1
|
Kaito Y, Imai Y. Evolution of natural killer cell-targeted therapy for acute myeloid leukemia. Int J Hematol 2024; 120:34-43. [PMID: 38693419 DOI: 10.1007/s12185-024-03778-0] [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: 03/15/2024] [Revised: 04/04/2024] [Accepted: 04/14/2024] [Indexed: 05/03/2024]
Abstract
In hematologic oncology, acute myeloid leukemia (AML) presents a significant challenge due to its complex genetic landscape and resistance to conventional therapies. Despite advances in treatment, including intensive chemotherapy and hematopoietic stem cell transplantation (HSCT), the prognosis for many patients with AML remains poor. Recently, immunotherapy has emerged as a promising approach to improve outcomes by augmenting existing treatments. Natural killer (NK) cells, a subset of innate lymphoid cells, have garnered attention for their potent cytotoxic capabilities against AML cells. In this review, we discuss the role of NK cells in AML immunosurveillance, their dysregulation in patients with AML, and various therapeutic strategies leveraging NK cells in AML treatment. We explore the challenges and prospects associated with NK cell therapy, including approaches to enhance NK cell function, overcome immune evasion mechanisms, and optimize treatment efficacy. Finally, we emphasize the importance of further research to validate and refine patient-first NK cell-based immunotherapies for AML.
Collapse
Affiliation(s)
- Yuta Kaito
- Department of Hematology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-Ku, Tokyo, 113-8602, Japan.
| | - Yoichi Imai
- Department of Hematology and Oncology, Dokkyo Medical University, Tochigi, Japan
| |
Collapse
|
2
|
Xie L, Wang L, Li L, Liu C, Guo L, Liao Y, Zhou S, Wu W, Duo Y, Shi L, Yuan M. Novel Carrier-Free Nanodrug Enhances Photodynamic Effects by Blocking the Autophagy Pathway and Synergistically Triggers Immunogenic Cell Death for the Efficient Treatment of Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2024; 16:5683-5695. [PMID: 38261396 DOI: 10.1021/acsami.3c17977] [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: 01/24/2024]
Abstract
Photosensitizers have been widely used to cause intratumoral generation of reactive oxygen species (ROS) for cancer therapy, but they are easily disturbed by the autophagy pathway, a self-protective mechanism by mitigating oxidative damage. Hereby, we reported a simple and effective strategy to construct a carrier-free nanodrug, Ce6@CQ namely, based on the self-assembly of the photosensitizer chlorin e6 (Ce6) and the autophagy inhibitor chloroquine (CQ). Specifically, Ce6@CQ avoided the unexpected toxicity caused by the regular nanocarrier and also ameliorated its stability in different conditions. Light-activated Ce6 generated cytotoxic ROS and elicited part of the immunogenic cell death (ICD). Moreover, CQ induced autophagy dysfunction, which hindered self-healing in tumor cells and enhanced photodynamic therapy (PDT) to exert a more potent killing effect and more efficient ICD. Also, Ce6@CQ could effectively accumulate in the xenograft breast tumor site in a mouse model through the enhanced permeability and retention (EPR) effect, and the growth of breast tumors was effectively inhibited by Ce6@CQ with light. Such a carrier-free nanodrug provided a new strategy to improve the efficacy of PDT via the suppression of autophagy to digest ROS-induced toxic substances.
Collapse
Affiliation(s)
- Luoyijun Xie
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Pharmacology, the Eighth Affiliated Hospital, Sun Yat-sen University, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Shenzhen 510275, China
| | - Li Wang
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ling Li
- Department of Pharmacology, the Eighth Affiliated Hospital, Sun Yat-sen University, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Shenzhen 510275, China
| | - Chutong Liu
- Department of Pharmacology, the Eighth Affiliated Hospital, Sun Yat-sen University, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Shenzhen 510275, China
| | - Lihao Guo
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- School of Advanced Materials and Nanotechnology, Interdisciplinary Research Center of Smart Sensors, Xidian University, Xi'an 710126, China
| | - Yingying Liao
- Department of Pharmacology, the Eighth Affiliated Hospital, Sun Yat-sen University, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Shenzhen 510275, China
| | - Shuyi Zhou
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weiwei Wu
- School of Advanced Materials and Nanotechnology, Interdisciplinary Research Center of Smart Sensors, Xidian University, Xi'an 710126, China
| | - Yanhong Duo
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02138, United States
| | - Leilei Shi
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Miaomiao Yuan
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Pharmacology, the Eighth Affiliated Hospital, Sun Yat-sen University, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Shenzhen 510275, China
| |
Collapse
|
3
|
Tan Y, Chen H, Gou X, Fan Q, Chen J. Tumor vaccines: Toward multidimensional anti-tumor therapies. Hum Vaccin Immunother 2023; 19:2271334. [PMID: 37905395 PMCID: PMC10760370 DOI: 10.1080/21645515.2023.2271334] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/12/2023] [Indexed: 11/02/2023] Open
Abstract
For decades, immunotherapies have offered hope for patients with advanced cancer. However, they show distinct benefits and limited clinical effects. Tumor vaccines have the potential to prime tumor-antigen-specific T cells and induce broad subsets of immune responses, ultimately eradicating tumor cells. Here, we classify tumor vaccines by their anti-tumor mechanisms, which include boosting the immune system, overcoming tumor immunosuppression, and modulating tumor angiogenesis. We focus on multidimensional tumor vaccine strategies using combinations of two or three of the above mechanisms, as these are superior to single-dimensional treatments. This review offers a perspective on tumor vaccine strategies and the future role of vaccine therapies in cancer treatment.
Collapse
Affiliation(s)
- Yuanfang Tan
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Huiyuan Chen
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xi Gou
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qiuying Fan
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Juanjuan Chen
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
4
|
Zhang N, Liu S, Zhang Z, Liu Y, Mi L, Xu K. Lung Transplantation: A Viable Option for Connective Tissue Disease? Arthritis Care Res (Hoboken) 2023; 75:2389-2398. [PMID: 37052523 DOI: 10.1002/acr.25133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/24/2023] [Accepted: 04/11/2023] [Indexed: 04/14/2023]
Abstract
Interstitial lung disease (ILD) and pulmonary hypertension (PH) caused by connective tissue disease (CTD) are one of the main causes of morbidity and death in patients. Although the International Society for Heart & Lung Transplant suggested that ILD and PH related to CTD are rare indications for lung transplantation in 2006, many lung transplantation centers are concerned that the multisystem involvement of CTD will affect survival outcomes after lung transplantation, and CTD is regarded as a relative contraindication for lung transplantation. However, long-term and short-term survival after lung transplantation in CTD patients is similar compared with survival in common indications for lung transplantation such as idiopathic pulmonary fibrosis (IPF), and no higher incidence of complications after transplantation in many lung transplant centers. This suggests that lung transplantation may be beneficial in CTD patients with disease that progresses to end-stage lung disease, and CTD should not be considered a contraindication for lung transplantation. In the future, more prospective studies are needed to analyze the risk factors of lung transplantation in CTD patients to improve survival rates and reduce the risk of complications. This narrative review summarizes the selection and evaluation of candidates for CTD before lung transplantation and describes the clinical outcomes in CTD after lung transplantation in large-capacity lung transplantation center. The purpose of this review is to help rheumatologists decide when to refer patients with CTD-related lung involvement to a lung transplantation center and the conditions to consider before transplantation and to provide confidence to lung transplant experts.
Collapse
Affiliation(s)
- Na Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China and Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shizhou Liu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China and Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaoliang Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Ying Liu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China and Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liangyu Mi
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Ke Xu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China and Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Locatelli F, Shah B, Thomas T, Velasco K, Adedokun B, Aldoss I, Gore L, Hoelzer D, Bassan R, Park JH, Boissel N, Kantarjian H. Incidence of CD19-negative relapse after CD19-targeted immunotherapy in R/R BCP acute lymphoblastic leukemia: a review. Leuk Lymphoma 2023; 64:1615-1633. [PMID: 37526512 DOI: 10.1080/10428194.2023.2232496] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/25/2023] [Indexed: 08/02/2023]
Abstract
There are inconsistencies in the reporting of CD19 antigen status following treatment with CD19-targeted therapies. A majority of evidence comes from studies reporting small sample sizes. In this review, we systematically summarize published studies that have reported rates of CD19-negative relapse after treatment with either blinatumomab or CD19-directed CAR T-cell therapy and report the rates of CD19-negative relapse when evaluated in a standardized way across trials. CD19-negative relapse appears to occur more commonly in relapses following CAR T-cell therapy compared with blinatumomab, whether proportions are calculated among all treated patients (8.7% vs 4.5%) or among patients who relapse (30% vs 22.5%). The median (range) duration of follow-up was 29.3 (17.4-50.8) and 20.4 (6.9-49.0) months for publications on blinatumomab (n = 10) and CAR T-cell therapies (n = 23), respectively. There is a need for standardized reporting of CD19 antigen status in the setting of relapse following novel immunotherapies to inform clinical practice.
Collapse
Affiliation(s)
- Franco Locatelli
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome, Italy
| | - Bijal Shah
- Moffitt Cancer Center, Tampa, Florida, USA
| | | | | | | | - Ibrahim Aldoss
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Lia Gore
- Children's Hospital Colorado and University of Colorado Cancer Center, Colorado, USA
| | | | - Renato Bassan
- Hematology Unit, Azienda Ulss3 Serenissima, Ospedale dell'Angelo, Venice, Italy
| | - Jae H Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Nicolas Boissel
- Hematology Adolescent and Young Adult Unit, Saint-Louis Hospital, AP-HP; URP-3518, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
6
|
Pagliuca S, Gurnari C, Hercus C, Hergalant S, Hong S, Dhuyser A, D'Aveni M, Aarnink A, Rubio MT, Feugier P, Ferraro F, Carraway HE, Sobecks R, Hamilton BK, Majhail NS, Visconte V, Maciejewski JP. Leukemia relapse via genetic immune escape after allogeneic hematopoietic cell transplantation. Nat Commun 2023; 14:3153. [PMID: 37258544 PMCID: PMC10232425 DOI: 10.1038/s41467-023-38113-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 04/13/2023] [Indexed: 06/02/2023] Open
Abstract
Graft-versus-leukemia (GvL) reactions are responsible for the effectiveness of allogeneic hematopoietic cell transplantation as a treatment modality for myeloid neoplasia, whereby donor T- effector cells recognize leukemia neoantigens. However, a substantial fraction of patients experiences relapses because of the failure of the immunological responses to control leukemic outgrowth. Here, through a broad immunogenetic study, we demonstrate that germline and somatic reduction of human leucocyte antigen (HLA) heterogeneity enhances the risk of leukemic recurrence. We show that preexistent germline-encoded low evolutionary divergence of class II HLA genotypes constitutes an independent factor associated with disease relapse and that acquisition of clonal somatic defects in HLA alleles may lead to escape from GvL control. Both class I and II HLA genes are targeted by somatic mutations as clonal selection factors potentially impairing cellular immune responses and response to immunomodulatory strategies. These findings define key molecular modes of post-transplant leukemia escape contributing to relapse.
Collapse
Affiliation(s)
- Simona Pagliuca
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Hematology, CHRU de Nancy, Vandœuvre-lès-Nancy, France
- CNRS UMR 7365, IMoPA, Biopole of University of Lorraine, Vandœuvre-lès-Nancy, France
| | - Carmelo Gurnari
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Biomedicine and Prevention, PhD in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome Tor Vergata, Rome, Italy
| | - Colin Hercus
- Novocraft Technologies Sdn Bhd, Kuala Lumpur, Malaysia
| | - Sébastien Hergalant
- Inserm UMR-S 1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500, Vandœuvre-lès-Nancy, France
| | - Sanghee Hong
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Adele Dhuyser
- CNRS UMR 7365, IMoPA, Biopole of University of Lorraine, Vandœuvre-lès-Nancy, France
- Histocompatibility Department, CHRU de Nancy, Vandœuvre-lès-Nancy, France
| | - Maud D'Aveni
- Department of Hematology, CHRU de Nancy, Vandœuvre-lès-Nancy, France
- CNRS UMR 7365, IMoPA, Biopole of University of Lorraine, Vandœuvre-lès-Nancy, France
| | - Alice Aarnink
- CNRS UMR 7365, IMoPA, Biopole of University of Lorraine, Vandœuvre-lès-Nancy, France
- Histocompatibility Department, CHRU de Nancy, Vandœuvre-lès-Nancy, France
| | - Marie Thérèse Rubio
- Department of Hematology, CHRU de Nancy, Vandœuvre-lès-Nancy, France
- CNRS UMR 7365, IMoPA, Biopole of University of Lorraine, Vandœuvre-lès-Nancy, France
| | - Pierre Feugier
- Department of Hematology, CHRU de Nancy, Vandœuvre-lès-Nancy, France
| | - Francesca Ferraro
- Division of Oncology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Hetty E Carraway
- Leukemia Program, Hematology Department, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ronald Sobecks
- Blood and Marrow Transplant Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Betty K Hamilton
- Blood and Marrow Transplant Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Navneet S Majhail
- Sarah Cannon Transplant and Cellular Therapy Network, Nashville, TN, USA
| | - Valeria Visconte
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jaroslaw P Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
7
|
Pagliuca S, Gurnari C, Hercus C, Hergalant S, Hong S, Dhuyser A, D'Aveni M, Aarnink A, Rubio MT, Feugier P, Ferraro F, Carraway HE, Sobecks R, Hamilton BK, Majhail NS, Visconte V, Maciejewski JP. Leukemia relapse via genetic immune escape after allogeneic hematopoietic cell transplantation. RESEARCH SQUARE 2023:rs.3.rs-2773498. [PMID: 37066269 PMCID: PMC10104200 DOI: 10.21203/rs.3.rs-2773498/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Graft-versus-leukemia (GvL) reactions are responsible for the effectiveness of allogeneic hematopoietic cell transplantation as a treatment modality for myeloid neoplasia, whereby donor T- effector cells recognize leukemia neoantigens. However, a substantial fraction of patients experience relapses because of the failure of the immunological responses to control leukemic outgrowth. Here, through a broad immunogenetic study, we demonstrate that germline and somatic reduction of human leucocyte antigen (HLA) heterogeneity enhances the risk of leukemic recurrence. We show that preexistent germline-encoded low evolutionary divergence of class II HLA genotypes constitutes an independent factor associated with disease relapse and that acquisition of clonal somatic defects in HLA alleles may lead to escape from GvL control. Both class I and II HLA genes are targeted by somatic mutations as clonal selection factors potentially impairing cellular immune reactions and response to immunomodulatory strategies. These findings define key molecular modes of post-transplant leukemia escape contributing to relapse.
Collapse
Affiliation(s)
| | - Carmelo Gurnari
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Colin Hercus
- Novocraft Technologies Sdn Bhd, Kuala Lumpur, Malaysia
| | - Sébastien Hergalant
- Inserm UMR S1256 Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine, 54500 Nancy, France
| | - Sanghee Hong
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Adele Dhuyser
- Histocompatibility department, CHRU Nancy, Nancy, France
| | - Maud D'Aveni
- Department of Clinical Hematology, CHRU Nancy, Nancy, France
| | - Alice Aarnink
- Histocompatibility department, CHRU Nancy, Nancy, France
| | | | - Pierre Feugier
- Department of Clinical Hematology, CHRU Nancy, Nancy, France
| | - Francesca Ferraro
- Division of Oncology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Hetty E Carraway
- Leukemia program, Hematology Department, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ronald Sobecks
- Blood and Marrow Transplant Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Betty K Hamilton
- Blood and Marrow Transplant Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Navneet S Majhail
- Sarah Cannon Transplant and Cellular Therapy Network, Nashville, TN, USA
| | - Valeria Visconte
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Jaroslaw P Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
8
|
Rahmani S, Yazdanpanah N, Rezaei N. Natural killer cells and acute myeloid leukemia: promises and challenges. Cancer Immunol Immunother 2022; 71:2849-2867. [PMID: 35639116 PMCID: PMC10991240 DOI: 10.1007/s00262-022-03217-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
Abstract
Acute myeloid leukemia (AML) is considered as one of the most malignant conditions of the bone marrow. Over the past few decades, despite substantial progresses in the management of AML, relapse remission remains a major problem. Natural killer cells (NK cells) are known as a unique component of the innate immune system. Due to swift tumor detection, distinct cytotoxic action, and extensive immune interaction, NK cells have been used in various cancer settings for decades. It has been a growing knowledge of therapeutic magnitudes ranging from adoptive NK cell transfer to chimeric antigen receptor NK cells, aiming to achieve better therapeutic responses in patients with AML. In this article, the potentials of NK cells for treatment of AML are highlighted, and challenges for such therapeutic methods are discussed. In addition, the clinical application of NK cells, mainly in patients with AML, is pictured according to the existing evidence.
Collapse
Affiliation(s)
- Shayan Rahmani
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloufar Yazdanpanah
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Pagliuca S, Gurnari C, Rubio MT, Visconte V, Lenz TL. Individual HLA heterogeneity and its implications for cellular immune evasion in cancer and beyond. Front Immunol 2022; 13:944872. [PMID: 36131910 PMCID: PMC9483928 DOI: 10.3389/fimmu.2022.944872] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/17/2022] [Indexed: 01/07/2023] Open
Abstract
Structural and functional variability of human leukocyte antigen (HLA) is the foundation for competent adaptive immune responses against pathogen and tumor antigens as it assures the breadth of the presented immune-peptidome, theoretically sustaining an efficient and diverse T cell response. This variability is presumably the result of the continuous selection by pathogens, which over the course of evolution shaped the adaptive immune system favoring the assortment of a hyper-polymorphic HLA system able to elaborate efficient immune responses. Any genetic alteration affecting this diversity may lead to pathological processes, perturbing antigen presentation capabilities, T-cell reactivity and, to some extent, natural killer cell functionality. A highly variable germline HLA genotype can convey immunogenetic protection against infections, be associated with tumor surveillance or influence response to anti-neoplastic treatments. In contrast, somatic aberrations of HLA loci, rearranging the original germline configuration, theoretically decreasing its variability, can facilitate mechanisms of immune escape that promote tumor growth and immune resistance. The purpose of the present review is to provide a unified and up-to-date overview of the pathophysiological consequences related to the perturbations of the genomic heterogeneity of HLA complexes and their impact on human diseases, with a special focus on cancer.
Collapse
Affiliation(s)
- Simona Pagliuca
- Translational Hematology and Oncology Research Department, Cleveland Clinic, Cleveland, OH, United States
- Service d’hématologie Clinique, Hôpital Brabois, CHRU Nancy and CNRS UMR 7365 IMoPa, Biopole de l’Université de Loarraine, Vandoeuvre les Nancy, France
| | - Carmelo Gurnari
- Translational Hematology and Oncology Research Department, Cleveland Clinic, Cleveland, OH, United States
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Marie Thérèse Rubio
- Service d’hématologie Clinique, Hôpital Brabois, CHRU Nancy and CNRS UMR 7365 IMoPa, Biopole de l’Université de Loarraine, Vandoeuvre les Nancy, France
| | - Valeria Visconte
- Translational Hematology and Oncology Research Department, Cleveland Clinic, Cleveland, OH, United States
| | - Tobias L. Lenz
- Research Unit for Evolutionary Immunogenomics, Department of Biology, University of Hamburg, Hamburg, Germany
| |
Collapse
|
10
|
Lau SP, Klaase L, Vink M, Dumas J, Bezemer K, van Krimpen A, van der Breggen R, Wismans LV, Doukas M, de Koning W, Stubbs AP, Mustafa DAM, Vroman H, Stadhouders R, Nunes JB, Stingl C, de Miranda NFCC, Luider TM, van der Burg SH, Aerts JG, van Eijck CHJ. Autologous dendritic cells pulsed with allogeneic tumour cell lysate induce tumour-reactive T-cell responses in patients with pancreatic cancer: A phase I study. Eur J Cancer 2022; 169:20-31. [PMID: 35490565 DOI: 10.1016/j.ejca.2022.03.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/09/2022] [Accepted: 03/16/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is notorious for its poor prognosis even after curative resection. Responses to immunotherapy are rare and related to inadequate T-cell priming. We previously demonstrated the potency of allogeneic lysate-dendritic cell (DC) vaccination in a preclinical model. Here we translate this concept to patients. METHODS In this phase I study, patients with resected PDAC were included when they demonstrated no radiologic signs of recurrence after standard-of-care treatment. Allogeneic tumour lysate-loaded autologous monocyte-derived DCs were injected at weeks 0, 2, 4 and at months 3 and 6. Objectives are feasibility, safety and immunogenicity of allogeneic tumour-DCs. The presence of tumour antigens shared between the vaccine and patient tumours was investigated. Immunological analyses were performed on peripheral blood, skin and tumour. RESULTS Ten patients were included. DC production and administration were successful. All patients experienced a grade 1 injection-site and infusion-related reaction. Two patients experienced a grade 2 fever and 1 patient experienced a grade 3 dyspnoea. No vaccine-related serious adverse events were observed. Shared tumour antigens were found between the vaccine and patient tumours. All evaluated patients displayed a vaccine-induced response indicated by increased frequencies of Ki67+ and activated PD-1+ circulating T-cells. In addition, treatment-induced T-cell reactivity to autologous tumour of study patients was detected. Seven out of ten patients have not experienced disease recurrence or progression at a median follow-up of 25 months (15-32 months). CONCLUSION Allogeneic tumour lysate-DC treatment is feasible, safe and induces immune reactivity to PDAC expressed antigens.
Collapse
Affiliation(s)
- S P Lau
- Department of Surgery, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands; Department of Pulmonary Medicine, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands
| | - L Klaase
- Department of Pulmonary Medicine, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands
| | - M Vink
- Department of Pulmonary Medicine, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands
| | - J Dumas
- Department of Pathology, The Tumor Immuno-Pathology Laboratory, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands
| | - K Bezemer
- Department of Pulmonary Medicine, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands; Amphera B.V., Onderwijsboulevard 225, 5223DE, 'S-Hertogenbosch, the Netherlands
| | - A van Krimpen
- Department of Pulmonary Medicine, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands
| | - R van der Breggen
- Department of Pathology, Leiden University Medical Center, P.O. Box 9600, 2300RC, Leiden, the Netherlands
| | - L V Wismans
- Department of Surgery, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands
| | - M Doukas
- Department of Pathology, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands
| | - W de Koning
- Department of Pathology, The Tumor Immuno-Pathology Laboratory, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands; Department of Pathology, Clinical Bioinformatics Unit, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands
| | - A P Stubbs
- Department of Pathology, Clinical Bioinformatics Unit, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands
| | - D A M Mustafa
- Department of Pathology, The Tumor Immuno-Pathology Laboratory, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands
| | - H Vroman
- Department of Pulmonary Medicine, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands
| | - R Stadhouders
- Department of Pulmonary Medicine, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands; Department of Cell Biology, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands
| | - J B Nunes
- Department of Pathology, Leiden University Medical Center, P.O. Box 9600, 2300RC, Leiden, the Netherlands
| | - C Stingl
- Department of Neurology, Clinical and Cancer Proteomics, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands
| | - N F C C de Miranda
- Department of Pathology, Leiden University Medical Center, P.O. Box 9600, 2300RC, Leiden, the Netherlands
| | - T M Luider
- Department of Neurology, Clinical and Cancer Proteomics, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands
| | - S H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, P.O. Box 9600, 2300RC, Leiden, the Netherlands
| | - J G Aerts
- Department of Pulmonary Medicine, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands; Erasmus MC Cancer Institute, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands
| | - C H J van Eijck
- Department of Surgery, Erasmus University Medical Center, 'S-Gravendijkwal 230, 3015CE, Rotterdam, the Netherlands.
| |
Collapse
|
11
|
Aquilani R, Brugnatelli S, Maestri R, Boschi F, Filippi B, Perrone L, Barbieri A, Buonocore D, Dossena M, Verri M. Peripheral Blood Lymphocyte Percentage May Predict Chemotolerance and Survival in Patients with Advanced Pancreatic Cancer. Association between Adaptive Immunity and Nutritional State. Curr Oncol 2021; 28:3280-3296. [PMID: 34449579 PMCID: PMC8395458 DOI: 10.3390/curroncol28050285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 01/14/2023] Open
Abstract
Pancreatic Carcinoma (PC) cells have the ability to induce patient immunosuppression and to escape immunosurveillance. Low circulating lymphocytes are associated with an advanced stage of PC and reduced survival. Blood lymphocytes expressed as a percentage of Total White Blood Cells (L% TWBC) could predict chemotolerance (n° of tolerated cycles), survival time and Body Weight (BW) more effectively than lymphocytes expressed as an absolute value (LAB > 1500 n°/mm3) or lymphocytes >22%, which is the lowest limit of normal values in our laboratory. Forty-one patients with advanced PC, treated with chemotherapy, were selected for this observational retrospective study. Patients were evaluated at baseline (pre-chemotherapy), and at 6, 12 and 18 months, respectively, after diagnosis of PC. The study found L ≥ 29.7% to be a better predictor of survival (COX model, using age, sex, BW, serum creatinine, bilirubin and lymphocytes as covariates), chemotolerance (r = +0.50, p = 0.001) and BW (r = +0.35, p = 0.027) than LAB > 1500 or L > 22%. BW did not significantly correlate with chemotolerance or survival. The preliminary results of this study suggest that L ≥ 29.7% is more effective than LAB > 1500 or L > 22% at predicting chemotolerance, survival time and nutritional status. A possible impact of nutritional status on chemotherapy and survival seems to be lymphocyte-mediated given the association between BW and L%. This study may serve as the basis for future research to explore whether nutritional interventions can improve lymphopenia, and if so, how this may be possible.
Collapse
Affiliation(s)
- Roberto Aquilani
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (R.A.); (D.B.); (M.D.); (M.V.)
| | - Silvia Brugnatelli
- Medical Oncology Division, Fondazione IRCCS, Policlinico San Matteo, 27100 Pavia, Italy; (S.B.); (B.F.); (L.P.)
| | - Roberto Maestri
- Department of Biomedical Engineering of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, 27040 Montescano, Italy;
| | - Federica Boschi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Beatrice Filippi
- Medical Oncology Division, Fondazione IRCCS, Policlinico San Matteo, 27100 Pavia, Italy; (S.B.); (B.F.); (L.P.)
| | - Lorenzo Perrone
- Medical Oncology Division, Fondazione IRCCS, Policlinico San Matteo, 27100 Pavia, Italy; (S.B.); (B.F.); (L.P.)
| | - Annalisa Barbieri
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Daniela Buonocore
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (R.A.); (D.B.); (M.D.); (M.V.)
| | - Maurizia Dossena
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (R.A.); (D.B.); (M.D.); (M.V.)
| | - Manuela Verri
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (R.A.); (D.B.); (M.D.); (M.V.)
| |
Collapse
|
12
|
Nelde A, Rammensee HG, Walz JS. The Peptide Vaccine of the Future. Mol Cell Proteomics 2021; 20:100022. [PMID: 33583769 PMCID: PMC7950068 DOI: 10.1074/mcp.r120.002309] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/27/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022] Open
Abstract
The approach of peptide-based anticancer vaccination has proven the ability to induce cancer-specific immune responses in multiple studies for various cancer entities. However, clinical responses remain so far limited to single patients and broad clinical applicability was not achieved. Therefore, further efforts are required to improve peptide vaccination in order to integrate this low-side-effect therapy into the clinical routine of cancer therapy. To design clinically effective peptide vaccines in the future, different issues have to be addressed and optimized comprising antigen target selection as well as choice of optimal adjuvants and vaccination schedules. Furthermore, the combination of peptide-based vaccines with other immuno- and molecular targeted therapies as well as the development of predictive biomarkers could further improve efficacy. In this review, current approaches in the development of peptide-based vaccines and critical implications for optimal vaccine design are discussed.
Collapse
Affiliation(s)
- Annika Nelde
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), University Hospital Tübingen, Tübingen, Germany; Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany
| | - Juliane S Walz
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), University Hospital Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany.
| |
Collapse
|
13
|
Du H, Xu T, Cui M. cGAS-STING signaling in cancer immunity and immunotherapy. Biomed Pharmacother 2020; 133:110972. [PMID: 33254021 DOI: 10.1016/j.biopha.2020.110972] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/25/2020] [Accepted: 11/01/2020] [Indexed: 12/17/2022] Open
Abstract
Recent studies have shown that the innate immune cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway may play an important role in antitumor immunity. Additionally, the cGAS-STING pathway promotes the senescence of cancer cells, induces apoptosis of cancer cells, and increases the protective effect of cytotoxic T cells and natural killer cell-mediated cytotoxicity. We believe that the combination of the cGAS-STING signaling pathway with other therapeutic methods provides a new perspective from which to overcome obstacles in the application of this review. Further, we highlight the antitumor mechanism of the cGAS-STING signaling pathway and the latest advances in monotherapy and combination therapy with related agonists.
Collapse
Affiliation(s)
- Huashan Du
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, No. 218, Ziqiang Road, Changchun, Jilin, 130041, People's Republic of China.
| | - Tianmin Xu
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, No. 218, Ziqiang Road, Changchun, Jilin, 130041, People's Republic of China.
| | - Manhua Cui
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, No. 218, Ziqiang Road, Changchun, Jilin, 130041, People's Republic of China.
| |
Collapse
|
14
|
Heterogeneity of Glucose Transport in Lung Cancer. Biomolecules 2020; 10:biom10060868. [PMID: 32517099 PMCID: PMC7356687 DOI: 10.3390/biom10060868] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
Increased glucose uptake is a known hallmark of cancer. Cancer cells need glucose for energy production via glycolysis and the tricarboxylic acid cycle, and also to fuel the pentose phosphate pathway, the serine biosynthetic pathway, lipogenesis, and the hexosamine pathway. For this reason, glucose transport inhibition is an emerging new treatment for different malignancies, including lung cancer. However, studies both in animal models and in humans have shown high levels of heterogeneity in the utilization of glucose and other metabolites in cancer, unveiling a complexity that is difficult to target therapeutically. Here, we present an overview of different levels of heterogeneity in glucose uptake and utilization in lung cancer, with diagnostic and therapeutic implications.
Collapse
|
15
|
Malignancy After Lung Transplantation: How to Manage Immunosuppression? Transplant Proc 2020; 52:315-320. [DOI: 10.1016/j.transproceed.2019.09.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/29/2019] [Accepted: 09/26/2019] [Indexed: 02/07/2023]
|
16
|
Prabhu AA, Kumar JP, Mandal BB, Veeranki VD. Glucose-methanol-based fed-batch fermentation for the production of recombinant human interferon gamma (rhIFN-γ) and evaluation of its antitumor potential. Biotechnol Appl Biochem 2019; 67:973-982. [PMID: 31811672 DOI: 10.1002/bab.1868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 12/02/2019] [Indexed: 11/09/2022]
Abstract
Squamous cell carcinoma (SCC) is nonmelanoma skin cancer, which is very common in patients having T-cell immunosuppressant drugs. Anticancerous agents such as cytokines showed effective response on SCC. Human interferon-gamma (hIFN-γ), a type II cytokines, are having potent antiproliferative and immunomodulatory effects. In the current study, the fed-batch cultivation of recombinant Pichia pastoris was carried out, and its effect on cell biomass production, recombinant human interferon-gamma (rhIFN-γ) production, and the overflow metabolites was estimated. P. pastoris GS115 strain coexpressed with 6-phosphogluconolactonase (SOL3) and ribulose-phosphate 3-epimerase (RPE1) gene (GS115/rhIFN-γ/SR) resulted in 60 mg L-1 of rhIFN-γ production, which was twofold higher as compared with the production from GS115/rhIFN-γ strain. The antiproliferative potential of rhIFN-γ was examined on the human squamous carcinoma (A431) cell lines. Cells treated with 80 ng mL-1 of rhIFN-γ exhibited 50% growth inhibition by enhancing the production of intracellular reactive oxygen species levels and disrupting membrane integrity. Our findings highlight a state of art process development strategy for the high-level production of rhIFN-γ and its potential application as a therapeutic drug in SCC therapy.
Collapse
Affiliation(s)
- Ashish A Prabhu
- Biochemical Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, India
| | - Jadi Praveen Kumar
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, India
| | - Biman B Mandal
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, India.,Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Venkata Dasu Veeranki
- Biochemical Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, India
| |
Collapse
|
17
|
Yu Y, Yang L, He C, Tai S, Ma C, Yang T, Wang D. Evaluation of riboflavin photochemical treatment for inactivation of HCT116 tumor cells mixed in simulative intraoperative salvage blood. Transfusion 2019; 59:3205-3213. [PMID: 31571260 PMCID: PMC6856795 DOI: 10.1111/trf.15499] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/11/2019] [Accepted: 07/13/2019] [Indexed: 02/03/2023]
Abstract
BACKGROUND Radiation and filtration have achieved satisfactory results in inactivation or removal of tumor cells mixed in salvage blood, but some drawbacks remain. This study evaluated the inactivation on HCT116 cells mixed in simulative salvage blood by riboflavin photochemical treatment. METHODS HCT116 cells were added to the whole blood to simulate contaminated salvaged blood. The mixed blood was added with riboflavin of 50 μmol/L final concentration and illuminated by ultraviolet light. The samples were divided into control group and Experimental Groups 1 (18 J/cm2 ), 2 (23.4 J/cm2 ), and 3 (28.8 J/cm2 ). An autotransfusion system (Cell Saver Elite, Haemonetics) was used to simulate the intraoperative blood salvage procedure to deal with whole blood. The apoptosis rate and tumorigenicity of HCT116 cells and the superimposed damage to red blood cells (RBCs) were evaluated. RESULTS The apoptosis rates of HCT116 in Experimental Groups 1, 2, and 3 were much higher than that in the control group. Tumor growth was found in the control group, but no tumor growth was found in the three experimental groups. The hemolysis rates in the three experimental groups were significantly higher than that in the control group, but much lower than the quality standard of RBCs at the end of preservation. The concentration of adenosine triphosphate in RBCs was comparable in the control and experimental groups. CONCLUSION Riboflavin at a 50 μmol/L final concentration and 18 J/cm2 ultraviolet illumination can effectively inactivate HCT116 cells in salvaged blood, with minimum damage to the structure and function of RBCs, and the main quality indexes of salvaged RBCs were within the standard range.
Collapse
Affiliation(s)
- Yang Yu
- The Medical School of Chinese PLA, Beijing, China.,Department of Blood Transfusion, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lu Yang
- Department of Blood Transfusion, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chunyu He
- Department of Blood Transfusion, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shengfei Tai
- Department of Blood Transfusion, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chunya Ma
- Department of Blood Transfusion, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Tianxin Yang
- Department of Blood Transfusion, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Deqing Wang
- The Medical School of Chinese PLA, Beijing, China.,Department of Blood Transfusion, First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
18
|
Ghaemdoust F, Keshavarz-Fathi M, Rezaei N. Natural killer cells and cancer therapy, what we know and where we are going. Immunotherapy 2019; 11:1231-1251. [DOI: 10.2217/imt-2019-0040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells are among the significant components of innate immune system and they have come to the first line of defense against tumor cells developing inside the body. CD56lo/CD16+ NK cells are highly cytotoxic and CD56hi NK cells can produce cytokines and perform a regulatory function. Specific features of NK cells have made them a unique choice for cancer immunotherapy. Simple interventions like cytokine-injection to boost the internal NK cells were the first trials to target these cells. Nowadays, many other types of intervention are under investigation, such as adoptive NK cell immunotherapy. In this paper, we will discuss the biology and function of NK cells in cancer immunosurveillance and therapeutic approaches against cancer via using NK cells.
Collapse
Affiliation(s)
- Faezeh Ghaemdoust
- School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, 1419733151, Iran
| | - Mahsa Keshavarz-Fathi
- School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, 1419733151, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, 1419733151, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, 1419733151, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| |
Collapse
|
19
|
Soh JE, Abu N, Sagap I, Mazlan L, Yahaya A, Mustangin M, Khoo TS, Saidin S, Ishak M, Ab Mutalib NS, Jamal R. Validation of immunogenic PASD1 peptides against HLA-A*24:02 colorectal cancer. Immunotherapy 2019; 11:1205-1219. [PMID: 31478431 DOI: 10.2217/imt-2019-0073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Colorectal cancer is the third commonest malignancy in Asia including Malaysia. The immunogenic cancer-testis antigens, which are expressed in a variety of cancers but with limited expression in normal tissues except the testis, represent an attractive approach to improve treatment options for colorectal cancer. We aimed to validate four PASD1 peptides as the immunotherapeutic targets in colorectal cancer. First, PASD1 mRNA and protein expression were determined via real-time polymerase chain reaction (RT-PCR) and immunohistochemistry. The PASD1 peptides specific to HLA-A*24:02 were investigated using IFN-y-ELISpot assay, followed by the cytolytic and granzyme-B-ELISpot assays to analyze the cytolytic effects of CD8+ T cells. Gene and protein expressions of PASD1 were detected in 20% and 17.3% of colorectal cancer samples, respectively. PASD1(4) peptide was shown to be immunogenic in colorectal cancer samples. CD8+ T cells raised against PASD1(4) peptide were able to lyze HLA-A*24:02+ PASD1+ cells. Our results reveal that PASD1(4) peptide represents a potential target for colorectal cancer.
Collapse
Affiliation(s)
- Joanne Ec Soh
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nadiah Abu
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ismail Sagap
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Luqman Mazlan
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Azyani Yahaya
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Muaatamarulain Mustangin
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Tze S Khoo
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Sazuita Saidin
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Muhiddin Ishak
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nurul S Ab Mutalib
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Rahman Jamal
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
20
|
Tai JA, Chang CY, Nishikawa T, Kaneda Y. Cancer immunotherapy using the Fusion gene of Sendai virus. Cancer Gene Ther 2019; 27:498-508. [PMID: 31383952 DOI: 10.1038/s41417-019-0126-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 07/17/2019] [Accepted: 07/20/2019] [Indexed: 12/17/2022]
Abstract
Inactivated Sendai virus particle (or hemagglutinating virus of Japan envelope; HVJ-E) has been previously reported to possess antitumour properties that activate antitumour immunity. Two glycoproteins, fusion (F) and hemagglutinin-neuraminidase (HN), are present on the surface of HVJ-E. HN is necessary for binding to receptors such as acidic gangliosides, and F induces membrane fusion by associating with membrane lipids. We previously reported that liposomes reconstituted with F but not HN showed antitumour activity by inducing IL-6 secretion in dendritic cells (DCs), suggesting that F protein is capable of eliciting antitumour activity. Here, we attempted to deliver F gene into tumour tissue in mice by electroporation and demonstrated that F gene therapy retarded tumour growth, increased CD4+ and CD8+ T-cell infiltration into tumours and induced tumour-specific IFN-γ T-cell response. However, neutralisation of IL-6R signalling did not impact F plasmid-mediated antitumour effect. Instead, we found that F plasmid treatment resulted in a significant increase in the secretion of the chemokine RANTES (regulated upon activation, normal T cell expressed and secreted) by tumour-infiltrating T cells. Neutralising antibody against RANTES abolished the antitumour effect of F plasmid treatment in a dose-dependent manner. Thus, F gene therapy may show promise as a novel therapeutic for single or combined cancer immunotherapy.
Collapse
Affiliation(s)
- Jiayu A Tai
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Chin Yang Chang
- Department of Device Application for Molecular Therapeutics, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Tomoyuki Nishikawa
- Department of Device Application for Molecular Therapeutics, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Yasufumi Kaneda
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
21
|
Zamkova M, Kalinina A, Silaeva Y, Persiyantseva N, Bruter A, Deikin A, Khromykh L, Kazansky D. Dominant role of the α-chain in rejection of tumor cells bearing a specific alloantigen in TCRα transgenic mice and in in vitro experiments. Oncotarget 2019; 10:4808-4821. [PMID: 31448049 PMCID: PMC6690675 DOI: 10.18632/oncotarget.27093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/29/2019] [Indexed: 12/17/2022] Open
Abstract
Both TCRα and TCRβ types of T-cell receptors contribute to antigen recognition. However, some TCRs have chain centricity, which means that either the α-chain or the β-chain dictates the peptide–MHC complex specificity. Most earlier reports investigated the role of well-studied β-chains in antigen recognition by TCRαβ. In a previous study, we identified TCRs specific to the H-2Kb molecule. In the present work, we generated transgenic mice carrying the α-chain of this TCR. We found that these transgenic mice rejected EL-4 tumor cells bearing alloantigen H-2Kb more effectively than wild-type mice and similarly to mice with established specific memory T cells. Moreover, we found that T cells transduced with this TCRα can inhibit EL-4 cell growth in vitro and in vivo. We also found that transgenic mice recruit fewer CD8 T cells into the peritoneal cavity at the peak of the immune response and had a significantly higher number of central memory CD8 T cells in the spleen of intact transgenic mice compared to intact wild-type control. These results indicate the ability of a single transgenic α-chain of the H-2Kb-specific TCR to determine specific recognition of the H-2Kb molecule by a repertoire of T lymphocytes and to rapidly reject H-2Kb-bearing lymphoma cells.
Collapse
Affiliation(s)
- Maria Zamkova
- "N. N. Blokhin National Medical Research Centre of Oncology" of the Health Ministry of Russia, Moscow, Russia
| | - Anastasiya Kalinina
- "N. N. Blokhin National Medical Research Centre of Oncology" of the Health Ministry of Russia, Moscow, Russia
| | - Yuliya Silaeva
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Nadezhda Persiyantseva
- "N. N. Blokhin National Medical Research Centre of Oncology" of the Health Ministry of Russia, Moscow, Russia
| | - Alexandra Bruter
- Russian Academy of Sciences, Engelhardt Institute of Molecular Biology, Moscow, Russia
| | - Alexey Deikin
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Ludmila Khromykh
- "N. N. Blokhin National Medical Research Centre of Oncology" of the Health Ministry of Russia, Moscow, Russia
| | - Dmitry Kazansky
- "N. N. Blokhin National Medical Research Centre of Oncology" of the Health Ministry of Russia, Moscow, Russia
| |
Collapse
|
22
|
Ahel J, Hudorović N, Vičić-Hudorović V, Nikles H. TGF-BETA IN THE NATURAL HISTORY OF PROSTATE CANCER. Acta Clin Croat 2019; 58:128-138. [PMID: 31363335 PMCID: PMC6629207 DOI: 10.20471/acc.2019.58.01.17] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
All transforming growth factors beta (TGFß) are cytokines that regulate several cellular functions such as cell growth, differentiation and motility. They may also have a role in immunosuppression. Their role is important for normal prostate development. TGFß is active in the regulation of balance between epithelial cell proliferation and apoptosis through stromal epithelia via the androgen receptor action. TGFß protects and maintains prostate stem cells, an important population necessary for prostate tissue regeneration. However, TGFß is shown to have a contrasting role in prostate tumor genesis. In the early stages of tumor development, TGFß acts as a tumor suppressor, whereas in the later stages, TGFß becomes a tumor promoter by inducing proliferation, invasion and metastasis. In this review, we outline complex interactions that TGFß-mediated signaling has on prostate tumor genesis, focusing on the role of these interactions during the course of prostate cancer and, in particular, during disease progression.
Collapse
Affiliation(s)
| | - Narcis Hudorović
- 1Dr Zaky Polyclinic for Internal Medicine and Urology, Zagreb, Croatia; 2Department of Vascular Surgery, Sestre milosrdnice University Hospital Centre, Zagreb, Croatia; 3Croatian Nursing Association, Zagreb, Croatia; 4Department of Abdominal Surgery, Sestre milosrdnice University Hospital Centre, Zagreb, Croatia
| | - Višnja Vičić-Hudorović
- 1Dr Zaky Polyclinic for Internal Medicine and Urology, Zagreb, Croatia; 2Department of Vascular Surgery, Sestre milosrdnice University Hospital Centre, Zagreb, Croatia; 3Croatian Nursing Association, Zagreb, Croatia; 4Department of Abdominal Surgery, Sestre milosrdnice University Hospital Centre, Zagreb, Croatia
| | - Hrvoje Nikles
- 1Dr Zaky Polyclinic for Internal Medicine and Urology, Zagreb, Croatia; 2Department of Vascular Surgery, Sestre milosrdnice University Hospital Centre, Zagreb, Croatia; 3Croatian Nursing Association, Zagreb, Croatia; 4Department of Abdominal Surgery, Sestre milosrdnice University Hospital Centre, Zagreb, Croatia
| |
Collapse
|
23
|
Konno-Kumagai T, Fujishima F, Nakamura Y, Nakano T, Nagai T, Kamei T, Sasano H. Programmed death-1 ligands and tumor infiltrating T lymphocytes in primary and lymph node metastasis of esophageal cancer patients. Dis Esophagus 2019; 32:5066745. [PMID: 30085020 DOI: 10.1093/dote/doy063] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neoadjuvant chemotherapy (NAC) is administered to many patients with esophageal squamous cell carcinoma (ESCC) prior to surgery, but it is also true that some of these patients demonstrated no response to the therapy following surgery. In addition, the prognosis of advanced case such as ESCC patients with lymph node metastasis has remained relatively low. Programmed death ligand-1 (PD-L1) in conjunction with tumor-infiltrating lymphocytes (TILs) has been studied as a potential mechanism of "immune escape" in several human malignancies. Therefore, in this study, we studied PD-L1 status in carcinoma cells and forkhead box protein 3 (FOXP3) and CD8 status among TILs in the residual tumors of primary and metastatic sites following NAC. We also studied the association of these factors with the clinicopathological findings in 44 patients with ESCC harboring lymph node metastasis. There was discordance in the pathological response to chemotherapy between the primary tumor and lymph node metastasis, and histologically identified resistance to NAC in lymph node metastases tended to be correlated with an adverse clinical outcome (P = 0.0765) than resistance in the primary tumor. Both univariate and multivariate analyses for disease-specific survival (DSS) revealed that the PD-L1 status of carcinoma cells in metastatic lymph nodes and a higher FOXP3/CD8 ratio in the primary tumor were both significantly correlated with an eventual adverse clinical outcome of the patients (P = 0.0178, P = 0.0463, respectively). These results all indicated that the PD-L1 status of carcinoma cells in metastatic lymph nodes and the FOXP3/CD8 ratio in primary tumors could predict eventual clinical outcomes in ESCC patients with NAC.
Collapse
Affiliation(s)
- T Konno-Kumagai
- Division of Advanced Surgical Science and Technology, Tohoku University Graduate School of Medicine.,Department of Pathology, Tohoku University Hospital
| | - F Fujishima
- Department of Pathology, Tohoku University Hospital
| | - Y Nakamura
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University
| | - T Nakano
- Division of Advanced Surgical Science and Technology, Tohoku University Graduate School of Medicine
| | - T Nagai
- School of Medicine, Tohoku University, Miyagi, Japan
| | - T Kamei
- Division of Advanced Surgical Science and Technology, Tohoku University Graduate School of Medicine
| | - H Sasano
- Department of Pathology, Tohoku University Hospital
| |
Collapse
|
24
|
Mousavi-Niri N, Naseroleslami M, Hadjati J. Anti-regulatory T cell vaccines in immunotherapy: focusing on FoxP3 as target. Hum Vaccin Immunother 2019; 15:620-624. [PMID: 30633616 PMCID: PMC6605713 DOI: 10.1080/21645515.2018.1545625] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/01/2018] [Accepted: 11/03/2018] [Indexed: 12/29/2022] Open
Abstract
Anti- tumor vaccination elicits imperfect immune responses against tumor cells; that is related to the presence of suppressive obstacles in the tumor microenvironment. The main members of suppressive milieu of tumor are heteroogenous groups of immune cells in which regulatory T cell is a substantial component. Tregs express different immunomodulatory molecules such as FoxP3. Transcription factor, FoxP3, is a specific intracellular marker of Treg and crucial for Treg development. Therefore it is an attractive target for cancer treatment. This article reviews some recent anti-Treg vaccine focusing on FoxP3 to ameliorate anti-tumor immune responses. Among them, fusion vaccine of FoxP3-Fc(IgG) recombinant DNA vaccine and its accordant protein vaccine represents effective results.
Collapse
Affiliation(s)
- Neda Mousavi-Niri
- Department of Medical Biotechnology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Naseroleslami
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Jamshid Hadjati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Immunotherapy: enhancing the efficacy of this promising therapeutic in multiple cancers. Clin Sci (Lond) 2019; 133:181-193. [PMID: 30659159 DOI: 10.1042/cs20181003] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/21/2018] [Accepted: 12/28/2018] [Indexed: 12/31/2022]
Abstract
Cancer treatments often reach a refractory period leading to treatment failure and patients developing disease recurrence. This can be due to tumour cells escaping the immune response and creating an immunosuppressive microenvironment enhancing cancer progression. Immunotherapy has become a promising tool for cancer treatment as it restores the anti-tumour response of the patient's immune system. Immune checkpoint inhibitors are the most widely studied immunotherapies worldwide and are now approved for multiple cancers. However, chimeric antigen receptor (CAR)-T cell therapy has also shown promise by targeting T lymphocytes that are genetically modified ex vivo to express CARs and this is now approved to treat some haematological cancers. Although immunotherapy has shown successful treatment outcomes in multiple cancers, some patients do not respond to this treatment. Therefore, approaches to enhance the efficacy of immunotherapies are likely to be the key to improve their effectiveness. Therefore, combination therapies of checkpoint inhibitors +/- chemotherapy are at the forefront of current research. Furthermore, biomarkers that predict treatment response are now beginning to emerge. Additionally, utilising nanoparticles as a newly targeted drug delivery system to enhance CAR-T cell therapy may enhance the efficacy of the cells when re-infused within the patient. Even if efficacy is enhanced, severe immune-related adverse events (irAEs) occur that are life-threatening and could lead to therapy being stopped. Therefore, predictive biomarkers for toxicity are also needed to improve both the patient's quality of life and treatment outcomes. This review will look at the current immunotherapies in clinical trials and discuss how to enhance their efficacy.
Collapse
|
26
|
Phung CD, Nguyen HT, Tran TH, Choi HG, Yong CS, Kim JO. Rational combination immunotherapeutic approaches for effective cancer treatment. J Control Release 2018; 294:114-130. [PMID: 30553850 DOI: 10.1016/j.jconrel.2018.12.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 12/18/2022]
Abstract
Immunotherapy is an important mode of cancer treatment. Over the past decades, immunotherapy has improved the clinical outcome for cancer patients. However, in many cases, mutations in cancer cells, lack of selectivity, insufficiency of tumor-reactive T cells, and host immunosuppression limit the clinical benefit of immunotherapy. Combination approaches in immunotherapy may overcome these obstacles. Accumulating evidence demonstrates that combination immunotherapy is the future of cancer treatment. However, designing safe and rational combinations of immunotherapy with other treatment modalities is critical. This review will discuss the optimal immunotherapy-based combinations mainly with respect to the mechanisms of action of individual therapeutic agents that target multiple steps in evasion and progression of tumor.
Collapse
Affiliation(s)
- Cao Dai Phung
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Hanh Thuy Nguyen
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Tuan Hiep Tran
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Viet Nam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Viet Nam
| | - Han-Gon Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 426-791, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
27
|
Abstract
Immune checkpoint therapy represents a new, revolutionary type of cancer therapy, but emerging evidence indicates that only a minority of patients will benefit from it. The issue of how to improve and widen the clinical response is a pivotal issue, and combining other types of therapy with immune checkpoint inhibitors is currently under development. A nanotechnology-based drug-delivery system (nano DDS) could be an important contribution to the development of an effective combination therapy. In this document, we review recent findings in the field of tumor immunology, which provide a strategy for an efficient combination therapy, and discuss nano DDS that are associated with cancer immunotherapy and nano DDS strategies based on the immune status in tumor microenvironments.
Collapse
|
28
|
Abstract
Lung transplantation remains a viable option for patients with endstage pulmonary disease. Despite removing the affected organ and replacing both lungs, the risk of lung malignancies still exists. Regardless of the mode of entry, lung cancer affects the prognosis in these patients and diligence is required.
Collapse
Affiliation(s)
- Timothy Brand
- Division of Cardiothoracic Surgery, Department of Surgery, University of North Carolina, Chapel Hill, NC, USA
| | - Benjamin Haithcock
- Division of Cardiothoracic Surgery, Department of Surgery, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
29
|
Nelde A, Kowalewski DJ, Backert L, Schuster H, Werner JO, Klein R, Kohlbacher O, Kanz L, Salih HR, Rammensee HG, Stevanović S, Walz JS. HLA ligandome analysis of primary chronic lymphocytic leukemia (CLL) cells under lenalidomide treatment confirms the suitability of lenalidomide for combination with T-cell-based immunotherapy. Oncoimmunology 2018; 7:e1316438. [PMID: 29632711 DOI: 10.1080/2162402x.2017.1316438] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 03/14/2017] [Accepted: 03/31/2017] [Indexed: 02/06/2023] Open
Abstract
Recent studies suggest that CLL is an immunogenic disease, which might be effectively targeted by antigen-specific T-cell-based immunotherapy. However, CLL is associated with a profound immune defect, which might represent a critical limitation for mounting clinically effective antitumor immune responses. As several studies have demonstrated that lenalidomide can reinforce effector T-cell responses in CLL, the combination of T-cell-based immunotherapy with the immunomodulatory drug lenalidomide represents a promising approach to overcome the immunosuppressive state in CLL. Antigen-specific immunotherapy also requires the robust presentation of tumor-associated HLA-presented antigens on target cells. We thus performed a longitudinal study of the effect of lenalidomide on the HLA ligandome of primary CLL cells in vitro. We showed that lenalidomide exposure does not affect absolute HLA class I and II surface expression levels on primary CLL cells. Importantly, semi-quantitative mass spectrometric analyses of the HLA peptidome of three CLL patient samples found only minor qualitative and quantitative effects of lenalidomide on HLA class I- and II-restricted peptide presentation. Furthermore, we confirmed stable presentation of previously described CLL-associated antigens under lenalidomide treatment. Strikingly, among the few HLA ligands showing significant modulation under lenalidomide treatment, we identified upregulated IKZF-derived peptides, which may represent a direct reflection of the cereblon-mediated effect of lenalidomide on CLL cells. Since we could not observe any relevant influence of lenalidomide on the established CLL-associated antigen targets of anticancer T-cell responses, this study validates the suitability of lenalidomide for the combination with antigen-specific T-cell-based immunotherapies.
Collapse
Affiliation(s)
- Annika Nelde
- Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.,Department of Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Daniel J Kowalewski
- Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.,Immatics Biotechnologies GmbH, Tübingen, Germany
| | - Linus Backert
- Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.,Applied Bioinformatics, Center for Bioinformatics and Department of Computer Science, University of Tübingen, Tübingen, Germany
| | - Heiko Schuster
- Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.,Immatics Biotechnologies GmbH, Tübingen, Germany
| | - Jan-Ole Werner
- Department of Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Reinhild Klein
- Department of Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Oliver Kohlbacher
- Applied Bioinformatics, Center for Bioinformatics and Department of Computer Science, University of Tübingen, Tübingen, Germany.,Quantitative Biology Center, University of Tübingen, Tübingen, Germany.,Biomolecular Interactions, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Lothar Kanz
- Department of Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Helmut R Salih
- Department of Hematology and Oncology, University of Tübingen, Tübingen, Germany.,Clinical Cooperation Unit Translational Immunology, German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
| | - Hans-Georg Rammensee
- Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
| | - Stefan Stevanović
- Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
| | - Juliane S Walz
- Department of Hematology and Oncology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
30
|
Berastegui C, LaPorta R, López-Meseguer M, Romero L, Gómez-Ollés S, Riera J, Monforte V, Sáez B, Bravo C, Roman A, Ussetti P. Epidemiology and Risk Factors for Cancer After Lung Transplantation. Transplant Proc 2017; 49:2285-2291. [DOI: 10.1016/j.transproceed.2017.09.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
31
|
Kosmides AK, Sidhom JW, Fraser A, Bessell CA, Schneck JP. Dual Targeting Nanoparticle Stimulates the Immune System To Inhibit Tumor Growth. ACS NANO 2017; 11:5417-5429. [PMID: 28589725 PMCID: PMC8635119 DOI: 10.1021/acsnano.6b08152] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
We describe the development of a nanoparticle platform that overcomes the immunosuppressive tumor microenvironment. These nanoparticles are coated with two different antibodies that simultaneously block the inhibitory checkpoint PD-L1 signal and stimulate T cells via the 4-1BB co-stimulatory pathway. These "immunoswitch" particles significantly delay tumor growth and extend survival in multiple in vivo models of murine melanoma and colon cancer in comparison to the use of soluble antibodies or nanoparticles separately conjugated with the inhibitory and stimulating antibodies. Immunoswitch particles enhance effector-target cell conjugation and bypass the requirement for a priori knowledge of tumor antigens. The use of the immunoswitch nanoparticles resulted in an increased density, specificity, and in vivo functionality of tumor-infiltrating CD8+ T cells. Changes in the T cell receptor repertoire against a single tumor antigen indicate immunoswitch particles expand an effective set of T cell clones. Our data show the potential of a signal-switching approach to cancer immunotherapy that simultaneously targets two stages of the cancer immunity cycle resulting in robust antitumor activity.
Collapse
Affiliation(s)
- Alyssa K. Kosmides
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - John-William Sidhom
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Andrew Fraser
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Catherine A. Bessell
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Jonathan P. Schneck
- Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Corresponding Author:
| |
Collapse
|
32
|
Hu X, Wu T, Bao Y, Zhang Z. Nanotechnology based therapeutic modality to boost anti-tumor immunity and collapse tumor defense. J Control Release 2017; 256:26-45. [PMID: 28434891 DOI: 10.1016/j.jconrel.2017.04.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/15/2017] [Accepted: 04/18/2017] [Indexed: 12/19/2022]
Abstract
Cancer is still the leading cause of death. While traditional treatments such as surgery, chemotherapy and radiotherapy play dominating roles, recent breakthroughs in cancer immunotherapy indicate that the influence of immune system on cancer development is virtually beyond our expectation. Manipulating the immune system to fight against cancer has been thriving in recent years. Further understanding of tumor anatomy provides opportunities to put a brake on immunosuppression by overcoming tumor intrinsic resistance or modulating tumor microenvironment. Nanotechnology which provides versatile engineered approaches to enhance therapeutic effects may potentially contribute to the development of future cancer treatment modality. In this review, we will focus on the application of nanotechnology both in boosting anti-tumor immunity and collapsing tumor defense.
Collapse
Affiliation(s)
| | | | - Yuling Bao
- Tongji School of Pharmacy, PR China; Department of Pharmacy, Tongji Hospital, PR China
| | - Zhiping Zhang
- Tongji School of Pharmacy, PR China; National Engineering Research Center for Nanomedicine, PR China; Hubei Engineering Research Center for Novel Drug Delivery System, HuaZhong University of Science and Technology, Wuhan 430030, PR China.
| |
Collapse
|
33
|
Tyagi RK, Parmar R, Patel N. A generic RNA pulsed DC based approach for developing therapeutic intervention against nasopharyngeal carcinoma. Hum Vaccin Immunother 2017; 13:854-866. [PMID: 27901642 PMCID: PMC5404382 DOI: 10.1080/21645515.2016.1256518] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The recurrent nasopharyngeal carcinoma of head-and-neck cancers pathology showed unique symptoms and clinical characteristics. The complexity of pathology poses challenges for developing therapeutic interventional approaches against nasopharyngeal carcinoma (NPC). The conventional treatment regimens offer limited local control and survival, which, leads to adverse delayed complications. Our study present a generic monocyte derived dendritic cell (MoDC) vaccine strategy for NPC in which RNA is used as a source of tumor-associated antigens (TAAgs). The RNA extracted from well-characterized highly immunogenic NPC cells (C666-1) was transfected into MoDCs. The formulated and characterized cationic liposomes were used to achieving efficient RNA transfection of immature DCs. Further, DCs were forcibly matured with a cytokine cocktail to achieve greater expression of MHC and co-stimulatory molecules. Moreover, our results did not see any effect of RNA or lipids on MoDCs phenotype or cytokine expression. RNA loaded DCs derived from HLA-A2-positive donors were shown to activate effector memory cytotoxic T lymphocytes (CTLs) specific for TAAg ligand expressed by C666-1 cells. Our results show the comparison of cytotoxic response mounted against RNA-loaded DCs with those directly stimulated by C666-1 tumor cells. Our findings suggest that DCs expressing tumor cell RNA primed naïve T cells show T cells priming with lesser cytotoxicity and cytokine secretion when exposed with with C666-1 tumor cells. These results surface the potential of DCs to deliver RNA in NPCs, sufficient presentation of RNA to provoke perdurable immune responses against nasopharyngeal carcinoma. Our results implies that DC based vaccine approach may be useful to develop therapeutic interventional approach in the form of vaccine to address NPCs.
Collapse
Affiliation(s)
- Rajeev K. Tyagi
- Department of Periodontics, College of Dental Medicine, Georgia Regents University, Augusta, GA, USA
- Institute of Science, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad, Gujarat, India
- CONTACT Rajeev K. Tyagi, Ph.D. ; Department of Periodontics, College of Dental Medicine, Georgia Regents University, Augusta, GA, USA
| | - Rajesh Parmar
- Institute of Science, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad, Gujarat, India
| | - Naisargee Patel
- Institute of Science, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad, Gujarat, India
| |
Collapse
|
34
|
Lin CF, Lin CM, Lee KY, Wu SY, Feng PH, Chen KY, Chuang HC, Chen CL, Wang YC, Tseng PC, Tsai TT. Escape from IFN-γ-dependent immunosurveillance in tumorigenesis. J Biomed Sci 2017; 24:10. [PMID: 28143527 PMCID: PMC5286687 DOI: 10.1186/s12929-017-0317-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 01/19/2017] [Indexed: 12/24/2022] Open
Abstract
Immune interferon (IFN), also known as IFN-γ, promotes not only immunomodulation but also antimicrobial and anticancer activity. After IFN-γ binds to the complex of IFN-γ receptor (IFNGR) 1-IFNGR2 and subsequently activates its downstream signaling pathways, IFN-γ immediately causes transcriptional stimulation of a variety of genes that are principally involved in its biological activities. Regarding IFN-γ-dependent immunosurveillance, IFN-γ can directly suppress tumorigenesis and infection and/or can modulate the immunological status in both cancer cells and infected cells. Regarding the anticancer effects of IFN-γ, cancer cells develop strategies to escape from IFN-γ-dependent cancer immunosurveillance. Immune evasion, including the recruitment of immunosuppressive cells, secretion of immunosuppressive factors, and suppression of cytotoxic T lymphocyte responses, is speculated to be elicited by the oncogenic microenvironment. All of these events effectively downregulate IFN-γ-expressing cells and IFN-γ production. In addition to these extrinsic pathways, cancer cells may develop cellular tolerance that manifests as hyporesponsiveness to IFN-γ stimulation. This review discusses the potential escape mechanisms from IFN-γ-dependent immunosurveillance in tumorigenesis.
Collapse
Affiliation(s)
- Chiou-Feng Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan. .,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.
| | - Chih-Ming Lin
- Department of Thoracic Surgery, Lotung Poh-Ai Hospital, Yilan, 265, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, 110, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Szu-Yuan Wu
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.,Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.,Department of Radiation Oncology, Wan Fang Hospital, Taipei Medical University, Taipei Medical University, Taipei, 110, Taiwan.,Department of Biotechnology, Hung Kuang University, Taichung, 433, Taiwan
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, 110, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, 110, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Chia-Ling Chen
- Translational Research Center, Taipei Medical University, Taipei, 110, Taiwan
| | - Yu-Chih Wang
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Po-Chun Tseng
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Tsung-Ting Tsai
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| |
Collapse
|
35
|
Soliman HH. nab-Paclitaxel as a potential partner with checkpoint inhibitors in solid tumors. Onco Targets Ther 2016; 10:101-112. [PMID: 28053544 PMCID: PMC5189972 DOI: 10.2147/ott.s122974] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Tumors recognized by the host immune system are associated with better survival. However, the immune system is often suppressed in patients with established tumor burden. Stimulating the immune system to detect and kill tumor cells has been a challenge in cancer therapy for some time. Recently, novel cancer immunotherapies, such as immune checkpoint inhibitors, monoclonal antibodies, and vaccine therapies, have emerged as promising therapeutic approaches for many solid tumors. However, for some tumors, immunotherapy alone has not provided significant benefits, and some may even be fully resistant to immunotherapy. It has been suggested that the immune system may require "priming" before an immunotherapy can elicit an immune response. Although chemotherapies are believed to be immunosuppressive, when given at the right dose and sequence these agents may provide this "priming" effect for the immune system. In addition to direct cytotoxic killing of tumor cells, standard chemotherapeutic agents can elicit immunogenicity through various mechanisms. This review highlights the general immunomodulatory properties of chemotherapy agents. It also provides a rationale for combined therapy with nab-paclitaxel and immune checkpoint inhibitors. Recent clinical trial data with these combination regimens in solid tumors are presented, along with a summary of ongoing trials.
Collapse
Affiliation(s)
- Hatem H Soliman
- Department of Oncologic Sciences, Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL, USA
| |
Collapse
|
36
|
Sato Y, Sakurai Y, Kajimoto K, Nakamura T, Yamada Y, Akita H, Harashima H. Innovative Technologies in Nanomedicines: From Passive Targeting to Active Targeting/From Controlled Pharmacokinetics to Controlled Intracellular Pharmacokinetics. Macromol Biosci 2016; 17. [PMID: 27797146 DOI: 10.1002/mabi.201600179] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/19/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Yusuke Sato
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Yu Sakurai
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Kazuaki Kajimoto
- Health Research Institute; National Institute of Advanced Industrial Science and Technology (AIST); 2217-14 Hayashi-cho Takamatsu, Kagawa 761-0395 Japan
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Hidetaka Akita
- Graduate School of Pharmaceutical Sciences; Chiba University; 1-8-1 Inohana Chuo-ku, Chiba-shi, Chiba 260-8675 Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| |
Collapse
|
37
|
Sato Y, Nakamura T, Yamada Y, Harashima H. Development of a multifunctional envelope-type nano device and its application to nanomedicine. J Control Release 2016; 244:194-204. [PMID: 27374187 DOI: 10.1016/j.jconrel.2016.06.042] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/21/2016] [Accepted: 06/28/2016] [Indexed: 02/06/2023]
Abstract
Successful nanomedicines should be based on sound drug delivery systems (DDS) the permit intracellular trafficking as well as the biodistribution of cargos to be controlled. We have been developing new types of DDS that are multifunctional envelope-type nano devices referred to as MENDs. First, we will focus the in vivo delivery of siRNA to hepatocytes using a YSK-MEND which is composed of pH-responsive cationic lipids. The YSK-MEND is capable of inducing efficient silencing activity in hepatocytes and can be used to cure mice that are infected with hepatitis C or B. The YSK-MEND can also be applied to cancer immunotherapy through the activation of immune cells by delivering different compounds such as cyclic-di-GMP, siRNA or alpha-galactosylceramide as a lipid antigen. The findings indicate that, as predicted, these compounds, when encapsulated in the YSK-MEND, can be delivered to the site of action and induced immune activation through different mechanisms. Finally, a MITO-Porter, a membrane fusion-based delivery system to mitochondria, is introduced as an organelle targeting DDS and a new strategy for cancer therapy is proposed by delivering gentamicin to mitochondria of cancer cells. These new technologies are expected to extend the therapeutic area of Nanomedicine by increasing the power of DDS, especially from the view point of controlled intracellular trafficking.
Collapse
Affiliation(s)
- Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
38
|
Mahasa KJ, Ouifki R, Eladdadi A, Pillis LD. Mathematical model of tumor-immune surveillance. J Theor Biol 2016; 404:312-330. [PMID: 27317864 DOI: 10.1016/j.jtbi.2016.06.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 06/09/2016] [Accepted: 06/09/2016] [Indexed: 12/26/2022]
Abstract
We present a novel mathematical model involving various immune cell populations and tumor cell populations. The model describes how tumor cells evolve and survive the brief encounter with the immune system mediated by natural killer (NK) cells and the activated CD8(+) cytotoxic T lymphocytes (CTLs). The model is composed of ordinary differential equations describing the interactions between these important immune lymphocytes and various tumor cell populations. Based on up-to-date knowledge of immune evasion and rational considerations, the model is designed to illustrate how tumors evade both arms of host immunity (i.e. innate and adaptive immunity). The model predicts that (a) an influx of an external source of NK cells might play a crucial role in enhancing NK-cell immune surveillance; (b) the host immune system alone is not fully effective against progression of tumor cells; (c) the development of immunoresistance by tumor cells is inevitable in tumor immune surveillance. Our model also supports the importance of infiltrating NK cells in tumor immune surveillance, which can be enhanced by NK cell-based immunotherapeutic approaches.
Collapse
Affiliation(s)
- Khaphetsi Joseph Mahasa
- DST/NRF Centre of Excellence in Epidemiological Modelling and Analysis (SACEMA), University of Stellenbosch, Stellenbosch, South Africa.
| | - Rachid Ouifki
- DST/NRF Centre of Excellence in Epidemiological Modelling and Analysis (SACEMA), University of Stellenbosch, Stellenbosch, South Africa
| | | | | |
Collapse
|
39
|
Programmed cell death protein 1 expression is an independent prognostic factor in gastric cancer after curative resection. Gastric Cancer 2016. [PMID: 26210691 DOI: 10.1007/s10120-015-0519-7] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Programmed cell death protein 1 (PD-1) and its ligand PD-L1 downregulate T cell activation and are related to immune tolerance. The aim of this study was to clarify the significance of PD-1 and PD-L1 expression and to analyze the relationships among PD-1, PD-L1, and Foxp3 expression in gastric cancer. METHODS A total of 105 patients who underwent curative gastrectomy for stage II/III gastric cancer were included in this study. PD-1, PD-L1, and Foxp3 expression were examined by immunohistochemistry and related to prognostic factors by univariate and multivariate analyses. RESULTS PD-1 expression was correlated with both PD-L1 and Foxp3 expression. Disease-free survival (DFS) was significantly poorer in PD-1-positive patients than in PD-1-negative patients (3-year DFS, 36.1 % vs. 64.7 %, respectively; p < 0.05). Overall survival also tended to be poorer in PD-L1-positive patients than in PD-L1-negative patients. Univariate analysis identified sex, T factor, lymphatic invasion, and PD-1 positivity as significant predictors of poor DFS. Multivariate analysis confirmed male sex, lymphatic invasion, and positive PD-1 expression as independent prognostic indicators. CONCLUSIONS PD-1 expression is associated with a poor prognosis and is correlated with PD-L1 and Foxp3 expression in patients with gastric cancer.
Collapse
|
40
|
Rainone V, Martelli C, Ottobrini L, Biasin M, Borelli M, Lucignani G, Trabattoni D, Clerici M. Immunological Characterization of Whole Tumour Lysate-Loaded Dendritic Cells for Cancer Immunotherapy. PLoS One 2016; 11:e0146622. [PMID: 26795765 PMCID: PMC4721657 DOI: 10.1371/journal.pone.0146622] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/18/2015] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Dendritic cells play a key role as initiators of T-cell responses, and even if tumour antigen-loaded dendritic cells can induce anti-tumour responses, their efficacy has been questioned, suggesting a need to enhance immunization strategies. MATHERIALS & METHODS We focused on the characterization of bone marrow-derived dendritic cells pulsed with whole tumour lysate (TAA-DC), as a source of known and unknown antigens, in a mouse model of breast cancer (MMTV-Ras). Dendritic cells were evaluated for antigen uptake and for the expression of MHC class I/II and costimulatory molecules and markers associated with maturation. RESULTS Results showed that antigen-loaded dendritic cells are characterized by a phenotypically semi-mature/mature profile and by the upregulation of genes involved in antigen presentation and T-cell priming. Activated dendritic cells stimulated T-cell proliferation and induced the production of high concentrations of IL-12p70 and IFN-γ but only low levels of IL-10, indicating their ability to elicit a TH1-immune response. Furthermore, administration of Antigen loaded-Dendritic Cells in MMTV-Ras mice evoked a strong anti-tumour response in vivo as demonstrated by a general activation of immunocompetent cells and the release of TH1 cytokines. CONCLUSION Data herein could be useful in the design of antitumoral DC-based therapies, showing a specific activation of immune system against breast cancer.
Collapse
Affiliation(s)
- Veronica Rainone
- Department of Biomedical and Clinical Sciences “L. Sacco”, Chair of Immunology, University of Milan, Milan, Italy
| | - Cristina Martelli
- Department of Pathophysiology and Transplantation, University of Milan, Segrate, Milan, Italy
- Centre of Molecular and Cellular Imaging—IMAGO, University of Milan, Milan, Italy
| | - Luisa Ottobrini
- Department of Pathophysiology and Transplantation, University of Milan, Segrate, Milan, Italy
- Centre of Molecular and Cellular Imaging—IMAGO, University of Milan, Milan, Italy
- Institute for Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Segrate, Milan, Italy
| | - Mara Biasin
- Department of Biomedical and Clinical Sciences “L. Sacco”, Chair of Immunology, University of Milan, Milan, Italy
| | - Manuela Borelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, Chair of Immunology, University of Milan, Milan, Italy
| | - Giovanni Lucignani
- Centre of Molecular and Cellular Imaging—IMAGO, University of Milan, Milan, Italy
- Departments of Health Sciences, University of Milan, Milan, Italy
- Department of Diagnostic Services, Unit of Nuclear Medicine, San Paolo Hospital, Milan, Italy
| | - Daria Trabattoni
- Department of Biomedical and Clinical Sciences “L. Sacco”, Chair of Immunology, University of Milan, Milan, Italy
| | - Mario Clerici
- Department of Pathophysiology and Transplantation, University of Milan, Segrate, Milan, Italy
- Don C. Gnocchi Foundation IRCCS, Milan, Italy
| |
Collapse
|
41
|
Vγ9Vδ2 T cells and zoledronate mediate antitumor activity in an orthotopic mouse model of human chondrosarcoma. Tumour Biol 2015; 37:7333-44. [DOI: 10.1007/s13277-015-4615-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/08/2015] [Indexed: 12/11/2022] Open
|
42
|
Nakamura T, Miyabe H, Hyodo M, Sato Y, Hayakawa Y, Harashima H. Liposomes loaded with a STING pathway ligand, cyclic di-GMP, enhance cancer immunotherapy against metastatic melanoma. J Control Release 2015; 216:149-57. [PMID: 26282097 DOI: 10.1016/j.jconrel.2015.08.026] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 07/16/2015] [Accepted: 08/12/2015] [Indexed: 01/31/2023]
Abstract
Malignant melanomas escape immunosurveillance via the loss/down-regulation of MHC-I expression. Natural killer (NK) cells have the potential to function as essential effector cells for eliminating melanomas. Cyclic di-GMP (c-di-GMP), a ligand of the stimulator of interferon genes (STING) signal pathway, can be thought of as a new class of adjuvant against cancer. However, it is yet to be tested, because technologies for delivering c-di-GMP to the cytosol are required. Herein, we report that c-di-GMP efficiently activates NK cells and induces antitumor effects against malignant melanomas when loaded in YSK05 lipid containing liposomes, by assisting in the efficient delivery of c-di-GMP to the cytosol. The intravenous administration of c-di-GMP encapsulated within YSK05-liposomes (c-di-GMP/YSK05-Lip) into mice efficiently induced the production of type I interferon (IFN) as well as the activation of NK cells, resulting in a significant antitumor effect in a lung metastasis mouse model using B16-F10. This antitumor effect was dominated by NK cells. The infiltration of NK cells was observed in the lungs with B16-F10 melanomas. These findings indicate that the c-di-GMP/YSK05-Lip induces MHC-I non-restricted antitumor immunity mediated by NK cells. Consequently, c-di-GMP/YSK05-Lip represents a potentially new adjuvant system for use in immunotherapy against malignant melanomas.
Collapse
Affiliation(s)
- Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Hiroko Miyabe
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Mamoru Hyodo
- Department of Applied Chemistry, Faculty of Engineering, Aichi Institute of Technology, Toyota, Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yoshihiro Hayakawa
- Department of Applied Chemistry, Faculty of Engineering, Aichi Institute of Technology, Toyota, Japan
| | | |
Collapse
|
43
|
Anzengruber F, Avci P, de Freitas LF, Hamblin MR. T-cell mediated anti-tumor immunity after photodynamic therapy: why does it not always work and how can we improve it? Photochem Photobiol Sci 2015; 14:1492-1509. [PMID: 26062987 PMCID: PMC4547550 DOI: 10.1039/c4pp00455h] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Photodynamic therapy (PDT) uses the combination of non-toxic photosensitizers and harmless light to generate reactive oxygen species that destroy tumors by a combination of direct tumor cell killing, vascular shutdown, and activation of the immune system. It has been shown in some animal models that mice that have been cured of cancer by PDT, may exhibit resistance to rechallenge. The cured mice can also possess tumor specific T-cells that recognize defined tumor antigens, destroy tumor cells in vitro, and can be adoptively transferred to protect naïve mice from cancer. However, these beneficial outcomes are the exception rather than the rule. The reasons for this lack of consistency lie in the ability of many tumors to suppress the host immune system and to actively evade immune attack. The presence of an appropriate tumor rejection antigen in the particular tumor cell line is a requisite for T-cell mediated immunity. Regulatory T-cells (CD25+, Foxp3+) are potent inhibitors of anti-tumor immunity, and their removal by low dose cyclophosphamide can potentiate the PDT-induced immune response. Treatments that stimulate dendritic cells (DC) such as CpG oligonucleotide can overcome tumor-induced DC dysfunction and improve PDT outcome. Epigenetic reversal agents can increase tumor expression of MHC class I and also simultaneously increase expression of tumor antigens. A few clinical reports have shown that anti-tumor immunity can be generated by PDT in patients, and it is hoped that these combination approaches may increase tumor cures in patients.
Collapse
Affiliation(s)
- Florian Anzengruber
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Pinar Avci
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Department of Dermatology, Dermatooncology and Venerology, Semmelweis University School of Medicine, Budapest, 1085, Hungary
| | - Lucas Freitas de Freitas
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Programa de Pos Graduacao Interunidades Bioengenharia – USP – Sao Carlos, Brazil
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
- Correspondence to: Michael R Hamblin, PhD, Wellman Center for Photomedicine, Massachusetts General Hospital, 50 Blossom Street, Boston, MA 02114, USA.
| |
Collapse
|
44
|
Lipson EJ, Sharfman WH, Chen S, McMiller TL, Pritchard TS, Salas JT, Sartorius-Mergenthaler S, Freed I, Ravi S, Wang H, Luber B, Sproul JD, Taube JM, Pardoll DM, Topalian SL. Safety and immunologic correlates of Melanoma GVAX, a GM-CSF secreting allogeneic melanoma cell vaccine administered in the adjuvant setting. J Transl Med 2015; 13:214. [PMID: 26143264 PMCID: PMC4491237 DOI: 10.1186/s12967-015-0572-3] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/11/2015] [Indexed: 02/04/2023] Open
Abstract
Background Limited adjuvant treatment options exist for patients with high-risk surgically resected melanoma. This first-in-human study investigated the safety, tolerability and immunologic correlates of Melanoma GVAX, a lethally irradiated granulocyte–macrophage colony stimulating factor (GM-CSF)-secreting allogeneic whole-cell melanoma vaccine, administered in the adjuvant setting. Methods Patients with stage IIB-IV melanoma were enrolled following complete surgical resection. Melanoma GVAX was administered intradermally once every 28 days for four cycles, at 5E7 cells/cycle (n = 3), 2E8 cells/cycle (n = 9), or 2E8 cells/cycle preceded by cyclophosphamide 200 mg/m2 to deplete T regulatory cells (Tregs; n = 8). Blood was collected before each vaccination and at 4 and 6 months after treatment initiation for immunologic studies. Vaccine injection site biopsies and additional blood samples were obtained 2 days after the 1st and 4th vaccines. Results Among 20 treated patients, 18 completed 4 vaccinations. Minimal treatment-related toxicity was observed. One patient developed vitiligo and patches of white hair during the treatment and follow-up period. Vaccine site biopsies demonstrated complex inflammatory infiltrates, including significant increases in eosinophils and PD-1+ lymphocytes from cycle 1 to cycle 4 (P < 0.05). Serum GM-CSF concentrations increased significantly in a dose-dependent manner 48 h after vaccination (P = 0.0086), accompanied by increased numbers of activated circulating monocytes (P < 0.0001) and decreased percentages of myeloid-derived suppressor cells among monocytes (CD14+ , CD11b+ , HLA-DR low or negative; P = 0.002). Cyclophosphamide did not affect numbers of circulating Tregs. No significant changes in anti-melanoma immunity were observed in peripheral T cells by interferon-gamma ELIPSOT, or immunoglobulins by serum Western blotting. Conclusion Melanoma GVAX was safe and tolerable in the adjuvant setting. Pharmacodynamic testing revealed complex vaccine site immune infiltrates and an immune-reactive profile in circulating monocytic cell subsets. These findings support the optimization of Melanoma GVAX with additional monocyte and dendritic cell activators, and the potential development of combinatorial treatment regimens with synergistic agents. Trial registration: NCT01435499 Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0572-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Evan J Lipson
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, 1550 Orleans Street, Room 507, Baltimore, MD, 21287, USA.
| | - William H Sharfman
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA.
| | - Shuming Chen
- Department of Surgery, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Tracee L McMiller
- Department of Surgery, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Theresa S Pritchard
- Department of Surgery, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - January T Salas
- Department of Surgery, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Susan Sartorius-Mergenthaler
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA.
| | - Irwin Freed
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA.
| | - Sowmya Ravi
- Department of Dermatology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Hao Wang
- Division of Biostatistics and Bioinformatics, Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Brandon Luber
- Division of Biostatistics and Bioinformatics, Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Janice Davis Sproul
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA.
| | - Janis M Taube
- Department of Dermatology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA. .,Department of Pathology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, 21287, USA.
| | - Suzanne L Topalian
- Department of Surgery, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| |
Collapse
|
45
|
Karimi S, Chattopadhyay S, Chakraborty NG. Manipulation of regulatory T cells and antigen-specific cytotoxic T lymphocyte-based tumour immunotherapy. Immunology 2015; 144:186-96. [PMID: 25243729 DOI: 10.1111/imm.12387] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 09/12/2014] [Accepted: 09/16/2014] [Indexed: 12/16/2022] Open
Abstract
The most potent killing machinery in our immune system is the cytotoxic T lymphocyte (CTL). Since the possibility for self-destruction by these cells is high, many regulatory activities exist to prevent autoimmune destruction by these cells. A tumour (cancer) grows from the cells of the body and is tolerated by the body's immune system. Yet, it has been possible to generate tumour-associated antigen (TAA) -specific CTL that are also self-antigen specific in vivo, to achieve a degree of therapeutic efficacy. Tumour-associated antigen-specific T-cell tolerance through pathways of self-tolerance generation represents a significant challenge to successful immunotherapy. CD4(+) CD25(+) FoxP3(+) T cells, referred to as T regulatory (Treg) cells, are selected in the thymus as controllers of the anti-self repertoire. These cells are referred to as natural T regulatory (nTreg) cells. According to the new consensus (Nature Immunology 2013; 14:307-308) these cells are to be termed as (tTreg). There is another class of CD4(+) Treg cells also involved in regulatory function in the periphery, also phenotypically CD4(+) CD25(±) , classified as induced Treg (iTreg) cells. These cells are to be termed as peripherally induced Treg (pTreg) cells. In vitro-induced Treg cells with suppressor function should be termed as iTreg. These different Treg cells differ in their requirements for activation and in their mode of action. The current challenges are to determine the degree of specificity of these Treg cells in recognizing the same TAA as the CTL population and to circumvent their regulatory constraints so as to achieve robust CTL responses against cancer.
Collapse
Affiliation(s)
- Shirin Karimi
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | | | | |
Collapse
|
46
|
InCVAX--a novel strategy for treatment of late-stage, metastatic cancers through photoimmunotherapy induced tumor-specific immunity. Cancer Lett 2015; 359:169-77. [PMID: 25633839 DOI: 10.1016/j.canlet.2015.01.029] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/20/2015] [Accepted: 01/21/2015] [Indexed: 12/31/2022]
Abstract
A novel, promising potential cancer vaccine strategy was proposed to use a two-injection procedure for solid tumors to prompt the immune system to identify and systemically eliminate primary and metastatic cancers. The two-injection procedure consists of local photothermal application on a selected tumor intended to liberate whole cell tumor antigens, followed by a local injection of an immunoadjuvant that consists of a semi-synthetic functionalized glucosamine polymer, N-dihydro-galacto-chitosan (GC), which is intended to activate antigen presenting cells and facilitate an increased uptake of tumor antigens. This strategy is thus proposed as an in situ autologous cancer vaccine (inCVAX) that may activate antigen presenting cells and expose them to tumor antigens in situ, with the intention of inducing a systemic tumor specific T-cell response. Here, the development of inCVAX for the treatment of metastatic cancers in the past decades is systematically reviewed. The antitumor immune responses of local photothermal treatment and immunological stimulation with GC are also discussed. This treatment approach is also commonly referred to as laser immunotherapy (LIT).
Collapse
|
47
|
Manufacture of T cells using the Sleeping Beauty system to enforce expression of a CD19-specific chimeric antigen receptor. Cancer Gene Ther 2015; 22:95-100. [PMID: 25591810 DOI: 10.1038/cgt.2014.69] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 10/20/2014] [Indexed: 01/10/2023]
Abstract
T cells can be reprogrammed to redirect specificity to tumor-associated antigens (TAAs) through the enforced expression of chimeric antigen receptors (CARs). The prototypical CAR is a single-chain molecule that docks with TAA expressed on the cell surface and, in contrast to the T-cell receptor complex, recognizes target cells independent of human leukocyte antigen. The bioprocessing to generate CAR(+) T cells has been reduced to clinical practice based on two common steps that are accomplished in compliance with current good manufacturing practice. These are (1) gene transfer to stably integrate the CAR using viral and nonviral approaches and (2) activating the T cells for proliferation by crosslinking CD3 or antigen-driven numeric expansion using activating and propagating cells (AaPCs). Here, we outline our approach to nonviral gene transfer using the Sleeping Beauty system and the selective propagation of CD19-specific CAR(+) T cells on AaPCs.
Collapse
|
48
|
Li X, Mei Q, Nie J, Fu X, Han W. Decitabine: a promising epi-immunotherapeutic agent in solid tumors. Expert Rev Clin Immunol 2015; 11:363-75. [DOI: 10.1586/1744666x.2015.1002397] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
49
|
Grewal AS, Padera RF, Boukedes S, Divo M, Rosas IO, Camp PC, Fuhlbrigge A, Goldberg H, El-Chemaly S. Prevalence and outcome of lung cancer in lung transplant recipients. Respir Med 2015; 109:427-33. [PMID: 25616348 DOI: 10.1016/j.rmed.2014.12.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 07/20/2014] [Accepted: 12/30/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Lung transplant is the only available therapy for patients with advanced lung disease. The goal of this study was to examine the prevalence, origin, management and outcome of lung cancer in recipients of lung transplant at our institution. METHODS After institutional review board approval, we conducted a retrospective chart review of all lung transplantations in our institution from January 1990 until June 2012. RESULTS The prevalence of lung cancer in the explanted lung was 6 (1.2%) of 462 and all cases were in subjects with lung fibrosis. All 4 subjects with lymph node involvement died of causes related to the malignancy. Nine (1.9%) of 462 patients were found to have bronchogenic carcinoma after lung transplant. The most common location was in the native lung in recipients of a single lung transplant (6 out of 9 patients). In one case, the tumor originated in the allograft and was potentially donor related. The median time to diagnosis after lung transplant was 28 months with a range from 9 months to 10 years. Median survival was 8 months, with tumors involving lymph nodes or distant metastases associated with a markedly worse prognosis (median survival 7 months) than stage I disease (median survival 27 months). CONCLUSIONS The prevalence of lung cancer in lung transplant recipients is low. Using accepted donor screening criteria, donor derived malignancy is exceptionally rare. While stage I disease is associated with improved survival in this cohort, survival is still not comparable to that of the general population, likely influenced by the need for aggressive immune suppression.
Collapse
Affiliation(s)
| | | | - Steve Boukedes
- Division of Pulmonary and Critical Care Medicine, Boston, MA 02115, USA
| | - Miguel Divo
- Division of Pulmonary and Critical Care Medicine, Boston, MA 02115, USA
| | - Ivan O Rosas
- Division of Pulmonary and Critical Care Medicine, Boston, MA 02115, USA
| | - Phillip C Camp
- Department of Thoracic Surgery Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Anne Fuhlbrigge
- Division of Pulmonary and Critical Care Medicine, Boston, MA 02115, USA
| | - Hilary Goldberg
- Division of Pulmonary and Critical Care Medicine, Boston, MA 02115, USA
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Boston, MA 02115, USA.
| |
Collapse
|
50
|
Conniot J, Silva JM, Fernandes JG, Silva LC, Gaspar R, Brocchini S, Florindo HF, Barata TS. Cancer immunotherapy: nanodelivery approaches for immune cell targeting and tracking. Front Chem 2014; 2:105. [PMID: 25505783 PMCID: PMC4244808 DOI: 10.3389/fchem.2014.00105] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 10/31/2014] [Indexed: 12/14/2022] Open
Abstract
Cancer is one of the most common diseases afflicting people globally. New therapeutic approaches are needed due to the complexity of cancer as a disease. Many current treatments are very toxic and have modest efficacy at best. Increased understanding of tumor biology and immunology has allowed the development of specific immunotherapies with minimal toxicity. It is important to highlight the performance of monoclonal antibodies, immune adjuvants, vaccines and cell-based treatments. Although these approaches have shown varying degrees of clinical efficacy, they illustrate the potential to develop new strategies. Targeted immunotherapy is being explored to overcome the heterogeneity of malignant cells and the immune suppression induced by both the tumor and its microenvironment. Nanodelivery strategies seek to minimize systemic exposure to target therapy to malignant tissue and cells. Intracellular penetration has been examined through the use of functionalized particulates. These nano-particulate associated medicines are being developed for use in imaging, diagnostics and cancer targeting. Although nano-particulates are inherently complex medicines, the ability to confer, at least in principle, different types of functionality allows for the plausible consideration these nanodelivery strategies can be exploited for use as combination medicines. The development of targeted nanodelivery systems in which therapeutic and imaging agents are merged into a single platform is an attractive strategy. Currently, several nanoplatform-based formulations, such as polymeric nanoparticles, micelles, liposomes and dendrimers are in preclinical and clinical stages of development. Herein, nanodelivery strategies presently investigated for cancer immunotherapy, cancer targeting mechanisms and nanocarrier functionalization methods will be described. We also intend to discuss the emerging nano-based approaches suitable to be used as imaging techniques and as cancer treatment options.
Collapse
Affiliation(s)
- João Conniot
- Faculdade de Farmácia, Instituto de Investigação do Medicamento (iMed.ULisboa), Universidade de Lisboa Lisboa, Portugal
| | - Joana M Silva
- Faculdade de Farmácia, Instituto de Investigação do Medicamento (iMed.ULisboa), Universidade de Lisboa Lisboa, Portugal
| | - Joana G Fernandes
- Faculdade de Farmácia, Instituto de Investigação do Medicamento (iMed.ULisboa), Universidade de Lisboa Lisboa, Portugal
| | - Liana C Silva
- Faculdade de Farmácia, Instituto de Investigação do Medicamento (iMed.ULisboa), Universidade de Lisboa Lisboa, Portugal
| | - Rogério Gaspar
- Faculdade de Farmácia, Instituto de Investigação do Medicamento (iMed.ULisboa), Universidade de Lisboa Lisboa, Portugal
| | - Steve Brocchini
- EPSRC Centre for Innovative Manufacturing in Emergent Macromolecular Therapies, UCL School of Pharmacy London, UK
| | - Helena F Florindo
- Faculdade de Farmácia, Instituto de Investigação do Medicamento (iMed.ULisboa), Universidade de Lisboa Lisboa, Portugal
| | - Teresa S Barata
- EPSRC Centre for Innovative Manufacturing in Emergent Macromolecular Therapies, UCL School of Pharmacy London, UK
| |
Collapse
|