1
|
Ljubimov VA, Sun T, Wang J, Li L, Wang PZ, Ljubimov AV, Holler E, Black KL, Kopeček J, Ljubimova JY, Yang J. Blood-brain barrier crossing biopolymer targeting c-Myc and anti-PD-1 activate primary brain lymphoma immunity: Artificial intelligence analysis. J Control Release 2025; 381:113611. [PMID: 40088978 DOI: 10.1016/j.jconrel.2025.113611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
Primary Central Nervous System Lymphoma is an aggressive central nervous system neoplasm with poor response to pharmacological treatment, partially due to insufficient drug delivery across blood-brain barrier. In this study, we developed a novel therapy for this lymphoma by combining a targeted nanopolymer treatment with an immune checkpoint inhibitor antibody (anti-PD-1). A N-(2-hydroxypropyl)methacrylamide copolymer-based nanoconjugate was designed to block tumor cell c-Myc oncogene expression by antisense oligonucleotide. Angiopep-2 peptide was conjugated to the copolymer to facilitate nanodrug crossing of the blood-brain barrier. Systemically administered polymeric nanodrug, alone or in combination with immune checkpoint inhibitor antibody anti-PD-1, was tested in syngeneic mouse model of A20 intracranial brain lymphoma. There was no significant survival difference between saline- and free anti-PD-1-treated groups. However, significant survival advantage vs. saline was observed upon treatment with nanodrug bearing Angiopep-2, H6 (6 histidines for endosome escape), and c-Myc antisense alone and especially when it was combined with anti-PD-1 antibody. Animal survival after combined treatment was also significantly increased vs. free anti-PD-1. Artificial Intelligence-assisted analysis of gene expression database after RNA-seq of tumors was used to find novel immune pathways, molecular targets and the most effective multifunctional drugs together with future drug prediction for brain lymphoma in vivo model. Spectral flow cytometry and RNA-seq analysis revealed a robust activation of tumor infiltrating T lymphocytes with enhanced interferon γ signaling and polarization to M1-type macrophages in treated tumors, which was confirmed by immunofluorescence staining. In summary, a new effective blood-brain barrier crossing nano immuno therapeutic system was developed that effectively blocked tumor c-Myc acting in combination with immune checkpoint inhibitor anti-PD-1 to treat primary brain lymphoma. The treatment improved survival of tumor-bearing animals through activation of both the adaptive and innate immune responses.
Collapse
Affiliation(s)
- Vladimir A Ljubimov
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, United States
| | - Tao Sun
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, United States
| | - Jiawei Wang
- Department of Molecular Pharmaceutics/CCCD, University of Utah, 20 S 2030 E, Salt Lake City, UT 84112, United States
| | - Lian Li
- Department of Molecular Pharmaceutics/CCCD, University of Utah, 20 S 2030 E, Salt Lake City, UT 84112, United States
| | - Paul Z Wang
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Alexander V Ljubimov
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, United States; Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States; Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Eggehard Holler
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, United States; Institut für Biophysik und Physikalische Biochemie Universität Regensburg, D-93040 Regensburg, Germany
| | - Keith L Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, United States; Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Jindřich Kopeček
- Department of Molecular Pharmaceutics/CCCD, University of Utah, 20 S 2030 E, Salt Lake City, UT 84112, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, United States
| | - Julia Y Ljubimova
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, United States.
| | - Jiyuan Yang
- Department of Molecular Pharmaceutics/CCCD, University of Utah, 20 S 2030 E, Salt Lake City, UT 84112, United States.
| |
Collapse
|
2
|
Huang CP, Liu LC, Chang CC, Wu CC, Shyr CR. Intratumoral xenogeneic tissue-specific cell immunotherapy inhibits tumor growth by increasing antitumor immunity in murine triple negative breast and pancreatic tumor models. Cancer Lett 2021; 545:115478. [PMID: 35902043 DOI: 10.1016/j.canlet.2021.10.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/18/2021] [Accepted: 10/31/2021] [Indexed: 12/13/2022]
Abstract
Low immunogenicity in tumors and the immunosuppressive tumor microenvironment (TME) represent major obstacles to the full success of immunotherapy in cancer patients. A novel intratumoral xenogeneic tissue-specific cell immunotherapeutic approach could overcome the obstacles. Murine 4T1 triple negative breast cancer (TNBC) cells and Pan18 pancreatic ductal adenocarcinoma (PDAC) cells were used for establishing syngeneic graft tumor models to evaluate antitumor effect of intratumoral injection of xenogeneic tissue-specific cells. Responses to treatment were assessed by measuring tumor growth and tumor weight of the tumor-bearing mice. To investigate the mechanisms of action, tumor histology and immunohistochemistry and cytokine gene expression were measured. Splenic lymphocytes proliferation, cytokine production and cytotoxicity activities were also assessed. The findings showed that intratumoral injection of xenogeneic tissue-specific cells in monotherapy and combination with chemotherapy inhibit tumor growth. The therapeutic efficacy of intratumoral xenogeneic cells was significantly enhanced by the addition of cytotoxic chemotherapeutic agents. Mice that received combined treatment showed maximal attenuation in tumor growth rate. The antitumor immunity was explained by altered immune cell infiltration in tumors and immune cell functions. Our findings demonstrate that xenogeneic tissue-specific cells given intratumorally, provide a potent antitumor effect in murine breast and pancreatic tumor models by enhancing recruitment and activation of immune cells in tumors for local and systemic antitumor effects. Moreover, intratumoral xenogeneic cell treatment turns immunologically "cold" tumors to "hot" ones, generates systemic antitumor immunity, and synergizes with chemotherapy. Thus, the intratumoral xenogeneic tissue-specific cell immunotherapy may represent a useful therapeutic option to difficult-to-treat cancers.
Collapse
Affiliation(s)
- Chi-Ping Huang
- Department of Medicine, Department of Urology, College of Medicine, China Medical University and Hospital, Taichung, 404, Taiwan
| | - Liang-Chih Liu
- Department of Medicine, Department of Surgery, College of Medicine, China Medical University and Hospital, Taichung, 404, Taiwan
| | - Chih-Chun Chang
- Sex Hormone Research Center, Department of Medical Laboratory Science and Biotechnology, China Medical University and Hospital, Taichung, 404, Taiwan
| | - Chun-Chie Wu
- Department of Medicine, Department of Urology, College of Medicine, China Medical University and Hospital, Taichung, 404, Taiwan
| | - Chih-Rong Shyr
- Sex Hormone Research Center, Department of Medical Laboratory Science and Biotechnology, China Medical University and Hospital, Taichung, 404, Taiwan.
| |
Collapse
|
3
|
Vile RG, Melcher A, Pandha H, Harrington KJ, Pulido JS. APOBEC and Cancer Viroimmunotherapy: Thinking the Unthinkable. Clin Cancer Res 2021; 27:3280-3290. [PMID: 33558423 PMCID: PMC8281496 DOI: 10.1158/1078-0432.ccr-20-1888] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/25/2020] [Accepted: 01/19/2021] [Indexed: 01/21/2023]
Abstract
The apolipoprotein B mRNA editing enzyme catalytic polypeptide (APOBEC) family protects against infection by degrading incoming viral genomes through cytosine deamination. Here, we review how the potential to unleash these potent DNA mutagens comes at a price as APOBEC DNA mutagenesis can contribute to development of multiple types of cancer. In addition, because viral infection induces its expression, APOBEC is seen as the enemy of oncolytic virotherapy through mutation of the viral genome and by generating virotherapy-resistant tumors. Therefore, overall APOBEC in cancer has received very poor press. However, we also speculate how there may be silver linings to the storm clouds (kataegis) associated with APOBEC activity. Thus, although mutagenic genomic chaos promotes emergence of ever more aggressive subclones, it also provides significant opportunity for cytotoxic and immune therapies. In particular, the superpower of cancer immunotherapy derives in part from mutation, wherein generation of tumor neoantigens-neoantigenesis-exposes tumor cells to functional T-cell repertoires, and susceptibility to immune checkpoint blockade. Moreover, APOBECs may be able to induce suprathreshold levels of cellular mutation leading to mitotic catastrophe and direct tumor cell killing. Finally, we discuss the possibility that linking predictable APOBEC-induced mutation with escape from specific frontline therapies could identify mutated molecules/pathways that can be targeted with small molecules and/or immunotherapies in a Trap and Ambush strategy. Together, these considerations lead to the counterintuitive hypothesis that, instead of attempting to expunge and excoriate APOBEC activity in cancer therapy, it might be exploited-and even, counterintuitively, encouraged.
Collapse
Affiliation(s)
- Richard G Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota.
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | - Alan Melcher
- The Institute of Cancer Research/Royal Marsden, National Institute for Health Research Biomedical Research Centre, London, United Kingdom
| | - Hardev Pandha
- Surrey Cancer Research Institute, Faculty of Health and Medical Sciences, University of Surrey Guildford, Surrey, United Kingdom
| | - Kevin J Harrington
- The Institute of Cancer Research/Royal Marsden, National Institute for Health Research Biomedical Research Centre, London, United Kingdom
| | - Jose S Pulido
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota
- Will's Eye Hospital, Philadelphia, Pennsylvania
| |
Collapse
|
4
|
Huang CP, Yang CY, Shyr CR. Utilizing Xenogeneic Cells As a Therapeutic Agent for Treating Diseases. Cell Transplant 2021; 30:9636897211011995. [PMID: 33975464 PMCID: PMC8120531 DOI: 10.1177/09636897211011995] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/19/2021] [Accepted: 04/05/2021] [Indexed: 12/27/2022] Open
Abstract
The utilization of biologically produced cells to treat diseases is a revolutionary invention in modern medicine after chemically synthesized small molecule drugs and biochemically made protein drugs. Cells are basic units of life with diverse functions in mature and developing organs, which biological properties could be utilized as a promising therapeutic approach for currently intractable and incurable diseases. Xenogeneic cell therapy utilizing animal cells other than human for medicinal purpose has been studied as a new way of treating diseases. Xenogeneic cell therapy is considered as a potential regenerative approach to fulfill current unmet medical needs because xenogeneic cells could be isolated from different animal organs and expanded ex vivo as well as maintain the characteristics of original organs, providing a versatile and plenty cell source for cell-based therapeutics beside autologous and allogeneic sources. The swine species is considered the most suitable source because of the similarity with humans in size and physiology of many organs in addition to the economic and ethical reasons plus the possibility of genetic modification. This review discusses the old proposed uses of xenogeneic cells such as xenogeneic pancreatic islet cells, hepatocytes and neuronal cells as a living drug for the treatment of degenerative and organ failure diseases. Novel applications of xenogeneic mesenchymal stroma cells and urothelial cells are also discussed. There are formidable immunological barriers toward successful cellular xenotransplantation in clinic despite major progress in the development of novel immunosuppression regimens and genetically multimodified donor pigs. However, immunological barriers could be turn into immune boosters by using xenogeneic cells of specific tissue types as a novel immunotherapeutic agent to elicit bystander antitumor immunity due to rejection immune responses. Xenogeneic cells have the potential to become a safe and efficacious option for intractable diseases and hard-to-treat cancers, adding a new class of cellular medicine in our drug armamentarium.
Collapse
Affiliation(s)
- Chi-Ping Huang
- Department of Urology, School of Medicine, China Medical University and Hospital, Taichung, Taiwan
| | - Chi-Yu Yang
- Animal Technology Research Center/Division of Animal Technology, Agriculture Technology Research Institute, Miaoli, Taiwan
| | - Chih-Rong Shyr
- Sex Hormone Research Center, Department of Medical Laboratory Science and Biotechnology, China Medical University and Hospital, Taichung, Taiwan
| |
Collapse
|
5
|
Combination of novel intravesical xenogeneic urothelial cell immunotherapy and chemotherapy enhances anti-tumor efficacy in preclinical murine bladder tumor models. Cancer Immunol Immunother 2020; 70:1419-1433. [PMID: 33156394 PMCID: PMC8053151 DOI: 10.1007/s00262-020-02775-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors induce robust and durable responses in advanced bladder cancer (BC), but only for a subset of patients. Xenovaccination has been proposed as an effective immunotherapeutic approach to induce anti-tumor immunity. Thus, we proposed a novel intravesical xenogeneic urothelial cell immunotherapy strategy to treat advanced BC based on the hypothesis that implanted xenogeneic urothelial cells not only provoke xeno-rejection immune responses but also elicit bystander anti-tumor immunity. METHODS Mouse advanced bladder cancer models were treated with vehicle control, intravesical xenogeneic urothelial cells, cisplatin + gemcitabine, or the combination and assessed for tumor responses to treatments. Tumors and spleens samples were collected for immunohistological staining, cellular and molecular analysis assessed by antibody staining, ELISA, cytotoxicity, and flow cytometry, respectively. RESULTS The combination treatment of xenogeneic urothelial cell immunotherapy with chemotherapy was more efficacious than either single therapy to extend survival time in MBT-2 graft bladder tumor model and to suppress tumor progression in murine carcinogen BBN-induced bladder tumor model. The single-cell immunotherapy and combined therapy increased more tumor-infiltrating immune cells in MBT-2 graft tumors compared to vehicle control and chemotherapy treatment groups. The activated T-cell proliferation, cytokine production, and cytotoxicity capacities were also higher in mice with xenogeneic urothelial cell immunotherapy and combination treatments. CONCLUSIONS Our results suggest the potential for a novel xenogeneic urothelial cell-based immunotherapy alone and synergy with chemotherapy in the combination therapy. Therefore, our study supports developing xenogeneic urothelial cells as an immunotherapeutic agent in combination with chemotherapy for BC treatment.
Collapse
|
6
|
Kachler K, Holzinger C, Trufa DI, Sirbu H, Finotto S. The role of Foxp3 and Tbet co-expressing Treg cells in lung carcinoma. Oncoimmunology 2018; 7:e1456612. [PMID: 30221050 DOI: 10.1080/2162402x.2018.1456612] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/14/2018] [Accepted: 03/15/2018] [Indexed: 01/21/2023] Open
Abstract
Despite the opposite roles of Tbet and Foxp3 in the immune system as well as in tumour biology, recent studies have demonstrated the presence of of CD4+ T cells, expressing both, Tbet and Foxp3. Although Tbet+Foxp3+ T cells are currently a subject of intense research, less is known about their biological function especially in cancer. Here we found a considerable accumulation of Tbet+Foxp3+CD4+ T cells, mediated by the immunosuppressive cytokine TGFβ in the lungs of tumour bearing mice. This is in line with previous studies, demonstrating the important role of TGFβ for the immunopathogenesis of cancer. By gathering results both in murine model and in human disease, we demonstrate that, the conversion of IFNγ producing anti-tumoral T-bet+Th1 CD4+ T cells into immunosuppressive Tbet and Foxp3-PD1 co-expressing regulatory cells could represent an additional important mechanism of TGFβ-mediated blockade of anti-tumour immunity.
Collapse
Affiliation(s)
- Katerina Kachler
- Department of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Corinna Holzinger
- Department of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Denis I Trufa
- Department of Thoracic Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Horia Sirbu
- Department of Thoracic Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Susetta Finotto
- Department of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
7
|
Perales MA. Timing Is Everything: Combining Post-Transplantation Adoptive Cell Therapy and Tumor Vaccines. Biol Blood Marrow Transplant 2016; 22:2113-2114. [PMID: 27721032 DOI: 10.1016/j.bbmt.2016.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 10/05/2016] [Indexed: 11/18/2022]
Affiliation(s)
- Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| |
Collapse
|
8
|
Nahas GR, Walker ND, Bryan M, Rameshwar P. A Perspective of Immunotherapy for Breast Cancer: Lessons Learned and Forward Directions for All Cancers. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2015; 9:35-43. [PMID: 26568682 PMCID: PMC4631157 DOI: 10.4137/bcbcr.s29425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/09/2015] [Accepted: 07/12/2015] [Indexed: 12/14/2022]
Abstract
Immunotherapy for cancer has been a focus 50 years ago. At the time, this treatment was developed prior to cloning of the cytokines, no knowledge of regulatory T-cells, and very little information that mesenchymal stem cells (MSCs) (originally colony forming unit-fibroblasts [CFU-F]) could be licensed by the inflammatory microenvironment to suppress an immune response. Given the information available at that time, mononuclear cells from the peripheral blood were activated ex vivo and then replaced in the patients with tumor. The intent was to harness these activated immune cells to target the cancer cells. These studies did not lead to long-term responses because the activated cells when reinfused into the patients were an advantage to the resident MSCs, which can home the tumor and then become suppressive in the presence of the immune cells. The immune suppression caused by MSCs would also expand regulatory T-cells, resulting instead in tumor protection. As time progressed, these different fields converged into a new approach to use immunotherapy for cancer. This article discusses these approaches and also reviews chimeric antigen receptor in the context of future treatments for solid tumors, including breast cancer.
Collapse
Affiliation(s)
| | - Nykia D Walker
- Rutgers New Jersey Medical School, Newark, NJ, USA. ; Rutgers Graduate School of Biomedical Sciences, Newark, NJ, USA
| | | | - Pranela Rameshwar
- Rutgers New Jersey Medical School, Newark, NJ, USA. ; Rutgers Graduate School of Biomedical Sciences, Newark, NJ, USA
| |
Collapse
|
9
|
Enhanced responses to tumor immunization following total body irradiation are time-dependent. PLoS One 2013; 8:e82496. [PMID: 24349298 PMCID: PMC3861406 DOI: 10.1371/journal.pone.0082496] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 10/25/2013] [Indexed: 12/25/2022] Open
Abstract
The development of successful cancer vaccines is contingent on the ability to induce effective and persistent anti-tumor immunity against self-antigens that do not typically elicit immune responses. In this study, we examine the effects of a non-myeloablative dose of total body irradiation on the ability of tumor-naïve mice to respond to DNA vaccines against melanoma. We demonstrate that irradiation followed by lymphocyte infusion results in a dramatic increase in responsiveness to tumor vaccination, with augmentation of T cell responses to tumor antigens and tumor eradication. In irradiated mice, infused CD8+ T cells expand in an environment that is relatively depleted in regulatory T cells, and this correlates with improved CD8+ T cell functionality. We also observe an increase in the frequency of dendritic cells displaying an activated phenotype within lymphoid organs in the first 24 hours after irradiation. Intriguingly, both the relative decrease in regulatory T cells and increase in activated dendritic cells correspond with a brief window of augmented responsiveness to immunization. After this 24 hour window, the numbers of dendritic cells decline, as does the ability of mice to respond to immunizations. When immunizations are initiated within the period of augmented dendritic cell activation, mice develop anti-tumor responses that show increased durability as well as magnitude, and this approach leads to improved survival in experiments with mice bearing established tumors as well as in a spontaneous melanoma model. We conclude that irradiation can produce potent immune adjuvant effects independent of its ability to induce tumor ablation, and that the timing of immunization and lymphocyte infusion in the irradiated host are crucial for generating optimal anti-tumor immunity. Clinical strategies using these approaches must therefore optimize such parameters, as the correct timing of infusion and vaccination may mean the difference between an ineffective treatment and successful tumor eradication.
Collapse
|
10
|
Baia GS, Caballero OL, Ho JSY, Zhao Q, Cohen T, Binder ZA, Salmasi V, Gallia GL, Quinones-Hinojosa A, Olivi A, Brem H, Burger P, Strausberg RL, Simpson AJG, Eberhart CG, Riggins GJ. NY-ESO-1 expression in meningioma suggests a rationale for new immunotherapeutic approaches. Cancer Immunol Res 2013; 1:296-302. [PMID: 24777967 DOI: 10.1158/2326-6066.cir-13-0029] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Meningiomas are the most common primary intracranial tumors. Surgical resection remains the treatment of choice for these tumors. However, a significant number of tumors are not surgically accessible, recur, or become malignant, necessitating the repetition of surgery and sometimes radiation. Chemotherapy is rarely used and is generally not recognized as an effective treatment. Cancer/testis (CT) genes represent a unique class of genes, which are expressed by germ cells, normally silenced in somatic cells, but activated in various cancers. CT proteins can elicit spontaneous immune responses in patients with cancer and this feature makes them attractive targets for immunotherapy-based approaches. We analyzed mRNA expression of 37 testis-restricted CT genes in a discovery set of 18 meningiomas by reverse transcription PCR. The overall frequency of expression of CT genes ranged from 5.6% to 27.8%. The most frequently expressed was NY-ESO-1, in 5 patients (27.8%). We subsequently analyzed NY-ESO-1 protein expression in a larger set of meningiomas by immunohistochemistry and found expression in 108 of 110 cases. In some cases, NY-ESO-1 expression was diffused and homogenous, but in most instances it was heterogeneous. Importantly, NY-ESO-1 expression was positively correlated with higher grade and patients presenting with higher levels of NY-ESO-1 staining had significantly worse disease-free and overall survival. We have also shown that NY-ESO-1 expression may lead to humoral immune response in patients with meningioma. Considering the limited treatment options for patients with meningioma, the potential of NY-ESO-1-based immunotherapy should be explored.
Collapse
Affiliation(s)
- Gilson S Baia
- Authors' Affiliations: New York Branch at Memorial Sloan-Kettering Cancer Center, New York, New York
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Chang DZ, Lomazow W, Joy Somberg C, Stan R, Perales MA. Granulocyte-Macrophage Colony Stimulating Factor: An Adjuvant for Cancer Vaccines. Hematology 2013; 9:207-15. [PMID: 15204102 DOI: 10.1080/10245330410001701549] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Granulocyte-macrophage colony stimulating factor (GM-CSF) enhances immune responses by inducing the proliferation, maturation, and migration of dendritic cells, and the expansion and differentiation of B and T lymphocytes. There is significant data in pre-clinical animal models demonstrating the adjuvant effects of GM-CSF in a variety of cancer vaccine approaches, including cellular vaccines, viral vaccines, peptide and protein vaccines, and DNA vaccines. GM-CSF is an attractive vaccine adjuvant because of its immune modulation effects and low toxicity profile. The results in animal models have been confirmed in pilot clinical trials and several clinical trials are currently ongoing.
Collapse
Affiliation(s)
- David Z Chang
- Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | | | | | | | |
Collapse
|
12
|
Lee KW, Jeong D, Na K. Doxorubicin loading fucoidan acetate nanoparticles for immune and chemotherapy in cancer treatment. Carbohydr Polym 2013; 94:850-6. [DOI: 10.1016/j.carbpol.2013.02.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 02/11/2013] [Accepted: 02/13/2013] [Indexed: 10/27/2022]
|
13
|
Andreev K, Graser A, Maier A, Mousset S, Finotto S. Therapeutical measures to control airway tolerance in asthma and lung cancer. Front Immunol 2012; 3:216. [PMID: 22855687 PMCID: PMC3405289 DOI: 10.3389/fimmu.2012.00216] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 07/03/2012] [Indexed: 01/28/2023] Open
Abstract
Airway tolerance is a specialized immunological surveillance which is activated by the cells of the lung to deal with and distinguish between innocuous and pathogenic inhalants. However, this distinction does not always occur. Airway tolerance is necessary to avoid the development of allergic disorders, such as asthma, which is dominated by a pathological expansion of Th2 and Th17 cells in the airways. By contrast, tumor cells induce tolerogenic factors in their microenvironment to evade T-cell mediated anti-tumor-immune responses. This review updates current understandings on the effect of the cytokines TGF-β, IL-10, and IL-17A on the lung immune responses to antigen, and analyzes their involvement in allergic asthma and lung cancer. The aim of the review is to evaluate where therapeutic intervention may be feasible and where it might fail. The multifunctional role of these cytokines further complicates the decision on the timing and concentration for their use as therapeutical targets. In fact, TGF-β has suppressive activity in early tumorigenesis, but may become tumor-promoting in the later stages of the disease. This dual behavior is sometimes due to changes in the cellular target of TGF-β, and to the expansion of the induced (i)-Tregs. Similarly, IL-17A has been found to elicit pro- as well as anti-tumor properties. Thus, this pro-inflammatory cytokine induces the production of IL-6 which interferes with Treg development. Yet IL-17A could promote tumor growth in conjunction with IL-6-dependent activation of Stat3. Thus, understanding the mechanisms of airway tolerance could help to improve the therapy to both, allergic asthma and lung cancer. Hereby, asthma therapy aims to induce and maintain tolerance to inhaled allergens and therapy against lung cancer tries to inhibit the tolerogenic response surrounding the tumor.
Collapse
Affiliation(s)
- Katerina Andreev
- Laboratory of Cellular and Molecular Lung Immunology, Institute of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg Erlangen, Germany
| | | | | | | | | |
Collapse
|
14
|
Page DB, Yuan J, Wolchok JD. Targeting cytotoxic T-lymphocyte antigen 4 in immunotherapies for melanoma and other cancers. Immunotherapy 2010; 2:367-79. [DOI: 10.2217/imt.10.21] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The immune system can simultaneously protect against tumor growth and sculpt resistant tumor strains. By a variety of mechanisms, anti-cytotoxic T-lymphocyte antigen (CTLA)-4 therapy may shift such opposing forces towards tumor elimination. In recent clinical trials, anti-CTLA-4 therapy induces durable responses that correlate with markers of immune activity, such as antigen-specific CD4+ or CD8+ cytokine release, antitumor antibody formation or cellular phenotype differentiation. However, some patients exhibit atypical responses to anti-CTLA-4 therapy, demonstrating transient/delayed responses or heterogeneity by lesion site. Such atypical responses may offer insight into the mechanism of anti-CTLA-4 therapy. The immunogram – a newly described graphical synthesis of treatment data and immune correlates in individual patients – may help us to confirm, reject or formulate new hypotheses regarding the mechanism of anti-CTLA-4 activity.
Collapse
Affiliation(s)
- David B Page
- Melanoma/Sarcoma Service, Memorial Sloan-Kettering Cancer Center, NY, USA
- Columbia University Medical Center, New York-Presbyterian Hospital, NY, USA
| | - Jianda Yuan
- Ludwig Center for Cancer Immunotherapy, Sloan-Kettering Institute, NY, USA; 1275 York Avenue, Box #340, NY 10065, USA
| | | |
Collapse
|
15
|
Avogadri F, Yuan J, Yang A, Schaer D, Wolchok JD. Modulation of CTLA-4 and GITR for cancer immunotherapy. Curr Top Microbiol Immunol 2010; 344:211-44. [PMID: 20563707 DOI: 10.1007/82_2010_49] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The rational manipulation of antigen-specific T cells to reignite a tumor-specific immune response in cancer patients is a challenge for cancer immunotherapy. Targeting coinhibitory and costimulatory T cell receptors with specific antibodies in cancer patients is an emerging approach to T cell manipulation, namely "immune modulation." Cytotoxic T-lymphocyte antigen-4 (CTLA-4) and glucocorticoid-induced tumor necrosis factor family receptor (GITR) are potential targets for immune modulation through anti-CTLA-4 blocking antibodies and anti-GITR agonistic antibodies, respectively. In this review, we first discuss preclinical findings key to the understanding of the mechanisms of action of these immunomodulatory antibodies and the preclinical evidence of antitumor activity which preceded translation into the clinic. We next describe the outcomes and immune related adverse effects associated with anti-CTLA-4 based clinical trials with particular emphasis on specific biomarkers used to elucidate the mechanisms of tumor immunity in patients. The experience with anti-CTLA-4 therapy and the durable clinical benefit observed provide proof of principle to effective antitumor immune modulation and the promise of future clinical immune modulatory antibodies.
Collapse
|
16
|
Wolchok JD, Saenger Y. The mechanism of anti-CTLA-4 activity and the negative regulation of T-cell activation. Oncologist 2009; 13 Suppl 4:2-9. [PMID: 19001145 DOI: 10.1634/theoncologist.13-s4-2] [Citation(s) in RCA: 183] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The survival rate of patients diagnosed with late-stage melanoma is poor--only 5%-10%. Enlisting the immune system in the fight against cancers such as melanoma could help improve the prognosis of these patients. Data have shown that melanocyte proteins make good targets for immune system-based therapy in this disease. However, self-tolerance, which develops to inhibit autoimmune attack, makes this strategy difficult. Two proteins on the surface of T cells--CD28 and cytotoxic T-lymphocyte antigen 4 (CTLA-4)--play important roles in the regulation of immune activation and tolerance. CD28 provides positive modulatory signals in the early stages of an immune response, while CTLA-4 signaling inhibits T-cell activation, particularly during strong T-cell responses. CTLA-4 blockade using anti-CTLA-4 monoclonal antibody therapy has great appeal because suppression of inhibitory signals results in the generation of an antitumor T-cell response. Both clinical and preclinical data indicate that CTLA-4 blockade results in direct activation of CD4+ and CD8+ effector cells, and anti-CTLA-4 monoclonal antibody therapy has shown promise in a number of cancers, particularly melanoma. Interestingly, the occurrence of adverse events among patients treated with CTLA-4 blockade helps shed light on the mechanism of action of anti-CTLA-4 monoclonal antibodies. Most adverse events involve immune-related toxicity to the skin and gastrointestinal tract. Major gastrointestinal toxicity develops in up to 21% of treated patients, and while an objective response occurs in approximately 36% of melanoma patients who develop enterocolitis with treatment, an objective response is found in only 11% of patients who do not experience this adverse reaction.
Collapse
Affiliation(s)
- Jedd D Wolchok
- Melanoma/Sarcoma Oncology Service and Ludwig Center for Cancer Immunotherapy, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Box #340, New York, New York 10065, USA.
| | | |
Collapse
|
17
|
Wang YS, Tsang YW, Chi CH, Chang CC, Chu RM, Chi KH. Synergistic anti-tumor effect of combination radio- and immunotherapy by electro-gene therapy plus intra-tumor injection of dendritic cells. Cancer Lett 2008; 266:275-85. [DOI: 10.1016/j.canlet.2008.02.063] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Revised: 02/26/2008] [Accepted: 02/28/2008] [Indexed: 11/26/2022]
|
18
|
Felicetti P, Mennecozzi M, Barucca A, Montgomery S, Orlandi F, Manova K, Houghton AN, Gregor PD, Concetti A, Venanzi FM. Tumor endothelial marker 8 enhances tumor immunity in conjunction with immunization against differentiation Ag. Cytotherapy 2008; 9:23-34. [PMID: 18236207 DOI: 10.1080/14653240601048369] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND We have previously shown that xenogeneic DNA vaccines encoding rat neu and melanosomal differentiation Ag induce tumor immunity. Others have developed vaccines targeting tumor neovasculature. Tumor endothelial marker 8 (TEM8) is expressed in the neovasculature of human tumors, and in the mouse melanoma B16, but its expression is limited in normal adult tissues. We describe a DNA vaccine combining xenogeneic tumor Ag and TEM8. METHODS In-situ hybridization was used to detect TEM8 RNA in mouse tumors. Mice transgenic for the rat neu proto-oncogene were immunized with DNA vaccines encoding TEM8 and the extracellular domain of rat neu and challenged with the 233-VSGA1 breast cancer cell line. In parallel experiments, C57BL/6 mice were immunized with TEM8 and human tyrosinase-related protein 1 (hTYRP1/hgp75) and challenged with B16F10 melanoma. RESULTS TEM8 was expressed in the stroma of transplantable mouse breast and melanoma tumors. In both model systems, TEM8 DNA had no activity as a single agent but significantly enhanced the anti-tumor immunity of neu and hTYRP1/hgp75 DNA vaccines when given in concert. The observed synergy was dependent upon CD8+ T cells, as depletion of this cell population just prior to tumor challenge obviated the effect of the TEM8 vaccine in both tumor models. DISCUSSION A local immune response to TEM8 may increase inflammation or tumor necrosis within the tumor, resulting in improved Ag presentation of HER2/neu and hTYRP1/hgp75. Alternatively, TEM8 expression in host APC may alter T-cell interactions or homing. In this way, TEM8 may act more as an adjuvant than an immunologic target.
Collapse
Affiliation(s)
- P Felicetti
- Swim Across America Laboratory of Tumor Immunology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Immunotherapy of Angiogenesis with DNA Vaccines. Angiogenesis 2008. [DOI: 10.1007/978-0-387-71518-6_39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
20
|
Perales MA, Diab A, Cohen AD, Huggins DW, Guevara-Patiño JA, Hubbard VM, Engelhorn ME, Kochman AA, Eng JM, Mortazavi F, Alpdogan O, Terwey TH, Heller G, Wolchok JD, Houghton AN, van den Brink MRM. DNA immunization against tissue-restricted antigens enhances tumor immunity after allogeneic hemopoietic stem cell transplantation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 177:4159-67. [PMID: 16951381 DOI: 10.4049/jimmunol.177.6.4159] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Malignant relapse remains a major problem for recipients of allogeneic hemopoietic stem cell transplantation (HSCT). We hypothesized that immunization of allogeneic HSCT recipients against tissue-restricted Ags using DNA vaccines would decrease the risk of relapse without enhancing graft-vs-host disease (GVHD). Using the mouse B16 melanoma model, we found that post-HSCT DNA immunization against a single tumor Ag induces tumor rejection that is significantly greater than HSCT alone in a T cell-depleted MHC-matched minor Ag-mismatched allogeneic HSCT model (LP --> B6). In treatment models, post-HSCT DNA immunization provides significantly greater overall survival than the vaccine alone. Donor leukocyte infusion further enhances tumor-free survival, including in treatment models. There was no GVHD in HSCT recipients treated with DNA vaccination and donor leukocyte infusion. Further analysis demonstrated that these effects are dependent on CD8+ T cells of donor origin that recognize multiple epitopes. These results demonstrate that DNA immunization against tissue-restricted Ags after allogeneic T cell-depleted HSCT can induce potent antitumor effects without causing GVHD.
Collapse
Affiliation(s)
- Miguel-Angel Perales
- Swim Across America Laboratory of Tumor Immunology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Bergman PJ, Camps-Palau MA, McKnight JA, Leibman NF, Craft DM, Leung C, Liao J, Riviere I, Sadelain M, Hohenhaus AE, Gregor P, Houghton AN, Perales MA, Wolchok JD. Development of a xenogeneic DNA vaccine program for canine malignant melanoma at the Animal Medical Center. Vaccine 2006; 24:4582-5. [PMID: 16188351 DOI: 10.1016/j.vaccine.2005.08.027] [Citation(s) in RCA: 181] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Canine malignant melanoma (CMM) is an aggressive neoplasm treated with surgery and/or fractionated RT; however, metastatic disease is common and chemoresistant. Preclinical and clinical studies by our laboratory and others have shown that xenogeneic DNA vaccination with tyrosinase family members can produce immune responses resulting in tumor rejection or protection and prolongation of survival. These studies provided the impetus for development of a xenogeneic DNA vaccine program in CMM. MATERIALS AND METHODS Cohorts of three dogs each received increasing doses of xenogeneic plasmid DNA encoding either human tyrosinase (huTyr; 100/500/1500 mcg), murine GP75 (muGP75; 100/500/1500 mcg), murine tyrosinase (muTyr; 5 dogs each at 100/500 mcg), muTyr+/-HuGM-CSF (9 dogs at 50 mcg muTyr, 3 dogs each at 100/400/800 mcg HuGM-CSF, or 3 dogs each at 50 mcg muTyr with 100/400/800 mcg HuGM-CSF), or 50 mcg MuTyr intramuscularly biweekly for a total of four vaccinations. RESULTS The Kaplan-Meier median survival time (KM MST) for all stage II-IV dogs treated with huTyr, muGP75 and muTyr are 389, 153 and 224 days, respectively. Preliminarily, the KM MST for stage II-IV dogs treated with 50 mcg MuTyr, 100/400/800 mcg HuGM-CSF or combination MuTyr/HuGM-CSF are 242, 148 and >402 (median not reached) days, respectively. Thirty-three stage II-III dogs with loco-regionally controlled CMM across the xenogeneic vaccine studies have a KM MST of 569 days. Minimal to mild pain was noted on vaccination and one dog experienced vitiligo. We have recently investigated antibody responses in dogs vaccinated with HuTyr and found 2- to 5-fold increases in circulating antibodies to human tyrosinase. CONCLUSIONS The results of these trials demonstrate that xenogeneic DNA vaccination in CMM: (1) is safe, (2) leads to the development of anti-tyrosinase antibodies, (3) is potentially therapeutic, and (4) is an attractive candidate for further evaluation in an adjuvant, minimal residual disease Phase II setting for CMM.
Collapse
Affiliation(s)
- P J Bergman
- Donaldson-Atwood Cancer Clinic & Flaherty Comparative Oncology Laboratory, The Animal Medical Center, 510 East 62nd Street, New York, NY 10021, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Rizzuto GA, Wolchok JD. Persistence makes perfect: the benefits of IL-2 in adoptive immunotherapy. Cytotherapy 2006; 7:391-2. [PMID: 16236627 DOI: 10.1080/14653240500318970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- G A Rizzuto
- Swim Across America Laboratory of Tumor Immunology, Memorial Sloan-Kettering Cancer Center, New York 10021, USA
| | | |
Collapse
|
23
|
Luo W, Hsu JCF, Tsao CY, Ko E, Wang X, Ferrone S. Differential Immunogenicity of Two Peptides Isolated by High Molecular Weight-Melanoma-Associated Antigen-Specific Monoclonal Antibodies with Different Affinities. THE JOURNAL OF IMMUNOLOGY 2005; 174:7104-10. [PMID: 15905554 DOI: 10.4049/jimmunol.174.11.7104] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Peptide mimics isolated from phage display peptide libraries by panning with self-tumor-associated Ag (TAA)-specific mAbs are being evaluated as immunogens to implement active specific immunotherapy. Although TAA-specific mAb are commonly used to isolate peptide mimics, no information is available regarding the Ab characteristics required to isolate immunogenic TAA peptide mimics. To address this question, we have used mAb 763.74 and mAb GH786, which recognize the same or spatially close antigenic determinant(s) of the human high m.w.-melanoma-associated Ag (HMW-MAA), although with different affinity. mAb 763.74 affinity is higher than that of mAb GH786. Panning of phage display peptide libraries with mAb 763.74 and mAb GH786 resulted in the isolation of peptides P763.74 and PGH786, respectively. When compared for their ability to induce HMW-MAA-specific immune responses in BALB/c mice, HMW-MAA-specific Ab titers were significantly higher in mice immunized with P763.74 than in those immunized with PGH786. The HMW-MAA-specific Ab titers were markedly increased by a booster with HMW-MAA-bearing melanoma cells, an effect that was significantly higher in mice primed with P763.74 than in those primed with PGH786. Lastly, P763.74, but not PGH786, induced a delayed-type hypersensitivity response to HMW-MAA-bearing melanoma cells. These findings suggest that affinity for TAA is a variable to take into account when selecting mAb to isolate peptide mimics from a phage display peptide library.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/pharmacology
- Adoptive Transfer
- Animals
- Antibodies, Neoplasm/metabolism
- Antigens, Neoplasm/administration & dosage
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Binding Sites, Antibody
- Cell Line, Tumor
- Humans
- Hypersensitivity, Delayed/immunology
- Immunization, Secondary
- Inovirus/genetics
- Melanoma/immunology
- Melanoma/metabolism
- Mice
- Mice, Inbred BALB C
- Molecular Mimicry/immunology
- Molecular Weight
- Peptide Fragments/administration & dosage
- Peptide Fragments/chemical synthesis
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Peptide Library
Collapse
Affiliation(s)
- Wei Luo
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | |
Collapse
|
24
|
|
25
|
Ochsenbein AF. Immunological ignorance of solid tumors. ACTA ACUST UNITED AC 2005; 27:19-35. [PMID: 15965711 DOI: 10.1007/s00281-004-0192-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2004] [Accepted: 12/15/2004] [Indexed: 01/21/2023]
Abstract
Many peripheral solid tumors such as sarcomas and carcinomas express tumor-specific antigens that can serve as targets for immune effector T cells. Nevertheless, the immune surveillance against clinically manifest carcinomas and sarcomas seems relatively inefficient. Naïve cytotoxic T cells are activated exclusively in secondary lymphoid organs including the spleen and lymph nodes. Tumor antigen might be either cross-presented to naïve cytotoxic T cells by professional antigen-presenting cells (pAPC), or presented directly by tumor cells that migrated to secondary lymphoid organs. Direct priming is quite inefficient during early tumor development because metastasis to lymphoid organs is usually limited to advanced stage diseases. Similarly, the process of cross-priming by pAPC seems to depend on relatively large antigen amounts and on maturation stimuli for dendritic cells, and both requirements may be limiting during initial tumorigenesis. Therefore, the immunosurveillance of solid tumors may fail because they are ignored for too long by the immune system. However, these situations may prove promising for the induction of tumor-specific T cell immunity by vaccination, as the T cell repertoire against these antigens has a naïve phenotype and is not yet affected by tolerance mechanisms.
Collapse
Affiliation(s)
- Adrian F Ochsenbein
- Department of Clinical Research, University of Berne, Murtenstrasse 35, 3010, Berne, Switzerland.
| |
Collapse
|
26
|
Perales MA, Chapman PB. Immunizing against partially defined antigen mixtures, gangliosides, or peptides to induce antibody, T cell, and clinical responses. ACTA ACUST UNITED AC 2005; 22:749-60. [PMID: 16110638 DOI: 10.1016/s0921-4410(04)22034-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Affiliation(s)
- Miguel A Perales
- Department of Medicine & Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
27
|
Luo W, Hsu JCF, Kieber-Emmons T, Wang X, Ferrone S. Human tumor associated antigen mimicry by xenoantigens, anti-idiotypic antibodies and peptide mimics: Implications for immunotherapy of malignant diseases. ACTA ACUST UNITED AC 2005; 22:769-87. [PMID: 16110640 DOI: 10.1016/s0921-4410(04)22036-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Affiliation(s)
- Wei Luo
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | |
Collapse
|
28
|
|
29
|
Perales MA, Wolchok JD. CD4 help and tumor immunity: beyond the activation of cytotoxic T lymphocytes. Ann Surg Oncol 2004; 11:881-2. [PMID: 15383423 DOI: 10.1245/aso.2004.08.911] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
30
|
Abstract
The field of immunotherapy holds clear promise not only for the development of new approaches to cancer and other diseases, but also for providing fundamental insight into the human immune response. In order for this promise to be realized, however, the scientific community must overcome an array of challenges. These challenges reflect not only the difficulties inherent in conducting investigations in human patients, but also difficulties created by the culture and practice of our own institutions, reward structure, and funding mechanisms. We suggest steps to be taken to reinvigorate basic research in human subjects as part of the mainstream of science.
Collapse
Affiliation(s)
- Ralph M Steinman
- Laboratory of Cellular Physiology and Immunology, Rockefeller University, New York, NY 10021-6399, USA.
| | | |
Collapse
|
31
|
McBride WH, Chiang CS, Olson JL, Wang CC, Hong JH, Pajonk F, Dougherty GJ, Iwamoto KS, Pervan M, Liao YP. A Sense of Danger from Radiation1. Radiat Res 2004; 162:1-19. [PMID: 15222781 DOI: 10.1667/rr3196] [Citation(s) in RCA: 242] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Tissue damage caused by exposure to pathogens, chemicals and physical agents such as ionizing radiation triggers production of generic "danger" signals that mobilize the innate and acquired immune system to deal with the intrusion and effect tissue repair with the goal of maintaining the integrity of the tissue and the body. Ionizing radiation appears to do the same, but less is known about the role of "danger" signals in tissue responses to this agent. This review deals with the nature of putative "danger" signals that may be generated by exposure to ionizing radiation and their significance. There are a number of potential consequences of "danger" signaling in response to radiation exposure. "Danger" signals could mediate the pathogenesis of, or recovery from, radiation damage. They could alter intrinsic cellular radiosensitivity or initiate radioadaptive responses to subsequent exposure. They may spread outside the locally damaged site and mediate bystander or "out-of-field" radiation effects. Finally, an important aspect of classical "danger" signals is that they link initial nonspecific immune responses in a pathological site to the development of specific adaptive immunity. Interestingly, in the case of radiation, there is little evidence that "danger" signals efficiently translate radiation-induced tumor cell death into the generation of tumor-specific immunity or normal tissue damage into autoimmunity. The suggestion is that radiation-induced "danger" signals may be inadequate in this respect or that radiation interferes with the generation of specific immunity. There are many issues that need to be resolved regarding "danger" signaling after exposure to ionizing radiation. Evidence of their importance is, in some areas, scant, but the issues are worthy of consideration, if for no other reason than that manipulation of these pathways has the potential to improve the therapeutic benefit of radiation therapy. This article focuses on how normal tissues and tumors sense and respond to danger from ionizing radiation, on the nature of the signals that are sent, and on the impact on the eventual consequences of exposure.
Collapse
Affiliation(s)
- William H McBride
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095-1714, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Strair RK, Schaar D, Medina D, Todd MB, Aisner J, DiPaola RS, Manago J, Knox B, Jenkinson A, Senzon R, Baker C, Liesel D, Ciardella M, Kuriyan M, Rubin A, Lattime EC. Antineoplastic Effects of Partially HLA-Matched Irradiated Blood Mononuclear Cells in Patients With Renal Cell Carcinoma. J Clin Oncol 2003; 21:3785-91. [PMID: 14551297 DOI: 10.1200/jco.2003.05.094] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose: Vaccines, cytokines, and other biologic-based therapies are being developed as antineoplastic agents. Many of these agents are designed to induce an autologous immune response directed against the malignancy. In contrast, hematopoietic stem-cell transplantation is being developed as a form of allogeneic immunotherapy. This study tests the tolerance and antineoplastic activity of sequential infusions of partially HLA-matched allogeneic blood mononuclear cells (obtained from relatives) when administered outside of the context of a hematopoietic stem-cell transplantation. The cells are irradiated to prevent graft-versus-host disease. Patients and Methods: Fifteen patients with relapsed or refractory malignancies for which no standard therapy was available were enrolled onto a clinical trial designed to assess the tolerability and antineoplastic effects of irradiated partially HLA-matched blood mononuclear cells obtained from relatives. Results: There was disease regression in three patients with metastatic renal cell carcinoma during treatment. There was disease progression in six patients with metastatic renal cell carcinoma and two patients with metastatic melanoma during treatment. There was no change in disease state in several other patients. Conclusion: Irradiated allogeneic blood mononuclear cells administered outside the context of hematopoietic stem-cell transplantation may induce disease responses in patients with relapsed or refractory malignancies. Transfusion of irradiated allogeneic blood mononuclear cells should be developed further as a novel therapeutic antineoplastic approach.
Collapse
Affiliation(s)
- Roger K Strair
- Division of Medical Oncology and Surgical Oncology, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Knutson KL, Curiel TJ, Salazar L, Disis ML. Immunologic principles and immunotherapeutic approaches in ovarian cancer. Hematol Oncol Clin North Am 2003; 17:1051-73. [PMID: 12959191 DOI: 10.1016/s0889-8588(03)00064-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Ovarian cancer is an immunogenic tumor, and numerous antigens have been identified in recent years. Several of these antigens are important in regulating tumor growth and may be ideal targets for the development of immune-based strategies. In the absence of immunologic intervention, tumors evade the immune system by several mechanisms, most notably tolerance and immunosuppression. As understanding of the immune response improves, strategies are being designed to circumvent T-cell tolerance to self-antigens through modulation of APC function. In addition, techniques are being developed to identify reverse ovarian cancer-induced immune evasion tactics. The type of the immune-based therapy to apply varies with disease burden. It is hoped that discoveries at the bench along with lessons learned in prior clinical trials soon will allow clinicians to develop rationally based immunologic strategies to treat and prevent ovarian cancer.
Collapse
Affiliation(s)
- Keith L Knutson
- Tumor Vaccine Group, 1959 Northeast Pacific Street, Box 356527, HSB BB1321, University of Washington, Seattle, WA 98195, USA.
| | | | | | | |
Collapse
|
34
|
Sato M, Chamoto K, Nishimura T. A novel tumor-vaccine cell therapy using bone marrow-derived dendritic cell type 1 and antigen-specific Th1 cells. Int Immunol 2003; 15:837-43. [PMID: 12807822 DOI: 10.1093/intimm/dxg081] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Dendritic cell (DC)-based tumor-vaccine therapy is a rational strategy for tumor immunotherapy. However, using this protocol, it is still difficult to induce long-term regression in established tumor-bearing mice. To overcome this problem we developed a novel tumor-vaccine therapy, combining inactivated tumor cells with bone marrow-derived DC type 1 (BMDC1) and antigen-specific T(h)1 cells. BALB/c mice were intradermally inoculated with A20-OVA tumor cells expressing ovalbumin (OVA) as a model tumor antigen. After A20-OVA tumor mass became palpable (6-8 mm), mice were treated with DC-based vaccine therapy in various protocols. A complete cure of tumor-bearing mice was induced only when mice were repeatedly vaccinated with inactivated A20-OVA cells, OVA-pulsed BMDC1 and OVA-specific T(h)1 cells. Regression of tumor cells was associated with induction of T(h)1/T(c)1-dominant antitumor immunity. Removal of one of these cellular components during vaccination resulted in failure to completely cure tumor-bearing mice. Moreover, BMDC2 cells could not replace the therapeutic effect of BMDC1 cells combined with T(h)1 cells. Thus, we propose a novel tumor-vaccine cell therapy using DC1 and T(h)1 cells.
Collapse
Affiliation(s)
- Marimo Sato
- Division of Immunoregulation, Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo 060-0815, Japan
| | | | | |
Collapse
|
35
|
Knutson KL, Salazar L, Schiffman K, Disis ML. Immunotherapy of glioblastoma multiforme. Expert Rev Neurother 2003; 3:511-23. [PMID: 19810935 DOI: 10.1586/14737175.3.4.511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Glioblastoma multiforme is immunogenic and several glioblastoma multiforme-related antigens have now been identified. In addition, the immunologic characteristics of the tumor microenvironment that may affect tumor growth are becoming increasingly understood. The type of immune-based approach selected to treat glioblastoma multiforme will depend on the tumor burden. For minimal disease states, active vaccination may be useful for generating adequate protection from relapse. However, for more advanced stage disease states, more rigorous strategies may need to be applied, such as adoptive T-cell therapy, antibody therapy or a combination of different techniques. The immunosuppressive environment observed during advanced malignancy may need to be reversed for improved efficacy of immune-based therapies.
Collapse
Affiliation(s)
- Keith L Knutson
- Tumor Vaccine Group, 1959 NE Pacific St, Box 356527, HSB BB1321, University of Washington, Seattle, WA 98195 USA.
| | | | | | | |
Collapse
|
36
|
Schirrmacher V, Feuerer M, Beckhove P, Ahlert T, Umansky V. T cell memory, anergy and immunotherapy in breast cancer. J Mammary Gland Biol Neoplasia 2002; 7:201-8. [PMID: 12463740 DOI: 10.1023/a:1020308104613] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
T cell immunity in breast cancer is suggested to play a role in tumor dormancy, a period of stability which can correspond to the time interval between primary treatment and tumor recurrence. Bone marrow in breast cancer patients seems to be particularly important because it is highly enriched with cancer specific memory T cells. Similar cells can be found in peripheral blood, but these appear to be functionally anergic. The immune system of primary operated breast cancer patients does not seem to be completely anergized. Bone marrow derived memory T cells can be reactivated ex vivo and show functional reactivity, including tumor rejection in NOD/SCID mice. Promising results were obtained from a postoperative phase-II active specific immunotherapy study. In this study, 32 patients treated with an optimal formulation of a virus-modified autologous tumor vaccine (ATV-NDV) appeared to have a significant 5-year survival benefit. Our results suggest that cancer reactive memory T cells which are enriched in the bone marrow of breast cancer patients, can be activated ex vivo via autologous dendritic cells pulsed with breast cancer tumor antigens, or they can be activated in situ via a tumor vaccine, which combines tumor antigens with virus infection. The findings should encourage further studies in breast cancer on active specific immunotherapy with tumor vaccines or adoptive immunotherapy with activated memory T cells.
Collapse
|