1
|
Sun W, Xie S, Liu SF, Hu X, Xing D. Evolving Tumor Characteristics and Smart Nanodrugs for Tumor Immunotherapy. Int J Nanomedicine 2024; 19:3919-3942. [PMID: 38708176 PMCID: PMC11070166 DOI: 10.2147/ijn.s453265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
Typical physiological characteristics of tumors, such as weak acidity, low oxygen content, and upregulation of certain enzymes in the tumor microenvironment (TME), provide survival advantages when exposed to targeted attacks by drugs and responsive nanomedicines. Consequently, cancer treatment has significantly progressed in recent years. However, the evolution and adaptation of tumor characteristics still pose many challenges for current treatment methods. Therefore, efficient and precise cancer treatments require an understanding of the heterogeneity degree of various factors in cancer cells during tumor evolution to exploit the typical TME characteristics and manage the mutation process. The highly heterogeneous tumor and infiltrating stromal cells, immune cells, and extracellular components collectively form a unique TME, which plays a crucial role in tumor malignancy, including proliferation, invasion, metastasis, and immune escape. Therefore, the development of new treatment methods that can adapt to the evolutionary characteristics of tumors has become an intense focus in current cancer treatment research. This paper explores the latest understanding of cancer evolution, focusing on how tumors use new antigens to shape their "new faces"; how immune system cells, such as cytotoxic T cells, regulatory T cells, macrophages, and natural killer cells, help tumors become "invisible", that is, immune escape; whether the diverse cancer-associated fibroblasts provide support and coordination for tumors; and whether it is possible to attack tumors in reverse. This paper discusses the limitations of targeted therapy driven by tumor evolution factors and explores future strategies and the potential of intelligent nanomedicines, including the systematic coordination of tumor evolution factors and adaptive methods, to meet this therapeutic challenge.
Collapse
Affiliation(s)
- Wenshe Sun
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
- Qingdao Cancer Institute, Qingdao University, Qingdao, 266071, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, People’s Republic of China
| | - Shaowei Xie
- Department of Ultrasound, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - Shi Feng Liu
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Xiaokun Hu
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
- Qingdao Cancer Institute, Qingdao University, Qingdao, 266071, People’s Republic of China
| |
Collapse
|
2
|
Li L, Zhu X, Qian Y, Yuan X, Ding Y, Hu D, He X, Wu Y. Chimeric Antigen Receptor T-Cell Therapy in Glioblastoma: Current and Future. Front Immunol 2020; 11:594271. [PMID: 33224149 PMCID: PMC7669545 DOI: 10.3389/fimmu.2020.594271] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is a highly aggressive glioma with an extremely poor prognosis after conventional treatment. Recent advances in immunotherapy offer hope for these patients with incurable GBM. Our present review aimed to provide an overview of immunotherapy for GBM, especially chimeric antigen receptor T-cell (CAR T) therapy. CAR T-cell immunotherapy, which involves the engineering of T cells to kill tumors by targeting cell surface-specific antigens, has been successful in eliminating B-cell leukemia by targeting CD19. IL-13Rα2, EGFRvIII, and HER2-targeted CAR T cells have shown significant clinical efficacy and safety in phase 1 or 2 clinical trials conducted in patients with GBM; these findings support the need for further studies to examine if this therapy can ultimately benefit this patient group. However, local physical barriers, high tumor heterogeneity, and antigen escape make the use of CAR T therapy, as a treatment for GBM, challenging. The potential directions for improving the efficacy of CAR T in GBM are to combine the existing traditional therapies and the construction of multi-target CAR T cells.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Brain Neoplasms/etiology
- Brain Neoplasms/therapy
- Combined Modality Therapy/methods
- Genetic Engineering
- Glioblastoma/etiology
- Glioblastoma/therapy
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Treatment Outcome
Collapse
Affiliation(s)
- Long Li
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiqun Zhu
- Department of Surgical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Qian
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangling Yuan
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Ding
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Desheng Hu
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin He
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yuan Wu
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Cao JX, Gao WJ, You J, Wu LH, Wang ZX. Assessment of the efficacy of passive cellular immunotherapy for glioma patients. Rev Neurosci 2020; 31:427-440. [PMID: 31926107 DOI: 10.1515/revneuro-2019-0102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/08/2019] [Indexed: 11/15/2022]
Abstract
To evaluate the therapeutic efficacy of passive cellular immunotherapy for glioma, a total of 979 patients were assigned to the meta-analysis. PubMed and the Cochrane Central Register of Controlled Trials were searched initially from February 2018 and updated in April 2019. The overall survival (OS) rates and Karnofsky performance status (KPS) values of patients who underwent passive cellular immunotherapy were compared to those of patients who did not undergo immunotherapy. The proportion of survival rates was also evaluated in one group of clinical trials. Pooled analysis was performed with random- or fixed-effects models. Clinical trials of lymphokine-activated killer cells, cytotoxic T lymphocytes, autologous tumor-specific T lymphocytes, chimeric antigen receptor T cells, cytokine-induced killer cells, cytomegalovirus-specific T cells, and natural killer cell therapies were selected. Results showed that treatment of glioma with passive cellular immunotherapy was associated with a significantly improved 0.5-year OS (p = 0.003) as well as improved 1-, 1.5-, and 3-year OS (p ≤ 0.05). A meta-analysis of 206 patients in one group of clinical trials with 12-month follow-up showed that the overall pooled survival rate was 37.9% (p = 0.003). Analysis of KPS values demonstrated favorable results for the immunotherapy arm (p < 0.001). Thus, the present meta-analysis showed that passive cellular immunotherapy prolongs survival and improves quality of life for glioma patients, suggesting that it has some clinical benefits.
Collapse
Affiliation(s)
- Jun-Xia Cao
- Biotherapy Center, The Seventh Medical Center of PLA General Hospital, No. 5 Nan Men Cang Road, Dongcheng District, Beijing 100700, China
| | - Wei-Jian Gao
- Biotherapy Center, The Seventh Medical Center of PLA General Hospital, No. 5 Nan Men Cang Road, Dongcheng District, Beijing 100700, China
| | - Jia You
- Biotherapy Center, The Seventh Medical Center of PLA General Hospital, No. 5 Nan Men Cang Road, Dongcheng District, Beijing 100700, China
| | - Li-Hua Wu
- Biotherapy Center, The Seventh Medical Center of PLA General Hospital, No. 5 Nan Men Cang Road, Dongcheng District, Beijing 100700, China
| | - Zheng-Xu Wang
- Biotherapy Center, The Seventh Medical Center of PLA General Hospital, No. 5 Nan Men Cang Road, Dongcheng District, Beijing 100700, China, e-mail:
| |
Collapse
|
4
|
Biomarkers for glioma immunotherapy: the next generation. J Neurooncol 2015; 123:359-72. [PMID: 25724916 DOI: 10.1007/s11060-015-1746-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 02/16/2015] [Indexed: 12/11/2022]
Abstract
The term "biomarker" historically refers to a single parameter, such as the expression level of a gene or a radiographic pattern, used to indicate a broader biological state. Molecular indicators have been applied to several aspects of cancer therapy: to describe the genotypic and phenotypic state of neoplastic tissue for prognosis, to predict susceptibility to anti-proliferative agents, to validate the presence of specific drug targets, and to evaluate responsiveness to therapy. For glioblastoma (GBM), immunohistochemical and radiographic biomarkers accessible to the clinical lab have informed traditional regimens, but while immunotherapies have emerged as potentially disruptive weapons against this diffusely infiltrating, heterogeneous tumor, biomarkers with strong predictive power have not been fully established. The cancer immunotherapy field, through the recently accelerated expansion of trials, is currently leveraging this wealth of clinical and biological data to define and revise the use of biomarkers for improving prognostic accuracy, personalization of therapy, and evaluation of responses across the wide variety of tumors. Technological advancements in DNA sequencing, cytometry, and microscopy have facilitated the exploration of more integrated, high-dimensional profiling of the disease system-incorporating both immune and tumor parameters-rather than single metrics, as biomarkers for therapeutic sensitivity. Here we discuss the utility of traditional GBM biomarkers in immunotherapy and how the impending transformation of the biomarker paradigm-from single markers to integrated profiles-may offer the key to bringing predictive, personalized immunotherapy to GBM patients.
Collapse
|
5
|
Glioma stem cells and immunotherapy for the treatment of malignant gliomas. ISRN ONCOLOGY 2013; 2013:673793. [PMID: 23762610 PMCID: PMC3671309 DOI: 10.1155/2013/673793] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 03/27/2013] [Indexed: 02/06/2023]
Abstract
Stem cell research has led to the discovery of glioma stem cells (GSCs), and because these cells are resistant to chemotherapy and radiotherapy, analysis of their properties has been rapidly pursued for targeted treatment of malignant glioma. Recent studies have also revealed complex crosstalk between GSCs and their specialized environment (niche). Therefore, targeting not only GSCs but also their niche may be a principle for novel therapies of malignant glioma. One possible novel strategy for targeting GSCs and their niches is immunotherapy with different antitumor mechanism(s) from those of conventional therapy. Recent clinical studies of immunotherapy using peptide vaccines and antibodies have shown promising results. This review describes the recent findings related to GSCs and their niches, as well as immunotherapies for glioma, followed by discussion of immunotherapies that target GSCs for the treatment of malignant glioma.
Collapse
|
6
|
Hishii M, Nitta T, Ebato M, Okumura K, Sato K. Targeting therapy for glioma by LAK cells coupled with bispecific antibodies. J Clin Neurosci 2012; 1:261-5. [PMID: 18638771 DOI: 10.1016/0967-5868(94)90067-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/1993] [Accepted: 11/19/1993] [Indexed: 11/29/2022]
Abstract
Targeting of T cells or natural killer (NK) cells to tumours by using bispecific antibodies has attracted increasing interest in the past few years. We treated 31 patients with malignant glioma using adoptive transfer of lymphokine-activated killer (LAK) cells coupled to a bispecific antibody (anti-CD3+anti-glioma) as post-operative adjuvant therapy. Although this study excluded patients with deeply-seated tumours or a poor performance status, approximately 50% of the patients remained alive after 3 years, and 40% were free of recurrence. Serial CT scans revealed disappearance of remnant tumours in some patients. In addition, CT-guided stereotactic biopsy of the tumour in 3 patients showed extensive necrosis and degeneration after specific targeting therapy (STT). Five patients developed acute infection, and one of them died of bacterial meningitis. Our results suggest that antibody-targeted LAK therapy can achieve a higher response rate in patients with standard LAK therapy or any type of conventional treatment.
Collapse
Affiliation(s)
- M Hishii
- Departments of Neurosurgery and Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | |
Collapse
|
7
|
|
8
|
Wainwright DA, Nigam P, Thaci B, Dey M, Lesniak MS. Recent developments on immunotherapy for brain cancer. Expert Opin Emerg Drugs 2012; 17:181-202. [PMID: 22533851 DOI: 10.1517/14728214.2012.679929] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Brain tumors are a unique class of cancers since they are anatomically shielded from normal immunosurveillance by the blood-brain barrier, lack a normal lymphatic drainage system and reside in a potently immunosuppressive environment. Of the primary brain cancers, glioblastoma multiforme (GBM) is the most common and aggressive in adults. Although treatment options include surgery, radiation and chemotherapy, the average lifespan of GBM patients remains at only 14.6 months post-diagnosis. AREAS COVERED A review of key cellular and molecular immune system mediators in the context of brain tumors including TGF-β, cytotoxic T cells, Tregs, CTLA-4, PD-1 and IDO is discussed. In addition, prognostic factors, currently utilized immunotherapeutic strategies, ongoing clinical trials and a discussion of new or potential immunotherapies for brain tumor patients are considered. EXPERT OPINION Current drugs that improve the quality of life and overall survival in patients with brain tumors, especially for GBM, are poorly effective. This disease requires a reanalysis of currently accepted treatment strategies, as well as newly designed approaches. Here, we review the fundamental aspects of immunosuppression in brain tumors, new and promising immunotherapeutic drugs as well as combinatorial strategies that focus on the simultaneous inhibition of immunosuppressive hubs, both in immune and brain tumor cells, which is critical to consider for achieving future success for the treatment of this devastating disease.
Collapse
|
9
|
Ishikawa E, Takano S, Ohno T, Tsuboi K. Adoptive cell transfer therapy for malignant gliomas. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 746:109-20. [PMID: 22639163 DOI: 10.1007/978-1-4614-3146-6_9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
To date, various adoptive immunotherapies have been attempted for treatment of malignant gliomas using nonspecific and/or specific effector cells. Since the late 1980s, with the development of rIL-2, the efficacy of lymphokine-activated killer (LAK) cell therapy with or without rIL-2 for malignant gliomas had been tested with some modifications in therapeutic protocols. With advancements in technology, ex vivo expanded tumor specific cytotoxic T-lymphocytes (CTL) or those lineages were used in clinical trials with higher tumor response rates. In addition, combinations of those adoptive cell transfer using LAK cells, CTLs or natural killer (NK) cells with autologous tumor vaccine (ATV) therapy were attempted. Also, a strategy of high-dose (or lymphodepleting) chemotherapy followed by adoptive cell transfer has been drawing attentions recently. The most important role of these clinical studies using cell therapy was to prove that these ex vivo expanded effector cells could kill tumor cells in vivo. Although recent clinical results could demonstrate radiologic tumor shrinkage in a number of cases, cell transfer therapy alone has been utilized less frequently, because of the high cost of ex vivo cell expansion, the short duration of antitumor activity in vivo, and the recent shift of interest to vaccine immunotherapy. Nevertheless, NK cell therapy using specific feeder cells or allergenic NK cell lines have potentials to be a good choice of treatment because of easy ex vivo expansion and their efficacy especially when combined with vaccine therapy as they are complementary to each other. Also, further studies are expected to clarify the efficacy of the high-dose chemotherapy followed by a large scale cell transfer therapy as a new therapeutic strategy for malignant gliomas.
Collapse
Affiliation(s)
- Eiichi Ishikawa
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | | | | | | |
Collapse
|
10
|
Marshall D, Mitchell DA, Graner MW, Bigner DD. Immunotherapy of brain tumors. HANDBOOK OF CLINICAL NEUROLOGY 2012; 104:309-30. [PMID: 22230450 DOI: 10.1016/b978-0-444-52138-5.00020-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
11
|
Han SJ, Kaur G, Yang I, Lim M. Biologic Principles of Immunotherapy for Malignant Gliomas. Neurosurg Clin N Am 2010; 21:1-16. [DOI: 10.1016/j.nec.2009.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
12
|
Abstract
Despite aggressive multi-modality therapy including surgery, radiation, and chemotherapy, the prognosis for patients with malignant primary brain tumors remains very poor. Moreover, the non-specific nature of conventional therapy for brain tumors often results in incapacitating damage to surrounding normal brain and systemic tissues. Thus, there is an urgent need for the development of therapeutic strategies that precisely target tumor cells while minimizing collateral damage to neighboring eloquent cerebral cortex. The rationale for using the immune system to target brain tumors is based on the premise that the inherent specificity of immunologic reactivity could meet the clear need for more specific and precise therapy. The success of this modality is dependent on our ability to understand the mechanisms of immune regulation within the central nervous system (CNS), as well as counter the broad defects in host cell-mediated immunity that malignant gliomas are known to elicit. Recent advances in our understanding of tumor-induced and host-mediated immunosuppressive mechanisms, the development of effective strategies to combat these suppressive effects, and a better understanding of how to deliver immunologic effector molecules more efficiently to CNS tumors have all facilitated significant progress toward the realization of true clinical benefit from immunotherapeutic treatment of malignant gliomas.
Collapse
Affiliation(s)
- Duane A Mitchell
- Division of Neurosurgery, Department of Surgery, The Preston Robert Tisch Brain Tumor Center, Duke, NC 27710, USA.
| | | | | |
Collapse
|
13
|
Abstract
Despite remarkable advancements in imaging modalities and treatment options available to patients diagnosed with malignant brain tumors, the prognosis for those with high-grade lesions remains poor. The imprecise mechanisms of currently available treatments to manage these tumors do not spare damage to the normal surrounding brain and often result in major cognitive and motor deficits. Immunotherapy holds the promise of offering a potent, yet targeted, treatment to patients with brain tumors, with the potential to eradicate the malignant tumor cells without damaging normal tissues. The T cells of the immune system are uniquely capable of recognizing the altered protein expression patterns within tumor cells and mediating their destruction through a variety of effector mechanisms. Adoptive T-cell therapy is an attempt to harness and amplify the tumor-eradicating capacity of a patients' own T cells and then return these effectors to the patient in such a state that they effectively eliminate residual tumor. Although this approach is not new to the field of tumor immunology, new advancements in our understanding of T-cell activation and function and breakthroughs in tumor antigen discovery hold great promise for the translation of this modality into a clinical success.
Collapse
Affiliation(s)
- Duane A Mitchell
- Department of Pathology and Department of Surgery, Duke University Medical Center, Box 3807, Durham, NC 27710, USA
| | | | | |
Collapse
|
14
|
Hayes RL, Arbit E, Odaimi M, Pannullo S, Scheff R, Kravchinskiy D, Zaroulis C. Adoptive cellular immunotherapy for the treatment of malignant gliomas. Crit Rev Oncol Hematol 2001; 39:31-42. [PMID: 11418300 DOI: 10.1016/s1040-8428(01)00122-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
UNLABELLED The median survival for adults with recurrent primary malignant gliomas is 56 weeks following surgery, radiation, and chemotherapy. Generally, reoperation can extend the median survival an additional 26-32 weeks. We have developed an aggressive treatment program that utilizes low doses of interleukin-2 (IL-2) combined with ex vivo activated killer cells (LAK) infused via an indwelling catheter placed into the surgical resection cavity. Autologous leukocytes were collected during a standard 3-4 h, outpatient leukapheresis procedure, then activated ex vivo for 4-5 days with high doses of IL-2. The treatment protocol consisted of two 2-week cycles of therapy over a 6-week period. Patients with stable disease or objective response on follow-up MRI scans were retreated at 3-month intervals. Acute and cumulative IL-2-related toxicities were observed, but limited, and included fever, headache and transient neurologic irritation. Corticosteroid levels and usage were strictly controlled during immunotherapy, although higher doses were used intermittently to mitigate toxicity. Biologic changes included lymphocytic infiltration, regional eosinophilia, tumor necrosis, and the localized production of IL-2, IFN-gamma and IL-12, demonstrated by in situ hybridization and immunohistochemistry. SUMMARY IL-2 plus autogeneic LAK cells can be safely administered intracavitary to treat high grade primary brain tumors with limited toxicity within the central nervous system. Six out of 28 patients had long-term survival of greater than 2 years post-reoperation plus immunotherapy with 2 patients alive over 8 years. The presence of a marked regional eosinophilia appeared to correlate with increased survival and may be predictive of a biologic and therapeutic response. Regional adoptive immune therapy was well tolerated and should be considered an option for patients with high-grade tumors refractive to standard therapeutic approaches.
Collapse
Affiliation(s)
- R L Hayes
- Department of Medicine, Immunotherapy Program, Sanford R. Nalitt Institute for Cancer and Blood-Related Diseases, Staten Island University Hospital, 256 Mason Avenue, Staten Island, NY 10305, USA.
| | | | | | | | | | | | | |
Collapse
|
15
|
Okada H, Pollack IF, Lieberman F, Lunsford LD, Kondziolka D, Schiff D, Attanucci J, Edington H, Chambers W, Kalinski P, Kinzler D, Whiteside T, Elder E, Potter D. Gene therapy of malignant gliomas: a pilot study of vaccination with irradiated autologous glioma and dendritic cells admixed with IL-4 transduced fibroblasts to elicit an immune response. Hum Gene Ther 2001; 12:575-95. [PMID: 11268289 DOI: 10.1089/104303401300042528] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- H Okada
- Department of Neurological Surgery, University of Pittsburgh Medical Center, PA 15213, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Okada H, Pollack IF, Lotze MT, Lunsford LD, Kondziolka D, Lieberman F, Schiff D, Attanucci J, Edington H, Chambers W, Robbins P, Baar J, Kinzler D, Whiteside T, Elder E. Gene therapy of malignant gliomas: a phase I study of IL-4-HSV-TK gene-modified autologous tumor to elicit an immune response. Hum Gene Ther 2000; 11:637-53. [PMID: 10724042 DOI: 10.1089/10430340050015824] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- H Okada
- University of Pittsburgh Cancer Institute, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Plautz GE, Barnett GH, Miller DW, Cohen BH, Prayson RA, Krauss JC, Luciano M, Kangisser DB, Shu S. Systemic T cell adoptive immunotherapy of malignant gliomas. J Neurosurg 1998; 89:42-51. [PMID: 9647171 DOI: 10.3171/jns.1998.89.1.0042] [Citation(s) in RCA: 96] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT To determine the feasibility, toxicity, and potential therapeutic benefits of systemic adoptive immunotherapy, 10 patients with progressive primary or recurrent malignant glioma received this treatment. Adoptive immunotherapy, the transfer of immune T lymphocytes, is capable of mediating the regression of experimental brain tumors in animal models. In animal models, lymph nodes (LNs) that drain the tumor vaccine site are a rich source of tumor-immune T cells. METHODS In this clinical study, patients were inoculated intradermally with irradiated autologous tumor cells and granulocyte macrophage-colony stimulating factor as an adjuvant. Cells from draining inguinal LNs, surgically resected 7 days after vaccination, were stimulated sequentially with staphylococcal enterotoxin A and anti-CD3, and a low dose of interleukin-2 (60 IU/ml) was used to expand the stimulated cells. The maximum cell proliferation was 350-fold over 10 days of culture. The activated cells were virtually all T cells consisting of various proportions of CD4 and CD8 cells. These cells were given to patients by intravenous infusion at doses ranging from 9 x 10(8) to 1.5 x 10(11). There were no Grade 3 or 4 toxicities associated with the treatment. Following T-cell transfer therapy, radiographic regression that lasted at least 6 months was demonstrated in two patients with recurrent tumors. One patient demonstrated stable disease that has lasted for more than 17 months. The remaining patients had progressive disease; however, four of the eight patients with recurrent tumor remain alive more than 1 year after surgery for recurrence. Three patients required intervention with corticosteroid agents or additional surgery approximately 1 month following cell transfer. CONCLUSIONS These intriguing clinical observations warrant further trials to determine whether this approach can provide therapeutic benefits and improve survival.
Collapse
Affiliation(s)
- G E Plautz
- Department of Neurological Surgery, Center for Surgery Research, The Cleveland Clinic Foundation, Ohio 44195, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Plautz GE, Barnett GH, Miller DW, Cohen BH, Prayson RA, Krauss JC, Luciano M, Shu S. Activated lymph node T cells for systemic adoptive immunotherapy of malignant glioma. Neurosurg Focus 1997. [DOI: 10.3171/foc.1997.3.5.8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Ten patients with progressive primary or recurrent malignant glioma received systemic adoptive immunotherapy to determine the feasibility, toxicity, and potential therapeutic benefits of this treatment. Adoptive immunotherapy, the transfer of immune T lymphocytes, is capable of mediating the regression of experimental brain tumors in animal models. A rich source of tumor-immune T cells are lymph nodes (LNs) draining the tumor vaccine site. In this clinical study, patients were inoculated intradermally with irradiated autologous tumor cells and granulocyte-macrophage colony stimulating factor as an adjuvant. Cells from draining inguinal LNs, surgically resected 7 days after vaccination, were stimulated sequentially with staphylococcal enterotoxin A and anti-CD3, and a low dose of interleukin-2 (60 IU/ml) was used to expand the stimulated cells. The maximum cell proliferation was 350-fold over 10 days of culture. The activated cells were virtually all T cells consisting of various proportions of CD4 and CD8 cells. These cells were given to patients by intravenous infusion at doses ranging from 9 X 108 to 1.5 X 1011. There were no Grade 3 or 4 toxicities associated with the treatment. Following T cell transfer therapy, radiographic regression that lasted at least 4 months was demonstrated in three patients with recurrent tumors, and four patients remain alive more than 11 months after surgery. The remaining patients had progressive disease, and three patients required intervention with corticosteroid agents or additional surgery approximately 1 month following cell transfer. These findings demonstrate that adoptive immunotherapy can be administered in patients with glioma without causing significant toxicity. It appears that this experimental regimen can provide therapeutic benefits for some patients.
Collapse
|
19
|
Yoshida J, Takaoka T, Mizuno M, Momota H, Okada H. Cytolysis of malignant glioma cells by lymphokine-activated killer cells combined with anti-CD3/antiglioma bifunctional antibody and tumor necrosis factor-alpha. J Surg Oncol 1996; 62:177-82. [PMID: 8667624 DOI: 10.1002/(sici)1096-9098(199607)62:3<177::aid-jso6>3.0.co;2-4] [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: 02/01/2023]
Abstract
With the aim of developing an effective immunotherapy for malignant glioma, glioma cells were incubated with tumor necrosis factor-alpha (TNF-alpha) to increase their susceptibility to lysis by lymphokine-activated killer (LAK) cells. Treatment with exogenous TNF-alpha induced the expression of intercellular adhesion molecule-1 (ICAM-1) on the surface of glioma cells. In addition, the cytolytic activity of LAK cells toward exogenous TNF-alpha treated glioma cells was significantly greater than LAK cell activity toward untreated glioma cells. This increase in cytolytic activity was blocked by anti-ICAM-1 monoclonal antibodies (MAb). Furthermore, co-treatment with a bifunctional antibody (BFA) composed of anti-CD3 (UCHT1) and antiglioma (G-22) antibodies synergistically increased the cytolytic activity of LAK cells towards TNF-alpha-treated glioma cells. These results indicate that a combination of exogenous TNF-alpha and anti-CD3/antiglioma BFA may provide an effective modified adoptive immunotherapy for patients with malignant glioma.
Collapse
MESH Headings
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/therapeutic use
- Antibodies, Monoclonal/pharmacology
- Antibodies, Neoplasm/immunology
- Antibodies, Neoplasm/therapeutic use
- CD3 Complex/immunology
- CD3 Complex/therapeutic use
- Cytotoxicity, Immunologic/drug effects
- Flow Cytometry
- Glioma/immunology
- Glioma/therapy
- Humans
- Immunotherapy, Adoptive/methods
- Intercellular Adhesion Molecule-1/drug effects
- Intercellular Adhesion Molecule-1/immunology
- Killer Cells, Lymphokine-Activated/drug effects
- Killer Cells, Lymphokine-Activated/immunology
- Recombinant Proteins/pharmacology
- Recombinant Proteins/therapeutic use
- Tumor Cells, Cultured
- Tumor Necrosis Factor-alpha/pharmacology
- Tumor Necrosis Factor-alpha/therapeutic use
Collapse
Affiliation(s)
- J Yoshida
- Department of Neurosurgery, Nagoya University School of Medicine, Japan
| | | | | | | | | |
Collapse
|
20
|
Sankhla SK, Nadkarni JS, Bhagwati SN. Adoptive immunotherapy using lymphokine-activated killer (LAK) cells and interleukin-2 for recurrent malignant primary brain tumors. J Neurooncol 1996; 27:133-40. [PMID: 8699235 DOI: 10.1007/bf00177476] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Ten patients with recurrent malignant primary brain neoplasms were treated with adoptive immunotherapy using lymphokine-activated killer (LAK) cells and interleukin-2 (IL-2). Nine patients had supratentorial glioma and they received multiple intratumoral instillations of LAK cells through reservoir-catheter system or burrhole. The other patients with disseminated subarachnoid metastases from posterior fossa medulloblastoma received immunotherapy via lumbar subarachnoid route. A partial and transient clinical response was observed in two patients. following the therapy, and a cystic transformation of the essentially solid tumour was noted on the CT scans of these two patients. No significant clinical or radiological response to the treatment was observed in the remaining 8 patients. The results of this preliminary study reveal limitations of the regional intratumoral adoptive immunotherapy using currently available techniques and provide sufficient evidence of its effectiveness to warrant further investigations.
Collapse
Affiliation(s)
- S K Sankhla
- Department of Neurosurgery, Bombay Hospital, India
| | | | | |
Collapse
|
21
|
Abstract
Cytokines are a group of molecules with an extremely broad range of activities on a variety of target cells. This review summarizes the known cytokine and cytokine receptor expression in primary brain tumors and derived cell lines. These expression patterns are compared with those occurring in other CNS diseases, such as virus or bacterial infections, experimental allergic encephalitis, multiple sclerosis, and trauma. A variety of cytokines are expressed during CNS neoplasia; their potential involvement in tumor growth through a variety of mechanisms, such as autocrine or paracrine growth stimulation, angiogenesis, and immune surveillance evasion, are discussed. Finally, results of preliminary therapeutic approaches with cytokines are critically evaluated.
Collapse
Affiliation(s)
- E G Van Meir
- Neurosurgery Department, University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
22
|
Takaoka T, Yoshida J, Mizuno M, Sugita K. Transfection-induced tumor necrosis factor-alpha increases the susceptibility of human glioma cells to lysis by lymphokine-activated killer cells: continuous expression of intercellular adhesion molecule-1 on the glioma cells. Jpn J Cancer Res 1994; 85:750-5. [PMID: 7915264 PMCID: PMC5919548 DOI: 10.1111/j.1349-7006.1994.tb02424.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
To develop more effective adoptive immunotherapy, we transfected the human tumor necrosis factor-alpha (TNF-alpha) gene into human glioma cells (U251-SP), which were used as target cells. TNF-alpha is known to increase both the expression of intercellular adhesion molecule-1 (ICAM-1) on the surface of glioma cells and the susceptibility of glioma cells to lymphokine-activated killer (LAK) cell cytolysis. We compared the expression of ICAM-1 induced by TNF-alpha generated by the TNF-alpha gene-transfected cells with that induced by exogenously added TNF-alpha. When the TNF-alpha gene was transfected into U251-SP cells, the expression of ICAM-1 was detected on the cell surface from 3 days after the transfection and continued until at least 9 days. In contrast, it was expressed only transiently in the case of exogenously added TNF-alpha. Also, the cytolytic activity of LAK cells induced by transfection-induced TNF-alpha was significantly stronger than that induced by exogenously added TNF-alpha. The increased susceptibility was quenched by anti-ICAM-1 monoclonal antibody. These data indicated that continuous expression of ICAM-1 induced by TNF-alpha gene transfection of glioma cells resulted in higher cytolytic activity of LAK cells.
Collapse
Affiliation(s)
- T Takaoka
- Department of Neurosurgery, Nagoya University School of Medicine
| | | | | | | |
Collapse
|
23
|
Vaquero J, Zurita M, Oya S, Morales C, Coca S. Inmunoterapia adoptiva con citoquinas y linfocitos activados en un modelo experimental de tumor neuroectodérmico primitivo. Neurocirugia (Astur) 1994. [DOI: 10.1016/s1130-1473(94)71104-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
Holladay FP, Wood GW. Generation of cellular immune responses against a glioma-associated antigen(s). J Neuroimmunol 1993; 44:27-32. [PMID: 8496337 DOI: 10.1016/0165-5728(93)90264-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The study demonstrated that RT2, a highly malignant anaplastic glioma, expresses antigens that make it susceptible to in vivo adoptive immunotherapy with cytotoxic T lymphocyte-containing immune cell populations. Rats were immunized with irradiated RT2 tumor cells and the adjuvant C. parvum. Lymphocytes from immunized rats were restimulated in vitro with irradiated RT2 tumor cells plus interleukin-2 (IL-2). The cells that proliferated and differentiated in vitro effectively killed RT2, but only low levels of cytotoxicity were observed against other histopathologically related and unrelated, syngeneic and allogeneic target cells. Adoptive transfer of immune cells combined with a 5-day course of systemic IL-2 produced specific regression of brain tumors growing as lung microfoci.
Collapse
Affiliation(s)
- F P Holladay
- Department of Surgery, University of Kansas Medical Center, Kansas City 66160-7410
| | | |
Collapse
|
25
|
Merlo A, Filgueira L, Zuber M, Juretic A, Harder F, Gratzl O, De Libero G, Heberer M, Spagnoli GC. T-cell receptor V-gene usage in neoplasms of the central nervous system. A comparative analysis in cultured tumor infiltrating and peripheral blood T cells. J Neurosurg 1993; 78:630-7. [PMID: 8450337 DOI: 10.3171/jns.1993.78.4.0630] [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: 01/30/2023]
Abstract
The use of tumor-infiltrating lymphocytes in the treatment of central nervous system (CNS) neoplasms has met with serious obstacles due to difficulty of culture and poor characterization. Since in other tumors the therapeutic effects of tumor-infiltrating lymphocytes have been shown to rely on T-cell receptor engagement, the authors addressed the question as to whether expression of T-cell receptor variable (V) domains in cultured tumor-infiltrating lymphocytes from CNS is different from that of autologous cultured peripheral blood mononuclear cells. Infiltrating lymphocytes from CNS neoplasms, including primary malignancies, metastatic cancers, and meningiomas, were cultured in the presence of interleukin-2 and anti-CD3 monoclonal antibodies (MoAb's) in order to obtain optimum growth of T cells. Autologous peripheral blood mononuclear cells from the same patients were similarly cultured. After 4 to 5 weeks of culture, 97.3% +/- 2.6% (mean +/- standard deviation) of the resulting cell populations were CD3-positive lymphocytes. The expression of T-cell receptor V domains was then studied by using a panel of 12 MoAb recognizing gene products from T-cell receptor V-alpha 2, V-beta 5, 6, 8, and 12, V-gamma 4 and 9 families, and from two subfamilies of V-delta 2. Remarkably, in over 70% of all paired measurements, percentages of T cells expressing discrete T-cell receptor V-gene products were found to be virtually identical in tumor- and peripheral blood-derived cultured cell populations, with differences never exceeding 1%. In contrast, a different expression of individual V-gene products, concerning both alpha/beta and gamma/delta T-cell receptors, could be detected between cultured tumor-infiltrating lymphocytes and autologous peripheral blood-derived T lymphocytes in seven of 12 patients. In two cases, significant differences between the two populations were also observed in the proliferative responses obtained upon stimulation with staphylococcal enterotoxins that trigger defined V-beta T-cell receptors. Altogether, these data suggest that the T-cell receptor repertoire of cultured tumor-infiltrating lymphocytes from CNS tumors, suitable for use in adoptive immunotherapies, differs from that of autologous cultured peripheral blood mononuclear cells.
Collapse
Affiliation(s)
- A Merlo
- Department of Surgery, University of Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Jeffes EW, Beamer YB, Jacques S, Silberman RS, Vayuvegula B, Gupta S, Coss JS, Yamamoto RS, Granger GA. Therapy of recurrent high grade gliomas with surgery, and autologous mitogen activated IL-2 stimulated killer (MAK) lymphocytes: I. Enhancement of MAK lytic activity and cytokine production by PHA and clinical use of PHA. J Neurooncol 1993; 15:141-55. [PMID: 8509819 DOI: 10.1007/bf01053935] [Citation(s) in RCA: 28] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nineteen patients with recurrent high grade gliomas were treated in a phase I/II trial with aggressive debulking of the tumor, mitogen activated IL-2 stimulated peripheral blood lymphocytes (MAK cells), and rIL-2. Phytohemagglutin (PHA) was introduced into the tumor site in 16 patients prior to implanting MAK cells and IL-2 in an attempt to trigger more effective lysis of the tumor in vivo. In vitro both TNF bioactivity and cytolytic activity of long term cultured MAK (LMAK) cells were dramatically enhanced by adding PHA to the cultures of these activated PBL. Three of eleven patients (27%) had a decrease in size of the enhancing lesion on CT and/or MRI. Seven (37%) patients clinically improved. Median survival after therapy was 30 weeks. PHA was shown to be safe in vivo and more effective than IL-2 triggering enhanced effector function in vitro.
Collapse
|
27
|
Lapeña P, Isasi C, Moltó L, Martínez R, Vaquero J, Alvarez-Mon M. Interleukin 2 and interferon alpha modulation of the lymphocyte non-major histocompatibility-restricted lytic activity in glioblastoma patients. INTERNATIONAL JOURNAL OF IMMUNOPHARMACOLOGY 1992; 14:1307-13. [PMID: 1464464 DOI: 10.1016/0192-0561(92)90001-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We studied the non-major histocompatibility-restricted cytotoxic activity induced by interleukin 2 (IL-2) and interferon alpha (IFN-alpha) in mononuclear cells of peripheral blood (PBMNC) from glioblastoma patients. We observed a depressed cytotoxic activity against natural killer (NK)-sensitive target cells in PBMNC from all the patients studied. Culture of these PBMNC with IFN-alpha for 5 days augmented the cytotoxic activity against NK-sensitive target cells in a small group of patients. Incubation with IL-2 for 5 days normalizes the decreased cytotoxic activity against NK-sensitive target cells of PBMNC from all the glioblastoma patients studied. When PBMNC from these patients were incubated with IL-2 for 5 days and IFN-alpha was added to the culture medium in the last 2 h of culture, an enhancement of non-major histocompatibility-restricted cytotoxic activity was observed compared with that obtained with either IL-2 or IFN-alpha alone. This improvement of the cytotoxic activity was more relevant when it was tested against NK-resistant target cells. The potential utility of the sequential use of the two cytokines in generating non-major histocompatibility cytotoxic activity in glioblastoma patients is discussed.
Collapse
Affiliation(s)
- P Lapeña
- Department of Medicine, Hospital Universitario Príncipe de Asturias, Universidad de Alcalá de Henares, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
28
|
Holladay FP, Heitz T, Wood GW. Antitumor activity against established intracerebral gliomas exhibited by cytotoxic T lymphocytes, but not by lymphokine-activated killer cells. J Neurosurg 1992; 77:757-62. [PMID: 1403119 DOI: 10.3171/jns.1992.77.5.0757] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Specific immune responses against malignant brain tumors have been difficult to demonstrate. Moreover, immunotherapy has met with little success, despite using lymphocytes with high levels of cytotoxicity against brain tumor cells. Lymphokine-activated killer (LAK) cells that nonspecifically kill brain tumor cells are produced by stimulating resting precursors with high concentrations of interleukin-2 (IL-2). Cytotoxic T lymphocytes that specifically kill brain tumor cells are produced by stimulating antigen receptor-positive immune-cell precursors with tumor cells. In an attempt to gain insight into immune cell function against brain tumors, the present study compared the in vitro and in vivo activities of LAK cells and cytotoxic T lymphocytes produced against RT2, a fast-growing rat glioma cell line. Lymphokine-activated killer cells were produced by stimulating normal rat spleen cells with 1000 units of IL-2, and RT2-specific cytotoxic T lymphocytes were produced by priming them in vivo with RT2 and Corynebacterium parvum and restimulating primed spleen cells with RT2 in vitro. Lymphokine-activated killer cells were highly cytotoxic for a panel of syngeneic and allogeneic brain tumor and non-brain tumor target cells, including RT2, as measured in a 4-hour 51Cr release assay. Cytotoxic T lymphocytes were highly cytotoxic only for syngeneic brain tumor target cells. Lymphokine-activated killer cells and cytotoxic T lymphocytes were tested for in vivo antitumor activity against intracerebral RT2 by intravenous adoptive transfer of activated lymphocytes. Untreated rats died in approximately 2 weeks. Lymphokine-activated killer cells plus IL-2 failed to affect survival when treatment was initiated as early as 1 day following tumor inoculation. Cytotoxic T lymphocytes and IL-2 administered as late as Day 5 rejected progressing intracerebral tumor. Thus, although both cytotoxic T lymphocytes and LAK cells exhibited high levels of in vitro killing of glioma cells, only cytotoxic T lymphocytes rejected progressing intracerebral tumors.
Collapse
Affiliation(s)
- F P Holladay
- Department of Pathology and Oncology, University of Kansas Medical Center, Kansas City
| | | | | |
Collapse
|
29
|
Holladay FP, Heitz T, Chen YL, Chiga M, Wood GW. Successful treatment of a malignant rat glioma with cytotoxic T lymphocytes. Neurosurgery 1992; 31:528-33. [PMID: 1407433 DOI: 10.1227/00006123-199209000-00015] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Brain tumors are highly resistant to therapy. Their diffuse infiltrative nature and the relative inaccessibility of brain tissue to blood and lymph are barriers to surgical and cytotoxic treatments alike. The purpose of this study was to produce immune cells specifically reactive with an anaplastic rat glioma (RT2) and determine whether those cells could affect tumor progression in the brain. RT2-specific cytotoxic cells were prepared by priming rats in vivo with RT2 tumor cells and Corynebacterium parvum and stimulating the primed lymphocytes in vitro with irradiated RT2 tumor cells and interleukin-2 (IL-2). Cultured cells exhibited a high level of cytotoxicity against RT2, but not C6 (an allogeneic glioma), 3M2N (a syngeneic mammary tumor), or CSE (a syngeneic fibrosarcoma) tumor cells. To generate a model for therapy, rats were injected intracerebrally with RT2, generating progressing brain tumors, which killed untreated rats in approximately 2 weeks. To test the therapeutic potential of the effector cells, tumor-bearing rats were treated by intravenous injection of lymphocytes on Day 5 of tumor growth. Treated rats also received a 5-day course of systemic IL-2 beginning on Day 5. Treatment with IL-2 alone, RT2-primed spleen cells, or RT2-primed spleen cells stimulated in vitro with C6 did not affect rat survival. However, tumor-bearing rats treated with RT2-stimulated lymphocytes exhibited increased survival or were cured. Systemic IL-2 was an essential adjunct, because survival was not affected by treatment with effector cells alone. Therapy initiated on Day 8 of tumor progression lacked effect on survival.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- F P Holladay
- Department of Pathology, University of Kansas Medical Center, Kansas City
| | | | | | | | | |
Collapse
|
30
|
|
31
|
Hall WA, Godal A, Juell S, Fodstad O. In vitro efficacy of transferrin-toxin conjugates against glioblastoma multiforme. J Neurosurg 1992; 76:838-44. [PMID: 1314294 DOI: 10.3171/jns.1992.76.5.0838] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The cytotoxic activity of immunotoxins constructed with human diferric transferrin (Tfn) as the carrier ligand and an abrin variant Pseudomonas exotoxin A (PE) and the diphtheria toxin mutant cross-reacting material (CRM) 107 as the toxin moieties were studied in vitro. Three malignant human cell lines, the glioblastomas multiforme SNB19 and SF295 and the LOX melanoma, and a nonhuman control murine melanoma cell line B16 were assessed. The presence of transferrin receptors on the cell lines was confirmed by direct 125I-Tfn binding assays. The 50% protein synthesis inhibitory concentration (IC50) values for all cell lines demonstrated that Tfn-abrin variant and Tfn-PE had comparable potency and were both more effective than Tfn-CRM 107. Monensin, a carboxylic ionophore, potentiated the effect of Tfn-abrin variant against glioma cells approximately 35-fold with IC50 values of 4.0 x 10(-13) M and 4.7 x 10(-12) M for SNB19 and SF295, respectively. Cytotoxic activity of Tfn-abrin variant (with or without monensin) and Tfn-PE was correlated with the degree of Tfn receptor expression measured on the cell lines. The exquisite in vitro cytotoxicity of Tfn-abrin variant and Tfn-PE immunotoxins against glioma and melanoma cells warrants further in vivo evaluation and future consideration of these agents for potential clinical application against glioblastoma multiforme and leptomeningeal neoplasia.
Collapse
Affiliation(s)
- W A Hall
- Department of Tumorbiology, Norwegian Radium Hospital, Oslo
| | | | | | | |
Collapse
|
32
|
Abstract
The poor prognosis associated with central nervous system (CNS) malignancy has led investigators to seek new, innovative treatment modalities. Immunotoxins, carrier molecules linked to toxic agents, combine high specificity for tumor-associated antigens with extreme potency. The rationale for both the development of these compounds and for their application to CNS neoplasia is explained. This report discusses the design and construction of immunoconjugates, using toxins that differ in their mechanism of action bound to ligands directed against various antigens. A comparison is made between the in vitro efficacy of standard chemotherapy and immunotoxins in glioblastoma- and medulloblastoma-derived cell lines. A review is included of the results of experiments in animals with leptomeningeal neoplasia, where prolongation of survival following intrathecal administration of immunotoxins has been reported. The obstacles encountered in clinical trials with other types of cancer are addressed and approaches to optimize the use of these novel agents in the context of treating malignant disease of the CNS are suggested.
Collapse
Affiliation(s)
- W A Hall
- Department of Neurosurgery, University of Minnesota Hospital and Clinic, Minneapolis
| | | |
Collapse
|
33
|
Yamamoto RS, Coss J, Vayuvegula B, Gupta S, Beamer Y, Jacques S, Jeffes EW, Carson WE, Jakowatz J, Granger GA. Generation of stimulated, lymphokine activated T killer (T-LAK) cells from the peripheral blood of normal donors and adult patients with recurrent glioblastoma. J Immunol Methods 1991; 137:225-35. [PMID: 2013699 DOI: 10.1016/0022-1759(91)90028-e] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Peripheral blood mononuclear cells (PBM) from normal donors and patients with recurrent glioma were activated initially for 48-72 h with phytohemagglutinin-P (PHA) and recombinant human interleukin-2 (IL-2), and then proliferated in vitro for up to 5 months with IL-2. These cells are termed mitogen-stimulated lymphokine-activated T killer (T-LAK) cells. We measured patterns of T-LAK cell growth, in vitro cytolytic activity on a panel of continuous and primary tumor cells, and the phenotypes of the cells in these cultures. Lymphocyte viability declined dramatically over the first 3-5 days; and then the remaining cells in these cultures began to divide and maintained a constant 30-36 h doubling time for long periods in vitro. Phenotyping revealed that cells in the initial few days of culture were heterogeneous, but became almost totally CD3 T cells after 7-10 days in culture. The T-LAK cells from individual normal donors and cancer patients demonstrated a non-genetically restricted cytolytic ability against a panel of both continuous cell lines and primary autologous and allogeneic glioblastoma cells in vitro. This technique provides a method of generating large numbers of autologous cytolytic T cells with non-restricted anti-tumor activity that can be derived from peripheral blood mononuclear cells.
Collapse
|
34
|
Matsuura H, Imaya H. Comparison of lymphokine-activated killer activities between thymocytes and splenocytes in rats with brain tumors. Cancer Immunol Immunother 1991; 33:50-3. [PMID: 2021958 PMCID: PMC11037996 DOI: 10.1007/bf01742528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/1990] [Accepted: 11/07/1990] [Indexed: 12/29/2022]
Abstract
We studied the lymphokine-activated killer (LAK) activity in splenocytes and thymocytes of rats with brain tumors chronologically from the early stage to the late stage, in order to clarify how much LAK activity would be developed at each stage. Simultaneously the natural killer (NK) activity in splenocytes, as one aspect of the host immunocompetence, was also determined. The splenic NK activity was significantly depressed in rats with brain tumors during the 2nd and 3rd weeks after tumor transplantation, as compared with normal controls. On the other hand, the splenocytes incubated with interleukin-2 showed the same killer activity in rats with brain tumors as in normal rats at all times. The LAK activity in thymocytes from rats with brain tumors was significantly higher than that of controls in the 1st and 2nd weeks and became equal to that of the controls during the 3rd week. The killer activity after incubation with interleukin-2 in thymocytes was superior to that in splenocytes throughout the experiment in both tumor-bearing rats and controls, which suggested that the precursor of LAK cells was not NK cells.
Collapse
Affiliation(s)
- H Matsuura
- Neurosurgery Service, Saitama Neurosurgical Institute, Kohnosu, Japan
| | | |
Collapse
|
35
|
Elliott LH, Brooks WH, Roszman TL. Inability of mitogen-activated lymphocytes obtained from patients with malignant primary intracranial tumors to express high affinity interleukin 2 receptors. J Clin Invest 1990; 86:80-6. [PMID: 2365829 PMCID: PMC296693 DOI: 10.1172/jci114719] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Patients with primary malignant brain tumors manifest a variety of abnormalities in cell-mediated and humoral immunity. Diminished T cell reactivity has been shown in these patients to be linked to deficiencies in interleukin 2 (IL-2) production that cannot be overcome by exogenous IL-2. In this study, specific binding of radiolabeled IL-2 to PHA-stimulated lymphocytes from brain tumor patients demonstrates that the number of high affinity interleukin 2 receptors (IL-2R) is greatly reduced. FACS analysis indicates that the relative density of the p55 protein (Tac protein) is lower on the mitogen-activated lymphocytes obtained from patients than on comparably treated lymphocytes from normal individuals. These data indicate that mitogen-stimulated lymphocytes obtained from patients have fewer functional high affinity IL-2R principally because of the failure to express sufficient levels of the p55 protein for association with the p75 protein. Northern analysis of total RNA isolated from mitogen-stimulated T cells from patients demonstrates normal levels of steady state mRNA, which codes for the p55 protein. Moreover, there is no defect in the postranslational processing of the primary translation product of this mRNA suggesting that normal levels of the p55 protein are produced in activated T cells from patients.
Collapse
Affiliation(s)
- L H Elliott
- Department of Microbiology and Immunology, University of Kentucky Medical Center, Lexington 40536-0084
| | | | | |
Collapse
|
36
|
Maleci A, Alterman RL, Sundstrom D, Kornblith PL, Moskal JR. Effect of phorbol esters on the susceptibility of a glioma cell line to lymphokine-activated killer cell activity. J Neurosurg 1990; 73:91-7. [PMID: 2352027 DOI: 10.3171/jns.1990.73.1.0091] [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: 12/31/2022]
Abstract
The mechanisms by which lymphokine-activated killer (LAK) cells exert their cytotoxic effects are not well understood. This study demonstrates that phorbol ester pretreatment of a LAK cell-sensitive glioma cell line (SNB-19) induced a significant decrease in the susceptibility of cells to LAK cell-mediated lysis. This effect was produced by low concentrations of the tumor-promoting phorbol ester, phorbol-12,13-myristate acetate (PMA), and was reversible. Protein kinase C (PKC) inhibitors failed to block this phenomenon. No apparent alteration in the ability of LAK cells to bind to their targets was observed. Thus, PMA may have exerted its effects by a mechanism that does not require PKC, or these glioma cells may possess an isozyme of PKC which is insensitive to the inhibitors used in these studies.
Collapse
Affiliation(s)
- A Maleci
- Department of Neurosurgery, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | | | | | | | | |
Collapse
|
37
|
Tratkiewicz JA, Szer J, Boyd RL. Lymphokine-activated killer cytotoxicity against leukaemic blast cells. Clin Exp Immunol 1990; 80:94-9. [PMID: 2323104 PMCID: PMC1535230 DOI: 10.1111/j.1365-2249.1990.tb06447.x] [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: 12/31/2022] Open
Abstract
While the sensitivity of cell lines and solid tumours to lymphokine-activated killer (LAK) cell cytotoxicity is extensively reported, the ability of these generated cytotoxic cells to lyse leukaemic blast cells is controversial. This study reports the successful generation of LAK cells with specificity for leukaemic blast cells. Control donors, patients in remission and patients with active leukaemia were capable of lysing allogeneic leukaemic blast cells. One patient was also capable of lysing autologous leukaemic blast cells. Successful LAK generation was achieved by the use of high dose recombinant interleukin-2 (rIL-2) while combinations of lymphokines did not improve LAK specificity or efficacy. These findings suggest that LAK immunotherapy may be considered for the treatment of patients with acute leukaemia.
Collapse
|
38
|
|
39
|
Abstract
With progress in cellular immunology and the development of hybridoma technology, the idea of manipulating host-tumor immune interactions to improve the prognosis of brain tumors has aroused renewed interest. Although no brain tumor-specific antigens have been found, and in spite of the wide antigenic heterogeneity of brain tumor cells, some monoclonal antibodies possessing restricted specificity have been isolated and their potential clinical applications investigated. One of the most pronounced changes in the cellular immune responses of brain tumor patients is a profound depression of the T4-helper lymphocytes. Clinical and laboratory trials are under way to assess the ability of lymphokines, such as gamma-interferon or interleukin-2, to restore immunologic competence in these patients and potentiate a specific anti-tumor immunologic response. Recent work suggests that the endothelium-astrocyte complex may have a pivotal role in assisting the escape of brain tumors from the host's immunologic responses, since it is responsible for the intracerebral sequestration of antigens and local anti-tumor responses. In this review, the data on the antigenic properties of central nervous system tumors and the host's humoral and cellular immune responses to them are analyzed and potential immunologic therapies are discussed.
Collapse
Affiliation(s)
- C de Micco
- Laboratoire de Neuropathologie, Faculté de Médecine, Marseilles, France
| |
Collapse
|
40
|
Yamasaki T, Kikuchi H, Paine JT, Yamashita J, Miyatake S, Iwasaki K, Kobayashi H, Namba Y, Hanaoka M. Murine intracerebral interleukin-2 injection: pathological and immunological effects. J Neurosurg 1989; 71:732-40. [PMID: 2809728 DOI: 10.3171/jns.1989.71.5.0732] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The authors have investigated whether specific pathological changes and antibodies against interleukin-2 (IL-2) are induced after intracerebral administration of recombinant IL-2 (rIL-2). In addition, IL-2 receptor (IL-2R) expression was checked on the cell surface of normal brain tissues before and after the intracerebral infusion. Reconstituted rIL-2 (specific activity 1.2 x 10(7) U/mg protein) was injected into the right cerebral hemisphere of normal adult C57BL/6 mice in three different dose groups, each receiving single or multiple infusions of 8, 32, or 80 U. In sham control experiments, mouse albumin purified by gel filtration and ion exchange chromatography and adjusted to the same concentration of protein as rIL-2 was injected into mice at various doses. Anti-IL-2 antibodies were measured by an enzyme-linked immunosorbent assay concurrently with assessment of IL-2 activity in serum. The IL-2R expression was determined by using immunofluorescence techniques with monoclonal antibodies against mouse IL-2R. Since histological alteration after rIL-2 injection did not differ from that in the sham control preparations, it seems that there is no direct toxic action of rIL-2 on normal brain tissues. Interleukin-2 antibodies were produced at low levels only in mice injected repeatedly at the maximum dose, and levels were insignificant in other groups. Serum levels of IL-2 activity remained low. The IL-2R expression within the brain was not enhanced within 8 weeks following the intracerebral administration of rIL-2, suggesting that direct intracerebral infusion of rIL-2 may be safely used in the immunotherapy of brain tumors.
Collapse
Affiliation(s)
- T Yamasaki
- Department of Neurosurgery, Medical School, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Sutton RC, Kennedy J, Duncan J, Conley FK. Toxicity of IL-2 and Corynebacterium parvum following direct intracranial injection. J Neurooncol 1989; 7:261-7. [PMID: 2795120 DOI: 10.1007/bf00172920] [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: 01/02/2023]
Abstract
The toxicity of IL-2 and/or C. Parvum when injected directly into the mouse brain was examined by survival and histopathology using different numbers of injections, different doses of IL-2, different solvents for the IL-2, and different doses and routes of administration of C. Parvum. Two injections was found significantly to increase mortality (19%) over a single injection (4%). Mortality from two injections of 30,000 U (23%) or 60,000 U (20%) was higher than from two injections of 15,000 U (12%). The mortality from two injections with normal saline as solvent was much higher (29%) than from two injections with sterile water (19%) or D5W (9%). Two injections of IL-2 given simultaneously with C. Parvum showed a much higher mortality (26%) than other doses and routes of C. Parvum administration. Mice dying acutely (6-24 days) of toxicity showed an extensive mononuclear infiltrate at the site of injection. The brains of surviving mice (sacrificed at 30 days) showed a mild residual mononuclear cell infiltrate with the exception of mice which had received IL-2 and C. Parvum simultaneously. Brains from this latter group had an extensive residual mononuclear cell infiltrate.
Collapse
Affiliation(s)
- R C Sutton
- Section of Neurosurgery, Palo Alto Veterans Administration Medical Center, CA
| | | | | | | |
Collapse
|
42
|
Naganuma H, Kimurat R, Sasaki A, Fukamachi A, Nukui H, Tasaka K. Complete remission of recurrent glioblastoma multiforme following local infusions of lymphokine activated killer cells. Case report. Acta Neurochir (Wien) 1989; 99:157-60. [PMID: 2549767 DOI: 10.1007/bf01402326] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We report the case of a 26-year-old man in whom glioblastoma multiforme had recurred six months following a subtotal resection. Despite radiotherapy and a course of interferon beta and ACNU, the tumour increased in size (to 3 cm) and there was neurological deterioration. Treatment was then initiated with LAK cells, together with ACNU and interferon beta. After three courses of LAK cells, tumour size was markedly reduced, and at about six months the tumour had nearly disappeared on computed tomographic (CT) scans. At one year, and after nine courses of LAK cell therapy (total dose of 2.7 x 10(9) cells) infused via an Ommaya reservoir and supplemented by ACNU and interferon beta, the tumour has disappeared and the patient is considered to be in complete remission since 6 months. This marked response is thought to be due chiefly to LAK cell therapy. The relatively low dose administered was well-tolerated.
Collapse
Affiliation(s)
- H Naganuma
- Department of Neurosurgery, Yamanashi Medical College, Japan
| | | | | | | | | | | |
Collapse
|
43
|
Bosnes V, Hirschberg H. Immunomagnetic separation of infiltrating T lymphocytes from brain tumors. J Neurosurg 1989; 71:218-23. [PMID: 2664096 DOI: 10.3171/jns.1989.71.2.0218] [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: 01/02/2023]
Abstract
Tumor-infiltrating lymphocytes (TIL's) were isolated from human glioma biopsy specimens by immunomagnetic separation using T cell-specific monoclonal antibodies coupled to paramagnetic beads, and were expanded in culture with feeder cells and interleukin-2 (IL-2). The infiltrating cells from five of seven patients proliferated in culture. When tested after 2 to 3 weeks of culture, virtually all of the cells stained with antibodies against the CD2 and CD3 antigens. Most cells also expressed human leukocyte antigen class II molecules, while varying percentages of cells stained with antibodies against the IL-2 receptor and the CD4 and CD8 antigens. The cytotoxicity of the cultured TIL's against autologous and allogeneic glioma cells and the K562 and Daudi cell lines was measured and compared with that of lymphokine-activated killer (LAK) cells from the same patients. None of the TIL's showed significant cytotoxicity against these targets, whereas LAK cells lysed all of the targets.
Collapse
Affiliation(s)
- V Bosnes
- Institute of Transplantation Immunology, National Hospital, Oslo, Norway
| | | |
Collapse
|
44
|
Saris SC, Patronas NJ, Rosenberg SA, Alexander JT, Frank J, Schwartzentruber DJ, Rubin JT, Barba D, Oldfield EH. The effect of intravenous interleukin-2 on brain water content. J Neurosurg 1989; 71:169-74. [PMID: 2787395 DOI: 10.3171/jns.1989.71.2.0169] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Parenteral treatment with interleukin-2 (IL-2) is effective against certain advanced cancers outside the central nervous system. Prior to commencement of Phase II trials in patients with brain tumors, the neurological and neuroradiological features of 10 patients treated with intravenous administration of repeated doses of IL-2 were studied. Three patients had malignant gliomas, and seven patients had extracranial cancer without evidence of intracranial metastasis. All were treated with intravenous doses of 10(5) U/kg three times daily for up to 5 days. The patients with gliomas received cranial computerized axial tomography (CT) scans before IL-2 therapy was initiated and during the later stages of treatment. The patients with extracranial cancer underwent T2-weighted magnetic resonance (MR) imaging before and later during therapy. After two to 11 doses of IL-2, the patients with gliomas had marked neurological deterioration that was associated with a mild to marked increase in peritumoral edema and mass effect visible on CT scans. With cessation of treatment and appropriate supportive care, all returned to their pretreatment state. The patients with extracranial cancer were either neurologically unchanged or underwent minor transient changes in mental status (lethargy and confusion). In these patients, the MR signal intensity was quantified and compared in eight anatomic regions of interest. In six of the seven patients, there were increases in gray and white matter signal intensity consistent with increased cerebral water content. The percentage changes (means +/- standard error of the means) were 12.6% +/- 7.3% in the gray matter and 17.0% +/- 6.2% in the white matter. This study demonstrates that treatment with a high parenteral dose of IL-2 is not tolerated by patients with gliomas due to increased cerebral edema. In patients with extracranial cancer but no brain disease, parenteral IL-2 induces an increase in the cerebral water content of both gray and white matter.
Collapse
Affiliation(s)
- S C Saris
- Clinical Neurosurgery Section, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Vaquero J, Martínez R, Oya S, Coca S, Barbolla L, Ramiro J, Salazar FG. Intratumoural injection of autologous lymphocytes plus human lymphoblastoid interferon for the treatment of glioblastoma. Acta Neurochir (Wien) 1989; 98:35-41. [PMID: 2472737 DOI: 10.1007/bf01407174] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Preliminary experience with a clinical trial of immunotherapy for glioblastoma, by means of intratumoural injection of autologous lymphocytes (AL) mixed with low doses of human lymphoblastoid interferon (HLI) is presented. In two of twelve patients, a transient reduction of tumoural volume was obtained. Morphological studies showed that injected lymphocytes remain within the tumour, and suggest tumoural lysis due to activity of natural killer (NK) cells. Clinically no significant prolongation of survival time could be achieved and, as in other series, patients with additional radiation therapy survived longer. But the morphological findings suggest that immunotherapy carrying NK-cells to contact with tumoural cells might be useful in some patients with glioblastoma. Actually no explanation can be given why only two of our cases responded positively. Regarding the otherwise poor prognosis it seems justified to continue these studies.
Collapse
Affiliation(s)
- J Vaquero
- Department of Neurosurgery, Puerta de Hierro Clinic, Autonomous University, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Autologous lymphocytes were instilled into the tumor in a girl 15 years of age who had presented with a recurrent left frontoparietal astrocytoma 1.5 years after surgery, radiation, and chemotherapy. She had remissions for 5 weeks, 5 months, and 20 months after the first, second, and third instillations, respectively. The usefulness of intratumoral instillation of autologous lymphocytes [or preferably lymphokine activated killer (LAK) cells with interleukin-2 is demonstrated in such recurrent astrocytomas.
Collapse
|
47
|
Barba D, Saris SC, Holder C, Rosenberg SA, Oldfield EH. Intratumoral LAK cell and interleukin-2 therapy of human gliomas. J Neurosurg 1989; 70:175-82. [PMID: 2643685 DOI: 10.3171/jns.1989.70.2.0175] [Citation(s) in RCA: 182] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Adoptive immunotherapy using lymphokine-activated killer (LAK) cells and interleukin-2 (IL-2) offers the possibility of a new treatment for patients with malignant glial tumors. In a clinical trial, the effectiveness of a 5-day treatment cycle of direct intratumoral administration of both LAK cells and IL-2 via a reservoir/catheter system in patients with recurrent malignant gliomas was studied. Ten patients were entered into the study, nine of whom were treated with 15 cycles of LAK cells (0.9 to 21.0 x 10(9) cells) and IL-2 (49 to 450 x 10(3) U/kg). The 10th patient in the study was not treated because of the onset of severe neurological deficits prior to beginning immunotherapy. Of the nine patients treated, one had a partial tumor response to immunotherapy as documented by computerized tomography. Neurological side effects occurred in all patients undergoing treatment and were related to increases in cerebral edema that appeared to be mediated by the immunotherapy. This report demonstrates the present limitations of regional adoptive immunotherapy with LAK cells and IL-2 in the treatment of human glial tumors.
Collapse
Affiliation(s)
- D Barba
- Clinical Neurosurgery Section, National Institute of Neurological and Communicative Disorders and Stroke, Bethesda, Maryland
| | | | | | | | | |
Collapse
|
48
|
Alexander JT, Saris SC, Oldfield EH. The effect of interleukin-2 on the blood-brain barrier in the 9L gliosarcoma rat model. J Neurosurg 1989; 70:92-6. [PMID: 2783343 DOI: 10.3171/jns.1989.70.1.0092] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Carbon-14-labeled aminoisobutyric acid was used to determine local blood-to-tissue transfer constants in 22 Fischer rats with intracerebral 9L gliosarcomas that received either high-dose parenteral interleukin-2 (IL-2) or a control injection. In tumor and peritumoral tissue, the transfer constants in the IL-2-treated animals (89.6 +/- 14.6 and 35.8 +/- 6.0, respectively, mean +/- standard error of the mean) were larger (p less than 0.05) than in control animals (61.4 +/- 6.4 and 14.6 +/- 2.2, respectively). In contrast, in normal frontal and occipital tissue contralateral to the tumor-bearing hemisphere, there was no significant difference between the transfer constants in IL-2-treated and control animals. Furthermore, treatment of animals with IL-2 excipient caused no change in permeability as compared to animals treated with Hanks' balanced salt solution. Parenteral injection of IL-2 increases blood-brain barrier disruption in tumor-bearing rat brain but does not increase the vascular permeability of normal brain. Methods to prevent this increased tumor vessel permeability are required before parenteral IL-2 can be used safely for the treatment of primary or metastatic brain tumors.
Collapse
Affiliation(s)
- J T Alexander
- Clinical Neurosurgery Section, National Institute of Neurological and Communicative Disorders and Stroke, Bethesda, Maryland
| | | | | |
Collapse
|
49
|
Ausiello C, Maleci A, Spagnoli GC, Antonelli G, Cassone A. Cell-mediated cytotoxicity in glioma-bearing patients: differential responses of peripheral blood mononuclear cells to stimulation with interleukin-2 and microbial antigen. J Neurooncol 1988; 6:329-38. [PMID: 3146618 DOI: 10.1007/bf00177428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Peripheral blood mononuclear cells (PBMC) of malignant glioma-bearing patients were stimulated in vitro with Interleukin-2 (IL-2) or a glucomannan-protein antigen of Candida albicans (GMP) then assayed for proliferation, production of IFN-gamma, and generation of cytotoxic effectors against either K562 tumor cell line or freshly-cultured allogenic glioma cells. PBMC of healthy, age and sex-matched subjects were the controls. PBMC of glioma-bearing patients did not differ, as a whole, from PBMC of healthy donors in IL-2 or GMP-induced proliferation. However, they showed a lesser ability to produce IFN as well as a substantial inability to generate cytotoxic effectors following GMP stimulation. PBMC of glioma patients were fully responsive to IL-2 in cytotoxicity generation, as were the PBMC from normal subjects. The results suggest that glioma patients may have a defective antigen-mediated activation of natural cytotoxic effectors. This hyporesponsiveness is not accompanied by depressed lymphoproliferation and does not apparently involve a reduced response to IL-2.
Collapse
Affiliation(s)
- C Ausiello
- Istituto CNR Tipizzazione Tissutale e Problemi della Dialisi, L'Aquila, Italy
| | | | | | | | | |
Collapse
|
50
|
Djeu JY, Blanchard DK. Interferon-gamma-induced alterations of monocyte susceptibility to lysis by autologous lymphokine-activated killer (LAK) cells. Int J Cancer 1988; 42:449-54. [PMID: 3138195 DOI: 10.1002/ijc.2910420323] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Interleukin-2 (IL-2)-activated killer (LAK) cells specifically lyse human monocytes, which may account for some of the toxicity seen during LAK/IL-2 immunotherapy of cancer patients. In an effort to protect autologous monocytes, we treated monolayer cultures of monocytes with various doses of recombinant human interferon-gamma (IFN-gamma) and assessed their sensitivity to LAK-mediated lysis. IFN-gamma lessens the sensitivity of monocytes to lysis in a dose-dependent manner. Treatment of FMEX, an NK-resistant melanoma tumor cell line, with IFN-gamma did not affect its susceptibility to LAK lysis. Kinetic studies demonstrated that as little as 2 hr incubation with IFN-gamma was sufficient for protection to occur, and that monocytes which were treated with IFN-gamma for 2 hr, washed, and then cultured in medium alone retained their resistance to lysis for at least 4 days. Cold target inhibition studies showed that IFN-treated and untreated monocytes could effectively compete with each other for binding sites on LAK cells. Finally, binding studies demonstrated that there was no significant difference between the number of conjugates formed using either IFN-treated or untreated monocytes. This indicates that resistance to lysis induced by IFN treatment affects a post-binding event and not an initial recognition signal.
Collapse
Affiliation(s)
- J Y Djeu
- University of South Florida College of Medicine, Department of Medical Microbiology and Immunology, Tampa 33612
| | | |
Collapse
|