1
|
Liu J, Cui Y, Cabral H, Tong A, Yue Q, Zhao L, Sun X, Mi P. Glucosylated Nanovaccines for Dendritic Cell-Targeted Antigen Delivery and Amplified Cancer Immunotherapy. ACS NANO 2024; 18:25826-25840. [PMID: 39196858 DOI: 10.1021/acsnano.4c09053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
Engineering nanovaccines capable of targeting dendritic cells (DCs) is desperately required to maximize antigen cross-presentation to effector immune cells, elicit strong immune responses, and avoid adverse reactions. Here, we showed that glucose transporter 1 (Glut-1) on DCs is a reliable target for delivering antigens to DCs, and thus, a versatile antigen delivery strategy using glucosylated nanovaccines was developed for DC-targeted antigen delivery and tumor immunotherapy. The developed glucosylated ovalbumin-loaded nanovaccines highly accumulated in lymph nodes and efficiently engaged with Glut-1 on DCs to accelerate intracellular antigen delivery and promote DC maturation and antigen presentation, which elicited potent antitumor immunity to prevent and inhibit ovalbumin-expressing melanoma. Moreover, immunotherapeutic experiments in DC- and macrophage-depleted animal models confirmed that the glucosylated nanovaccines functioned mainly through DCs. In addition, the neoantigen-delivering glucosylated nanovaccines were further engineered to elicit tumor-specific immune responses against MC38 tumors. This study offers a DC-targeted antigen delivery strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiology, State Key Laboratory of Biotherapy and Huaxi MR Research Center (HMRRC), West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yongsheng Cui
- Department of Radiology, State Key Laboratory of Biotherapy and Huaxi MR Research Center (HMRRC), West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Aiping Tong
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Yue
- Department of Radiology, State Key Laboratory of Biotherapy and Huaxi MR Research Center (HMRRC), West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lihong Zhao
- Department of Radiology, State Key Laboratory of Biotherapy and Huaxi MR Research Center (HMRRC), West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xun Sun
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Peng Mi
- Department of Radiology, State Key Laboratory of Biotherapy and Huaxi MR Research Center (HMRRC), West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
2
|
Liu D, Liu L, Li X, Wang S, Wu G, Che X. Advancements and Challenges in Peptide-Based Cancer Vaccination: A Multidisciplinary Perspective. Vaccines (Basel) 2024; 12:950. [PMID: 39204073 PMCID: PMC11359700 DOI: 10.3390/vaccines12080950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/09/2024] [Accepted: 08/21/2024] [Indexed: 09/03/2024] Open
Abstract
With the continuous advancements in tumor immunotherapy, researchers are actively exploring new treatment methods. Peptide therapeutic cancer vaccines have garnered significant attention for their potential in improving patient outcomes. Despite its potential, only a single peptide-based cancer vaccine has been approved by the U.S. Food and Drug Administration (FDA). A comprehensive understanding of the underlying mechanisms and current development status is crucial for advancing these vaccines. This review provides an in-depth analysis of the production principles and therapeutic mechanisms of peptide-based cancer vaccines, highlights the commonly used peptide-based cancer vaccines, and examines the synergistic effects of combining these vaccines with immunotherapy, targeted therapy, radiotherapy, and chemotherapy. While some studies have yielded suboptimal results, the potential of combination therapies remains substantial. Additionally, we addressed the management and adverse events associated with peptide-based cancer vaccines, noting their relatively higher safety profile compared to traditional radiotherapy and chemotherapy. Lastly, we also discussed the roles of adjuvants and targeted delivery systems in enhancing vaccine efficacy. In conclusion, this review comprehensively outlines the current landscape of peptide-based cancer vaccination and underscores its potential as a pivotal immunotherapy approach.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (L.L.); (S.W.)
| | - Lei Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (L.L.); (S.W.)
| | - Xinghan Li
- Department of Stomatology, General Hospital of Northern Theater Command, Shenyang 110016, China;
| | - Shijin Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (L.L.); (S.W.)
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (L.L.); (S.W.)
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (L.L.); (S.W.)
| |
Collapse
|
3
|
Kono M, Wakisaka R, Komatsuda H, Hayashi R, Kumai T, Yamaki H, Sato R, Nagato T, Ohkuri T, Kosaka A, Ohara K, Kishibe K, Kobayashi H, Hayashi T, Takahara M. Immunotherapy targeting tumor-associated antigen in a mouse model of head and neck cancer. Head Neck 2024; 46:2056-2067. [PMID: 38390628 DOI: 10.1002/hed.27703] [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: 10/19/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND The identification of epitope peptides from tumor-associated antigens (TAAs) is informative for developing tumor-specific immunotherapy. However, only a few epitopes have been detected in mouse TAAs of head and neck cancer (HNSCC). METHODS Novel mouse c-Met-derived T-cell epitopes were predicted by computer-based algorithms. Mouse HNSCC cell line-bearing mice were treated with a c-Met peptide vaccine. The effects of CD8 and/or CD4 T-cell depletion, and vaccine combination with immune checkpoint inhibitors (ICIs) were evaluated. Tumor re-inoculation was performed to assess T-cell memory. RESULTS We identified c-Met-derived short and long epitopes that elicited c-Met-reactive antitumor CD8 and/or CD4 T-cell responses. Vaccination using these peptides showed remarkable antitumor responses via T cells in which ICIs were not required. The c-Met peptide-vaccinated mice rejected the re-inoculated tumors. CONCLUSIONS We demonstrated that novel c-Met peptide vaccines can induce antitumor T-cell response, and could be a potent immunotherapy in a syngeneic mouse HNSCC model.
Collapse
Affiliation(s)
- Michihisa Kono
- Department of Otolaryngology-Head & Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Risa Wakisaka
- Department of Otolaryngology-Head & Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Hiroki Komatsuda
- Department of Otolaryngology-Head & Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Ryusuke Hayashi
- Department of Otolaryngology-Head & Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Takumi Kumai
- Department of Otolaryngology-Head & Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
- Department of Innovative Head & Neck Cancer Research and Treatment, Asahikawa Medical University, Asahikawa, Japan
| | - Hidekiyo Yamaki
- Department of Otolaryngology-Head & Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Ryosuke Sato
- Department of Otolaryngology-Head & Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Toshihiro Nagato
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Takayuki Ohkuri
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Akemi Kosaka
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Kenzo Ohara
- Department of Otolaryngology-Head & Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Kan Kishibe
- Department of Otolaryngology-Head & Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Hiroya Kobayashi
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Tatsuya Hayashi
- Department of Otolaryngology-Head & Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Miki Takahara
- Department of Otolaryngology-Head & Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
- Department of Innovative Head & Neck Cancer Research and Treatment, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
4
|
Butterfield LH, Najjar YG. Immunotherapy combination approaches: mechanisms, biomarkers and clinical observations. Nat Rev Immunol 2024; 24:399-416. [PMID: 38057451 PMCID: PMC11460566 DOI: 10.1038/s41577-023-00973-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2023] [Indexed: 12/08/2023]
Abstract
The approval of the first immune checkpoint inhibitors provided a paradigm shift for the treatment of malignancies across a broad range of indications. Whereas initially, single-agent immune checkpoint inhibition was used, increasing numbers of patients are now treated with combination immune checkpoint blockade, where non-redundant mechanisms of action of the individual agents generally lead to higher response rates. Furthermore, immune checkpoint therapy has been combined with various other therapeutic modalities, including chemotherapy, radiotherapy and other immunotherapeutics such as vaccines, adoptive cellular therapies, cytokines and others, in an effort to maximize clinical efficacy. Currently, a large number of clinical trials test combination therapies with an immune checkpoint inhibitor as a backbone. However, proceeding without inclusion of broad, if initially exploratory, biomarker investigations may ultimately slow progress, as so far, few combinations have yielded clinical successes based on clinical data alone. Here, we present the rationale for combination therapies and discuss clinical data from clinical trials across the immuno-oncology spectrum. Moreover, we discuss the evolution of biomarker approaches and highlight the potential new directions that comprehensive biomarker studies can yield.
Collapse
Affiliation(s)
- Lisa H Butterfield
- University of California San Francisco, Microbiology and Immunology, San Francisco, CA, USA.
| | | |
Collapse
|
5
|
Xing H, Li X. Engineered Nanomaterials for Tumor Immune Microenvironment Modulation in Cancer Immunotherapy. Chemistry 2024:e202400425. [PMID: 38576219 DOI: 10.1002/chem.202400425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 04/06/2024]
Abstract
Tumor immunotherapy, represented by immune checkpoint blocking and chimeric antigen receptor (CAR) T cell therapy, has achieved promising results in clinical applications. However, it faces challenges that hinder its further development, such as limited response rates and poor tumor permeability. The efficiency of tumor immunotherapy is also closely linked to the structure and function of the immune microenvironment where the tumor resides. Recently, nanoparticle-based tumor immune microenvironment (TIME) modulation strategies have attracted a great deal of attention in cancer immunotherapy. This is primarily due to the distinctive physical characteristics of nanoparticles, which enable them to effectively infiltrate the TIME and selectively modulate its key constituents. This paper reviews recent advances in nanoparticle engineering to improve anti-cancer immunotherapy. Emerging nanoparticle-based approaches for modulating immune cells, tumor stroma, cytokines and immune checkpoints are discussed, aiming to overcome current challenges in the clinic. In addition, integrating immunotherapy with various treatment modalities such as chemotherapy and photodynamic therapy can be facilitated through the utilization of nanoparticles, thereby enhancing the efficacy of cancer treatment. The future challenges and opportunities of using nanomaterials to reeducate the suppressive immune microenvironment of tumors are also discussed, with the aim of anticipating further advancements in this growing field.
Collapse
Affiliation(s)
- Hao Xing
- Department of General Surgery, Naval Medical Center, Naval Medical University, 200052, Shanghai, China
- The First Affiliated Hospital of Naval Medical University, 200433, Shanghai, China
| | - Xiaomin Li
- Department of Chemistry, Laboratory of Advanced Materials, College of Chemistry and Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, 200438, Shanghai, China
| |
Collapse
|
6
|
Stern LJ, Clement C, Galluzzi L, Santambrogio L. Non-mutational neoantigens in disease. Nat Immunol 2024; 25:29-40. [PMID: 38168954 PMCID: PMC11075006 DOI: 10.1038/s41590-023-01664-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/29/2023] [Indexed: 01/05/2024]
Abstract
The ability of mammals to mount adaptive immune responses culminating with the establishment of immunological memory is predicated on the ability of the mature T cell repertoire to recognize antigenic peptides presented by syngeneic MHC class I and II molecules. Although it is widely believed that mature T cells are highly skewed towards the recognition of antigenic peptides originating from genetically diverse (for example, foreign or mutated) protein-coding regions, preclinical and clinical data rather demonstrate that novel antigenic determinants efficiently recognized by mature T cells can emerge from a variety of non-mutational mechanisms. In this Review, we describe various mechanisms that underlie the formation of bona fide non-mutational neoantigens, such as epitope mimicry, upregulation of cryptic epitopes, usage of non-canonical initiation codons, alternative RNA splicing, and defective ribosomal RNA processing, as well as both enzymatic and non-enzymatic post-translational protein modifications. Moreover, we discuss the implications of the immune recognition of non-mutational neoantigens for human disease.
Collapse
Affiliation(s)
- Lawrence J Stern
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbiology Program, UMass Chan Medical School, Worcester, MA, USA
| | - Cristina Clement
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Yi M, Li T, Niu M, Mei Q, Zhao B, Chu Q, Dai Z, Wu K. Exploiting innate immunity for cancer immunotherapy. Mol Cancer 2023; 22:187. [PMID: 38008741 PMCID: PMC10680233 DOI: 10.1186/s12943-023-01885-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/23/2023] [Indexed: 11/28/2023] Open
Abstract
Immunotherapies have revolutionized the treatment paradigms of various types of cancers. However, most of these immunomodulatory strategies focus on harnessing adaptive immunity, mainly by inhibiting immunosuppressive signaling with immune checkpoint blockade, or enhancing immunostimulatory signaling with bispecific T cell engager and chimeric antigen receptor (CAR)-T cell. Although these agents have already achieved great success, only a tiny percentage of patients could benefit from immunotherapies. Actually, immunotherapy efficacy is determined by multiple components in the tumor microenvironment beyond adaptive immunity. Cells from the innate arm of the immune system, such as macrophages, dendritic cells, myeloid-derived suppressor cells, neutrophils, natural killer cells, and unconventional T cells, also participate in cancer immune evasion and surveillance. Considering that the innate arm is the cornerstone of the antitumor immune response, utilizing innate immunity provides potential therapeutic options for cancer control. Up to now, strategies exploiting innate immunity, such as agonists of stimulator of interferon genes, CAR-macrophage or -natural killer cell therapies, metabolic regulators, and novel immune checkpoint blockade, have exhibited potent antitumor activities in preclinical and clinical studies. Here, we summarize the latest insights into the potential roles of innate cells in antitumor immunity and discuss the advances in innate arm-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ming Yi
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Qi Mei
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China
| | - Bin Zhao
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China.
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
8
|
Yang W, Pan X, Zhang P, Yang X, Guan H, Dou H, Lu Q. Defeating Melanoma Through a Nano-Enabled Revision of Hypoxic and Immunosuppressive Tumor Microenvironment. Int J Nanomedicine 2023; 18:3711-3725. [PMID: 37435153 PMCID: PMC10332423 DOI: 10.2147/ijn.s414882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/19/2023] [Indexed: 07/13/2023] Open
Abstract
Rationale Reversing the hypoxic and immunosuppressive tumor microenvironment (TME) is crucial for treating malignant melanoma. Seeking a robust platform for the effective reversion of hypoxic and immunosuppressive TME may be an excellent solution to revolutionizing the current landscape of malignant melanoma treatment. Here, we demonstrated a transdermal and intravenous dual-administration paradigm. A tailor-made Ato/cabo@PEG-TK-PLGA NPs were administrated transdermally to melanoma with the help of a gel spray containing a skin-penetrating material borneol. Nanoparticles encased Ato and cabo were released and thereby reversed the hypoxic and immunosuppressive tumor microenvironment (TME). Methods Ato/cabo@PEG-TK-PLGA NPs were synthesized through a self-assembly emulsion process, and the transdermal ability was assessed using Franz diffusion cell assembly. The inhibition effect on cell respiration was measured by OCR, ATP, and pO2 detection and in vivo photoacoustic (PA) imaging. The reversing of the immunosuppressive was detected through flow cytometry analysis of MDSCs and T cells. At last, the in vivo anti-tumor efficacy and histopathology, immunohistochemical analysis and safety detection were performed using tumor-bearing mice. Results The transdermally administrated Ato/cabo@PEG-TK-PLGA NPs successfully spread to the skin surface of melanoma and then entered deep inside the tumor with the help of a gel spray and a skin puncturing material borneol. Atovaquone (Ato, a mitochondrial-respiration inhibitor) and cabozantinib (cabo, a MDSCs eliminator) were concurrently released in response to the intratumorally overexpressed H2O2. The released Ato and cabo respectively reversed the hypoxic and immunosuppressive TME. The reversed hypoxic TME offered sufficient O2 for the intravenously administrated indocyanine green (ICG, an FDA-approved photosensitizer) to produce adequate amount of ROS. In contrast, the reversed immunosuppressive TME conferred amplified systemic immune responses. Conclusion Taken together, we developed a transdermal and intravenous dual-administration paradigm, which effectively reversed the hypoxic and immunosuppressive tumor microenvironment in the treatment of the malignant melanoma. We believe our study will open a new path for the effective elimination of the primary tumors and the real-time control of tumor metastasis.
Collapse
Affiliation(s)
- Wenzhe Yang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong Province, People’s Republic of China
- Marine Traditional Chinese Medicine R&D Laboratory, Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong Province, People’s Republic of China
| | - Xue Pan
- Marine Traditional Chinese Medicine R&D Laboratory, Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong Province, People’s Republic of China
| | - Peng Zhang
- Marine Traditional Chinese Medicine R&D Laboratory, Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong Province, People’s Republic of China
| | - Xue Yang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong Province, People’s Republic of China
- Marine Traditional Chinese Medicine R&D Laboratory, Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong Province, People’s Republic of China
| | - Huashi Guan
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong Province, People’s Republic of China
- Marine Traditional Chinese Medicine R&D Laboratory, Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong Province, People’s Republic of China
| | - Huan Dou
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu Province, People’s Republic of China
| | - Qian Lu
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong Province, People’s Republic of China
- Marine Traditional Chinese Medicine R&D Laboratory, Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong Province, People’s Republic of China
| |
Collapse
|
9
|
Dillman RO, Nistor GI, Keirstead HS. Autologous dendritic cells loaded with antigens from self-renewing autologous tumor cells as patient-specific therapeutic cancer vaccines. Hum Vaccin Immunother 2023:2198467. [PMID: 37133853 DOI: 10.1080/21645515.2023.2198467] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023] Open
Abstract
A promising personal immunotherapy is autologous dendritic cells (DC) loaded ex vivo with autologous tumor antigens (ATA) derived from self-renewing autologous cancer cells. DC-ATA are suspended in granulocyte-macrophage colony stimulating factor at the time of each subcutaneous injection. Previously, irradiated autologous tumor cell vaccines have produced encouraging results in 150 cancer patients, but the DC-ATA vaccine demonstrated superiority in single-arm and randomized trials in metastatic melanoma. DC-ATA have been injected into more than 200 patients with melanoma, glioblastoma, and ovarian, hepatocellular, and renal cell cancers. Key observations include: [1] greater than 95% success rates for tumor cell cultures and monocyte collection for dendritic cell production; [2] injections are well-tolerated; [3] the immune response is rapid and includes primarily TH1/TH17 cellular responses; [4] efficacy has been suggested by delayed but durable complete tumor regressions in patients with measurable disease, by progression-free survival in glioblastoma, and by overall survival in melanoma.
Collapse
Affiliation(s)
| | - Gabriel I Nistor
- Research and Development, AIVITA Biomedical Inc, Irvine, CA, USA
| | | |
Collapse
|
10
|
Ren Y, Bäcker H, Müller M, Kienzle A. The role of myeloid derived suppressor cells in musculoskeletal disorders. Front Immunol 2023; 14:1139683. [PMID: 36936946 PMCID: PMC10020351 DOI: 10.3389/fimmu.2023.1139683] [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: 01/07/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The immune system is closely linked to bone homeostasis and plays a pivotal role in several pathological and inflammatory conditions. Through various pathways it modulates various bone cells and subsequently sustains the physiological bone metabolism. Myeloid-derived suppressor cells (MDSCs) are a group of heterogeneous immature myeloid-derived cells that can exert an immunosuppressive function through a direct cell-to-cell contact, secretion of anti-inflammatory cytokines or specific exosomes. These cells mediate the innate immune response to chronic stress on the skeletal system. In chronic inflammation, MDSCs act as an inner offset to rebalance overactivation of the immune system. Moreover, they have been found to be involved in processes responsible for bone remodeling in different musculoskeletal disorders, autoimmune diseases, infection, and cancer. These cells can not only cause bone erosion by differentiating into osteoclasts, but also alleviate the immune reaction, subsequently leading to long-lastingly impacted bone remodeling. In this review, we discuss the impact of MDSCs on the bone metabolism under several pathological conditions, the involved modulatory pathways as well as potential therapeutic targets in MDSCs to improve bone health.
Collapse
Affiliation(s)
- Yi Ren
- Center for Musculoskeletal Surgery, Clinic for Orthopedics, Charité University Hospital, Berlin, Germany
| | - Henrik Bäcker
- Department of Orthopedics, Auckland City Hospital, Auckland, New Zealand
| | - Michael Müller
- Center for Musculoskeletal Surgery, Clinic for Orthopedics, Charité University Hospital, Berlin, Germany
| | - Arne Kienzle
- Center for Musculoskeletal Surgery, Clinic for Orthopedics, Charité University Hospital, Berlin, Germany
- BIH Charité Clinician Scientist Program, BIH Biomedical Innovation Academy, Berlin Institute of Health, Charité — Universitätsmedizin Berlin, Berlin, Germany
- *Correspondence: Arne Kienzle,
| |
Collapse
|
11
|
Wu Y, Yi M, Niu M, Mei Q, Wu K. Myeloid-derived suppressor cells: an emerging target for anticancer immunotherapy. Mol Cancer 2022; 21:184. [PMID: 36163047 PMCID: PMC9513992 DOI: 10.1186/s12943-022-01657-y] [Citation(s) in RCA: 132] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/19/2022] [Indexed: 02/07/2023] Open
Abstract
The clinical responses observed following treatment with immune checkpoint inhibitors (ICIs) support immunotherapy as a potential anticancer treatment. However, a large proportion of patients cannot benefit from it due to resistance or relapse, which is most likely attributable to the multiple immunosuppressive cells in the tumor microenvironment (TME). Myeloid-derived suppressor cells (MDSCs), a heterogeneous array of pathologically activated immature cells, are a chief component of immunosuppressive networks. These cells potently suppress T-cell activity and thus contribute to the immune escape of malignant tumors. New findings indicate that targeting MDSCs might be an alternative and promising target for immunotherapy, reshaping the immunosuppressive microenvironment and enhancing the efficacy of cancer immunotherapy. In this review, we focus primarily on the classification and inhibitory function of MDSCs and the crosstalk between MDSCs and other myeloid cells. We also briefly summarize the latest approaches to therapies targeting MDSCs.
Collapse
Affiliation(s)
- Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Ming Yi
- Department of Breast Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, 310003, China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Qi Mei
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China. .,Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
12
|
Abstract
The clinical responses observed following treatment with immune checkpoint inhibitors (ICIs) support immunotherapy as a potential anticancer treatment. However, a large proportion of patients cannot benefit from it due to resistance or relapse, which is most likely attributable to the multiple immunosuppressive cells in the tumor microenvironment (TME). Myeloid-derived suppressor cells (MDSCs), a heterogeneous array of pathologically activated immature cells, are a chief component of immunosuppressive networks. These cells potently suppress T-cell activity and thus contribute to the immune escape of malignant tumors. New findings indicate that targeting MDSCs might be an alternative and promising target for immunotherapy, reshaping the immunosuppressive microenvironment and enhancing the efficacy of cancer immunotherapy. In this review, we focus primarily on the classification and inhibitory function of MDSCs and the crosstalk between MDSCs and other myeloid cells. We also briefly summarize the latest approaches to therapies targeting MDSCs.
Collapse
Affiliation(s)
- Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Ming Yi
- Department of Breast Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, 310003, China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Qi Mei
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
13
|
Mandula JK, Rodriguez PC. Tumor-directed dysregulation of erythroid progenitors drives immunosuppressive myeloid cells. Cancer Cell 2022; 40:597-599. [PMID: 35594864 DOI: 10.1016/j.ccell.2022.04.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
In this issue of Cancer Cell, Long et al. demonstrate that tumors can reprogram erythroid progenitors into myeloid-erythroid cells that promote immunosuppression. Erythroid-differentiated myeloid cells (EDMCs) expand in cancer-bearing individuals, resemble the functionality of myeloid-derived suppressor cells (MDSCs), and correlate with poor response to immune-checkpoint inhibitors (ICIs) and tumor-related anemia.
Collapse
Affiliation(s)
- Jessica K Mandula
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA.
| |
Collapse
|
14
|
Mortezaee K, Majidpoor J. (Im)maturity in Tumor Ecosystem. Front Oncol 2022; 11:813897. [PMID: 35145911 PMCID: PMC8821092 DOI: 10.3389/fonc.2021.813897] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/27/2021] [Indexed: 01/10/2023] Open
Abstract
Tumors have special features that make them distinct from their normal counterparts. Immature cells in a tumor mass and their critical contributions to the tumorigenesis will open new windows toward cancer therapy. Incomplete cellular development brings versatile and unique functionality in the cellular tumor ecosystem, such as what is seen for highly potential embryonic cells. There is evidence that maturation of certain types of cells in this ecosystem can recover the sensitivity of the tumor. Therefore, understanding more about the mechanisms that contributed to this immaturity will render new therapeutic approaches in cancer therapy. Targeting such mechanisms can be exploited as a supplementary to the current immunotherapeutic treatment schedules, such as immune checkpoint inhibitor (ICI) therapy. The key focus of this review is to discuss the impact of (im)maturity in cellular tumor ecosystems on cancer progression, focusing mainly on immaturity in the immune cell compartment of the tumor, as well as on the stemness of tumor cells.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
15
|
Langellotto F, Dellacherie MO, Yeager C, Ijaz H, Yu J, Cheng C, Dimitrakakis N, Seiler BT, Gebre MS, Gilboa T, Johnson R, Storm N, Bardales S, Graveline A, White D, Tringides CM, Cartwright MJ, Doherty EJ, Honko A, Griffiths A, Barouch DH, Walt DR, Mooney DJ. A Modular Biomaterial Scaffold-Based Vaccine Elicits Durable Adaptive Immunity to Subunit SARS-CoV-2 Antigens. Adv Healthc Mater 2021; 10:e2101370. [PMID: 34605223 PMCID: PMC8652677 DOI: 10.1002/adhm.202101370] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/07/2021] [Indexed: 12/14/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic demonstrates the importance of generating safe and efficacious vaccines that can be rapidly deployed against emerging pathogens. Subunit vaccines are considered among the safest, but proteins used in these typically lack strong immunogenicity, leading to poor immune responses. Here, a biomaterial COVID-19 vaccine based on a mesoporous silica rods (MSRs) platform is described. MSRs loaded with granulocyte-macrophage colony-stimulating factor (GM-CSF), the toll-like receptor 4 (TLR-4) agonist monophosphoryl lipid A (MPLA), and SARS-CoV-2 viral protein antigens slowly release their cargo and form subcutaneous scaffolds that locally recruit and activate antigen-presenting cells (APCs) for the generation of adaptive immunity. MSR-based vaccines generate robust and durable cellular and humoral responses against SARS-CoV-2 antigens, including the poorly immunogenic receptor binding domain (RBD) of the spike (S) protein. Persistent antibodies over the course of 8 months are found in all vaccine configurations tested and robust in vitro viral neutralization is observed both in a prime-boost and a single-dose regimen. These vaccines can be fully formulated ahead of time or stored lyophilized and reconstituted with an antigen mixture moments before injection, which can facilitate its rapid deployment against emerging SARS-CoV-2 variants or new pathogens. Together, the data show a promising COVID-19 vaccine candidate and a generally adaptable vaccine platform against infectious pathogens.
Collapse
Affiliation(s)
- Fernanda Langellotto
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Maxence O. Dellacherie
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
- Harvard John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMA02138USA
| | - Chyenne Yeager
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Hamza Ijaz
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Jingyou Yu
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMA02115USA
| | - Chi‐An Cheng
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
- Department of PathologyBrigham and Women's HospitalBostonMA02115USA
- Harvard Medical SchoolBostonMA02115USA
| | - Nikolaos Dimitrakakis
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Benjamin T. Seiler
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Makda S. Gebre
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMA02115USA
| | - Tal Gilboa
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
- Department of PathologyBrigham and Women's HospitalBostonMA02115USA
- Harvard Medical SchoolBostonMA02115USA
| | - Rebecca Johnson
- Department of MicrobiologyBoston University School of Medicine and National Emerging Infectious Diseases LaboratoriesBostonMA02118USA
| | - Nadia Storm
- Department of MicrobiologyBoston University School of Medicine and National Emerging Infectious Diseases LaboratoriesBostonMA02118USA
| | - Sarai Bardales
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Amanda Graveline
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Des White
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Christina M. Tringides
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
- Harvard Program in BiophysicsHarvard UniversityCambridgeMA02138USA
- Harvard–MIT Division in Health Sciences and TechnologyMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Mark J. Cartwright
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Edward J. Doherty
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Anna Honko
- Department of MicrobiologyBoston University School of Medicine and National Emerging Infectious Diseases LaboratoriesBostonMA02118USA
| | - Anthony Griffiths
- Department of MicrobiologyBoston University School of Medicine and National Emerging Infectious Diseases LaboratoriesBostonMA02118USA
| | - Dan H. Barouch
- Center for Virology and Vaccine ResearchBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMA02115USA
- Ragon Institute of MGHMIT, and HarvardCambridgeMA02139USA
- Massachusetts Consortium on Pathogen ReadinessBostonMA02215USA
| | - David R. Walt
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
- Department of PathologyBrigham and Women's HospitalBostonMA02115USA
- Harvard Medical SchoolBostonMA02115USA
| | - David J. Mooney
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
- Harvard John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMA02138USA
| |
Collapse
|
16
|
Zhu Y, Zhu X, Tang C, Guan X, Zhang W. Progress and challenges of immunotherapy in triple-negative breast cancer. Biochim Biophys Acta Rev Cancer 2021; 1876:188593. [PMID: 34280474 DOI: 10.1016/j.bbcan.2021.188593] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/26/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022]
Abstract
Triple-negative breast cancer (TNBC), a subtype of breast cancer, is defined as lacking estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2) expression. Compared with other subtypes in breast cancer, TNBC is more likely to recur and metastasize, with a lower survival rate. Due to the absence of definitive targets, there was limited novel therapeutic interventions and chemotherapy remained the primary treatment in the past decades. Following the development of immune checkpoint inhibition (ICI) in solid tumors and validation of the immunogenicity in TNBC, immunotherapy has attracted more and more attentions. On basis of accumulating clinical studies, we reviewed the current progress targeting different immune checkpoints in several-lines treatment for TNBC, including programmed death-1 (PD-1)/programmed death ligand-1 (PD-L1) inhibitors, cytotoxic T-lymphocyte associated antigen-4 (CTLA-4) inhibitor, and other novel immunotherapeutic approaches (e.g., individualized peptide vaccine, cancer-testis antigen (CTA), new antigen vaccine, RNA vaccine and chimeric antigen receptor modified T cells (CAR-T)). In order to improve the survival outcome of TNBC populations, we further discussed potential predictive biomarkers for immunotherapy (e.g., PD-L1 expression, tumor mutational burden (TMB), tumor-infiltrating lymphocytes (TILs), microsatellite instability (MSI)/mismatch repair (MMR) deficiency) and challenges in the future treatment of TNBC.
Collapse
Affiliation(s)
- Yinxing Zhu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xuedan Zhu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Cuiju Tang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China.
| | - Xiaoxiang Guan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing 211166, China.
| | - Wenwen Zhang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China.
| |
Collapse
|
17
|
Tarhini AA, Joshi I, Garner F. Sargramostim and immune checkpoint inhibitors: combinatorial therapeutic studies in metastatic melanoma. Immunotherapy 2021; 13:1011-1029. [PMID: 34157863 DOI: 10.2217/imt-2021-0119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The use of immune checkpoint inhibitors in patients with metastatic melanoma generates clinical benefit, including improved survival. Yet disease resistance and immune-related adverse events persist as unmet needs. Sargramostim, a yeast-derived recombinant human GM-CSF, has shown clinical activity against diverse solid tumors, including metastatic melanoma. Here we review the use of sargramostim for treatment of advanced melanoma. Potential sargramostim applications in melanoma draw on the unique ability of GM-CSF to link innate and adaptive immune responses. We review preclinical and translational data describing the mechanism of action of sargramostim and synergy with immune checkpoint inhibitors to enhance efficacy and reduce treatment-related toxicity.
Collapse
Affiliation(s)
- Ahmad A Tarhini
- Cutaneous Oncology & Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Ila Joshi
- Pre-Clinical & Translational Research & Development, Partner Therapeutics, 19 Muzzey Street, Lexington, MA 02421, USA
| | - Fiona Garner
- Immuno-Oncology Clinical Development & Translational Medicine, Partner Therapeutics, 19 Muzzey Street, Lexington, MA 02421, USA
| |
Collapse
|
18
|
Wang X, Yu Z, Liu W, Tang H, Yi D, Wei M. Recent progress on MHC-I epitope prediction in tumor immunotherapy. Am J Cancer Res 2021; 11:2401-2416. [PMID: 34249407 PMCID: PMC8263640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/13/2021] [Indexed: 06/13/2023] Open
Abstract
Tumor immunotherapy has now become one of the most potential therapy for those intractable cancer diseases. The antigens on the cancer cell surfaces are the keys for the immune system to recognize and eliminate them. As reported, the immunogenicity of the tumor antigens could be determined by the binding between the key epitope peptides and MHC molecules. In recent years, the approaches to anticipate the peptides from the candidate epitopes have gradually changed into more efficient methods. Including the improved conventional methods, more diverse methods were coming into view. Here we review the anticipated methods of the tumor associated epitopes that specifically bind with major histocompatibility complex (MHC) class I molecules, and the recent advances and applications of those epitope prediction methods.
Collapse
Affiliation(s)
- Xiangyi Wang
- Department of Pharmacology, School of Pharmacy, China Medical UniversityNo. 77 Puhe Road, Shenyang North New District, Shenyang, Liaoning, P. R. China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical UniversityNo. 77 Puhe Road, Shenyang North New District, Shenyang, Liaoning, P. R. China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical UniversityNo. 77 Puhe Road, Shenyang North New District, Shenyang, Liaoning, P. R. China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical UniversityNo. 77 Puhe Road, Shenyang North New District, Shenyang, Liaoning, P. R. China
| | - Wensi Liu
- Department of Pharmacology, School of Pharmacy, China Medical UniversityNo. 77 Puhe Road, Shenyang North New District, Shenyang, Liaoning, P. R. China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical UniversityNo. 77 Puhe Road, Shenyang North New District, Shenyang, Liaoning, P. R. China
| | - Haichao Tang
- Department of Pharmacology, School of Pharmacy, China Medical UniversityNo. 77 Puhe Road, Shenyang North New District, Shenyang, Liaoning, P. R. China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical UniversityNo. 77 Puhe Road, Shenyang North New District, Shenyang, Liaoning, P. R. China
| | - Dongxu Yi
- The Affiliated Reproductive Hospital of China Medical UniversityNo. 10 Puhe Street, Huanggu District Shenyang, Liaoning, P. R. China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical UniversityNo. 77 Puhe Road, Shenyang North New District, Shenyang, Liaoning, P. R. China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical UniversityNo. 77 Puhe Road, Shenyang North New District, Shenyang, Liaoning, P. R. China
| |
Collapse
|
19
|
Mortezaee K. Myeloid-derived suppressor cells in cancer immunotherapy-clinical perspectives. Life Sci 2021; 277:119627. [PMID: 34004256 DOI: 10.1016/j.lfs.2021.119627] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/19/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are heterogeneous and poorly mature cells of innate immunity that their population is increased substantially in cancer patients. MDSCs represent three subsets including CD14+ monocytic (M), CD15+ granulocytic (G) and Lin- early precursor (e) cells. MDSCs release a number of factors that direct several tumorigenic-related events including immune evasion, angiogenesis and metastasis. Assessment of MDSCs can provide valuable information from cancer immunity state, and it can be an indicator of tumor prognosis. The cells can be targeted in combination with current immunotherapeutic schedules, and the outcomes were promising. The focus of this review is to provide an overview of MDSCs, their involvement in tumor-related immunosuppression, and their impact on cancer immunotherapy. Then, strategies are proposed to boost the power of immune system against MDSCs.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
20
|
Petrina M, Martin J, Basta S. Granulocyte macrophage colony-stimulating factor has come of age: From a vaccine adjuvant to antiviral immunotherapy. Cytokine Growth Factor Rev 2021; 59:101-110. [PMID: 33593661 PMCID: PMC8064670 DOI: 10.1016/j.cytogfr.2021.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/28/2020] [Accepted: 01/04/2021] [Indexed: 12/19/2022]
Abstract
GM-CSF acts as a pro-inflammatory cytokine and a key growth factor produced by several immune cells such as macrophages and activated T cells. In this review, we discuss recent studies that point to the crucial role of GM-CSF in the immune response against infections. Upon induction, GM-CSF activates four main signalling networks including the JAK/STAT, PI3K, MAPK, and NFκB pathways. Many of these transduction pathways such as JAK/STAT signal via proteins commonly activated with other antiviral signalling cascades, such as those induced by IFNs. GM-CSF also helps defend against respiratory infections by regulating alveolar macrophage differentiation and enhancing innate immunity in the lungs. Here, we also summarize the numerous clinical trials that have taken advantage of GM-CSF's mechanistic attributes in immunotherapy. Moreover, we discuss how GM-CSF is used as an adjuvant in vaccines and how its activity is interfered with to reduce inflammation such as in the case of COVID-19. This review brings forth the current knowledge on the antiviral actions of GM-CSF, the associated signalling cascades, and its application in immunotherapy.
Collapse
Affiliation(s)
- Maria Petrina
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Jacqueline Martin
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Sameh Basta
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada.
| |
Collapse
|
21
|
Wang Y, Xiang Y, Xin VW, Wang XW, Peng XC, Liu XQ, Wang D, Li N, Cheng JT, Lyv YN, Cui SZ, Ma Z, Zhang Q, Xin HW. Dendritic cell biology and its role in tumor immunotherapy. J Hematol Oncol 2020. [PMID: 32746880 DOI: 10.1186/s13045-020-00939-6.pmid:32746880;pmcid:pmc7397618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
As crucial antigen presenting cells, dendritic cells (DCs) play a vital role in tumor immunotherapy. Taking into account the many recent advances in DC biology, we discuss how DCs (1) recognize pathogenic antigens with pattern recognition receptors through specific phagocytosis and through non-specific micropinocytosis, (2) process antigens into small peptides with proper sizes and sequences, and (3) present MHC-peptides to CD4+ and CD8+ T cells to initiate immune responses against invading microbes and aberrant host cells. During anti-tumor immune responses, DC-derived exosomes were discovered to participate in antigen presentation. T cell microvillar dynamics and TCR conformational changes were demonstrated upon DC antigen presentation. Caspase-11-driven hyperactive DCs were recently reported to convert effectors into memory T cells. DCs were also reported to crosstalk with NK cells. Additionally, DCs are the most important sentinel cells for immune surveillance in the tumor microenvironment. Alongside DC biology, we review the latest developments for DC-based tumor immunotherapy in preclinical studies and clinical trials. Personalized DC vaccine-induced T cell immunity, which targets tumor-specific antigens, has been demonstrated to be a promising form of tumor immunotherapy in patients with melanoma. Importantly, allogeneic-IgG-loaded and HLA-restricted neoantigen DC vaccines were discovered to have robust anti-tumor effects in mice. Our comprehensive review of DC biology and its role in tumor immunotherapy aids in the understanding of DCs as the mentors of T cells and as novel tumor immunotherapy cells with immense potential.
Collapse
Affiliation(s)
- Yingying Wang
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
- Department of Gynaecology, Comprehensive Cancer Center, Hannover Medical School, 30625, Hannover, Germany
| | - Ying Xiang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | | | - Xian-Wang Wang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
- Department of Laboratory Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
| | - Xiao-Chun Peng
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
- Department of Pathophysiology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Xiao-Qin Liu
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
- Department of Medical Imaging, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Dong Wang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Na Li
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Jun-Ting Cheng
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Yan-Ning Lyv
- Institute for Infectious Diseases and Endemic Diseases Prevention and Control, Beijing Center for Diseases Prevention and Control, Beijing, 100013, China
| | - Shu-Zhong Cui
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Zhaowu Ma
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China.
| | - Qing Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
- Institute of Sun Yat-sen University in Shenzhen, Shenzhen, China.
| | - Hong-Wu Xin
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China.
- People's Hospital of Lianjiang, Lianjiang, 524400, Guangdong, China.
| |
Collapse
|
22
|
Wang Y, Xiang Y, Xin VW, Wang XW, Peng XC, Liu XQ, Wang D, Li N, Cheng JT, Lyv YN, Cui SZ, Ma Z, Zhang Q, Xin HW. Dendritic cell biology and its role in tumor immunotherapy. J Hematol Oncol 2020; 13:107. [PMID: 32746880 PMCID: PMC7397618 DOI: 10.1186/s13045-020-00939-6] [Citation(s) in RCA: 231] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022] Open
Abstract
As crucial antigen presenting cells, dendritic cells (DCs) play a vital role in tumor immunotherapy. Taking into account the many recent advances in DC biology, we discuss how DCs (1) recognize pathogenic antigens with pattern recognition receptors through specific phagocytosis and through non-specific micropinocytosis, (2) process antigens into small peptides with proper sizes and sequences, and (3) present MHC-peptides to CD4+ and CD8+ T cells to initiate immune responses against invading microbes and aberrant host cells. During anti-tumor immune responses, DC-derived exosomes were discovered to participate in antigen presentation. T cell microvillar dynamics and TCR conformational changes were demonstrated upon DC antigen presentation. Caspase-11-driven hyperactive DCs were recently reported to convert effectors into memory T cells. DCs were also reported to crosstalk with NK cells. Additionally, DCs are the most important sentinel cells for immune surveillance in the tumor microenvironment. Alongside DC biology, we review the latest developments for DC-based tumor immunotherapy in preclinical studies and clinical trials. Personalized DC vaccine-induced T cell immunity, which targets tumor-specific antigens, has been demonstrated to be a promising form of tumor immunotherapy in patients with melanoma. Importantly, allogeneic-IgG-loaded and HLA-restricted neoantigen DC vaccines were discovered to have robust anti-tumor effects in mice. Our comprehensive review of DC biology and its role in tumor immunotherapy aids in the understanding of DCs as the mentors of T cells and as novel tumor immunotherapy cells with immense potential.
Collapse
Affiliation(s)
- Yingying Wang
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China.,Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.,Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China.,Department of Gynaecology, Comprehensive Cancer Center, Hannover Medical School, 30625, Hannover, Germany
| | - Ying Xiang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.,Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | | | - Xian-Wang Wang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.,Department of Laboratory Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
| | - Xiao-Chun Peng
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.,Department of Pathophysiology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Xiao-Qin Liu
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.,Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China.,Department of Medical Imaging, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Dong Wang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.,Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Na Li
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Jun-Ting Cheng
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.,Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Yan-Ning Lyv
- Institute for Infectious Diseases and Endemic Diseases Prevention and Control, Beijing Center for Diseases Prevention and Control, Beijing, 100013, China
| | - Shu-Zhong Cui
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Zhaowu Ma
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China. .,Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China.
| | - Qing Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China. .,Institute of Sun Yat-sen University in Shenzhen, Shenzhen, China.
| | - Hong-Wu Xin
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China. .,Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China. .,People's Hospital of Lianjiang, Lianjiang, 524400, Guangdong, China.
| |
Collapse
|
23
|
Abstract
GM-CSF drives the differentiation of granulocytes and monocyte/macrophages from hematopoietic stem cell progenitors. It is required for differentiating monocytes into dendritic cells (DC). Although approved for recovery of granulocytes/monocytes in patients receiving chemotherapy, G-CSF is preferred. Enthusiasm for GM-CSF monotherapy as a melanoma treatment was dampened by two large randomized trials. Although GM-CSF has been injected into tumors for many years, the efficacy of this has not been tested. There is a strong rationale for GM-CSF as a vaccine adjuvant, but it appears of benefit only for strategies that directly involve DCs, such as intratumor talimogene laherparepvec and vaccines in which DCs are loaded with antigen ex vivo and injected admixed with GM-CSF.
Collapse
Affiliation(s)
- Robert O Dillman
- Chief Medical Officer, AIVITA Biomedical, Inc. Irvine, CA 92612, USA.,Clinical Professor Medicine, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
24
|
Baetz TD, Fletcher GG, Knight G, McWhirter E, Rajagopal S, Song X, Petrella TM. Systemic adjuvant therapy for adult patients at high risk for recurrent melanoma: A systematic review. Cancer Treat Rev 2020; 87:102032. [PMID: 32473511 DOI: 10.1016/j.ctrv.2020.102032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 12/27/2022]
Abstract
Cutaneous melanoma is typically treated with wide local excision and, when appropriate, a sentinel node biopsy. Many patients are cured with this approach but for patients who have cancers with high risk features there is a significant risk of local and distant relapse and death. Interferon-based adjuvant therapy was recommended in the past but had modest results with significant toxicity. Recently, new therapies (immune checkpoint inhibitors and targeted therapies) have been found to be effective in the treatment of patients with metastatic melanoma and many of these therapies have been evaluated and found to be effective in the adjuvant treatment of high risk patients with melanoma. This systematic review of adjuvant therapies for cutaneous and mucosal melanoma was conducted for Ontario Health (Cancer Care Ontario) as the basis of a clinical practice guideline to address the question of whether patients with completely resected melanoma should be considered for adjuvant systemic therapy and which adjuvant therapy should be used.
Collapse
Affiliation(s)
- Tara D Baetz
- Department of Oncology, Queen's University, Kingston, ON, Canada; Cancer Centre of Southeastern Ontario/Kingston General Hospital, Kingston, ON, Canada.
| | - Glenn G Fletcher
- Program in Evidence-Based Care, McMaster University, Hamilton, ON, Canada
| | - Gregory Knight
- Department of Oncology, McMaster University, Hamilton, ON, Canada; Grand River Regional Cancer Centre, Kitchener, ON, Canada
| | - Elaine McWhirter
- Department of Oncology, McMaster University, Hamilton, ON, Canada; Juravinski Cancer Centre, Hamilton, ON, Canada
| | | | - Xinni Song
- Department of Internal Medicine, Division of Medical Oncology, University of Ottawa, Ottawa, ON, Canada; The Ottawa Hospital Cancer Centre, Ottawa, ON, Canada
| | - Teresa M Petrella
- University of Toronto, Toronto, ON, Canada; Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| |
Collapse
|
25
|
Chulpanova DS, Kitaeva KV, Green AR, Rizvanov AA, Solovyeva VV. Molecular Aspects and Future Perspectives of Cytokine-Based Anti-cancer Immunotherapy. Front Cell Dev Biol 2020; 8:402. [PMID: 32582698 PMCID: PMC7283917 DOI: 10.3389/fcell.2020.00402] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 05/01/2020] [Indexed: 12/11/2022] Open
Abstract
Cytokine-based immunotherapy is a promising field in the cancer treatment, since cytokines, as proteins of the immune system, are able to modulate the host immune response toward cancer cell, as well as directly induce tumor cell death. Since a low dose monotherapy with some cytokines has no significant therapeutic results and a high dose treatment leads to a number of side effects caused by the pleiotropic effect of cytokines, the problem of understanding the influence of cytokines on the immune cells involved in the pro- and anti-tumor immune response remains a pressing one. Immune system cells carry CD makers on their surface which can be used to identify various populations of cells of the immune system that play different roles in pro- and anti-tumor immune responses. This review discusses the functions and specific CD markers of various immune cell populations which are reported to participate in the regulation of the immune response against the tumor. The results of research studies and clinical trials investigating the effect of cytokine therapy on the regulation of immune cell populations and their surface markers are also discussed. Current trends in the development of cancer immunotherapy, as well as the role of cytokines in combination with other therapeutic agents, are also discussed.
Collapse
Affiliation(s)
- Daria S Chulpanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Kristina V Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, United Kingdom
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, United Kingdom
| | - Valeriya V Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
26
|
Nistor GI, Dillman RO. Cytokine network analysis of immune responses before and after autologous dendritic cell and tumor cell vaccine immunotherapies in a randomized trial. J Transl Med 2020; 18:176. [PMID: 32316978 PMCID: PMC7171762 DOI: 10.1186/s12967-020-02328-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/02/2020] [Indexed: 02/08/2023] Open
Abstract
Background In a randomized phase II trial conducted in patients with metastatic melanoma, patient-specific autologous dendritic cell vaccines (DCV) were associated with longer survival than autologous tumor cell vaccines (TCV). Both vaccines presented antigens from cell-renewing autologous tumor cells. The current analysis was performed to better understand the immune responses induced by these vaccines, and their association with survival. Methods 110 proteomic markers were measured at a week-0 baseline, 1 week before the first of 3 weekly vaccine injections, and at week-4, 1 week after the third injection. Data was presented as a deviation from normal controls. A two-component principal component (PC) statistical analysis and discriminant analysis were performed on this data set for all patients and for each treatment cohort. Results At baseline PC-1 contained 64.4% of the variance and included the majority of cytokines associated with Th1 and Th2 responses, which positively correlated with beta-2-microglobulin (B2M), programmed death protein-1 (PD-1) and transforming growth factor beta (TGFβ1). Results were similar at baseline for both treatment cohorts. After three injections, DCV-treated patients showed correlative grouping among Th1/Th17 cytokines on PC-1, with an inverse correlation with B2M, FAS, and IL-18, and correlations among immunoglobulins in PC-2. TCV-treated patients showed a positive correlation on PC-1 among most of the cytokines and tumor markers B2M and FAS receptor. There were also correlative changes of IL12p40 with both Th1 and Th2 cytokines and TGFβ1. Discriminant analysis provided additional evidence that DCV was associated with innate, Th1/Th17, and Th2 responses while TCV was only associated with innate and Th2 responses. Conclusions These analyses confirm that DCV induced a different immune response than that induced by TCV, and these immune responses were associated with improved survival. Trial registration Clinical trials.gov NCT004936930 retrospectively registered 28 July 2009
Collapse
Affiliation(s)
- Gabriel I Nistor
- AIVITA Biomedical, Inc., 18301 Von Karman, Suite 130, Irvine, CA, 92612, USA
| | - Robert O Dillman
- AIVITA Biomedical, Inc., 18301 Von Karman, Suite 130, Irvine, CA, 92612, USA.
| |
Collapse
|
27
|
Ma M, Liu J, Jin S, Wang L. Development of tumour peptide vaccines: From universalization to personalization. Scand J Immunol 2020; 91:e12875. [PMID: 32090366 DOI: 10.1111/sji.12875] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/08/2020] [Accepted: 02/20/2020] [Indexed: 12/19/2022]
Abstract
In recent years, relying on the human immune system to kill tumour cells has become an effective means of cancer treatment. The development of peptide vaccines, which not only break the immune tolerance of a tumour but also attack malignant cells via specific antitumour immunity, has received increased attention in tumour immunization therapy due to their safety and easy preparation. The use of large-scale sequencing technology enables the continuous discovery of new tumour antigens. With improved accuracy of epitope prediction by computer simulation and the usage of a tetramer assay, cytotoxic lymphocyte epitopes can be screened and identified more easily. Transmembrane peptide and nanoparticle technologies promote more effective intake and delivery of antigens. Consequently, considerable evolution from universal to personalized peptide vaccines has taken place, and such vaccines induce an efficient and specific immune response targeting tumour neoantigens. Recently, genomic analysis and bioinformatics approaches have greatly facilitated the breakthrough of personalized peptide vaccines targeting neoantigens, resulting in a renewed interest in this field. Further, the combination of tumour peptide vaccines with checkpoint blockades may improve patient outcomes. In this review, we discuss the development of tumour peptide vaccines and the new technological progress, from universalization to personalization, to highlight the substantial promise of tumour peptide vaccines in clinical cancer immunotherapy.
Collapse
Affiliation(s)
- Minjun Ma
- Department of Gastrology, The First People's Hospital of Fuyang of Hangzhou, Hangzhou, China
| | - Jingwen Liu
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shenghang Jin
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lan Wang
- Linhai Center for Disease Control and Prevention, Linhai, China
| |
Collapse
|
28
|
Hsu CH, Hsiao CW, Sun CA, Wu WC, Yang T, Hu JM, Huang CH, Liao YC, Chen CY, Lin FH, Chou YC. Novel methylation gene panel in adjacent normal tissues predicts poor prognosis of colorectal cancer in Taiwan. World J Gastroenterol 2020; 26:154-167. [PMID: 31988582 PMCID: PMC6962436 DOI: 10.3748/wjg.v26.i2.154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/14/2019] [Accepted: 12/22/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND It is evident that current clinical criteria are suboptimal to accurately estimate patient prognosis. Studies have identified epigenetic aberrant changes as novel prognostic factors for colorectal cancer (CRC).
AIM To estimate whether a methylation gene panel in different clinical stages can reflect a different prognosis.
METHODS We enrolled 120 CRC patients from Tri-Service General Hospital in Taiwan and used the candidate gene approach to select six genes involved in carcinogenesis pathways. Patients were divided into two groups based on the methylation status of the six evaluated genes, namely, the < 3 aberrancy group and ≥ 3 aberrancy group. Various tumor stages were divided into two subgroups (local and advanced stages) on the basis of the pathological type of the following tissues: Tumor and adjacent normal tissues (matched normal). We assessed DNA methylation in tumors and adjacent normal tissues from CRC patients and analyzed the association between DNA methylation with different cancer stages and the prognostic outcome including time to progression (TTP) and overall survival.
RESULTS We observed a significantly increasing trend of hazard ratio as the number of hypermethylated genes increased both in normal tissue and tumor tissue. The 5-year TTP survival curves showed a significant difference between the ≥ 3 aberrancy group and the < 3 aberrancy group. Compared with the < 3 aberrancy group, a significantly shorter TTP was observed in the ≥ 3 aberrancy group. We further analyzed the interaction between CRC prognosis and different cancer stages (local and advanced) according to the methylation status of the selected genes in both types of tissues. There was a significantly shorter 5-year TTP for tumors at advanced stages with the promoter methylation status of selected genes than for those with local stages. We found an interaction between cancer stages and the promoter methylation status of selected genes in both types of tissues.
CONCLUSION Our data provide a significant association between the methylation markers in normal tissues with advanced stage and prognosis of CRC. We recommend using these novel markers to assist in clinical decision-making.
Collapse
Affiliation(s)
- Chih-Hsiung Hsu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan
- Teaching Office, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Cheng-Wen Hsiao
- Division of Colorectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chien-An Sun
- Department of Public Health, College of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan
- Big Data Research Center, College of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan
| | - Wen-Chih Wu
- School of Public Health, National Defense Medical Center, Taipei 114, Taiwan
- Department of Surgery, Suao and Yuanshan Branches of Taipei Veterans General Hospital, Yilan County 264, Taiwan
| | - Tsan Yang
- Department of Health Business Administration, Meiho University, Pingtung County 912, Taiwan
| | - Je-Ming Hu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan
- Division of Colorectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Adjunct Instructor, School of Medicine, National Defense Medical Center, Taipei 114, Taiwan
| | - Chi-Hua Huang
- School of Public Health, National Defense Medical Center, Taipei 114, Taiwan
| | - Yu-Chan Liao
- School of Public Health, National Defense Medical Center, Taipei 114, Taiwan
| | - Chao-Yang Chen
- Division of Colorectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Adjunct Instructor, School of Medicine, National Defense Medical Center, Taipei 114, Taiwan
| | - Fu-Huang Lin
- School of Public Health, National Defense Medical Center, Taipei 114, Taiwan
| | - Yu-Ching Chou
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan
- School of Public Health, National Defense Medical Center, Taipei 114, Taiwan
| |
Collapse
|
29
|
Hsu CH, Hsiao CW, Sun CA, Wu WC, Yang T, Hu JM, Liao YC, Huang CH, Chen CY, Lin FH, Chou YC. Multiple gene promoter methylation and clinical stage in adjacent normal tissues: Effect on prognosis of colorectal cancer in Taiwan. Sci Rep 2020; 10:145. [PMID: 31924802 PMCID: PMC6954240 DOI: 10.1038/s41598-019-56691-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/16/2019] [Indexed: 12/16/2022] Open
Abstract
This study provide an insight that the panel genes methylation status in different clinical stage tended to reflect a different prognosis even in matched normal tissues, to clinical recommendation. We enrolled 153 colorectal cancer patients from a medical center in Taiwan and used the candidate gene approach to select five genes involved in carcinogenesis pathways. We analyzed the relationship between DNA methylation with different cancer stages and the prognostic outcome. There were significant trends of increasing risk of 5-year time to progression and event-free survival of subjects with raising number of hypermethylation genes both in normal tissue and tumor tissue. The group with two or more genes with aberrant methylation in the advanced cancer stages (Me/advanced) had lower 5-year event-free survival among patients with colorectal cancer in either normal or tumor tissue. The adjusted hazard ratios in the group with two or more genes with aberrant methylation with advanced cancer stages (Me/advanced) were 8.04 (95% CI, 2.80–23.1; P for trend <0.01) and 8.01 (95% CI, 1.92–33.4; P for trend <0.01) in normal and tumor tissue, respectively. DNA methylation status was significantly associated with poor prognosis outcome. This finding in the matched normal tissues of colorectal cancer patients could be an alternative source of prognostic markers to assist clinical decision making.
Collapse
Affiliation(s)
- Chih-Hsiung Hsu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Teaching Office, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Cheng-Wen Hsiao
- Division of Colorectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chien-An Sun
- Department of Public Health, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, Republic of China.,Big Data Research Center, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, Republic of China
| | - Wen-Chih Wu
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Department of Surgery, Suao and Yuanshan branches of Taipei Veterans General Hospital, Yilan County, Taiwan, Republic of China
| | - Tsan Yang
- Department of Health Business Administration, Meiho University, Pingtung County, Taiwan, Republic of China
| | - Je-Ming Hu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Division of Colorectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Adjunct Instructor, School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Yu-Chan Liao
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chi-Hua Huang
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chao-Yang Chen
- Division of Colorectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Adjunct Instructor, School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Fu-Huang Lin
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Yu-Ching Chou
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China. .,School of Public Health, National Defense Medical Center, Taipei, Taiwan, Republic of China.
| |
Collapse
|
30
|
Irradiated Bladder Cancer Cells Expressing both GM-CSF and IL-21 versus Either GM-CSF or IL-21 Alone as Tumor Vaccine in a Mouse Xenograft Model. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8262989. [PMID: 31467912 PMCID: PMC6699310 DOI: 10.1155/2019/8262989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/19/2019] [Accepted: 07/11/2019] [Indexed: 12/15/2022]
Abstract
Previous studies have established the efficacy of irradiated cancer cells overexpressing GM-CSF or IL-21 as a vaccine. Here we examined whether the vaccine efficacy was greater when both factors were overexpressed together. MB49 bladder cancer cells were transfected with expression plasmid pT7TS encoding mouse GM-CSF and human IL-21, and then irradiated with 100 Gy at 4 days later. The cells (1×107 per animal) were injected subcutaneously into C57BL/6 mice at 0, 4, 8, and 12 days after inoculation with MB49 tumor xenografts. Control animals were injected with MB49 cells transfected with pT7TS encoding GM-CSF or IL-21 on its own. Tumor growth was monitored for 45 days and compared among the groups using repeated-measures ANOVA. Vaccination with irradiated MB49 cells did not affect xenograft growth. Vaccination with irradiated cells overexpressing GM-CSF or IL-21 alone significantly inhibited tumor growth and led to significantly more CD4+ CD8+ T cells and fewer CD4+ Foxp3+ T cells in the spleen and xenograft. These effects were even greater following vaccination with irradiated cells overexpressing both GM-CSF and IL-21. Irradiated bladder cancer cells overexpressing both GM-CSF and IL-21 are more effective than cells expressing either factor alone as a vaccine against bladder cancer.
Collapse
|
31
|
Mastio J, Condamine T, Dominguez G, Kossenkov AV, Donthireddy L, Veglia F, Lin C, Wang F, Fu S, Zhou J, Viatour P, Lavilla-Alonso S, Polo AT, Tcyganov EN, Mulligan C, Nam B, Bennett J, Masters G, Guarino M, Kumar A, Nefedova Y, Vonderheide RH, Languino LR, Abrams SI, Gabrilovich DI. Identification of monocyte-like precursors of granulocytes in cancer as a mechanism for accumulation of PMN-MDSCs. J Exp Med 2019; 216:2150-2169. [PMID: 31239386 PMCID: PMC6719429 DOI: 10.1084/jem.20181952] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/11/2019] [Accepted: 05/30/2019] [Indexed: 01/08/2023] Open
Abstract
Mastio et al. describe monocytic precursors of granulocytes. These precursors are barely detectable in steady state conditions and are not consequential for differentiation of granulocytes. However, they accumulate in cancer and substantially contribute to PMN-MDSC expansion. We have identified a precursor that differentiates into granulocytes in vitro and in vivo yet belongs to the monocytic lineage. We have termed these cells monocyte-like precursors of granulocytes (MLPGs). Under steady state conditions, MLPGs were absent in the spleen and barely detectable in the bone marrow (BM). In contrast, these cells significantly expanded in tumor-bearing mice and differentiated to polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). Selective depletion of monocytic cells had no effect on the number of granulocytes in naive mice but decreased the population of PMN-MDSCs in tumor-bearing mice by 50%. The expansion of MLPGs was found to be controlled by the down-regulation of Rb1, but not IRF8, which is known to regulate the expansion of PMN-MDSCs from classic granulocyte precursors. In cancer patients, putative MLPGs were found within the population of CXCR1+CD15−CD14+HLA-DR−/lo monocytic cells. These findings describe a mechanism of abnormal myelopoiesis in cancer and suggest potential new approaches for selective targeting of MDSCs.
Collapse
Affiliation(s)
- Jérôme Mastio
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA
| | - Thomas Condamine
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA
| | - George Dominguez
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA.,Anixa Diagnostic Corporation, San Jose, CA
| | - Andrew V Kossenkov
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA
| | | | - Filippo Veglia
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA
| | - Cindy Lin
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA
| | - Fang Wang
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA
| | - Shuyu Fu
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA.,Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jie Zhou
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Patrick Viatour
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Sergio Lavilla-Alonso
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA
| | | | - Evgenii N Tcyganov
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA
| | - Charles Mulligan
- Helen F Graham Cancer Center at Christiana Care Health System, Newark, DE
| | - Brian Nam
- Helen F Graham Cancer Center at Christiana Care Health System, Newark, DE
| | - Joseph Bennett
- Helen F Graham Cancer Center at Christiana Care Health System, Newark, DE
| | - Gregory Masters
- Helen F Graham Cancer Center at Christiana Care Health System, Newark, DE
| | - Michael Guarino
- Helen F Graham Cancer Center at Christiana Care Health System, Newark, DE
| | - Amit Kumar
- Anixa Diagnostic Corporation, San Jose, CA
| | - Yulia Nefedova
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA
| | - Robert H Vonderheide
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA
| | | | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Dmitry I Gabrilovich
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA
| |
Collapse
|
32
|
Wang X, Qi Y, Kong X, Zhai J, Li Y, Song Y, Wang J, Feng X, Fang Y. Immunological therapy: A novel thriving area for triple-negative breast cancer treatment. Cancer Lett 2018; 442:409-428. [PMID: 30419345 DOI: 10.1016/j.canlet.2018.10.042] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/25/2018] [Accepted: 10/25/2018] [Indexed: 11/19/2022]
Abstract
Triple-negative breast cancer (TNBC) refers to cancers that are low in expression of the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). TNBC tends to behave more aggressively than other types of breast cancer. Unlike other breast cancer subtypes (ie, ER-positive, HER2-positive subtypes), there are no approved targeted treatments available, other than the administration of chemotherapy. Immunotherapy is a new kind of treatment approach for TNBC when compared with the surgical treatment, chemotherapy, endocrine therapy, and molecular targeting therapy. The present article reviews the research progresses of immunotherapy for TNBC in recent years. The full text structure covers molecular classification of TNBC, active immunotherapy of TNBC, passive immunotherapy of TNBC, oncolytic immunotherapy and the prospect of immunotherapy for TNBC.
Collapse
Affiliation(s)
- Xiangyu Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; Department of Laboratory Medicine, Mayo Clinic, Rochester, MN, 55902, USA
| | - Yihang Qi
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jie Zhai
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yalun Li
- Department of Breast Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, 264000, China
| | - Yan Song
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Xiaoli Feng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
33
|
Bezu L, Kepp O, Cerrato G, Pol J, Fucikova J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Peptide-based vaccines in anticancer therapy. Oncoimmunology 2018; 7:e1511506. [PMID: 30524907 PMCID: PMC6279318 DOI: 10.1080/2162402x.2018.1511506] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Indexed: 12/15/2022] Open
Abstract
Peptide-based anticancer vaccination aims at stimulating an immune response against one or multiple tumor-associated antigens (TAAs) following immunization with purified, recombinant or synthetically engineered epitopes. Despite high expectations, the peptide-based vaccines that have been explored in the clinic so far had limited therapeutic activity, largely due to cancer cell-intrinsic alterations that minimize antigenicity and/or changes in the tumor microenvironment that foster immunosuppression. Several strategies have been developed to overcome such limitations, including the use of immunostimulatory adjuvants, the co-treatment with cytotoxic anticancer therapies that enable the coordinated release of damage-associated molecular patterns, and the concomitant blockade of immune checkpoints. Personalized peptide-based vaccines are also being explored for therapeutic activity in the clinic. Here, we review recent preclinical and clinical progress in the use of peptide-based vaccines as anticancer therapeutics.Abbreviations: CMP: carbohydrate-mimetic peptide; CMV: cytomegalovirus; DC: dendritic cell; FDA: Food and Drug Administration; HPV: human papillomavirus; MDS: myelodysplastic syndrome; MHP: melanoma helper vaccine; NSCLC: non-small cell lung carcinoma; ODD: orphan drug designation; PPV: personalized peptide vaccination; SLP: synthetic long peptide; TAA: tumor-associated antigen; TNA: tumor neoantigen
Collapse
Affiliation(s)
- Lucillia Bezu
- Faculty of Medicine, University of Paris Sud/Paris XI, Le Kremlin-Bicêtre, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers,Paris, France.,U1138, INSERM, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers,Paris, France.,U1138, INSERM, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Giulia Cerrato
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers,Paris, France.,U1138, INSERM, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Jonathan Pol
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers,Paris, France.,U1138, INSERM, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Jitka Fucikova
- Sotio, Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio, Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- Faculty of Medicine, University of Paris Sud/Paris XI, Le Kremlin-Bicêtre, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,INSERM, U1015, Gustave Roussy Cancer Campus, Villejuif, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers,Paris, France.,U1138, INSERM, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Paris, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
34
|
Okla K, Wertel I, Wawruszak A, Bobiński M, Kotarski J. Blood-based analyses of cancer: Circulating myeloid-derived suppressor cells - is a new era coming? Crit Rev Clin Lab Sci 2018; 55:376-407. [PMID: 29927668 DOI: 10.1080/10408363.2018.1477729] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Progress in cancer treatment made by the beginning of the 21st century has shifted the paradigm from one-size-fits-all to tailor-made treatment. The popular vision, to study solid tumors through the relatively noninvasive sampling of blood, is one of the most thrilling and rapidly advancing fields in global cancer diagnostics. From this perspective, immune-cell analysis in cancer could play a pivotal role in oncology practice. This approach is driven both by rapid technological developments, including the analysis of circulating myeloid-derived suppressor cells (cMDSCs), and by the increasing application of (immune) therapies, the success or failure of which may depend on effective and timely measurements of relevant biomarkers. Although the implementation of these powerful noninvasive diagnostic capabilities in guiding precision cancer treatment is poised to change the ways in which we select and monitor cancer therapy, challenges remain. Here, we discuss the challenges associated with the analysis and clinical aspects of cMDSCs and assess whether the problems in implementing tumor-evolution monitoring as a global tool in personalized oncology can be overcome.
Collapse
Affiliation(s)
- Karolina Okla
- a 1st Chair and Department of Oncological Gynaecology and Gynaecology, Tumor Immunology Laboratory , Medical University of Lublin , Lublin , Poland
| | - Iwona Wertel
- a 1st Chair and Department of Oncological Gynaecology and Gynaecology, Tumor Immunology Laboratory , Medical University of Lublin , Lublin , Poland
| | - Anna Wawruszak
- b Department of Biochemistry and Molecular Biology , Medical University of Lublin , Lublin , Poland
| | - Marcin Bobiński
- a 1st Chair and Department of Oncological Gynaecology and Gynaecology, Tumor Immunology Laboratory , Medical University of Lublin , Lublin , Poland
| | - Jan Kotarski
- a 1st Chair and Department of Oncological Gynaecology and Gynaecology, Tumor Immunology Laboratory , Medical University of Lublin , Lublin , Poland
| |
Collapse
|
35
|
Robinson TM, Prince GT, Thoburn C, Warlick E, Ferguson A, Kasamon YL, Borrello IM, Hess A, Smith BD. Pilot trial of K562/GM-CSF whole-cell vaccination in MDS patients. Leuk Lymphoma 2018; 59:2801-2811. [PMID: 29616857 DOI: 10.1080/10428194.2018.1443449] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal hematopoietic stem cell malignancies. Currently, approved drugs are given with non-curative intent as the only known cure is allogeneic bone marrow transplantation, which relies on the donor's immune system driving an allogeneic effect. Previous efforts to harness the endogenous immune system have been less successful. We present the results of a pilot study of K562/GM-CSF (GVAX) whole-cell vaccination in MDS patients. The primary objective of safety was met as there were no serious adverse events. One patient had a decrease in transfusion requirements and another demonstrated hematologic improvement suggesting a signal for clinical activity. In vitro correlative studies indicated biological effects on immune cells following vaccination. Although only a pilot study, results are encouraging that an immunotherapeutic approach with a whole-cell vaccine may be feasible in MDS patients.
Collapse
Affiliation(s)
- Tara M Robinson
- a Department of Medical Oncology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Gabrielle T Prince
- a Department of Medical Oncology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Chris Thoburn
- b Department of Pathology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Erica Warlick
- c Department of Medicine , University of Minnesota Medical Center , St. Paul/Minneapolis , MN , USA
| | - Anna Ferguson
- a Department of Medical Oncology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Yvette L Kasamon
- a Department of Medical Oncology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Ivan M Borrello
- a Department of Medical Oncology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Allan Hess
- a Department of Medical Oncology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - B Douglas Smith
- a Department of Medical Oncology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| |
Collapse
|
36
|
García-Martínez E, Smith M, Buqué A, Aranda F, de la Peña FA, Ivars A, Cánovas MS, Conesa MAV, Fucikova J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunostimulation with recombinant cytokines for cancer therapy. Oncoimmunology 2018; 7:e1433982. [PMID: 29872569 PMCID: PMC5980390 DOI: 10.1080/2162402x.2018.1433982] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 01/24/2018] [Indexed: 12/15/2022] Open
Abstract
Cytokines regulate virtually aspects of innate and adaptive immunity, including the initiation, execution and extinction of tumor-targeting immune responses. Over the past three decades, the possibility of using recombinant cytokines as a means to elicit or boost clinically relevant anticancer immune responses has attracted considerable attention. However, only three cytokines have been approved so far by the US Food and Drug Administration and the European Medicines Agency for use in cancer patients, namely, recombinant interleukin (IL)-2 and two variants of recombinant interferon alpha 2 (IFN-α2a and IFN-α2b). Moreover, the use of these cytokines in the clinics is steadily decreasing, mostly as a consequence of: (1) the elevated pleiotropism of IL-2, IFN-α2a and IFN-α2b, resulting in multiple unwarranted effects; and (2) the development of highly effective immunostimulatory therapeutics, such as immune checkpoint blockers. Despite this and other obstacles, research in the field continues as alternative cytokines with restricted effects on specific cell populations are being evaluated. Here, we summarize research preclinical and clinical developments on the use of recombinant cytokines for immunostimulation in cancer patients.
Collapse
Affiliation(s)
- Elena García-Martínez
- Hematology and Oncology Department, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Melody Smith
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Fernando Aranda
- Immunoreceptors of the Innate and Adaptive System, IDIBAPS, Barcelona, Spain
| | | | - Alejandra Ivars
- Hematology and Oncology Department, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Manuel Sanchez Cánovas
- Hematology and Oncology Department, Hospital Universitario Morales Meseguer, Murcia, Spain
| | | | - Jitka Fucikova
- Sotio, Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio, Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, France
- Université Pierre et Marie Curie/Paris VI, Paris
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
- Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, Paris, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Université Paris Descartes/Paris V, France
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
37
|
Myeloid-derived suppressor cells coming of age. Nat Immunol 2018; 19:108-119. [PMID: 29348500 DOI: 10.1038/s41590-017-0022-x] [Citation(s) in RCA: 1234] [Impact Index Per Article: 205.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells generated during a large array of pathologic conditions ranging from cancer to obesity. These cells represent a pathologic state of activation of monocytes and relatively immature neutrophils. MDSCs are characterized by a distinct set of genomic and biochemical features, and can, on the basis of recent findings, be distinguished by specific surface molecules. The salient feature of these cells is their ability to inhibit T cell function and thus contribute to the pathogenesis of various diseases. In this Review, we discuss the origin and nature of these cells; their distinctive features; and their biological roles in cancer, infectious diseases, autoimmunity, obesity and pregnancy.
Collapse
|
38
|
Sadozai H, Gruber T, Hunger RE, Schenk M. Recent Successes and Future Directions in Immunotherapy of Cutaneous Melanoma. Front Immunol 2017; 8:1617. [PMID: 29276510 PMCID: PMC5727014 DOI: 10.3389/fimmu.2017.01617] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 11/08/2017] [Indexed: 12/14/2022] Open
Abstract
The global health burden associated with melanoma continues to increase while treatment options for metastatic melanoma are limited. Nevertheless, in the past decade, the field of cancer immunotherapy has witnessed remarkable advances for the treatment of a number of malignancies including metastatic melanoma. Although the earliest observations of an immunological antitumor response were made nearly a century ago, it was only in the past 30 years, that immunotherapy emerged as a viable therapeutic option, in particular for cutaneous melanoma. As such, melanoma remains the focus of various preclinical and clinical studies to understand the immunobiology of cancer and to test various tumor immunotherapies. Here, we review key recent developments in the field of immune-mediated therapy of melanoma. Our primary focus is on therapies that have received regulatory approval. Thus, a brief overview of the pathophysiology of melanoma is provided. The purported functions of various tumor-infiltrating immune cell subsets are described, in particular the recently described roles of intratumoral dendritic cells. The section on immunotherapies focuses on strategies that have proved to be the most clinically successful such as immune checkpoint blockade. Prospects for novel therapeutics and the potential for combinatorial approaches are delineated. Finally, we briefly discuss nanotechnology-based platforms which can in theory, activate multiple arms of immune system to fight cancer. The promising advances in the field of immunotherapy signal the dawn of a new era in cancer treatment and warrant further investigation to understand the opportunities and barriers for future progress.
Collapse
Affiliation(s)
- Hassan Sadozai
- Institute of Pathology, Experimental Pathology, University of Bern, Bern, Switzerland
| | - Thomas Gruber
- Institute of Pathology, Experimental Pathology, University of Bern, Bern, Switzerland
| | | | - Mirjam Schenk
- Institute of Pathology, Experimental Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
39
|
Stern PL. Is immunity in cancer the key to improving clinical outcome?: Report on the International Symposium on Immunotherapy, The Royal Society, London, UK, 12-13 May 2017. THERAPEUTIC ADVANCES IN VACCINES 2017; 5:55-68. [PMID: 28794878 DOI: 10.1177/2051013617720659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 06/19/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Peter L Stern
- Division of Molecular & Clinical Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Paterson Building, Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|