1
|
Yaraghi P, Kheyri A, Mikaeili N, Boroumand A, Abbasifard M, Farhangnia P, Rezagholizadeh F, Khorramdelazad H. Nanoparticle-mediated enhancement of DNA Vaccines: Revolutionizing immunization strategies. Int J Biol Macromol 2025; 302:140558. [PMID: 39900152 DOI: 10.1016/j.ijbiomac.2025.140558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/25/2025] [Accepted: 01/30/2025] [Indexed: 02/05/2025]
Abstract
DNA vaccines are a novel form of vaccination that aims to harness genetic material to produce targeted immune responses. Nevertheless, their therapeutic application is hampered by low transfection efficacy, immunogenicity, and instability. Nanoparticle (NP) - based delivery systems are beneficial in enhancing DNA stability, increasing DNA uptake by antigen-presenting cells (APCs), and controlling antigen release. Some key progress includes the polymeric, lipid-based, and hybrid NPs and biocompatible carriers with inherent adjuvant effects. These systems have helped to enhance the antigen cross-presentation and T-cell activation significantly. In addition, biocompatible hybrid nanocarriers, antigen cross-presentation strategies, and next-generation sequencing (NGS) technologies are speeding up the identification of new antigens, while AI and machine learning are facilitating the development of efficient delivery systems. This review aims to assess how NPs have contributed to improving the effectiveness of DNA vaccines for treating diseases, cancer, and emerging diseases, as well as advancing the next generation of DNA vaccines.
Collapse
Affiliation(s)
- Pegah Yaraghi
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Abbas Kheyri
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Narges Mikaeili
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Armin Boroumand
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mitra Abbasifard
- Department of Internal Medicine, School of Medicine, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Pooya Farhangnia
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Rezagholizadeh
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
2
|
Gianneschi G, Scolpino A, Oleske J. Risk of autoimmunity, cancer seeding, and adverse events in human trials of whole-tissue autologous therapeutic vaccines. CANCER PATHOGENESIS AND THERAPY 2025; 3:129-134. [PMID: 40182122 PMCID: PMC11963168 DOI: 10.1016/j.cpt.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 04/05/2025]
Abstract
Background Whole-tissue autologous therapeutic vaccines (WATVs) are a form of cancer immunotherapy that use a patient's own pathological tissue. Concerns exist regarding the potential of WATVs to induce autoimmunity or the spread of cancer; however, their adverse events (AEs) have not been adequately studied. This literature review primarily aimed to evaluate the risks of autoimmunity and cancer seeding associated with using WATVs in human clinical trials. Its secondary objectives included assessing the incidence of AEs graded 1-5 using the Common Terminology Criteria for Adverse Events v5.0. Methods The inclusion criteria were any clinical trial using human subjects in which at least part of the cancer vaccine was derived from the patient's own tumor tissue, which likely preserved the unique tumor-associated antigens (TAAs) present in the patient's tumor (i.e., whole-tissue). Tumor vaccine trials that used limited TAAs or highly processed tumor antigens were excluded. Published clinical trials were searched using Google Scholar until March 2024. The authors elaborated on the risk of bias in such cases, as indicated. All reviewed publications were searched for evidence of autoimmunity, cancer seeding, and other AEs. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses 2020 statement guided the review. Results Data from 55 human clinical trials, abstracts, case reports, and unpublished data were analyzed, including 3323 patients treated with WATVs for various cancers. The primary outcomes were: (1) no documented cases of WATV-induced autoimmunity, (2) no documented cases of WATV-induced spreading or seeding of noninfectious cancers, and (3) the observed 0.24% (2/838) risk of spreading or seeding infectious cancers was attributed to inadequate sterilization. The secondary outcomes were: (1) no deaths were attributed to WATV therapy, (2) 0.18% (6/3323) incidence of grade 4 AEs, (3) 0.42% (14/3323) incidence of grade 3 AEs, (4) the incidence of grades 1-2 AEs was 52.21% (478/916). Conclusions WATVs carry no risk of inducing autoimmunity and essentially no risk of cancer seeding if properly sterilized. WATVs also exhibit a side effect profile comparable to that of routine vaccinations, with common, mild, and transient adverse effects. The combined risk of grade 3 and 4 AEs was 0.60% (20/3323). No deaths were causally associated with WATV treatment.
Collapse
Affiliation(s)
- Garrett Gianneschi
- Department of Neurology, Rutgers University - New Jersey Medical School, Newark, NJ 07103, USA
| | - Anthony Scolpino
- Division of Immunology, Department of Pediatrics, Rutgers University - New Jersey Medical School, Newark, NJ 07103, USA
| | - James Oleske
- Division of Immunology, Department of Pediatrics, Rutgers University - New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
3
|
Baldi GG, Gronchi A, Tazzari M, Stacchiotti S. Immunotherapy in soft tissue sarcoma: current evidence and future perspectives in a variegated family of different tumour. Expert Rev Anticancer Ther 2022; 22:491-503. [PMID: 35412415 DOI: 10.1080/14737140.2022.2065986] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION In the last few years steps forward in the knowledge of the biology of soft tissue sarcomas (STS) has led to the development of new therapeutic strategies, including immunotherapy. AREAS COVERED This review outlines the recent findings on immunological features and provides a synopsis of the results of clinical trials with different immunotherapy approaches in STS, discussing criticisms and how the efficacy of immunotherapy could be improved. EXPERT OPINION The heterogeneity of STS has limited generalized approaches of immunotherapy in the disease. Clinical decisions should encompass a comprehensive characterization of the tumour microenvironment (TME), marked by intra-histotype diversity. Profiling of immune cells, checkpoint molecules and antigen target/HLA expression is deemed to re-shape the classical histotype classification for a selection of the most appropriate immune-based treatment. In a synergistic view, tumour-directed treatments, designed on the genetic and epigenetic histotype make-up, should be monitored for their immunomodulant effect and applied to ensure or amplify immunotherapy response. In light of the dynamic nature of the TME, this immunomonitoring should be conducted at baseline and during treatment, for improved therapeutic decisions and rational sequence of treatment combination, pursuing an immunological marker approach by histotype guidance.
Collapse
Affiliation(s)
- Giacomo G Baldi
- Department of Medical Oncology, Hospital of Prato, Prato, Italy
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marcella Tazzari
- Immunotherapy, Cell Therapy and Biobank Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Silvia Stacchiotti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
4
|
Atherton MJ, Lenz JA, Mason NJ. Sarcomas-A barren immunological wasteland or field of opportunity for immunotherapy? Vet Comp Oncol 2020; 18:447-470. [PMID: 32246517 DOI: 10.1111/vco.12595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/13/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022]
Abstract
Key advances in our understanding of immunobiology and the immunosuppressive mechanisms of the tumour microenvironment have led to significant breakthroughs in manipulating the immune system to successfully treat cancer. Remarkable therapeutic responses have occurred with tumours that carry a high mutational burden. In these cases, pre-existing tumour-specific T cells can be rejuvenated via checkpoint inhibition to eliminate tumours. Furthermore, durable remissions have been achieved in haematological malignancies following adoptive transfer of T cells that specifically target cell surface proteins where expression is restricted to the malignancy's cell of origin. Soft tissue sarcomas and bone sarcomas have a paucity of non-synonymous somatic mutations and do not commonly express known, targetable, tumour-specific antigens. Historically, soft tissue sarcomas have been considered immunologically 'cold' and as such, unlikely candidates for immune therapy. Here, we review the immune landscape of canine and feline sarcomas and the immunotherapeutic strategies that have been employed in veterinary clinical trials to improve patient outcome. We also provide insight into immunotherapeutic approaches being used to treat human sarcomas. Together, current data indicates that, rather than a barren immunological wasteland, sarcomas represent a field of opportunities for immunotherapies. Furthermore, we and others would suggest that strategic combinations of immunotherapeutic approaches may hold promise for more effective treatments for high grade soft tissue sarcomas and bone sarcomas.
Collapse
Affiliation(s)
- Matthew J Atherton
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jennifer A Lenz
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nicola J Mason
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
Abstract
Soft-tissue sarcoma (sts) is a rare mesenchymal malignancy that accounts for less than 1% of all adult tumours. Despite the successful advancement of localized therapies such as surgery and radiotherapy, these tumours can, for many, recur-often with metastatic disease. In the advanced setting, the role of systemic therapies is modest and is associated with poor survival. With the discovery of immunotherapies in other tumour types such as melanoma and lung cancer, interest has been renewed in exploring immunotherapy in sts. The biology of some stss makes them ripe for immunotherapy intervention; for example, some stss might have chromosomal translocations resulting in pathognomonic fusion products that have been shown to express cancer/testis antigens. Here, we present a targeted review of the published data and ongoing clinical trials for immunotherapies in patients with sarcoma, which comprise immune checkpoint inhibitors, adoptive cell therapies, and cancer vaccines.
Collapse
Affiliation(s)
- O Ayodele
- Toronto Sarcoma Program, Princess Margaret Cancer Centre and Mount Sinai Hospital, Toronto, ON
| | - A R Abdul Razak
- Toronto Sarcoma Program, Princess Margaret Cancer Centre and Mount Sinai Hospital, Toronto, ON
| |
Collapse
|
6
|
Nathenson MJ, Conley AP, Sausville E. Immunotherapy: A New (and Old) Approach to Treatment of Soft Tissue and Bone Sarcomas. Oncologist 2017; 23:71-83. [PMID: 28935774 DOI: 10.1634/theoncologist.2016-0025] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 07/14/2017] [Indexed: 12/11/2022] Open
Abstract
Soft tissue and bone sarcomas are a rare and heterogeneous form of cancer. With standard of care treatment options including surgery, radiation, and chemotherapy, the long-term survival is still low for high-risk soft tissue sarcoma patients. New treatment strategies are needed. Immunotherapy offers a new potential treatment paradigm with great promise. Immunotherapy of soft tissue sarcomas dates back to Dr. Coley's first use of toxins in the late 1800s. A variety of strategies of immunotherapy have been tried in soft tissue and bone sarcomas, including various vaccines and cytokines, with limited success. Results of these early clinical trials with vaccines and cytokines were disappointing, but there are reasons to be optimistic. Recent advances, particularly with the use of adoptive T-cell therapy and immune checkpoint inhibitors, have led to a resurgence of this field for all cancer patients. Clinical trials utilizing adoptive T-cell therapy and immune checkpoint inhibitors in soft tissue and bone sarcomas are under way. This paper reviews the current state of evidence for the use of immunotherapy, as well as current immunotherapy strategies (vaccines, adopative T-cell therapy, and immune checkpoint blockade), in soft tissue and bone sarcomas. By understanding the tumor microenviroment of sarcomas and how it relates to their immunoresponsiveness, better immunotherapy clinical trials can be designed, hopefully with improved outcomes for soft tissue and bone sarcoma patients. IMPLICATIONS FOR PRACTICE Immunotherapy is a promising treatment paradigm that is gaining acceptance for the management of several cancers, including melanoma, renal cell carcinoma, prostate cancer, and lung cancer. There is a long history of immunotherapy in the treatment of soft tissue and bone sarcomas, although with little success. It is important to understand past failures to develop future immunotherapy treatment strategies with an improved possibility of success. This article reviews the history of and current state of immunotherapy research in the treatment of soft tissue and bone sarcomas, with particular regard to vaccine trials, adoptive T-cell therapy, and immune checkpoint blockade.
Collapse
Affiliation(s)
- Michael J Nathenson
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Anthony P Conley
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Edward Sausville
- Department of Medicine and Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Ghosn M, El Rassy E, Kourie HR. Immunotherapies in sarcoma: Updates and future perspectives. World J Clin Oncol 2017; 8:145-150. [PMID: 28439495 PMCID: PMC5385435 DOI: 10.5306/wjco.v8.i2.145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/15/2016] [Accepted: 01/16/2017] [Indexed: 02/06/2023] Open
Abstract
Sarcomas are malignant tumors that are characterized by a wide diversity of subtypes with various cytogenetic profiles. Despite major treatment breakthroughs, standard treatment modalities combining chemotherapy, radiotherapy, and surgery failed to improve overall survival. Therefore, high expectations are foreseen with immunotherapy upon its maturation and better understanding of its mechanism of action. This paper presents a targeted review of the published data and ongoing clinical trials in immunotherapies of sarcomas, mainly adoptive cell therapies, cancer vaccines and immune checkpoint inhibitors.
Collapse
|
8
|
Current Immunotherapies for Sarcoma: Clinical Trials and Rationale. Sarcoma 2016; 2016:9757219. [PMID: 27703409 PMCID: PMC5039267 DOI: 10.1155/2016/9757219] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 08/24/2016] [Indexed: 12/21/2022] Open
Abstract
Sarcoma tumors are rare and heterogeneous, yet they possess many characteristics that may facilitate immunotherapeutic responses. Both active strategies including vaccines and passive strategies involving cellular adoptive immunotherapy have been applied clinically. Results of these clinical trials indicate a distinct benefit for select patients. The recent breakthrough of immunologic checkpoint inhibition is being rapidly introduced to a variety of tumor types including sarcoma. It is anticipated that these emerging immunotherapies will exhibit clinical efficacy for a variety of sarcomas. The increasing ability to tailor immunologic therapies to sarcoma patients will undoubtedly generate further enthusiasm and clinical research for this treatment modality.
Collapse
|
9
|
Abstract
Immunologic approaches to cancer are over a century old. Over the years, the strategy has been fine-tuned from inciting infections in subjects to inhibiting negative regulatory signals from the innate immune system. Sarcomas are among the first tumors to be considered for immune interventions. From Coley's toxin to cytokine-based therapies to adoptive cell therapy, there have been numerous immunotherapeutic investigations in this patient population. A promising strategy includes adoptive T cell therapy which has been studied in small cohorts of synovial sarcoma, a subtype that is known to widely express the cancer testis antigen, NY-ESO-1. Additionally, recent data in metastatic melanoma and renal cell carcinoma demonstrate the utility and tremendous efficacy of immune checkpoint blockade with increased rates of durable responses compared to standard therapies. Responses in traditionally "non-immunogenic" tumors, such as lung and bladder cancers, provide ample rationale for the study of immune checkpoint inhibitors in sarcoma. While immunotherapy has induced some responses in sarcomas, further research will help clarify optimal patient selection for future clinical trials and new combinatorial immunotherapeutic strategies.
Collapse
|
10
|
Tseng WW, Somaiah N, Engleman EG. Potential for immunotherapy in soft tissue sarcoma. Hum Vaccin Immunother 2015; 10:3117-24. [PMID: 25625925 DOI: 10.4161/21645515.2014.983003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Soft tissue sarcomas (STS) are rare, heterogeneous tumors of mesenchymal origin. Despite optimal treatment, a large proportion of patients will develop recurrent and metastatic disease. For these patients, current treatment options are quite limited. Significant progress has been made recently in the use of immunotherapy for the treatment of other solid tumors (e.g. prostate cancer, melanoma). There is a strong rationale for immunotherapy in STS, based on an understanding of disease biology. For example, STS frequently have chromosomal translocations which result in unique fusion proteins and specific subtypes have been shown to express cancer testis antigens. In this review, we discuss the current status of immunotherapy in STS, including data from human studies with cancer vaccines, adoptive cell therapy, and immune checkpoint blockade. Further research into STS immunology is needed to help design logical, subtype-specific immunotherapeutic strategies.
Collapse
Affiliation(s)
- William W Tseng
- a Section of Surgical Oncology; Division of Upper GI/General Surgery; Department of Surgery ; University of Southern California; Keck School of Medicine ; Los Angeles , CA USA
| | | | | |
Collapse
|
11
|
Aravindaram K, Yin SY, Yang NS. Biolistic transfection of tumor tissue samples. Methods Mol Biol 2013; 940:133-143. [PMID: 23104339 DOI: 10.1007/978-1-62703-110-3_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
A nonviral method for gene transfer into mammalian cells has been developed using physical force which accelerates plasmid DNA-coated gold particles to high -speed and penetrate the mammalian cells. This technology of gene transfer via a biolistic transfection method has been shown to have multiple applications to mammalian gene transfer systems. This method has also been adapted for delivery of other macromolecules like RNA, microRNA, and proteins. A broad range of somatic cell types, including primary cell cultures and established cell lines, have been successfully transfected ex vivo or in vitro by using the gene gun technology, either as suspension or adherent cells in cultures. This chapter describes the general procedures for in vitro DNA transfection by particle-mediated delivery to nonadherent and adherent cells. These procedures can be readily employed by using the Helios gene gun system (Bio-Rad, Hercules, CA) based on the Accell design.
Collapse
Affiliation(s)
- Kandan Aravindaram
- Agricultural Biotechnology Research Center, Academia Sinica, Nankang, Taipei, Taiwan, ROC
| | | | | |
Collapse
|
12
|
De Souza Rebouças J, Esparza I, Ferrer M, Sanz ML, Irache JM, Gamazo C. Nanoparticulate adjuvants and delivery systems for allergen immunotherapy. J Biomed Biotechnol 2012; 2012:474605. [PMID: 22496608 PMCID: PMC3303624 DOI: 10.1155/2012/474605] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 10/19/2011] [Accepted: 10/25/2011] [Indexed: 12/21/2022] Open
Abstract
In the last decades, significant progress in research and clinics has been made to offer possible innovative therapeutics for the management of allergic diseases. However, current allergen immunotherapy shows limitations concerning the long-term efficacy and safety due to local side effects and risk of anaphylaxis. Thus, effective and safe vaccines with reduced dose of allergen have been developed using adjuvants. Nevertheless, the use of adjuvants still has several disadvantages, which limits its use in human vaccines. In this context, several novel adjuvants for allergen immunotherapy are currently being investigated and developed. Currently, nanoparticles-based allergen-delivery systems have received much interest as potential adjuvants for allergen immunotherapy. It has been demonstrated that the incorporation of allergens into a delivery system plays an important role in the efficacy of allergy vaccines. Several nanoparticles-based delivery systems have been described, including biodegradable and nondegradable polymeric carriers. Therefore, this paper provides an overview of the current adjuvants used for allergen immunotherapy. Furthermore, nanoparticles-based allergen-delivery systems are focused as a novel and promising strategy for allergy vaccines.
Collapse
Affiliation(s)
- Juliana De Souza Rebouças
- Adjuvant Unit, Department of Pharmacy and Pharmaceutical Technology, and Department of Microbiology, University of Navarra, 31008 Pamplona, Spain
| | - Irene Esparza
- Adjuvant Unit, Department of Pharmacy and Pharmaceutical Technology, and Department of Microbiology, University of Navarra, 31008 Pamplona, Spain
| | - Marta Ferrer
- Department of Allergy and Clinical Immunology, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - María Luisa Sanz
- Department of Allergy and Clinical Immunology, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Juan Manuel Irache
- Adjuvant Unit, Department of Pharmacy and Pharmaceutical Technology, and Department of Microbiology, University of Navarra, 31008 Pamplona, Spain
| | - Carlos Gamazo
- Adjuvant Unit, Department of Pharmacy and Pharmaceutical Technology, and Department of Microbiology, University of Navarra, 31008 Pamplona, Spain
| |
Collapse
|
13
|
Tschoep-Lechner K, Drexler I, Hammer D, Neumann D, Pohla H, Sutter G, Noessner E, Issels RD. Modified vaccinia virus Ankara delivers a robust surrogate marker for immune monitoring to sarcoma cells even if cells are being exposed to chemotherapy and heat treatment. Int J Hyperthermia 2012; 28:33-42. [PMID: 22235783 PMCID: PMC9476112 DOI: 10.3109/02656736.2011.626834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Katharina Tschoep-Lechner
- Department of Internal Medicine III, Klinikum Grosshadern Medical Centre, Ludwig Maximilians University, Marchioninistrasse 25, Munich, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Francescutti V, Skitzki JJ. Sarcomas and the immune system: implications for therapeutic strategies. Surg Oncol Clin N Am 2011; 21:341-55. [PMID: 22365524 DOI: 10.1016/j.soc.2011.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Soft-tissue sarcomas are a heterogeneous group of tumors that are capable of generating host immune responses. Historically the role of antitumor immunity was first studied in soft-tissue sarcomas. Subsequent in vitro studies, preclinical models, and clinical observations have provided ample evidence for an immunologic approach to sarcoma treatment. Initial clinical trials involving vaccines and adoptive immunotherapy have demonstrated promising results. The continued search for sarcoma tumor-associated antigens as specific targets is central to the clinical translation of effective immunotherapies.
Collapse
Affiliation(s)
- Valerie Francescutti
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | |
Collapse
|
15
|
Xia J, Martinez A, Daniell H, Ebert SN. Evaluation of biolistic gene transfer methods in vivo using non-invasive bioluminescent imaging techniques. BMC Biotechnol 2011; 11:62. [PMID: 21635760 PMCID: PMC3125329 DOI: 10.1186/1472-6750-11-62] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Accepted: 06/02/2011] [Indexed: 01/01/2023] Open
Abstract
Background Gene therapy continues to hold great potential for treating many different types of disease and dysfunction. Safe and efficient techniques for gene transfer and expression in vivo are needed to enable gene therapeutic strategies to be effective in patients. Currently, the most commonly used methods employ replication-defective viral vectors for gene transfer, while physical gene transfer methods such as biolistic-mediated ("gene-gun") delivery to target tissues have not been as extensively explored. In the present study, we evaluated the efficacy of biolistic gene transfer techniques in vivo using non-invasive bioluminescent imaging (BLI) methods. Results Plasmid DNA carrying the firefly luciferase (LUC) reporter gene under the control of the human Cytomegalovirus (CMV) promoter/enhancer was transfected into mouse skin and liver using biolistic methods. The plasmids were coupled to gold microspheres (1 μm diameter) using different DNA Loading Ratios (DLRs), and "shot" into target tissues using a helium-driven gene gun. The optimal DLR was found to be in the range of 4-10. Bioluminescence was measured using an In Vivo Imaging System (IVIS-50) at various time-points following transfer. Biolistic gene transfer to mouse skin produced peak reporter gene expression one day after transfer. Expression remained detectable through four days, but declined to undetectable levels by six days following gene transfer. Maximum depth of tissue penetration following biolistic transfer to abdominal skin was 200-300 μm. Similarly, biolistic gene transfer to mouse liver in vivo also produced peak early expression followed by a decline over time. In contrast to skin, however, liver expression of the reporter gene was relatively stable 4-8 days post-biolistic gene transfer, and remained detectable for nearly two weeks. Conclusions The use of bioluminescence imaging techniques enabled efficient evaluation of reporter gene expression in vivo. Our results demonstrate that different tissues show different expression kinetics following gene transfer of the same reporter plasmid to different mouse tissues in vivo. We evaluated superficial (skin) and abdominal organ (liver) targets, and found that reporter gene expression peaked within the first two days post-transfer in each case, but declined most rapidly in the skin (3-4 days) compared to liver (10-14 days). This information is essential for designing effective gene therapy strategies in different target tissues.
Collapse
Affiliation(s)
- Jixiang Xia
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL 32827, USA
| | | | | | | |
Collapse
|
16
|
Abstract
Current treatment of malignant melanoma exemplifies not only the need for translational research but also many of the challenges of moving from bench to bedside. Melanoma remains unique among solid tumors in that its treatment primarily is surgical. Radiation is of limited benefit, and chemotherapy has been disappointing in both the adjuvant and metastatic settings. This leaves clinicians with few options for reducing the chance of recurrence after surgery and for treating unresectable disease. With this in mind, there has been a fervent attempt to identify novel approaches to melanoma therapy and translate them into clinical use.
Collapse
|
17
|
Peek LJ, Middaugh CR, Berkland C. Nanotechnology in vaccine delivery. Adv Drug Deliv Rev 2008; 60:915-28. [PMID: 18325628 PMCID: PMC7103321 DOI: 10.1016/j.addr.2007.05.017] [Citation(s) in RCA: 371] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Accepted: 05/01/2007] [Indexed: 01/19/2023]
Abstract
With very few adjuvants currently being used in marketed human vaccines, a critical need exists for novel immunopotentiators and delivery vehicles capable of eliciting humoral, cellular and mucosal immunity. Such crucial vaccine components could facilitate the development of novel vaccines for viral and parasitic infections, such as hepatitis, HIV, malaria, cancer, etc. In this review, we discuss clinical trial results for various vaccine adjuvants and delivery vehicles being developed that are approximately nanoscale (< 1000 nm) in size. Humoral immune responses have been observed for most adjuvants and delivery platforms while only viral vectors, ISCOMs and Montanide™ ISA 51 and 720 have shown cytotoxic T cell responses in the clinic. MF59 and MPL® have elicited Th1 responses, and virus-like particles, non-degradable nanoparticles and liposomes have also generated cellular immunity. Such vaccine components have also been evaluated for alternative routes of administration with clinical successes reported for intranasal delivery of viral vectors and proteosomes and oral delivery of a VLP vaccine.
Collapse
|
18
|
Cutaneous Melanoma. Oncology 2007. [DOI: 10.1007/0-387-31056-8_59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
19
|
Cassaday RD, Sondel PM, King DM, Macklin MD, Gan J, Warner TF, Zuleger CL, Bridges AJ, Schalch HG, Kim KM, Hank JA, Mahvi DM, Albertini MR. A phase I study of immunization using particle-mediated epidermal delivery of genes for gp100 and GM-CSF into uninvolved skin of melanoma patients. Clin Cancer Res 2007; 13:540-9. [PMID: 17255276 DOI: 10.1158/1078-0432.ccr-06-2039] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We examined in vivo particle-mediated epidermal delivery (PMED) of cDNAs for gp100 and granulocyte macrophage colony-stimulating factor (GM-CSF) into uninvolved skin of melanoma patients. The aims of this phase I study were to assess the safety and immunologic effects of PMED of these genes in melanoma patients. EXPERIMENTAL DESIGN Two treatment groups of six patients each were evaluated. Group I received PMED with cDNA for gp100, and group II received PMED with cDNA for GM-CSF followed by PMED for gp100 at the same site. One vaccine site per treatment cycle was biopsied and divided for protein extraction and sectioning to assess transgene expression, gold-bead penetration, and dendritic cell infiltration. Exploratory immunologic monitoring of HLA-A2(+) patients included flow cytometric analyses of peripheral blood lymphocytes and evaluation of delayed-type hypersensitivity to gp100 peptide. RESULTS Local toxicity in both groups was mild and resolved within 2 weeks. No systemic toxicity could be attributed to the vaccines. Monitoring for autoimmunity showed no induction of pathologic autoantibodies. GM-CSF transgene expression in vaccinated skin sites was detected. GM-CSF and gp100 PMED yielded a greater infiltration of dendritic cells into vaccine sites than did gp100 PMED only. Exploratory immunologic monitoring suggested modest activation of an antimelanoma response. CONCLUSIONS PMED with cDNAs for gp100 alone or in combination with GM-CSF is well tolerated by patients with melanoma. Moreover, pathologic autoimmunity was not shown. This technique yields biologically active transgene expression in normal human skin. Although modest immune responses were observed, additional investigation is needed to determine how to best utilize PMED to induce antimelanoma immune responses.
Collapse
Affiliation(s)
- Ryan D Cassaday
- Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW To discuss advances in immunotherapy as pertaining to systemic therapy for sarcomas. RECENT FINDINGS Developments in immunology will have direct relevance to studies in sarcomas and other cancers in the near future. Vaccines employing peptides are the backbone of many studies today due to issues of cost and ease of production, but intact or transfected whole tumor cells or antigen-pulsed or transfected dendritic cells are now being investigated as the immunogenic principle in sarcomas and other cancers. The importance of dendritic cells in generating immune responses is better appreciated than ever, as is the role of CD4+ CD25+ regulatory T cells in mediating immune responses. Enhancing costimulatory signals to T cells using anti-CD152 (CTLA4) and other antibodies or expanding anti-tumor cytotoxic T lymphocytes ex vivo are other means to enhance immunity to sarcoma-specific antigens. SUMMARY As of 2006, few studies are examining the utility of immunotherapy in sarcomas. However, a detailed understanding of the remarkable mechanics of how an immune response is mounted and how T cell activation and/or proliferation can be halted by the tumor will be central to properly design new studies of immunological agents against sarcomas and other cancers.
Collapse
Affiliation(s)
- Robert G Maki
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10021-6007, USA.
| |
Collapse
|
21
|
Dammeyer P, Jaramillo MC, Pipes BL, Badowski MS, Tsang TC, Harris DT. Heat-inducible amplifier vector for high-level expression of granulocyte-macrophage colony-stimulating factor. Int J Hyperthermia 2006; 22:407-19. [PMID: 16891243 DOI: 10.1080/02656730600765312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
PURPOSE In cytokine immunotherapy of cancer it is critical to deliver sufficiently high local cytokine concentrations in order to reach the therapeutic threshold needed for clinical efficacy. Simultaneously, for optimal clinical safety adverse effects caused by high systemic cytokine levels must be minimized. One of the most promising anti-cancer therapeutic cytokines, granulocyte-macrophage colony-stimulating factor (GM-CSF), has elicited anti-tumour immune responses in animal studies and clinical trials. However, the clinical efficacy has been limited, with local GM-CSF levels being therapeutically insufficient and systemic toxicity being a limiting factor. METHODS To address these problems we have developed a novel GM-CSF expression vector, pAD-HotAmp-GM-CSF, which can provide high levels of GM-CSF expression, and induction of cytokine expression to limited tissue areas. This expression system combines inducible and amplifying elements in a single multi-genic construct. The first transcriptional unit contains the inducible element, the heat shock protein 70B (HSP70B) promoter that regulates expression of the transcription-activating factor tat. RESULTS Upon the binding of tat to the second promoter, the HIV2 long terminal repeat amplifies downstream gene expression of the therapeutic cytokine GM-CSF. Moderate hyperthermia at 42 degrees C for 30 min induced GM-CSF expression in pAD-HotAmp-GM-CSF that was over 2.5- and 2.8-fold higher than levels reached with HSP70B promoter alone and the prototypical human cytomegalovirus promoter. CONCLUSIONS Thus, the inducible amplifier vector, pAD-HotAmp-GM-CSF, represents a novel system for regulated and enhanced GM-CSF expression, which enables both greater efficacy and safety in cytokine immunotherapy of cancer.
Collapse
Affiliation(s)
- Pascal Dammeyer
- Department of Microbiology and Immunology, University of Arizona, Tucson, AZ, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Saupe A, McBurney W, Rades T, Hook S. Immunostimulatory colloidal delivery systems for cancer vaccines. Expert Opin Drug Deliv 2006; 3:345-54. [PMID: 16640495 DOI: 10.1517/17425247.3.3.345] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Cancer vaccine delivery is a multidisciplinary scientific field that is currently undergoing rapid development. An important component of cancer vaccines is the development of novel vaccine delivery strategies, such as colloidal immunostimulatory delivery systems. The importance of formulation strategies for cancer vaccines can be explained by the poor immunogenicity of tumour antigens. Colloidal vaccine delivery systems modify the kinetics, body distribution, uptake and release of the vaccine. This review explores recent research that is directed towards more targeted treatments of cancer through to colloidal vaccine delivery systems. Widely investigated carrier systems include polymeric micro- and nanoparticles, liposomes, archaeal lipid liposomes (archaeosomes), immune-stimulating complexes and virus-like particles. These systems are evaluated in terms of their formulation techniques, immunological mechanisms of action as well as the potential and limitations of such colloidal systems in the field of cancer vaccines.
Collapse
Affiliation(s)
- Anne Saupe
- School of Pharmacy, University of Otago, PO Box 913, Dunedin, New Zealand.
| | | | | | | |
Collapse
|
23
|
Mocellin S, Rossi CR, Brandes A, Nitti D. Adult soft tissue sarcomas: Conventional therapies and molecularly targeted approaches. Cancer Treat Rev 2006; 32:9-27. [PMID: 16338075 DOI: 10.1016/j.ctrv.2005.10.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Revised: 09/21/2005] [Indexed: 01/12/2023]
Abstract
The therapeutic approach to soft tissue sarcomas (STS) has evolved over the past two decades based on the results from randomized controlled trials, which are guiding physicians in the treatment decision-making process. Despite significant improvements in the control of local disease, a significant number of patients ultimately die of recurrent/metastatic disease following radical surgery due to a lack of effective adjuvant treatments. In addition, the characteristic chemoresistance of STS has compromised the therapeutic value of conventional antineoplastic agents in cases of unresectable advanced/metastatic disease. Therefore, novel therapeutic strategies are urgently needed to improve the prognosis of patients with STS. Recent advances in STS biology are paving the way to the development of molecularly targeted therapeutic strategies, the efficacy of which relies not only on the knowledge of the molecular mechanisms underlying cancer development/progression but also on the personalization of the therapeutic regimen according to the molecular features of individual tumours. In this work, we review the state-of-the-art of conventional treatments for STS and summarize the most promising findings in the development of molecularly targeted therapeutic approaches.
Collapse
Affiliation(s)
- Simone Mocellin
- Surgery Branch, Department of Oncological and Surgical Sciences, University of Padova, Via Giustiniani 2, 35128 Padua, Italy.
| | | | | | | |
Collapse
|
24
|
Campoli M, Ferrone S. T-cell-based immunotherapy of melanoma: what have we learned and how can we improve? Expert Rev Vaccines 2004; 3:171-87. [PMID: 15056043 DOI: 10.1586/14760584.3.2.171] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The lack of effective treatment for advanced stage melanoma by conventional therapies, such as radiation and chemotherapy, has highlighted the need to develop alternative therapeutic strategies. Among them, immunotherapy has attracted much attention because of the potential role played by immunological events in the clinical course of melanoma and the availability of well-characterized melanoma antigens to target melanoma lesions with immunological effector mechanisms. In recent years, T-cell-based immunotherapy has been emphasized, in part because of the disappointing results of the antibody-based trials conducted in the early 1980s, and in part because of the postulated major role played by T-cells in tumor growth control. In this review, the characteristics of antibody and T-cell-defined melanoma antigens will first be described, with emphasis on those used in clinical trials. Following a review of the current immunization and immunomonitoring strategies, the results from the T-cell-based immunotherapy clinical trials conducted to date will be reviewed.
Collapse
Affiliation(s)
- Michael Campoli
- Department of Immunology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| | | |
Collapse
|
25
|
Santini SM, Belardelli F. Advances in the use of dendritic cells and new adjuvants for the development of therapeutic vaccines. Stem Cells 2004; 21:495-505. [PMID: 12832702 DOI: 10.1634/stemcells.21-4-495] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The recent advances in immunology and biotechnology have opened new perspectives for the development of immunotherapy strategies against cancer and infectious diseases. The understanding of the pivotal role of dendritic cells in the initiation and regulation of the immune response has led to an ensemble of preclinical studies and pilot clinical trials, which have provided some evidence on the potential advantages of using dendritic cells as cellular adjuvants for the development of therapeutic vaccines against infectious diseases and malignancies. Current research efforts are focused on the definition of optimal protocols for dendritic cell-based therapies in patients. An additional area of emerging importance in the field of immunotherapy is the identification of safe, selective, and more powerful adjuvants, capable not only of enhancing immune protection against pathogens, but also of breaking tolerance against certain tumor-associated antigens, which is the critical issue for the development of cancer vaccines. The recent recognition of the key role of certain cytokines, such as type I interferons, in linking the innate and adaptive immunity through their action on dendritic cells opens new perspectives for using these natural factors as adjuvants for the development of therapeutic vaccines. We review some of the emerging research aspects in immunotherapy, with special attention to the perspectives of using new adjuvants and dendritic cell-based vaccines for the treatment of cancer and infectious diseases.
Collapse
|
26
|
Abstract
The treatment of patients with metastatic soft tissue sarcomas (STS) is complex. There are limited agents available and many are associated with significant toxicity. When evaluating a patient with metastatic disease, physicians should ask themselves whether there is a role for surgery to render the patient free of disease. Combination chemotherapy in patients who have not received chemotherapy in the adjuvant setting is one option, particularly in a young patient with a good performance status. Sequential single-agent therapy for patients who are more elderly or debilitated by their disease may be more appropriate. Gemcitabine appears to be an agent with activity, particularly in patients with leiomyosarcomas. The data regarding prolonged gemcitabine infusions suggest improved activity that was predicted based on prolonged intracellular gemcitabine levels. Because of these data, the prolonged infusion schedule should be used. In addition, because of the paucity of effective agents, consideration of clinical trial participation for patients with newly diagnosed metastatic disease is appropriate, particularly in chemotherapy-insensitive histologies. The role of the newer agents (eg, ecteinascidin-743, epothilones, and mammalian target of rapamycin) is undefined. Ecteinascidin-743 has been the most extensively tested agent, and its ability to slow growth kinetics of a tumor and stabilize it clinically is intriguing. Data regarding the response to BMS-247550 will be published shortly and will help define the further role of epothilones in this disease. There is a preclinical rationale that makes the mammalian target of rapamycin inhibitors attractive for the treatment of muscle-derived neoplasms. In addition, there are cell-line data suggesting activity in rhabdomyosarcoma. These agents are being tested in adult STS and will likely be tested in pediatric histologies when there are more safety data available in that population. SU11248 will continue to be tested in patients refractory to imatinib mesylate and may well prove to be another active agent for patients with gastrointestinal stromal tumors. As depicted by the analysis of gemcitabine efficacy, agents with activity in a subgroup of STS may be overlooked by the "come one come all" approach to clinical trials in STS. Identifying key targets in specific STS will be helpful in the testing of newer molecularly targeted agents. Biologic differences will support histology-specific trials to better understand the activity of an agent in a specific disease site or specifically target a biologic pathway with relevance to the malignant potential of the disease. For future clinical trials in STS to achieve the goal of histology-specific trials, cooperative group and multi-institutional trials will be required to obtain the appropriate patients with these rare histologies. It will also be increasingly important to be committed to obtaining tumor tissue in these patients to validate hypotheses regarding tumor biology and the effectiveness of therapeutic agents.
Collapse
Affiliation(s)
- Margaret von Mehren
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA.
| |
Collapse
|
27
|
Yuan SF, Wang L, Li KZ, Yan Z, Han W, Zhang YQ. Inhibitory effect of MUC1 gene immunization on H22 hepatocellular carcinoma growth. Shijie Huaren Xiaohua Zazhi 2003; 11:1322-1325. [DOI: 10.11569/wcjd.v11.i9.1322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the special anti-H22 hepatocellular carcinoma growth effect of the MUC1 gene immunization.
METHODS Balb/c mice were immunized intramuscularly with 100 mg MUC1 cDNA 3 times at 3-weekly intervals. Three weeks after the last immunization, tumor challenge experiments were performed by using MUC1 expressing tumor cell line H22. Tumor growth inhibition and body protection were observed two weeks later. After 43d of challenge experiments, all mice were killed and tumors were weighed. Histological analysis of tumor tissue was carried out with HE staining.
RESULTS After 43 d of challenge experiments, the volumes of H22 hepatocellular carcinoma in MUC1cDNA, pcDNA3.1(+) and NS groups were 547±59 mm3, 1 185±84 mm3 and 1 220±95 mm3 (P <0.01), respectively. The average mass of H22 hepatocellular carcinoma in the three groups was 1.87±0.96 g, 4.19±1.34 g and 4.23±1.32 g (P <0.01), respectively . Tumorigenic rate was only 50 % in MUC1cDNA group, and was 100 % in pcDNA3.1(+) and NS group. H22 hepatocellular carcinoma growth in mice of MUC1cDNA group was significantly suppressed (P <0.01), and a significant body protective effect was found in mice of MUC1cDNA group (P <0.05), compared with control group. Histological analysis showed that the H22 hepatocellular carcinoma tissues were markedly necrosed in mice of MUC1cDNA group compared with that in control group.
CONCLUSION MUC1 gene immunization can significantly suppress H22 hepatocellular carcinoma growth.
Collapse
Affiliation(s)
- Shi-Fang Yuan
- Department of Vascular and Endocrine Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710033, Shaanxi Provice, China
| | - Ling Wang
- Department of Vascular and Endocrine Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710033, Shaanxi Provice, China
| | - Kai-Zong Li
- Department of Hepato-biliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710033, Shaanxi Provice, China
| | - Zhen Yan
- Biotechnology Center, Fourth Military Medical University, Xi'an 710033, Shaanxi Provice, China
| | - Wei Han
- Biotechnology Center, Fourth Military Medical University, Xi'an 710033, Shaanxi Provice, China
| | - Ying-Qi Zhang
- Biotechnology Center, Fourth Military Medical University, Xi'an 710033, Shaanxi Provice, China
| |
Collapse
|
28
|
Abstract
Recent advances in basic medical sciences have led to a deeper understanding of the molecular characteristics of soft-tissue sarcomas. Likewise, novel technologies have led to a better appreciation of the relationship between an antigenic stimulus and the subsequent immune response against the antigen. In the past few years, the intersection of the understanding of the immune system and the knowledge of sarcoma biology has become apparent. As seen with other forms of cancer, there is a detectable autologous immune response against sarcomas. It is the hope of many investigators that the hints of a tumor-specific immune response will be enough to generate a signal that can be amplified and directed against the host sarcoma. The data regarding the initial evidence of immune responses against sarcomas are reviewed in the context of current or potential clinical studies.
Collapse
Affiliation(s)
- Robert G Maki
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Box 223, New York, NY 10021-6007, USA.
| |
Collapse
|