1
|
Šošić L, Paolucci M, Flory S, Jebbawi F, Kündig TM, Johansen P. Allergen immunotherapy: progress and future outlook. Expert Rev Clin Immunol 2023:1-25. [PMID: 37122076 DOI: 10.1080/1744666x.2023.2209319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
INTRODUCTION Allergy, the immunological hypersensitivity to innocuous environmental compounds, is a global health problem. The disease triggers, allergens, are mostly proteins contained in various natural sources such as plant pollen, animal dander, dust mites, foods, fungi and insect venoms. Allergies can manifest with a wide range of symptoms in various organs, and be anything from just tedious to life-threatening. A majority of all allergy patients are self-treated with symptom-relieving medicines, while allergen immunotherapy (AIT) is the only causative treatment option. AREAS COVERED This review will aim to give an overview of the state-of-the-art allergy management, including the use of new biologics and the application of biomarkers, and a special emphasis and discussion on current research trends in the field of AIT. EXPERT OPINION Conventional AIT has proven effective, but the years-long treatment compromises patient compliance. Moreover, AIT is typically not offered in food allergy. Hence, there is a need for new, effective and safe AIT methods. Novel routes of administration (e.g. oral and intralymphatic), hypoallergenic AIT products and more effective adjuvants holds great promise. Most recently, the development of allergen-specific monoclonal antibodies for passive immunotherapy may also allow treatment of patients currently not treated or treatable.
Collapse
Affiliation(s)
- Lara Šošić
- Department of Dermatology, University of Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Marta Paolucci
- Department of Dermatology, University of Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Stephan Flory
- Department of Dermatology, University of Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Fadi Jebbawi
- Department of Dermatology, University of Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Thomas M Kündig
- Department of Dermatology, University of Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Pål Johansen
- Department of Dermatology, University of Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| |
Collapse
|
2
|
Peng X, Wang J, Zhou F, Liu Q, Zhang Z. Nanoparticle-based approaches to target the lymphatic system for antitumor treatment. Cell Mol Life Sci 2021; 78:5139-5161. [PMID: 33963442 PMCID: PMC11072902 DOI: 10.1007/s00018-021-03842-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 03/14/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023]
Abstract
Immunotherapies have been established as safe and efficient modalities for numerous tumor treatments. The lymphatic system, which is an important system, can modulate the immune system via a complex network, which includes lymph nodes, vessels, and lymphocytes. With the deepening understanding of tumor immunology, a plethora of immunotherapies, which include vaccines, photothermal therapy, and photodynamic therapy, have been established for antitumor treatments. However, the deleterious off-target effects and nonspecific targeting of therapeutic agents result in low efficacy of immunotherapy. Fortunately, nanoparticle-based approaches for targeting the lymphatic system afford a unique opportunity to manufacture drugs that can simultaneously tackle both aspects, thereby improving tumor treatments. Over the past decades, great strides have been made in the development of DC vaccines and nanomedicine as antitumor treatments in the field of lymphatic therapeutics and diagnosis. In this review, we summarize the current strategies through which nanoparticle technology has been designed to target the lymphatic system and describe applications of lymphatic imaging for the diagnosis and image-guided surgery of tumor metastasis. Moreover, improvements in the tumor specificity of nanovaccines and medicines, which have been realized through targeting or stimulating the lymphatic system, can provide amplified antitumor immune responses and reduce side effects, thereby promoting the paradigm of antitumor treatment into the clinic to benefit patients.
Collapse
Affiliation(s)
- Xingzhou Peng
- School of Biomedical Engineering, Hainan University, Haikou, 570228, Hainan, China
| | - Junjie Wang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Feifan Zhou
- School of Biomedical Engineering, Hainan University, Haikou, 570228, Hainan, China
| | - Qian Liu
- School of Biomedical Engineering, Hainan University, Haikou, 570228, Hainan, China.
| | - Zhihong Zhang
- School of Biomedical Engineering, Hainan University, Haikou, 570228, Hainan, China.
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.
| |
Collapse
|
3
|
Gregg JR, Thompson TC. Considering the potential for gene-based therapy in prostate cancer. Nat Rev Urol 2021; 18:170-184. [PMID: 33637962 DOI: 10.1038/s41585-021-00431-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2021] [Indexed: 01/31/2023]
Abstract
Therapeutic gene manipulation has been at the forefront of popular scientific discussion and basic and clinical research for decades. Basic and clinical research applications of CRISPR-Cas9-based technologies and ongoing clinical trials in this area have demonstrated the potential of genome editing to cure human disease. Evaluation of research and clinical trials in gene therapy reveals a concentration of activity in prostate cancer research and practice. Multiple aspects of prostate cancer care - including anatomical considerations that enable direct tumour injections and sampling, the availability of preclinical immune-competent models and the delineation of tumour-related antigens that might provide targets for an induced immune system - make gene therapy an appealing treatment option for this common malignancy. Vaccine-based therapies that induce an immune response and new technologies exploiting CRISPR-Cas9-assisted approaches, including chimeric antigen receptor (CAR) T cell therapies, are very promising and are currently under investigation both in the laboratory and in the clinic. Although laboratory and preclinical advances have, thus far, not led to oncologically relevant outcomes in the clinic, future studies offer great promise for gene therapy to become established in prostate cancer care.
Collapse
Affiliation(s)
- Justin R Gregg
- Department of Urology and Health Disparities Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Timothy C Thompson
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
4
|
Intranodal administration of mRNA encoding nucleoprotein provides cross-strain immunity against influenza in mice. J Transl Med 2019; 17:242. [PMID: 31345237 PMCID: PMC6659201 DOI: 10.1186/s12967-019-1991-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/17/2019] [Indexed: 01/25/2023] Open
Abstract
Background Current human influenza vaccines lack the adaptability to match the mutational rate of the virus and therefore require annual revisions. Because of extensive manufacturing times and the possibility that antigenic alterations occur during viral vaccine strain production, an inherent risk exists for antigenic mismatch between the new influenza vaccine and circulating viruses. Targeting more conserved antigens such as nucleoprotein (NP) could provide a more sustainable vaccination strategy by inducing long term and heterosubtypic protection against influenza. We previously demonstrated that intranodal mRNA injection can induce potent antigen-specific T-cell responses. In this study, we investigated whether intranodal administration of mRNA encoding NP can induce T-cell responses capable of protecting against a heterologous influenza virus challenge. Methods BALB/c mice were immunized in the inguinal lymph nodes with different vaccination regimens of mRNA encoding NP. Immune responses were compared with NP DNA vaccination via IFN-γ ELISPOT and in vivo cytotoxicity. For survival experiments, mice were prime-boost vaccinated with 17 µg NP mRNA and infected with 1LD50 of H1N1 influenza virus 8 weeks after boost. Weight was monitored and viral titers, cytokines and immune cell populations in the bronchoalveolar lavage, and IFN-γ responses in the spleen were analyzed. Results Our results demonstrate that NP mRNA induces superior systemic T-cell responses against NP compared to classical DNA vaccination. These responses were sustained for several weeks even at low vaccine doses. Upon challenge infection, vaccination with NP mRNA resulted in reduced lung viral titers and improved recovery from infection. Finally, we show that vaccination with NP mRNA affects the immune response in infected lungs by lowering immune cell infiltration while increasing the fraction of T cells, monocytes and MHC II+ alveolar macrophages within immune infiltrates. This change was associated with altered levels of both pro- and anti-inflammatory cytokines. Conclusions These findings suggest that intranodal vaccination with NP mRNA induces cross-strain immunity against influenza, but also highlight a paradox of influenza immunity, whereby robust immune responses can provide protection, but can also transiently exacerbate symptoms during infection. Electronic supplementary material The online version of this article (10.1186/s12967-019-1991-3) contains supplementary material, which is available to authorized users.
Collapse
|
5
|
Schudel A, Francis DM, Thomas SN. Material design for lymph node drug delivery. NATURE REVIEWS. MATERIALS 2019; 4:415-428. [PMID: 32523780 PMCID: PMC7286627 DOI: 10.1038/s41578-019-0110-7] [Citation(s) in RCA: 325] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
A significant fraction of the total immune cells in the body are located in several hundred lymph nodes, in which lymphocyte accumulation, activation and proliferation are organized. Therefore, targeting lymph nodes provides the possibility to directly deliver drugs to lymphocytes and lymph node-resident cells and thus to modify the adaptive immune response. However, owing to the structure and anatomy of lymph nodes, as well as the distinct localization and migration of the different cell types within the lymph node, it is difficult to access specific cell populations by delivering free drugs. Materials can be used as instructive delivery vehicles to achieve accumulation of drugs in the lymph nodes and to target specific lymph node-resident cell subtypes. In this Review, we describe the compartmental architecture of lymph nodes and the cell and fluid transport mechanisms to and from lymph nodes. We discuss the different entry routes into lymph nodes and how they can be explored for drug delivery, including the lymphatics, blood capillaries, high endothelial venules, cell-mediated pathways, homing of circulating lymphocytes and direct lymph node injection. We examine different nanoscale and microscale materials for the targeting of specific immune cells and highlight their potential for the treatment of immune dysfunction and for cancer immunotherapy. Finally, we give an outlook to the field, exploring how lymph node targeting can be improved by the use of materials.
Collapse
Affiliation(s)
- Alex Schudel
- School of Materials Science and Engineering, Georgia institute of Technology, Atlanta, GA, USA
- Parker H. Petit institute for Bioengineering and Bioscience, Georgia institute of Technology, Atlanta, GA, USA
- These authors contributed equally: Alex Schudel, David M. Francis
| | - David M Francis
- Parker H. Petit institute for Bioengineering and Bioscience, Georgia institute of Technology, Atlanta, GA, USA
- School of Chemical and Biomolecular Engineering, Georgia institute of Technology, Atlanta, GA, USA
- These authors contributed equally: Alex Schudel, David M. Francis
| | - Susan N Thomas
- Parker H. Petit institute for Bioengineering and Bioscience, Georgia institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia institute of Technology and Emory University, Atlanta, GA, USA
- Winship Cancer institute, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
6
|
Abstract
Gold Standard allergen-specific immunotherapy is associated with low efficacy because it requires either many subcutaneous injections of allergen or even more numerous sublingual allergen administrations to achieve amelioration of symptoms. Intralymphatic vaccination can maximize immunogenicity and hence efficacy. We and others have demonstrated that as few as three low dose intralymphatic allergen administrations are sufficient to effectively alleviate symptoms. Results of recent prospective and controlled trials suggest that this strategy may be an effective form of allergen immunotherapy.
Collapse
Affiliation(s)
- Gabriela Senti
- Clinical Trials Center, University Hospital Zurich, Raemistrasse 100/MOU2, CH-8091 Zurich, Switzerland
| | - Thomas M Kündig
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
7
|
Therapeutic DNA vaccination against colorectal cancer by targeting the MYB oncoprotein. Clin Transl Immunology 2015; 4:e30. [PMID: 25671128 PMCID: PMC4318491 DOI: 10.1038/cti.2014.29] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 12/02/2014] [Accepted: 12/03/2014] [Indexed: 12/18/2022] Open
Abstract
Cancers can be addicted to continued and relatively high expression of nuclear oncoproteins. This is evident in colorectal cancer (CRC) where the oncoprotein and transcription factor MYB is over expressed and essential to continued proliferation and tumour cell survival. Historically, targeting transcription factors in the context of cancer has been very challenging. Nevertheless, we formulated a DNA vaccine to generate a MYB-specific immune response in the belief MYB peptides might be aberrantly presented on the cell surface of CRC cells. MYB, like many tumour antigens, is weakly immunogenic as it is a 'self' antigen and is subject to tolerance. To break tolerance, a fusion vaccine was generated comprising a full-length MYB complementary DNA (cDNA) flanked by two potent CD4-epitopes derived from tetanus toxoid. Vaccination was achieved against tumours initiated by two distinct highly aggressive, syngeneic cancer cell lines (CT26 and MC38) that express MYB. This was done in BALB/c and C57BL/6 mouse strains respectively. We introduced multiple inactivating mutations into the oncogene sequence for safety and sub-cloned the cDNA into a Food and Drug Administration (FDA)-compliant vector. We used low dose cyclophosphamide (CY) to overcome T-regulatory cell immune suppression, and anti-program cell death receptor 1 (anti-PD-1) antibodies to block T-cell exhaustion. Anti-PD-1 administered alone slightly delayed tumour growth in MC38 and more effectively in CT26 bearing mice, while CY treatment alone did not. We found that therapeutic vaccination elicits protection when MC38 tumour burden is low, mounts tumour-specific cell killing and affords enhanced protection when MC38 and CT26 tumour burden is higher but only in combination with anti-PD-1 antibody or low dose CY, respectively.
Collapse
|
8
|
Andorko JI, Hess KL, Jewell CM. Harnessing biomaterials to engineer the lymph node microenvironment for immunity or tolerance. AAPS JOURNAL 2014; 17:323-38. [PMID: 25533221 PMCID: PMC4365095 DOI: 10.1208/s12248-014-9708-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 11/27/2014] [Indexed: 01/06/2023]
Abstract
Nanoparticles, microparticles, and other biomaterials are advantageous in vaccination because these materials provide opportunities to modulate specific characteristics of immune responses. This idea of “tuning” immune responses has recently been used to combat infectious diseases and cancer, and to induce tolerance during organ transplants or autoimmune disease. Lymph nodes and other secondary lymphoid organs such as the spleen play crucial roles in determining if and how these responses develop following vaccination or immunotherapy. Thus, by manipulating the local microenvironments within these immunological command centers, the nature of systemic immune response can be controlled. This review provides recent examples that harness the interactions between biomaterials and lymph nodes or other secondary lymphoid organs to generate immunity or promote tolerance. These strategies draw on mechanical properties, surface chemistry, stability, and targeting to alter the interactions of cells, signals, and vaccine components in lymph nodes. While there are still many unanswered questions surrounding how best to design biomaterial-based vaccines to promote specific structures or functions in lymph nodes, features such as controlled release and targeting will help pave the way for the next generation of vaccines and immunotherapies that generate immune responses tuned for specific applications.
Collapse
Affiliation(s)
- James I Andorko
- Fischell Department of Bioengineering, University of Maryland, 2212 Jeong H. Kim Engineering Building, College Park, Maryland, 20742, USA
| | | | | |
Collapse
|
9
|
Abstract
Vaccines are typically injected subcutaneously or intramuscularly for stimulation of immune responses. The success of this requires efficient drainage of vaccine to lymph nodes where antigen presenting cells can interact with lymphocytes for generation of the wanted immune responses. The strength and the type of immune responses induced also depend on the density or frequency of interactions as well as the microenvironment, especially the content of cytokines. As only a minute fraction of peripherally injected vaccines reaches the lymph nodes, vaccinations of mice and humans were performed by direct injection of vaccine into inguinal lymph nodes, i.e. intralymphatic injection. In man, the procedure is guided by ultrasound. In mice, a small (5-10 mm) incision is made in the inguinal region of anesthetized animals, the lymph node is localized and immobilized with forceps, and a volume of 10-20 μl of the vaccine is injected under visual control. The incision is closed with a single stitch using surgical sutures. Mice were vaccinated with plasmid DNA, RNA, peptide, protein, particles, and bacteria as well as adjuvants, and strong improvement of immune responses against all type of vaccines was observed. The intralymphatic method of vaccination is especially appropriate in situations where conventional vaccination produces insufficient immunity or where the amount of available vaccine is limited.
Collapse
Affiliation(s)
- Pål Johansen
- Department of Dermatology, University Hospital Zurich
| | | |
Collapse
|
10
|
DeMuth PC, Garcia-Beltran WF, Ai-Ling ML, Hammond PT, Irvine DJ. Composite dissolving microneedles for coordinated control of antigen and adjuvant delivery kinetics in transcutaneous vaccination. ADVANCED FUNCTIONAL MATERIALS 2013; 23:161-172. [PMID: 23503923 PMCID: PMC3595545 DOI: 10.1002/adfm.201201512] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Indexed: 05/17/2023]
Abstract
Transcutaneous administration has the potential to improve therapeutics delivery, providing an approach that is safer and more convenient than traditional alternatives, while offering the opportunity for improved therapeutic efficacy through sustained/controlled drug release. To this end, we demonstrate a microneedle materials platform for rapid implantation of controlled-release polymer depots into the cutaneous tissue. Arrays of microneedles comprised of drug-loaded poly(lactide-co-glycolide) (PLGA) microparticles or solid PLGA tips were prepared with a supporting and rapidly water-soluble poly(acrylic acid) (PAA) matrix. Upon application of microneedle patches to the skin of mice, the microneedles perforated the stratum corneum and epidermis. Penetration of the outer skin layers was followed by rapid dissolution of the PAA binder on contact with the interstitial fluid of the epidermis, implanting the microparticles or solid polymer microneedles in the tissue, which were retained following patch removal. These polymer depots remained in the skin for weeks following application and sustained the release of encapsulated cargos for systemic delivery. To show the utility of this approach we demonstrated the ability of these composite microneedle arrays to deliver a subunit vaccine formulation. In comparison to traditional needle-based vaccination, microneedle delivery gave improved cellular immunity and equivalent generation of serum antibodies, suggesting the potential of this approach for vaccine delivery. However, the flexibility of this system should allow for improved therapeutic delivery in a variety of diverse contexts.
Collapse
Affiliation(s)
- Peter C DeMuth
- Department of Biological Engineering, Massachusetts Institute of Technology77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Wilfredo F Garcia-Beltran
- Program in Health Sciences and Technology, Massachusetts Institute of Technology77 Massachusetts Ave., Cambridge, MA 02139, USA
| | | | - Paula T Hammond
- Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Darrell J Irvine
- Department of Biological Engineering, Massachusetts Institute of Technology77 Massachusetts Ave., Cambridge, MA 02139, USA
- Department of Materials Science and Engineering, Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology77 Massachusetts Ave., Cambridge, MA 02139, USA
- Ragon Institute of MIT, MGH, and HarvardBoston, MA 02139, USA
- Howard Hughes Medical Institute4000 Jones Bridge Rd., Chevy Chase, MD 20815, USA
| |
Collapse
|
11
|
Johansen P, von Moos S, Mohanan D, Kündig TM, Senti G. New routes for allergen immunotherapy. Hum Vaccin Immunother 2012; 8:1525-33. [PMID: 23095873 PMCID: PMC3660774 DOI: 10.4161/hv.21948] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 08/15/2012] [Accepted: 08/23/2012] [Indexed: 12/16/2022] Open
Abstract
IgE-mediated allergy is a highly prevalent disease in the industrialized world. Allergen-specific immunotherapy (SIT) should be the preferred treatment, as it has long lasting protective effects and can stop the progression of the disease. However, few allergic patients choose to undergo SIT, due to the long treatment time and potential allergic adverse events. Since the beneficial effects of SIT are mediated by antigen presenting cells inducing Th1, Treg and antibody responses, whereas the adverse events are caused by mast cells and basophils, the therapeutic window of SIT may be widened by targeting tissues rich in antigen presenting cells. Lymph nodes and the epidermis contain high density of dendritic cells and low numbers of mast cells and basophils. The epidermis has the added benefit of not being vascularised thereby reducing the chances of anaphylactic shock due to leakage of allergen. Hence, both these tissues represent highly promising routes for SIT and are the focus of discussion in this review.
Collapse
Affiliation(s)
- Pål Johansen
- Department of Dermatology; University Hospital Zurich; Zurich, Switzerland
| | - Seraina von Moos
- Clinical Trials Center; University Hospital Zurich; Zurich, Switzerland
| | - Deepa Mohanan
- Department of Dermatology; University Hospital Zurich; Zurich, Switzerland
| | - Thomas M. Kündig
- Department of Dermatology; University Hospital Zurich; Zurich, Switzerland
| | - Gabriela Senti
- Clinical Trials Center; University Hospital Zurich; Zurich, Switzerland
| |
Collapse
|
12
|
Geng S, Zhong Y, Wang S, Liu H, Zou Q, Xie X, Li C, Yu Q, He Z, Wang B. Amiloride enhances antigen specific CTL by faciliting HBV DNA vaccine entry into cells. PLoS One 2012; 7:e33015. [PMID: 22438887 PMCID: PMC3306379 DOI: 10.1371/journal.pone.0033015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 02/03/2012] [Indexed: 12/30/2022] Open
Abstract
The induction of relatively weak immunity by DNA vaccines in humans can be largely attributed to the low efficiency of transduction of somatic cells. Although formulation with liposomes has been shown to enhance DNA transduction of cultured cells, little, if any, effect is observed on the transduction of somatic tissues and cells. To improve the rate of transduction, DNA vaccine delivery by gene gun and the recently developed electroporation techniques have been employed. We report here that to circumvent requirement for such equipment, amiloride, a drug that is prescribed for hypertension treatment, can accelerate plasmid entry into antigen presenting cells (APCs) both in vitro and in vivo. The combination induced APCs more dramatically in both maturation and cytokine secretion. Amiloride enhanced development of full CD8 cytolytic function including induction of high levels of antigen specific CTL and expression of IFN-γ+perforin+granzymeB+ in CD8+ T cells. Thus, amiloride is a facilitator for DNA transduction into host cells which in turn enhances the efficiency of the immune responses.
Collapse
MESH Headings
- Adaptive Immunity/drug effects
- Amiloride/pharmacology
- Animals
- Cell Line
- Female
- Granzymes/metabolism
- Hepatitis B Vaccines/genetics
- Hepatitis B Vaccines/immunology
- Hepatitis B Vaccines/pharmacokinetics
- Immunity, Cellular/drug effects
- Immunity, Innate/drug effects
- Interferon-gamma/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Plasmids/genetics
- Plasmids/pharmacokinetics
- Pore Forming Cytotoxic Proteins/metabolism
- Sodium Channel Blockers/pharmacology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Transduction, Genetic
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vaccines, DNA/pharmacokinetics
Collapse
Affiliation(s)
- Shuang Geng
- Key laboratory of Medical Molecular Virology of MOH and MOE, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yiwei Zhong
- State Key Laboratory for Agro-Biotechnology, China Agricultural University, Beijing, China
| | - Shuang Wang
- State Key Laboratory for Agro-Biotechnology, China Agricultural University, Beijing, China
| | - Hu Liu
- State Key Laboratory for Agro-Biotechnology, China Agricultural University, Beijing, China
| | - Qiang Zou
- State Key Laboratory for Agro-Biotechnology, China Agricultural University, Beijing, China
| | - Xiaoping Xie
- State Key Laboratory for Agro-Biotechnology, China Agricultural University, Beijing, China
| | - Chaofan Li
- Beijing Advaccine Biotechnology, Beijing, China
| | - Qingling Yu
- Beijing Advaccine Biotechnology, Beijing, China
| | - Zhonghuai He
- Key laboratory of Medical Molecular Virology of MOH and MOE, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bin Wang
- State Key Laboratory for Agro-Biotechnology, China Agricultural University, Beijing, China
- Key laboratory of Medical Molecular Virology of MOH and MOE, Shanghai Medical College, Fudan University, Shanghai, China
- * E-mail:
| |
Collapse
|
13
|
Elnekave M, Furmanov K, Hovav AH. Intradermal naked plasmid DNA immunization: mechanisms of action. Expert Rev Vaccines 2012; 10:1169-82. [PMID: 21854310 DOI: 10.1586/erv.11.66] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Plasmid DNA is a promising vaccine modality that is regularly examined in prime-boost immunization regimens. Recent advances in skin immunity increased our understanding of the sophisticated cutaneous immune network, which revived scientific interest in delivering vaccines to the skin. Intradermal administration of plasmid DNA via needle injection is a simple and inexpensive procedure that exposes the plasmid and its encoded antigen to the dermal immune surveillance system. This triggers unique mechanisms for eliciting local and systemic immunity that can confer protection against pathogens and tumors. Understanding the mechanisms of intradermal plasmid DNA immunization is essential for enhancing and modulating its immunogenicity. With regard to vaccination, this is of greater importance as this routine injection technique is highly desirable for worldwide immunization. This article will focus on the current understanding of the mechanisms involved in antigen expression and presentation during primary and secondary syringe and needle intradermal plasmid DNA immunization.
Collapse
Affiliation(s)
- Mazal Elnekave
- Institute of Dental Sciences, Hebrew University-Hadassah School of Dental Medicine, PO Box 122722, Jerusalem 91120, Israel
| | | | | |
Collapse
|
14
|
Weber JS, Vogelzang NJ, Ernstoff MS, Goodman OB, Cranmer LD, Marshall JL, Miles S, Rosario D, Diamond DC, Qiu Z, Obrocea M, Bot A. A phase 1 study of a vaccine targeting preferentially expressed antigen in melanoma and prostate-specific membrane antigen in patients with advanced solid tumors. J Immunother 2011; 34:556-67. [PMID: 21760528 PMCID: PMC3709852 DOI: 10.1097/cji.0b013e3182280db1] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Preferentially expressed antigen in melanoma (PRAME) and prostate-specific membrane antigen (PSMA) are tumor-associated antigens implicated in cellular differentiation, genetic stability, and angiogenesis. MKC1106-PP is an immunotherapeutic regimen cotargeting PRAME and PSMA, comprised of a recombinant plasmid (pPRA-PSM encoding fragments derived from both antigens) and 2 peptides (E-PRA and E-PSM derived from PRAME and PSMA, respectively). This multicenter study evaluated MKC1106-PP with a fixed plasmid dose and 2 different peptide doses, administered by intralymph node injection in a prime-boost sequence in human leukocyte antigen-A*0201 and tumor-antigen-positive patients with progressing metastatic solid tumors who had failed standard therapy. Immune monitoring was done by tetramer and enzymatic-linked immune spot analysis. The treatment was well tolerated, with no significant differences in safety, immune response, and clinical outcome relative to peptide doses. Fifteen of 24 evaluable patients showed an immune response, as defined by the expansion of PRAME-specific or PSMA-specific T cells in the blood. There were no partial or complete responses by the Response Evaluation Criteria in Solid Tumors. Seven patients showed stable disease (SD) for 6 months or longer, or prostate specific antigen decline: 4 of 10 with prostate carcinoma, 2 of 2 with renal clear cell carcinoma, and 1 of 10 with metastatic melanoma. In addition, there was an association between the induction and persistence of antigen-specific T cells in blood above baseline levels and disease control, defined as SD for 6 months or longer. These results support further development of MKC1106-PP in specific clinical indications.
Collapse
Affiliation(s)
- Jeffrey S Weber
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL 33612.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
von Moos S, Kündig TM, Senti G. Novel Administration Routes for Allergen-Specific Immunotherapy: A Review of Intralymphatic and Epicutaneous Allergen-Specific Immunotherapy. Immunol Allergy Clin North Am 2011; 31:391-406, xi. [DOI: 10.1016/j.iac.2011.02.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
16
|
Ribas A, Weber JS, Chmielowski B, Comin-Anduix B, Lu D, Douek M, Ragavendra N, Raman S, Seja E, Rosario D, Miles S, Diamond DC, Qiu Z, Obrocea M, Bot A. Intra–Lymph Node Prime-Boost Vaccination against Melan A and Tyrosinase for the Treatment of Metastatic Melanoma: Results of a Phase 1 Clinical Trial. Clin Cancer Res 2011; 17:2987-96. [DOI: 10.1158/1078-0432.ccr-10-3272] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
17
|
Senti G, Johansen P, Kündig TM. Intralymphatic immunotherapy: from the rationale to human applications. Curr Top Microbiol Immunol 2011; 352:71-84. [PMID: 21725898 DOI: 10.1007/82_2011_133] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Allergen specific immunotherapy (SIT) is the only treatment of IgE mediated allergies that is causative and has a long-term effect. Classically, SIT requires numerous subcutaneous injections of the allergen during 3-5 years. Over the last decade sublingual allergen applications have established as an alternative, but treatment duration could not be shortened. This review focuses on direct administration of vaccines in general and of allergens in particular into lymph nodes with the aim to enhance immunotherapy. Several studies have found that direct injection of antigens into lymph nodes enhanced immune responses. Recently we have focused on intralymphatic allergen administration in order to enhance SIT. Data in mouse models and in clinical trials showed that intralymphatic allergen administration strongly enhanced SIT, so that the number of allergen injections could be reduced to three, and the allergen dose could be reduced 10-100 fold. Intralymphatic injections proved easy, practically painless and safe. In mice and men, intralymphatic immunotherapy injecting allergens into a subcutaneous lymph node markedly enhances the protective immune response, so that both the dose and number of allergen injections can be reduced, making SIT safer and faster, which enhances patient convenience and compliance.
Collapse
Affiliation(s)
- Gabriela Senti
- Clinical Trials Center, Center for Clinical Research, University and University Hospital of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland.
| | | | | |
Collapse
|
18
|
Bot A, Qiu Z, Wong R, Obrocea M, Smith KA. Programmed cell death-1 (PD-1) at the heart of heterologous prime-boost vaccines and regulation of CD8+ T cell immunity. J Transl Med 2010; 8:132. [PMID: 21144062 PMCID: PMC3012026 DOI: 10.1186/1479-5876-8-132] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Accepted: 12/14/2010] [Indexed: 01/24/2023] Open
Abstract
Developing new vaccination strategies and optimizing current vaccines through heterologous prime-boost carries the promise of integrating the benefits of different yet synergistic vectors. It has been widely thought that the increased immunity afforded by heterologous prime-boost vaccination is mainly due to the minimization of immune responses to the carrier vectors, which allows a progressive build up of immunity against defined epitopes and the subsequent induction of broader immune responses against pathogens. Focusing on CD8+ T cells, we put forward a different yet complementary hypothesis based primarily on the systematic analysis of DNA vaccines as priming agents. This hypothesis relies on the finding that during the initiation of immune response, acquisition of co-inhibitory receptors such as programmed cell death-1 (PD-1) is determined by the pattern of antigen exposure in conjunction with Toll-like receptor (TLR)-dependent stimulation, critically affecting the magnitude and profile of secondary immunity. This hypothesis, based upon the acquisition and co-regulation of pivotal inhibitory receptors by CD8+ T cells, offers a rationale for gene-based immunization as an effective priming strategy and, in addition, outlines a new dimension to immune homeostasis during immune reaction to pathogens. Finally, this model implies that new and optimized immunization approaches for cancer and certain viral infections must induce highly efficacious T cells, refractory to a broad range of immune-inhibiting mechanisms, rather than solely or primarily focusing on the generation of large pools of vaccine-specific lymphocytes.
Collapse
Affiliation(s)
- Adrian Bot
- MannKind Corporation, 28903 North Avenue Paine, Valencia, CA 91355, USA.
| | | | | | | | | |
Collapse
|
19
|
Kreiter S, Selmi A, Diken M, Koslowski M, Britten CM, Huber C, Türeci O, Sahin U. Intranodal vaccination with naked antigen-encoding RNA elicits potent prophylactic and therapeutic antitumoral immunity. Cancer Res 2010; 70:9031-40. [PMID: 21045153 DOI: 10.1158/0008-5472.can-10-0699] [Citation(s) in RCA: 228] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although naked antigen-encoding RNA has entered clinical testing, basic knowledge on how to apply this promising novel vaccine format is still pending. By comparing different administration routes, we observed surprisingly potent antigen-specific T-cell immunity upon intranodal injection of naked antigen-encoding RNA. RNA was selectively uptaken by resident dendritic cells, propagated a T-cell attracting and stimulatory intralymphatic milieu, and led to efficient expansion of antigen-specific CD8+ as well as CD4+ T cells. By intranodal treatment of mice with repeated cycles of RNA, we achieved de novo priming of naïve T cells, which became potent cytolytic effectors capable of homing to primary and secondary lymphatic tissues as well as memory T cells. In tumor-bearing mice intralymphatic RNA vaccination elicited protective and therapeutic antitumor immune responses, resulting in a remarkable survival benefit as compared with other treatment regimens. This is the first report of strong systemic antigen-specific Th1-type immunity and cancer cure achieved with naked antigen-encoding RNA in preclinical animal models.
Collapse
Affiliation(s)
- Sebastian Kreiter
- Center for Translational Oncology and Immunology (TRON), Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Smith KA, Qiu Z, Wong R, Tam VL, Tam BL, Joea DK, Quach A, Liu X, Pold M, Malyankar UM, Bot A. Multivalent immunity targeting tumor-associated antigens by intra-lymph node DNA-prime, peptide-boost vaccination. Cancer Gene Ther 2010; 18:63-76. [PMID: 20725097 DOI: 10.1038/cgt.2010.45] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Active immunotherapy of cancer has yet to yield effective therapies in the clinic. To evaluate the translatability of DNA-based vaccines we analyzed the profile of T-cell immunity by plasmid vaccination in a murine model, using transcriptome microarray analysis and flow cytometry. DNA vaccination resulted in specific T cells expressing low levels of co-inhibitory molecules (most notably PD-1), strikingly different from the expression profile elicited by peptide immunization. In addition, the T-cell response primed through this dual-antigen-expressing plasmid (MART-1/Melan-A and tyrosinase) translated into a substantial proliferation capacity and functional conversion to antitumor effector cells after tyrosinase and MART-1/Melan-A peptide analog boost. Furthermore, peptide boost rescued the immune response against the subdominant tyrosinase epitope. This immunization approach could be adapted to elicit potent immunity against multiple tumor antigens, resulting in a broader immune response that was more effective in targeting human tumor cells. Finally, this study sheds light on a novel mechanism of immune homeostasis through synchronous regulation of co-inhibitory molecules on T cells, highly relevant to heterologous prime boost approaches involving DNA vaccines as priming agents.
Collapse
Affiliation(s)
- K A Smith
- Department of Research and Development, Mannkind Corporation, Valencia, CA 91355, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Kachala SS, Servais EL, Park BJ, Rusch VW, Adusumilli PS. Therapeutic sentinel lymph node imaging. Semin Thorac Cardiovasc Surg 2010; 21:327-38. [PMID: 20226346 DOI: 10.1053/j.semtcvs.2009.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2009] [Indexed: 11/11/2022]
Abstract
Improving existing means of sentinel lymph node identification in non-small cell lung cancer will allow for molecular detection of occult micrometastases that may cause recurrence in early stage non-small cell lung cancer. Furthermore, targeted application of chemical and biological cytotoxic agents can potentially improve outcomes in patients with lymph node (LN) metastases. "Therapeutic Sentinel Lymph Node Imaging" incorporates these modalities into a single agent thereby identifying which LNs harbor tumor cells and simultaneously eradicating metastatic disease. In this review, we summarize the novel preclinical agents for identification and treatment of tumor bearing LNs and discuss their potential for clinical translation.
Collapse
Affiliation(s)
- Stefan S Kachala
- Division of Thoracic Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | | | | | | | | |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW IgE-mediated allergy can be treated by subcutaneous allergen-specific immunotherapy (SCIT). However, the percentage of allergic patients undergoing SCIT is low, mainly due to the long duration of the therapy and the risk of severe systemic allergic reactions associated with the allergen administration. Typically, SCIT requires dozens of subcutaneous allergen injections that stretch over 3-5 years. Over the last decade, sublingual immunotherapy has been established as an alternative to SCIT, but treatment duration and dosing frequencies could not be reduced. Recently, immunotherapy by direct administration of the allergen into lymph nodes [intralymphatic immunotherapy (ILIT)] has proven a promising alternative and this method is the focus of the present review. RECENT FINDINGS Several studies on animals and on humans have shown that direct injection into lymph nodes enhanced immune responses to protein, peptide, and naked DNA vaccines. Moreover, ILIT strongly improved allergen immunotherapy, so that the number of allergen administrations as well as the allergen dose could be reduced. As ILIT was also well tolerated, practically painless, and easy to perform, patient compliance was improved as compared with SCIT. SUMMARY Direct ILIT into a subcutaneous lymph node markedly enhances protective immune responses, so that both the dose and the number of allergen injections can be reduced, making ILIT safer and faster than other forms of immunotherapy, and most importantly, this enhances patient convenience and compliance.
Collapse
|
23
|
Synthesis of biodegradable polymer–mesoporous silica composite microspheres for DNA prime-protein boost vaccination. Eur J Pharm Sci 2010; 39:412-20. [DOI: 10.1016/j.ejps.2010.01.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 01/12/2010] [Accepted: 01/22/2010] [Indexed: 11/20/2022]
|
24
|
Current World Literature. Curr Opin Allergy Clin Immunol 2009; 9:574-8. [DOI: 10.1097/aci.0b013e328333c13c] [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]
|
25
|
Smith KA, Meisenburg BL, Tam VL, Pagarigan RR, Wong R, Joea DK, Lantzy L, Carrillo MA, Gross TM, Malyankar UM, Chiang CS, Da Silva DM, Kündig TM, Kast WM, Qiu Z, Bot A. Lymph node-targeted immunotherapy mediates potent immunity resulting in regression of isolated or metastatic human papillomavirus-transformed tumors. Clin Cancer Res 2009; 15:6167-76. [PMID: 19789304 DOI: 10.1158/1078-0432.ccr-09-0645] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE The goal of this study was to investigate the therapeutic potential of a novel immunotherapy strategy resulting in immunity to localized or metastatic human papillomavirus 16-transformed murine tumors. EXPERIMENTAL DESIGN Animals bearing E7-expressing tumors were coimmunized by lymph node injection with E7 49-57 antigen and TLR3-ligand (synthetic dsRNA). Immune responses were measured by flow cytometry and antitumor efficacy was evaluated by tumor size and survival. In situ cytotoxicity assays and identification of tumor-infiltrating lymphocytes and T regulatory cells were used to assess the mechanisms of treatment resistance in bulky disease. Chemotherapy with cyclophosphamide was explored to augment immunotherapy in late-stage disease. RESULTS In therapeutic and prophylactic settings, immunization resulted in a considerable expansion of E7 49-57 antigen-specific T lymphocytes in the range of 1/10 CD8(+) T cells. The resulting immunity was effective in suppressing disease progression and mortality in a pulmonary metastatic disease model. Therapeutic immunization resulted in control of isolated tumors up to a certain volume, and correlated with antitumor immune responses measured in blood. In situ analysis showed that within bulky tumors, T-cell function was affected by negative regulatory mechanisms linked to an increase in T regulatory cells and could be overcome by cyclophosphamide treatment in conjunction with immunization. CONCLUSIONS This study highlights a novel cancer immunotherapy platform with potential for translatability to the clinic and suggests its potential usefulness for controlling metastatic disease, solid tumors of limited size, or larger tumors when combined with cytotoxic agents that reduce the number of tumor-infiltrating T regulatory cells.
Collapse
Affiliation(s)
- Kent A Smith
- Department of Research and Development, MannKind Corporation, Valencia, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wong RM, Smith KA, Tam VL, Pagarigan RR, Meisenburg BL, Quach AM, Carrillo MA, Qiu Z, Bot AI. TLR-9 signaling and TCR stimulation co-regulate CD8(+) T cell-associated PD-1 expression. Immunol Lett 2009; 127:60-7. [PMID: 19751765 DOI: 10.1016/j.imlet.2009.09.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Revised: 08/10/2009] [Accepted: 09/04/2009] [Indexed: 11/28/2022]
Abstract
Elevated Programmed Death-1 (PD-1) expression can inhibit T cell activity and is a potential barrier to achieving persisting and optimal immunity via therapeutic vaccination. Using a direct lymph node-targeted vaccination procedure that enabled uncoupling of synthetic peptide (signal 1, TCR-mediated) and adjuvant (signal 2, non-TCR-mediated), we evaluated the impact of varied doses of Toll-like receptor (TLR)-9 ligand CpG oligodeoxynucleotide (ODN) adjuvant on epitope-specific CD8(+) T cell-associated PD-1 expression. Peptide vaccination without adjuvant yielded CD8(+) T cells with significantly elevated PD-1 expression. This conferred impaired function ex vivo, but was reversible by antibody-mediated PD-1 blockade. By comparison, peptide vaccination with escalating doses of CpG ODN adjuvant yielded higher magnitudes of CD8(+) T cells with progressively lower PD-1 expression and greater ex vivo function. CpG ODN adjuvant in context of titrated peptide doses for vaccination yielded the lowest overall PD-1 expression levels, demonstrating that fine-tuning both TCR-independent (adjuvant dose) and -dependent (antigen dose) stimuli can synergize to co-regulate PD-1 expression on epitope-specific CD8(+) T cells. These data hint at strategies to elicit PD-1(low) CD8(+) T cells using TLR-9 ligand adjuvants, and also shed light on the PD-1-regulated homeostasis of CD8(+) T cells.
Collapse
Affiliation(s)
- Raymond M Wong
- MannKind Corporation, 28903 Avenue Paine, Valencia, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|