce the incidence of spontaneous lung metastasis of syngeneic renal carcinoma (RENCA) cells. Alveolar macrophages (a) incubated in vitro with MLV containing CGP 31362 (MLV-31362) and (b) harvested from mice injected i.v. with MLV-31362 were rendered cytotoxic against the RENCA cells. Maximum cytotoxic activity of the macrophages was induced by injecting 5 mumol MLV consisting of 250 mg phospholipids and 0.5 mg CGP 31362. The single i.v. injection of 5 mumol MLV-31362 produced activation of macrophages that lasted for up to 4 days. Repeated i.v. injections of MLV-31362 produced a continuous antitumor activity in alveolar macrophages. To study the lipopeptide's effects on metastasis, we injected the left kidneys of BALB/c mice with RENCA cells. The kidney with growing tumor was resected 10 days later and, after a further 2 days, groups of mice were injected i.v. with MLV-31362 or with MLV-HBSS (twice weekly for 3 weeks). Treatment with MLV-31362 significantly decreased the median number of spontaneous lung metastases. These data demonstrate that the systemic administration of MLV-31362 can activate murine lung macrophages in situ and reduce the incidence of spontaneous RENCA lung metastases.
Collapse
Affiliation(s)
- T Utsugi
- Department of Cell Biology, University of Texas M.D. Anderson Cancer Center, Houston 77030
| | | | | | | |
Collapse
|
29
|
Fidler IJ, Naito S, Pathak S. Orthotopic implantation is essential for the selection, growth and metastasis of human renal cell cancer in nude mice [corrected]. Cancer Metastasis Rev 1990; 9:149-65. [PMID: 2253314 DOI: 10.1007/bf00046341] [Citation(s) in RCA: 78] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human neoplasms are heterogeneous for a variety of biological properties that include invasion and metastasis. The presence of a small subpopulation of cells with a highly metastatic phenotype has important clinical implications for diagnosis and therapy of cancer. For this reason, it is important to develop an animal model for the selection and isolation of metastatic variants from human neoplasms and for testing the metastatic potential of human tumor cells. We have implanted human renal cell carcinoma (HRCC) cells (obtained from a surgical specimen) into different organs of nude mice and then recovered the tumors and established each in culture. The 5 established lines differed in their biological-metastatic properties and had a unique karyotype, indicating that growth at different organs selects for different subpopulations of HRCC. Moreover, the HRCC did not metastasize unless they were implanted orthotopically. These findings indicate that the appropriate nude mouse model for studying the biology and therapy of HRCC must be based on the orthotopic implantation of tumor cells.
Collapse
Affiliation(s)
- I J Fidler
- Department of Cell Biology, University of Texas M.D. Anderson Cancer Center, Houston 77030
| | | | | |
Collapse
|
30
|
Gregorian SK, Battisto JR. Immunosuppression in murine renal cell carcinoma. I. Characterization of extent, severity and sources. Cancer Immunol Immunother 1990; 31:325-34. [PMID: 2386978 PMCID: PMC11038414 DOI: 10.1007/bf01741403] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/1989] [Accepted: 03/07/1990] [Indexed: 12/31/2022]
Abstract
Four cell-mediated immunological responses related to tumor elimination have been examined in mice injected with a transplantable renal cell carcinoma (Renca). Lymphokine-activated killer (LAK) cells generated in vitro from spleen cells of normal mice were capable of attacking Renca, EL-4, P815 and YAC-1 targets, but those from mice bearing Renca for 3 weeks could not. Natural killer activity, stimulated in vivo by administering poly(I) poly(C), was less than 50% of normal in Renca-bearing hosts. In addition, development of cytotoxic T lymphocytes to allogeneic targets was markedly inhibited in mice possessing the renal tumor. Finally, the delayed hypersensitivity response to a dermally applied hapten was approximately 70% less than normal in tumor-bearing mice, no matter whether the tumor existed subcutaneously or intrarenally. A kinetic study of the development of non-responsiveness using the LAK assay showed onset of poor response at 1 week, which became maximal within 3 weeks following receipt of tumor subcutaneously. The immunological depression was seen to be attributable in part to suppressor cells present among spleen cells but not bone marrow cells of tumor-bearing hosts. The suppressor cells prevented in vitro LAK generation by normal spleen cells and, when adoptively transferred to normal mice, they inhibited natural killer stimulation and delayed hypersensitivity generation. Another source of immunological down-regulation was provided by Renca cells themselves. Incorporation of Renca cells that had been X-irradiated with 30,000 rad into cultures of normal and Renca-derived splenic cells suppressed replication of both almost completely. Furthermore, the presence of X-irradiated Renca cells in cultures of normal spleen cells prevented development of LAK cells. Thus, the suppression seen in Renca-bearing mice derives from multiple sources and whether each is in any way related to the other has been discussed. Identification of the phenotypes of cells responsible for the lymphoid cell-mediated suppression and examination of its elimination are communicated in the companion paper.
Collapse
Affiliation(s)
- S K Gregorian
- Department of Chemistry, Cleveland State University, Ohio 44115
| | | |
Collapse
|
31
|
Gregorian SK, Battisto JR. Immunosuppression in murine renal cell carcinoma. II. Identification of responsible lymphoid cell phenotypes and examination of elimination of suppression. Cancer Immunol Immunother 1990; 31:335-41. [PMID: 1974826 PMCID: PMC11038562 DOI: 10.1007/bf01741404] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/1989] [Accepted: 03/22/1990] [Indexed: 12/29/2022]
Abstract
In our companion paper we have reported that cell-mediated immunity of mice bearing renal cell carcinoma is profoundly suppressed. The non-responsiveness of such animals was found to be attributable to Renca cells themselves and to splenic lymphoid cells that down-regulate other fully capable lymphoid cells. In this communication the lymphoid cell source of suppression within Renca-bearing mice has been explored with the aim of identifying phenotypes of the responsible cells, the manner by which suppression is mediated, and initial ways by which suppression may be eliminated. A plastic-adherent cell bearing the Thy1.2 surface marker as well as the Lyt1 and Lyt2 antigens has been found to operate, perhaps in conjunction with macrophages, to down-regulate lymphokine-activated killer (LAK) cell development for natural killer (NK) and non-NK targets that include Renca cells themselves. The splenic suppressor cells lost the capacity to suppress the NK response of normal recipient mice upon shallow irradiation (250 rad) prior to adoptive transfer. Spleen cells, presumably macrophages, from Renca-bearing mice were found to suppress the generation of LAK and NK cells in vitro by synthesizing prostaglandins. Indomethacin, a prostaglandin synthetase inhibitor, blocked the induction of suppression both in vitro and in vivo, suggesting the presence of endogenous prostaglandins in Renca-bearing mice. The suppression seen in Renca-bearing mice that derives from multiple sources and has been prevented by two separate methods has been discussed from the viewpoint of the inter-relatedness of the sources.
Collapse
Affiliation(s)
- S K Gregorian
- Department of Chemistry, Cleveland State University, Ohio 44115
| | | |
Collapse
|
32
|
Shimizu T, Pelletier H, Hammann A, Olsson NO, Martin MS, Martin F. Effects of a single injection of anti-asialo GM1 serum on natural cytotoxicity and the growth of a regressive colonic tumor in syngeneic rats. Int J Cancer 1987; 40:676-80. [PMID: 3316050 DOI: 10.1002/ijc.2910400518] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The REGb tumor cell line is a cloned variant of the DHD-K12 cell line, established from a colon carcinoma chemically induced in the rat. Unlike the parent DHD-K12 cell line, or other clones, which give progressive tumors when inoculated to the syngeneic rat, REGb cells produce tumors which regress in 3 to 5 weeks and never cause metastasis. In order to explore the role of natural killer (NK) cells in REGb tumor regression, each rat was given one injection of anti-asialoGM1 (anti-asGM1) serum, a known inhibitor of NK activity. This injection was done 24 hr before REGb cell challenge. This injection significantly depressed the in vitro cytotoxicity of peripheral blood lymphocytes on REGb cells for 2 weeks. REGb tumors grew larger and regressed later in the treated animals than those in the controls. Furthermore, a progressive or recurrent tumor was observed in 4 out of 10 treated rats, giving lung and/or lymph-node metastases in 2 cases. Immuno-histological study of the cells infiltrating the REGb tumors in control and treated animals showed a decrease number of asGM1+ and OX8+ lymphocytes, presumably NK cells, after anti-asGM1 treatment. An increase in number of macrophages was demonstrated in the progressive tumors of treated animals. These results suggest that NK cells play an important role in the initial stage of the regression TSb tumors in untreated syngeneic rats.
Collapse
Affiliation(s)
- T Shimizu
- INSERM U.252, Faculty of Medicine, University of Dijon, France
| | | | | | | | | | | |
Collapse
|
33
|
Salup RR, Wiltrout RH. Treatment of adenocarcinoma in the peritoneum of mice: chemoimmunotherapy with IL-2-stimulated cytotoxic lymphocytes as a model for treatment of minimal residual disease. Cancer Immunol Immunother 1986; 22:31-6. [PMID: 3486715 PMCID: PMC11038753 DOI: 10.1007/bf00205713] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/1985] [Accepted: 10/14/1985] [Indexed: 01/06/2023]
Abstract
We have used a transplantable murine adenocarcinoma of renal origin (Renca) introduced to the abdomen by i.p. injection of a tumor cell suspension, to study the therapeutic potential of adoptive immunotherapy and/or biological response modifiers (BRMs). This tumor model is therapeutically challenging since the tumor grows progressively resulting in extensive peritoneal carcinomatosis, with hemorrhagic ascites, metastases to abdominal lymph nodes, liver, most serous membranes, spleen, and in some animals, pulmonary metastases. Without therapy, death occurs invariably in 36 +/- 3 days. In vitro, the tumor is lysed by lymphocytes obtained from the peritoneal cavity of mice treated with human recombinant interleukin-2 (rIL-2) and by cytotoxic lymphocytes stimulated by in vitro culture with human rIL-2. Treatment of i.p. Renca with a single i.p. injection of the chemotherapeutic agent doxorubicin hydrochloride (DOX), or adoptive transfer of in vitro stimulated cytotoxic lymphocytes together with rIL-2 cured 50% and 20% of the tumor-bearing mice, respectively. In contrast, combined therapy with DOX and adoptive transfer of in vitro stimulated cytotoxic lymphocytes and rIL-2 cured the majority (90%) of tumor-bearing mice. These results suggest that administration of immunotherapy with in vitro activated cytotoxic cells together with human rIL-2 substantially enhances the effectiveness of chemotherapy.
Collapse
|