1
|
Shin HS, Kim H, Kwon SG, Lee H, Lee JO, Kim YS. Tumor cells ectopically expressing the membrane-bound form of IL-7 develop an antitumor immune response efficiently in a colon carcinoma model. Mol Cells 2025; 48:100175. [PMID: 39743142 PMCID: PMC11873615 DOI: 10.1016/j.mocell.2024.100175] [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: 09/26/2024] [Revised: 12/20/2024] [Accepted: 12/25/2024] [Indexed: 01/04/2025] Open
Abstract
Various approaches employing cytokines and cytokine gene-modified tumor cells have been explored to induce antitumor responses, yet their widespread application has been limited due to efficacy concerns and adverse effects. In this study, interleukin-7 was engineered for expression both as a natural secretory form (sIL-7) and as a membrane-bound form fused with the B7.1 type I transmembrane protein (mbIL-7/B7) on CT26 colon cancer cells. Analysis of the resulting cell clones demonstrated that ectopically expressed sIL-7 and mbIL-7/B7 both retained similar capacities to induce the expansion and activation of CD8+ T cells and to enhance antitumor responses in vitro. While the sIL-7 or mbIL-7/B7 clones showed similar growth in culture, the mbIL-7/B7 clone exhibited lower tumorigenicity in mice compared with the sIL-7 clone or wild-type CT26 cells. Specifically, the mbIL-7/B7 clone failed to form tumors in approximately 60% of the mice injected with it. Moreover, 80% of mice that rejected the mbIL-7/B7 clone developed long-term systemic immunity against CT26 cells. Analysis of immune cells within the tumor masses revealed significant increases in CD4+ T cells, CD8+ T cells, and dendritic cells in tumors formed by the mbIL-7/B7 clone compared to those formed by the sIL-7 clone. These findings suggest that the membrane-bound form of IL-7 with B7.1 is more effective than the secretory form in establishing antitumor immunity within the tumor microenvironment. Our strategy of expressing the mbIL-7/B7 chimera holds promise as a novel approach for tumor therapy, particularly in cases requiring IL-7 supplementation.
Collapse
Affiliation(s)
- Hee-Su Shin
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 34134, Korea
| | - Hyejin Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 34134, Korea
| | - Soon-Gyu Kwon
- Department of Life Sciences and Institute of Membrane Proteins, POSTECH, Pohang 37673, Korea
| | - Hayyoung Lee
- Department of Life Sciences and Postech Biotech Center, POSTECH, Pohang 37673, Korea
| | - Jie-Oh Lee
- Department of Life Sciences and Institute of Membrane Proteins, POSTECH, Pohang 37673, Korea.
| | - Young Sang Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 34134, Korea.
| |
Collapse
|
2
|
Lei W, Zhou K, Lei Y, Li Q, Zhu H. Cancer vaccines: platforms and current progress. MOLECULAR BIOMEDICINE 2025; 6:3. [PMID: 39789208 PMCID: PMC11717780 DOI: 10.1186/s43556-024-00241-8] [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: 08/02/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
Cancer vaccines, crucial in the immunotherapeutic landscape, are bifurcated into preventive and therapeutic types, both integral to combating oncogenesis. Preventive cancer vaccines, like those against HPV and HBV, reduce the incidence of virus-associated cancers, while therapeutic cancer vaccines aim to activate dendritic cells and cytotoxic T lymphocytes for durable anti-tumor immunity. Recent advancements in vaccine platforms, such as synthetic peptides, mRNA, DNA, cellular, and nano-vaccines, have enhanced antigen presentation and immune activation. Despite the US Food and Drug Administration approval for several vaccines, the full therapeutic potential remains unrealized due to challenges such as antigen selection, tumor-mediated immunosuppression, and optimization of delivery systems. This review provides a comprehensive analysis of the aims and implications of preventive and therapeutic cancer vaccine, the innovative discovery of neoantigens enhancing vaccine specificity, and the latest strides in vaccine delivery platforms. It also critically evaluates the role of adjuvants in enhancing immunogenicity and mitigating the immunosuppressive tumor microenvironment. The review further examines the synergistic potential of combining cancer vaccines with other therapies, such as chemotherapy, radiotherapy, and immune checkpoint inhibitors, to improve therapeutic outcomes. Overcoming barriers such as effective antigen identification, immunosuppressive microenvironments, and adverse effects is critical for advancing vaccine development. By addressing these challenges, cancer vaccines can offer significant improvements in patient outcomes and broaden the scope of personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Wanting Lei
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Kexun Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ye Lei
- College of Liberal Arts, Neijiang Normal University, Neijiang, 641100, Sichuan, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
3
|
Gupta DS, Gupta DS, Abjani NK, Dave Y, Apte K, Kaur G, Kaur D, Saini AK, Sharma U, Haque S, Tuli HS. Vaccine-based therapeutic interventions in lung cancer management: A recent perspective. Med Oncol 2024; 41:249. [PMID: 39316239 DOI: 10.1007/s12032-024-02489-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/24/2024] [Indexed: 09/25/2024]
Abstract
The incidence of lung cancer continues to grow globally, contributing to an ever-increasing load on healthcare systems. Emerging evidence has indicated lowered efficacy of conventional treatment strategies, such as chemotherapy, surgical interventions and radiotherapy, prompting the need for exploring alternative interventions. A growing focus on immunotherapy and the development of personalized medicine has paved the way for vaccine-based delivery in lung cancer. With various prominent targets such as CD8+T cells and PD-L1, immune-targeted, anti-cancer vaccines have been evaluated in both, pre-clinical and clinical settings, to improve therapeutic outcomes. However, there are a number of challenges that must be addressed, including the scalability of such delivery systems, heterogeneity of lung cancers, and long-term safety as well as efficacy. In addition to this, natural compounds, in combination with immunotherapy, have gained considerable research interest in recent times. This makes it necessary to explore their role in synergism with immune-targeted agents. The authors of this review aim to offer an overview of recent advances in our understanding of lung cancer pathogenesis, detection and management strategies, and the emergence of immunotherapy with a special focus on vaccine delivery. This finding is supported with evidence from testing in non-human and human models, showcasing promising results. Prospects for phytotherapy have also been discussed, in order to combat some pitfalls and limitations. Finally, the future perspectives of vaccine usage in lung cancer management have also been discussed, to offer a holistic perspective to readers, and to prompt further research in the domain.
Collapse
Affiliation(s)
- Dhruv Sanjay Gupta
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle-West, Mumbai, 56, India
| | - Daksh Sanjay Gupta
- Vivekanand Education Society's College of Pharmacy, Chembur, Mumbai, Maharashtra, 400074, India
| | - Nosheen Kamruddin Abjani
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle-West, Mumbai, 56, India
| | - Yash Dave
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle-West, Mumbai, 56, India
| | - Ketaki Apte
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle-West, Mumbai, 56, India
| | - Ginpreet Kaur
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle-West, Mumbai, 56, India.
| | - Damandeep Kaur
- University Center for Research & Development (UCRD), Chandigarh University, Gharuan, Mohali, Punjab, 140413, India
| | - Adesh Kumar Saini
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, India
| | - Ujjawal Sharma
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bhatinda, 151001, India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, India.
| |
Collapse
|
4
|
Huang X, Zhu X, Yang H, Li Q, Gai L, Sui X, Lu H, Feng J. Nanomaterial Delivery Vehicles for the Development of Neoantigen Tumor Vaccines for Personalized Treatment. Molecules 2024; 29:1462. [PMID: 38611742 PMCID: PMC11012694 DOI: 10.3390/molecules29071462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/16/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Tumor vaccines have been considered a promising therapeutic approach for treating cancer in recent years. With the development of sequencing technologies, tumor vaccines based on neoantigens or genomes specifically expressed in tumor cells, mainly in the form of peptides, nucleic acids, and dendritic cells, are beginning to receive widespread attention. Therefore, in this review, we have introduced different forms of neoantigen vaccines and discussed the development of these vaccines in treating cancer. Furthermore, neoantigen vaccines are influenced by factors such as antigen stability, weak immunogenicity, and biosafety in addition to sequencing technology. Hence, the biological nanomaterials, polymeric nanomaterials, inorganic nanomaterials, etc., used as vaccine carriers are principally summarized here, which may contribute to the design of neoantigen vaccines for improved stability and better efficacy.
Collapse
Affiliation(s)
- Xiaoyu Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Xiaolong Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Huan Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Qinyi Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Lizhi Gai
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, and Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China;
| | - Xinbing Sui
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Hua Lu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, and Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China;
| | - Jiao Feng
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| |
Collapse
|
5
|
Gurunathan S, Thangaraj P, Wang L, Cao Q, Kim JH. Nanovaccines: An effective therapeutic approach for cancer therapy. Biomed Pharmacother 2024; 170:115992. [PMID: 38070247 DOI: 10.1016/j.biopha.2023.115992] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Cancer vaccines hold considerable promise for the immunotherapy of solid tumors. Nanomedicine offers several strategies for enhancing vaccine effectiveness. In particular, molecular or (sub) cellular vaccines can be delivered to the target lymphoid tissues and cells by nanocarriers and nanoplatforms to increase the potency and durability of antitumor immunity and minimize negative side effects. Nanovaccines use nanoparticles (NPs) as carriers and/or adjuvants, offering the advantages of optimal nanoscale size, high stability, ample antigen loading, high immunogenicity, tunable antigen presentation, increased retention in lymph nodes, and immunity promotion. To induce antitumor immunity, cancer vaccines rely on tumor antigens, which are administered in the form of entire cells, peptides, nucleic acids, extracellular vesicles (EVs), or cell membrane-encapsulated NPs. Ideal cancer vaccines stimulate both humoral and cellular immunity while overcoming tumor-induced immune suppression. Herein, we review the key properties of nanovaccines for cancer immunotherapy and highlight the recent advances in their development based on the structure and composition of various (including synthetic and semi (biogenic) nanocarriers. Moreover, we discuss tumor cell-derived vaccines (including those based on whole-tumor-cell components, EVs, cell membrane-encapsulated NPs, and hybrid membrane-coated NPs), nanovaccine action mechanisms, and the challenges of immunocancer therapy and their translation to clinical applications.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641 021, Tamil Nadu, India.
| | - Pratheep Thangaraj
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641 021, Tamil Nadu, India
| | - Lin Wang
- Research and Development Department, Qingdao Haier Biotech Co., Ltd., Qingdao, China
| | - Qilong Cao
- Research and Development Department, Qingdao Haier Biotech Co., Ltd., Qingdao, China
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
6
|
Zhang X, Cui H, Zhang W, Li Z, Gao J. Engineered tumor cell-derived vaccines against cancer: The art of combating poison with poison. Bioact Mater 2022; 22:491-517. [PMID: 36330160 PMCID: PMC9619151 DOI: 10.1016/j.bioactmat.2022.10.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/07/2022] [Accepted: 10/13/2022] [Indexed: 12/23/2022] Open
Abstract
Tumor vaccination is a promising approach for tumor immunotherapy because it presents high specificity and few side effects. However, tumor vaccines that contain only a single tumor antigen can allow immune system evasion by tumor variants. Tumor antigens are complex and heterogeneous, and identifying a single antigen that is uniformly expressed by tumor cells is challenging. Whole tumor cells can produce comprehensive antigens that trigger extensive tumor-specific immune responses. Therefore, tumor cells are an ideal source of antigens for tumor vaccines. A better understanding of tumor cell-derived vaccines and their characteristics, along with the development of new technologies for antigen delivery, can help improve vaccine design. In this review, we summarize the recent advances in tumor cell-derived vaccines in cancer immunotherapy and highlight the different types of engineered approaches, mechanisms, administration methods, and future perspectives. We discuss tumor cell-derived vaccines, including whole tumor cell components, extracellular vesicles, and cell membrane-encapsulated nanoparticles. Tumor cell-derived vaccines contain multiple tumor antigens and can induce extensive and potent tumor immune responses. However, they should be engineered to overcome limitations such as insufficient immunogenicity and weak targeting. The genetic and chemical engineering of tumor cell-derived vaccines can greatly enhance their targeting, intelligence, and functionality, thereby realizing stronger tumor immunotherapy effects. Further advances in materials science, biomedicine, and oncology can facilitate the clinical translation of tumor cell-derived vaccines.
Collapse
Affiliation(s)
- Xinyi Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Hengqing Cui
- Department of Burns and Plastic Surgery, Shanghai Changzheng Hospital, Shanghai, 200003, China
| | - Wenjun Zhang
- Department of Burns and Plastic Surgery, Shanghai Changzheng Hospital, Shanghai, 200003, China
| | - Zhaoshen Li
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Corresponding author. Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China,Corresponding author. Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200444, China.
| |
Collapse
|
7
|
|
8
|
Kittler JM, Sommer J, Fischer A, Britting S, Karg MM, Bock B, Atreya I, Heindl LM, Mackensen A, Bosch JJ. Characterization of CD4+ T cells primed and boosted by MHCII primary uveal melanoma cell-based vaccines. Oncotarget 2019; 10:1812-1828. [PMID: 30956760 PMCID: PMC6442993 DOI: 10.18632/oncotarget.26737] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 02/09/2019] [Indexed: 02/03/2023] Open
Abstract
Uveal melanoma is the most common primary malignancy of the eye in adults. Despite significant improvements in treatment of the primary tumor, to date none of these therapies prevent metastatic disease or improve overall survival. We are exploring immunotherapeutic options for metastatic uveal melanoma using MHC II uveal melanoma cell-based vaccines that target the activation of tumor-reactive CD4+ T cells. Previously, we showed that these uveal melanoma cell-based vaccines activate CD4+ T cells within total peripheral blood lymphocytes (PBMC). Since PBMC include professional antigen presenting cells, we now demonstrate that Mel202/DR1/CD80 vaccine cells directly activate a diverse repertoire of purified, naïve CD4+ T cells. The activated CD4+ T cells proliferated, secreted high amounts of interferon gamma (IFNγ) and produced a heterogeneous profile of Th1, Th2 and Th17 cytokines. Analysis of the TCR-Vβ-repertoire showed that a polyclonal T cell response was induced, suggesting the capacity of vaccine-activated CD4+ T cells to target multiple tumor (neo)antigens. In addition, a subset of the responding CD4+ T cells expressed forkhead box protein P3 (FoxP3), indicating that although a regulatory component of the vaccine-activated CD4+ T cell response was induced, the anti-tumor vaccine response was not limited by these regulatory CD4+ T cells. Finally, Mel202/DR1/CD80 uveal melanoma vaccine cells expressed the intercellular adhesion molecule 1 (ICAM-1) that was pivotal for CD4+ T cell activation via lymphocyte function-associated antigen 1(LFA-1). In conclusion, MHC II uveal melanoma vaccines activate purified CD4+ T cells and may serve as a novel immunotherapy for uveal melanoma patients.
Collapse
Affiliation(s)
- Julia M Kittler
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Jonas Sommer
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Anika Fischer
- Department of Internal Medicine 1 - Gastroenterology, Pneumology and Endocrinology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Sabine Britting
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Margarete M Karg
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Barbara Bock
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Imke Atreya
- Department of Internal Medicine 1 - Gastroenterology, Pneumology and Endocrinology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Ludwig M Heindl
- Department of Ophthalmology and Center for Integrated Oncology (CIO) Cologne-Bonn, University of Cologne, Cologne, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Jacobus J Bosch
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| |
Collapse
|
9
|
Maccalli C, Rasul KI, Elawad M, Ferrone S. The role of cancer stem cells in the modulation of anti-tumor immune responses. Semin Cancer Biol 2018; 53:189-200. [DOI: 10.1016/j.semcancer.2018.09.006] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/12/2018] [Accepted: 09/17/2018] [Indexed: 02/07/2023]
|
10
|
Ravindranathan S, Nguyen KG, Kurtz SL, Frazier HN, Smith SG, Koppolu BP, Rajaram N, Zaharoff DA. Tumor-derived granulocyte colony-stimulating factor diminishes efficacy of breast tumor cell vaccines. Breast Cancer Res 2018; 20:126. [PMID: 30348199 PMCID: PMC6198508 DOI: 10.1186/s13058-018-1054-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 09/25/2018] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Although metastasis is ultimately responsible for about 90% of breast cancer mortality, the vast majority of breast-cancer-related deaths are due to progressive recurrences from non-metastatic disease. Current adjuvant therapies are unable to prevent progressive recurrences for a significant fraction of patients with breast cancer. Autologous tumor cell vaccines (ATCVs) are a safe and potentially useful strategy to prevent breast cancer recurrence, in a personalized and patient-specific manner, following standard-of-care tumor resection. Given the high intra-patient and inter-patient heterogeneity in breast cancer, it is important to understand which factors influence the immunogenicity of breast tumor cells in order to maximize ATCV effectiveness. METHODS The relative immunogenicity of two murine breast carcinomas, 4T1 and EMT6, were compared in a prophylactic vaccination-tumor challenge model. Differences in cell surface expression of antigen-presentation-related and costimulatory molecules were compared along with immunosuppressive cytokine production. CRISPR/Cas9 technology was used to modulate tumor-derived cytokine secretion. The impacts of cytokine deletion on splenomegaly, myeloid-derived suppressor cell (MDSC) accumulation and ATCV immunogenicity were assessed. RESULTS Mice vaccinated with an EMT6 vaccine exhibited significantly greater protective immunity than mice vaccinated with a 4T1 vaccine. Hybrid vaccination studies revealed that the 4T1 vaccination induced both local and systemic immune impairments. Although there were significant differences between EMT6 and 4T1 in the expression of costimulatory molecules, major disparities in the secretion of immunosuppressive cytokines likely accounts for differences in immunogenicity between the cell lines. Ablation of one cytokine in particular, granulocyte-colony stimulating factor (G-CSF), reversed MDSC accumulation and splenomegaly in the 4T1 model. Furthermore, G-CSF inhibition enhanced the immunogenicity of a 4T1-based vaccine to the extent that all vaccinated mice developed complete protective immunity. CONCLUSIONS Breast cancer cells that express high levels of G-CSF have the potential to diminish or abrogate the efficacy of breast cancer ATCVs. Fortunately, this study demonstrates that genetic ablation of immunosuppressive cytokines, such as G-CSF, can enhance the immunogenicity of breast cancer cell-based vaccines. Strategies that combine inhibition of immunosuppressive factors with immune stimulatory co-formulations already under development may help ATCVs reach their full potential.
Collapse
Affiliation(s)
| | - Khue G Nguyen
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, USA.,Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Samantha L Kurtz
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Haven N Frazier
- Honors College, University of Arkansas, Fayetteville, AR, USA
| | - Sean G Smith
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA.,Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC and North Carolina State University, Raleigh, NC, USA
| | - Bhanu Prasanth Koppolu
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA.,Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC and North Carolina State University, Raleigh, NC, USA
| | - Narasimhan Rajaram
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - David A Zaharoff
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA. .,Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, USA. .,Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA. .,Honors College, University of Arkansas, Fayetteville, AR, USA. .,Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC and North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
11
|
Addressing cancer immunotherapy research in Iran: adoptive cell therapy on the horizon. Cytotherapy 2018; 20:1227-1237. [DOI: 10.1016/j.jcyt.2018.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/04/2018] [Accepted: 08/14/2018] [Indexed: 12/15/2022]
|
12
|
Pilla L, Ferrone S, Maccalli C. Methods for improving the immunogenicity and efficacy of cancer vaccines. Expert Opin Biol Ther 2018; 18:765-784. [PMID: 29874943 PMCID: PMC8670419 DOI: 10.1080/14712598.2018.1485649] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/04/2018] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Cancer vaccines represent one of the oldest immunotherapy strategies. A variety of tumor-associated antigens have been exploited to investigate their immunogenicity as well as multiple strategies for vaccine administration. These efforts have led to the development of several clinical trials in tumors with different histological origins to test the clinical efficacy of cancer vaccines. However, suboptimal clinical results have been reported mainly due to the lack of optimized strategies to induce strong and sustained systemic tumor antigen-specific immune responses. AREAS COVERED We provide an overview of different types of cancer vaccines that have been developed and used in the context of clinical studies. Moreover, we review different preclinical and clinical strategies pursued to enhance the immunogenicity, stability, and targeting at tumor site of cancer vaccines. EXPERT OPINION Additional and appropriate preclinical studies are warranted to optimize the immunogenicity and delivery of cancer vaccines. The appropriate choice of target antigens is challenging; however, the exploitation of neoantigens generated from somatic mutations of tumor cells represents a promising approach to target highly immunogenic tumor-specific antigens. Remarkably, the investigation of the combination of cancer vaccines with immunomodulating agents able to skew the tumor microenvironment from immunosuppressive to immunostimulating will dramatically improve their clinical efficacy.
Collapse
Affiliation(s)
- Lorenzo Pilla
- Medical Oncology Unit, San Gerardo Hospital, Monza, Italy
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina Maccalli
- Clinical Research Center, Division of Translational Medicine, Sidra Medicine, Doha, Qatar
| |
Collapse
|
13
|
Volz B, Schmidt M, Heinrich K, Kapp K, Schroff M, Wittig B. Design and characterization of the tumor vaccine MGN1601, allogeneic fourfold gene-modified vaccine cells combined with a TLR-9 agonist. Mol Ther Oncolytics 2016; 3:15023. [PMID: 27119114 PMCID: PMC4824560 DOI: 10.1038/mto.2015.23] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/27/2015] [Accepted: 12/01/2015] [Indexed: 11/20/2022] Open
Abstract
The tumor vaccine MGN1601 was designed and developed for treatment of metastatic renal cell carcinoma (mRCC). MGN1601 consists of a combination of fourfold gene-modified cells with the toll-like receptor 9 agonist dSLIM, a powerful connector of innate and adaptive immunity. Vaccine cells originate from a renal cell carcinoma cell line (grown from renal cell carcinoma tissue), express a variety of known tumor-associated antigens (TAA), and are gene modified to transiently express two co-stimulatory molecules, CD80 and CD154, and two cytokines, GM-CSF and IL-7, aimed to support immune response. Proof of concept of the designed vaccine was shown in mice: The murine homologue of the vaccine efficiently (100%) prevented tumor growth when used as prophylactic vaccine in a syngeneic setting. Use of the vaccine in a therapeutic setting showed complete response in 92% of mice as well as synergistic action and necessity of the components. In addition, specific cellular and humoral immune responses in mice were found when used in an allogeneic setting. Immune response to the vaccine was also shown in mRCC patients treated with MGN1601: Peptide array analysis revealed humoral CD4-based immune response to TAA expressed on vaccine cells, including survivin, cyclin D1, and stromelysin.
Collapse
Affiliation(s)
- Barbara Volz
- Foundation Institute for Molecular Biology and Bioinformatics, Freie Universitaet Berlin, Berlin, Germany
- Mologen AG, Berlin, Germany
| | | | - Kerstin Heinrich
- Foundation Institute for Molecular Biology and Bioinformatics, Freie Universitaet Berlin, Berlin, Germany
- Mologen AG, Berlin, Germany
| | | | | | - Burghardt Wittig
- Foundation Institute for Molecular Biology and Bioinformatics, Freie Universitaet Berlin, Berlin, Germany
| |
Collapse
|
14
|
Seledtsov VI, Goncharov AG, Seledtsova GV. Clinically feasible approaches to potentiating cancer cell-based immunotherapies. Hum Vaccin Immunother 2016; 11:851-69. [PMID: 25933181 DOI: 10.1080/21645515.2015.1009814] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The immune system exerts both tumor-destructive and tumor-protective functions. Mature dendritic cells (DCs), classically activated macrophages (M1), granulocytes, B lymphocytes, aβ and ɣδ T lymphocytes, natural killer T (NKT) cells, and natural killer (NK) cells may be implicated in antitumor immunoprotection. Conversely, tolerogenic DCs, alternatively activated macrophages (M2), myeloid-derived suppressor cells (MDSCs), and regulatory T (Tregs) and B cells (Bregs) are capable of suppressing antitumor immune responses. Anti-cancer vaccination is a useful strategy to elicit antitumor immune responses, while overcoming immunosuppressive mechanisms. Whole tumor cells or lysates derived thereof hold more promise as cancer vaccines than individual tumor-associated antigens (TAAs), because vaccinal cells can elicit immune responses to multiple TAAs. Cancer cell-based vaccines can be autologous, allogeneic or xenogeneic. Clinical use of xenogeneic vaccines is advantageous in that they can be most effective in breaking the preexisting immune tolerance to TAAs. To potentiate immunotherapy, vaccinations can be combined with other modalities that target different immune pathways. These modalities include 1) genetic or chemical modification of cell-based vaccines; 2) cross-priming TAAs to T cells by engaging dendritic cells; 3) T-cell adoptive therapy; 4) stimulation of cytotoxic inflammation by non-specific immunomodulators, toll-like receptor (TLR) agonists, cytokines, chemokines or hormones; 5) reduction of immunosuppression and/or stimulation of antitumor effector cells using antibodies, small molecules; and 6) various cytoreductive modalities. The authors envisage that combined immunotherapeutic strategies will allow for substantial improvements in clinical outcomes in the near future.
Collapse
Key Words
- ADCC, antibody-dependent cell cytotoxicity
- APC, antigen-presenting cell
- Ab, antibodies
- BCG, Bacillus Calmette-Guérin
- Breg, regulatory B cell
- CAR, chimeric antigen receptor
- COX, cyclooxygenase
- CTA, cancer/testis antigen
- CTL, cytotoxic T lymphocyte
- CTLA-4, cytotoxic T lymphocyte antigen-4
- DC, dendritic cell
- DTH, delayed-type hypersensitivity
- GITR, glucocorticoid-induced tumor necrosis factor receptor
- GM-CSF, granulocyte-macrophage colony stimulating factor
- HIFU, high-intensity focused ultrasound
- IDO, indoleamine-2, 3-dioxygenase
- IFN, interferon
- IL, interleukin
- LAK, lymphokine-activated killer
- M, macrophage
- M1, classically activated macrophage
- M2, alternatively activated macrophage, MDSC, myeloid-derived suppressor cell
- MHC, major histocompatibility complex
- NK, natural killer (cell)
- PD-1, programmed death-1
- PGE2, prostaglandin E2
- RFA, radiofrequency ablation
- RNS, reactive nitrogen species
- ROS
- TAA, tumor-associated antigen
- TGF, transforming growth factor
- TLR, toll-like receptor
- TNF, tumor necrosis factor
- Th, T-helper cell
- Treg, regulatory T cell
- VEGF, vascular endothelial growth factor
- antitumor immunoprotection
- cancer cell-based vaccines
- combined immunotherapy
- immunosuppression
- reactive oxygen species
Collapse
Affiliation(s)
- V I Seledtsov
- a lmmanuel Kant Baltic Federal University ; Kaliningrad , Russia
| | | | | |
Collapse
|
15
|
Parmiani G, Russo V, Maccalli C, Parolini D, Rizzo N, Maio M. Peptide-based vaccines for cancer therapy. Hum Vaccin Immunother 2015; 10:3175-8. [PMID: 25483658 DOI: 10.4161/hv.29418] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Interest for cancer vaccination started more than 30 years ago after the demonstration that both in animal models and later on in patients it is possible to generate anti-tumor immune responses. The clinical application of this knowledge, however, was disappointing. In this review we summarize results on peptides epitopes recognized by T cells that have been studied thanks to their easy synthesis and the lack of significant side effects when administered in-vivo. To improve the clinical efficacy, peptides were modified in their aminoacid sequence to augment their immunogenicity. Peptides vaccines were recently shown to induce a high frequency of immune response in patients that were accompanied by clinical efficacy. These data are discussed at the light of recent progression of immunotherapy caused by the addition of check-point antibodies thus providing a general picture of the potential therapeutic efficacy of the peptide-based vaccines and their combination with other biological agents.
Collapse
|
16
|
Kazemi T, Younesi V, Jadidi-Niaragh F, Yousefi M. Immunotherapeutic approaches for cancer therapy: An updated review. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 44:769-79. [PMID: 25801036 DOI: 10.3109/21691401.2015.1019669] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In spite of specific immune effector mechanisms raised against tumor cells, there are mechanisms employed by the tumor cells to keep them away from immune recognition and elimination; some of these mechanisms have been identified, while others are still poorly understood. Manipulation or augmentation of specific antitumor immune responses are now the preferred approaches for treatment of malignancies, and traditional therapeutic approaches are being replaced by the use of agents which potentiate immune effector mechanisms, broadly called "immunotherapy". Cancer immunotherapy is generally classified into two main classes including active and passive methods. Interventions to augment the immune system of the patient, for example, vaccination or adjuvant therapy, actively promote antitumor effector mechanisms to improve cancer elimination. On the other hand, administration of specific monoclonal antibodies (mAbs) against different tumor antigens and adoptive transfer of genetically-modified specific T cells are currently the most rapidly developing approaches for cancer targeted therapy. In this review, we will discuss the different modalities for active and passive immunotherapy for cancer.
Collapse
Affiliation(s)
- Tohid Kazemi
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Vahid Younesi
- c Department of Immunology , School of Public Health, Tehran University of Medical Sciences , Tehran , Iran
| | - Farhad Jadidi-Niaragh
- c Department of Immunology , School of Public Health, Tehran University of Medical Sciences , Tehran , Iran
| | - Mehdi Yousefi
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
17
|
Bukhtoyarov OV, Samarin DM. Pathogenesis of Cancer: Cancer Reparative Trap. JOURNAL OF CANCER THERAPY 2015; 06:399-412. [DOI: 10.4236/jct.2015.65043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
18
|
Abstract
Despite major advances in the treatment of ovarian cancer over the past two decades, it is still an incurable disease and requires the development of better treatment strategies. In recent years, we have developed a greater understanding of tumor immunology and the interactions between tumors and the immune system. This has led to the emergence of cancer immunotherapy as the fourth treatment modality in cancer. In this article, we address the principles of immunotherapy and different approaches that have been investigated over the past decade and discuss the future of immune therapy in ovarian cancer.
Collapse
Affiliation(s)
- Yousef Zakharia
- a Georgia Regents University Cancer Center, Augusta, Georgia; and
| | | | | |
Collapse
|
19
|
Mackiewicz A, Mackiewicz J, Wysocki PJ, Wiznerowicz M, Kapcinska M, Laciak M, Rose-John S, Izycki D, Burzykowski T, Karczewska-Dzionk A. Long-term survival of high-risk melanoma patients immunized with a Hyper-IL-6-modified allogeneic whole-cell vaccine after complete resection. Expert Opin Investig Drugs 2012; 21:773-83. [DOI: 10.1517/13543784.2012.684753] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
20
|
Abstract
Attempts to generate an anticancer immune response in vivo in patients with cancer have taken several forms. Although to date there have been relatively few published studies describing the effects of the approach in hematologic malignancy, that circumstance is expected to change rapidly during the next few years. In solid tumors, it is not known which, if any, of the approaches being explored will be able to produce responses of sufficient effectiveness and duration to be of general clinical value. Despite the documented increase in survival of patients developing an immune response to tumor immunization, no randomized clinical trial has been entirely convincing. As knowledge of the molecular basis of the immune response and of the immune defenses used by cancer cells improves, it is reasonable to expect to see increasing benefits from tumor vaccines, which are likely to complement, long before they replace, conventional therapies.
Collapse
Affiliation(s)
- Peter J. DeMaria
- Genitourinary Malignancies Branch, Center for Cancer Research, National
Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marijo Bilusic
- Genitourinary Malignancies Branch, Center for Cancer Research, National
Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|