Clin Exp Vaccine Res. 2015 Jul;4(2):145-158. English.
Published online Jul 29, 2015.
© Korean Vaccine Society.
Review

Application of radiation technology in vaccines development

Ho Seong Seo
    • Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Korea.
Received May 13, 2015; Revised June 10, 2015; Accepted June 20, 2015.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

One of the earliest methods used in the manufacture of stable and safe vaccines is the use of chemical and physical treatments to produce inactivated forms of pathogens. Although these types of vaccines have been successful in eliciting specific humoral immune responses to pathogen-associated immunogens, there is a large demand for the development of fast, safe, and effective vaccine manufacturing strategies. Radiation sterilization has been used to develop a variety of vaccine types, because it can eradicate chemical contaminants and penetrate pathogens to destroy nucleic acids without damaging the pathogen surface antigens. Nevertheless, irradiated vaccines have not widely been used at an industrial level because of difficulties obtaining the necessary equipment. Recent successful clinical trials of irradiated vaccines against pathogens and tumors have led to a reevaluation of radiation technology as an alternative method to produce vaccines. In the present article, we review the challenges associated with creating irradiated vaccines and discuss potential strategies for developing vaccines using radiation technology.

Keywords
Gamma-radiation; Irradiated vaccine; GVAX; Killed vaccine

Radiation and Vaccines

Radiation is the emission or transmission of energy in the form of waves (ionizing radiation) or electron particles (non-ionizing radiation) [1]. The use of ionizing radiation, including X-rays and gamma rays, has increased substantially over the last 30 years in both medicine and industry [2, 3, 4, 5]. In addition to its initial applications in the diagnosis and treatment of disease, radiation technology has expanded into other areas such as crop breeding, sterilization of food, polymer processing, and processing of environmental pollutants [6, 7, 8, 9]. Radiation technology has also been used in the development of human and animal vaccines, especially in the sterilization and generation of random mutations.

Many vaccines used today rely on technologies developed over 100 years ago, and involve some form of attenuation (i.e., the use of an alternative or mutant strain of pathogenic organism with reduced virulence that maintains its immunogenicity, or inactivation, where chemical or physical methods are used to kill virulent pathogenic strains) [10, 11, 12, 13]. These vaccines have been extremely successful in protecting against animal and human diseases caused by viruses and bacteria. Smallpox and Rinderpest have now been successfully eradicated throughout the world since the introduction of vaccines [14, 15, 16, 17]. Nevertheless, the aim remains to maximize the effectiveness and quality of currently available or new vaccines, because current methods of vaccine manufacture are not cost-effective, are susceptible to chemical contamination, are difficult to match to current circulating strains, and are susceptible to other manufacturing issues.

Radiation technology is of interest to vaccine manufacturers, because it can remove chemical contaminants and penetrate pathogens to damage the DNA [18, 19]. However, the development of irradiated vaccines has not been pursued avidly over the past 20 years for two main reasons. First, the development of new radiation techniques has been considered impractical or difficult due to issues accessing the radiation equipment. Second, it has been thought that modern subunit vaccines would provide a solution, as they can be developed more easily [20]. However, there are several reasons to reevaluate the use of radiated inactivation and attenuation for the production of vaccines [20, 21]. The recent successful development of irradiated vaccines for human malaria and influenza have demonstrated the feasibility and practicality of this technique, and have shown that technical problems can be overcome using existing expertise, without needing to resort to sophisticated technology [22, 23]. Moreover, this technology has been used to produce an anti-cancer vaccine by inactivating cancer cells [24, 25, 26]. In the present review, we discuss several promising candidates for irradiated vaccines that have undergone clinical trials, and assess recent advances in radiation vaccine technologies.

Vaccines Based on Inactivated Microorganisms and Tumor Cells

Inactivated vaccines are produced by killing the pathogens with chemicals, heat, or radiation. These vaccines are more stable and safer than live vaccines, as they can be stored and transported in a freeze-dried form that makes them accessible to people in developing countries [27, 28]. Formaldehyde, the most common chemical used in vaccine production, was first tested in a vaccine by Madsen [29], and was later shown to be successful in preventing several infectious diseases such as typhoid, cholera, poliovirus, hepatitis A, Japanese encephalitis, and tick-borne encephalitis virus [28]. However, it can cause irreversible modifications by cross-linking antigens that can damage key antigenic epitopes, leading to reduced immunogenicity or even exacerbated disease following a microbial infection [30].

β-Propiolactone (BPL) is also a common inactivation method that was first described in 1955 [20]. It is used in the production of influenza and rabies vaccines, and is also used in vaccines currently under development, such as those for Streptococcus pneumoniae. The advantages of this chemical are that it is rapidly neutralized into a nontoxic, noncarcinogenic product by the addition of thiosulphate, and it interacts directly with nucleic acids by inducing DNA double helix cross-linking [31]. However, BPL may also interact with amino acids, which contain nucleophilic moieties that induce conformational changes on surface antigens [32]. Thus, inactivation of pathogens with BPL may also trigger adverse immune reactions, including the induction of allergic responses through chemical modification of the vaccine components [33, 34]. Therefore, there is an increasing demand to identify safe and effective strategies to produce inactivated vaccines, which are crucial for the future of vaccine development.

Radiation inactivation of pathogens has potential applications in sterilization and the manufacture of biological reagents and laboratory supplies [35]. Since the 1940s, when ionizing radiation (e.g., gamma rays and X-rays) was introduced for the sterilization of pathogens, vaccine development using irradiation has been extensively investigated [36, 37]. The major advantages of ionizing radiation in vaccine development compared to ultraviolet light or chemical agents are its ability to penetrate through most biological materials, and the fact that it targets both double and single stranded nucleic acids while causing less damage to surface antigenic proteins. Moreover, there is no need to remove any chemical residue after inactivation. Although the argument remains that irradiated vaccines elicit different immune responses than those generated by heat-killed or chemically killed methods, the demand for the application of gamma radiation is increasing for the development safe vaccines [38, 39, 40].

Irradiated Viral Vaccines

Trivalent inactivated or live attenuated influenza vaccines are commonly used worldwide [11, 41, 42, 43]. Inactivated influenza vaccine is prepared by treating the virus with a chemical agent that contains either the whole inactivated virus or the active part (split or subunit vaccines) [11, 43, 44]. The live attenuated influenza virus has the reduced ability to replicate in human cells, but can still stimulate immune responses [45, 46]. Each year, predictions are made of three potential influenza strains for the coming season, which are based on a continuous worldwide surveillance program by the World Health Organization [42]. However, most influenza vaccines against any predicted seasonal flu provide only modest protection for the given strains, and have little efficacy in the elderly [47, 48, 49, 50]. This is because the influenza vaccine is highly dependent on how well the vaccine strain matches the newly emerging virus. Research is underway to develop a universal vaccine that has a broad coverage of influenza antigenic drift that will not require annual modification [42, 43].

Over the last 60 years, the development of new methods of virus inactivation has been explored [51, 52, 53, 54, 55, 56, 57, 58, 59]. Gamma radiation inactivation has been suggested as an alternative method for inactivation of virus reproduction, primarily by damaging the nucleic acid while preserving immunogenicity. Mullbacher et al. [60] first demonstrated a high cross-protective immune response of irradiated influenza A virus against other influenza A strains. Gamma irradiated influenza vaccine was more effective at priming cross reactive cytotoxic T cells, and protected mice against a heterologous influenza virus [60, 61, 62]. Alsharifi and Mullbacher [63] showed that a single dose of nonadjuvanted intranasal gamma-irradiated influenza A vaccine (GammaFlu) provided robust protection in mice, which was mainly mediated by cytotoxic T cells. Unlike the chemical inactivation method, gamma irradiation preserved the functional domains of the viral proteins, which facilitated uptake and presentation on major histocompatibility complex class I (MHC-I) of antigen presenting cells. This approach has been tested in pre-clinical studies by Gamma Vaccines Pty (Manuka, ACT, Australia), and is now moving towards a full clinical trial.

A vaccine against the human immunodeficiency virus (HIV) would be highly effective for preventing acquired immunodeficiency syndrome. Because HIV was identified in 1983, significant progress has been made in the development of an HIV vaccine worldwide. However, to date, no vaccine has been fully successful. Initially, subunit vaccines, GP120 (VaxGen Inc., San Francisco, CA, USA) and Ad5 (Merck & Co., Kenilworth, NJ, USA), were approved in clinical trials, but have not shown sufficient efficacy in human subjects. In a recent clinical trial conducted by Sanofi Pasteur (Lyon, France), GP120 carrying the Canarypox virus vaccine showed ~25% improved protection compared to a non-vaccinated group of 60,000 human subjects; however, it was dropped at clinical phase III. Over the past 15 years, several groups have initiated the pre-clinical development of inactivated HIV or simian immunodeficiency virus (SIV) vaccines, which conferred potent serological responses against host cell components incorporated into HIV/SIV virions. Currently, the most promising result has come from two gamma-irradiated whole-killed attenuated HIV vaccines, SAV0001 and Remune [64, 65].

Remune, invented by Jonas Salk in 1987 in collaboration with Dr. Dennis Carlo of the Immune Response Corporation (http://www.immuneresponsebiopharma.com) [66, 67], was the first to go to large national trials to ascertain whether it could assist current antivirals by enabling the immune system to control HIV more effectively. This vaccine, derived from an intersubtype recombinant of clade A envelope and clade G Cag, is inactivated through the sequential application of BPL and gamma irradiation. The HIV envelope gp120 glycoprotein is depleted during preparation and inactivation [68]. Although a large-scale multi-center phase III trial with HIV patients on antiretroviral therapy showed no significant differences in the incidence of opportunistic infections or death, a statistically significant decline in viral load, increased CD4+ T cell counts, and enhanced HIV-1 specific antibody responses were observed in the subjects treated with Remune.

SAV001 was developed by Dr. Chil-Yong Kang at Western University, Canada [69]. This represents the first and only preventive HIV vaccine tested in clinical trials, and is based on a genetically modified killed whole-virus. The nef and vpu genes were deleted in the HIV-1 strain to make the attenuated strain, and the env signal peptide was replaced with the honey bee antimicrobial peptide melittin to enhance viral replication and production. Thus, this genetically modified HIV-1 strain is non-pathogenic, and can be produced in large quantities in a cell culture-based system. It is manufactured as a killed vaccine, by harvesting HIV-1 that is completely inactivated by aldrithiol-2 and sequential gamma irradiation [70, 71]. The phase I clinical trial (ClinicalTrials.gov Identifier: NCT01546818) was completed in 2013, and resulted in significant increases in the levels of gp120-specific and P24-specific antibodies, whereas no adverse effects were observed. Phase II/III large multi-center clinical trials on higher risk HIV patients will be conducted shortly.

Irradiated Bacterial Whole Cell Vaccines

Since the typhoid vaccine was first introduced as an inactivated bacterial vaccine at the end of the 19th century, the administration of inactivated whole cell bacterial vaccines is one of the most well-studied methods of vaccination against bacterial infections [20]. This approach offers several advantages. First, they are naturally occurring microparticles that can carry multiple antigens that can be important in providing protection. Second, this approach is relatively quick and inexpensive to manufacture. Although pertussis and anthrax vaccines are the only current licensed inactivated bacterial vaccine used for immunization of the general public, the demand to develop new inactivated bacterial vaccines for emerging pathogens is increasing [72, 73, 74]. Irradiated bacterial vaccines, which prevent replication but retain their metabolic activity, generate higher humoral immune responses and protection against extracellular and intracellular bacteria, including human and animal pathogens (Table 1) [75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86]. Inactivated bacterial vaccines were originally intended for intranasal or oral administration to activate a mucosal immune response [87, 88, 89, 90]. Intranasal immunization with inactivated bacterial vaccines elicits serotype independent humoral as well as cellular immune responses [91, 92].

Leprosy and tuberculosis are the most common mycobacterial diseases representing a major cause of death worldwide [93, 94, 95]. The most effective strategy for treating tuberculosis (TB) is vaccination. Live bacille Calmette-Guerin (BCG), which was introduced in 1921, is the only available vaccine against both diseases [96]. Although it provides immunization protection for infants and young adults, it has had inconsistent and unpredictable results in adults, sometimes causing severe allergenic reactions in the skin, and offers less durable protection that often requires a second boosting immunization [96, 97]. More than 10 TB vaccines are in the early development stages [98, 99, 100]. The most effective clinical results have being obtained using the heat-killed inactivated Mycobacterium obuense vaccine, DAR-901 (ClinicalTrials.gov Identifier: NCT02063555) [101, 102, 103]. A trial in Tanzania of >2,000 HIV positive subjects showed it to be both safe and effective. Irradiated killed TB vaccines were first reported by Olson et al. in 1947 [104] and Paterson et al. in 1949 [105]. Although the irradiated-killed TB vaccine gave a similar degree of protection as the live BCG vaccine in animal models, the allergenic effect was markedly reduced. Because many previous studies have shown that irradiated TB elicits the robust production of antibodies and protection against the challenge of infectious TB, this strategy should be considered an alternative inactivation method for TB whole-cell vaccines (WCVs).

Streptococcus pneumoniae is a causative agent in children and older adults. Currently the available polysaccharide conjugate vaccine (PCV) generates serotype-specific antibody responses [106, 107]. Phase IV surveillance studies in many countries found an increasing number of non-vaccine serotypes and the appearance of new serotypes [108]. Thus, alternative approaches are being considered, such as protein-based vaccines and WCVs. An inactivated whole-cell pneumococcal vaccine has been sought since the beginning of the 20th century. More recently, a collaboration between PATH Vaccine Solutions (Seattle, WA, USA) and Malley's group [109, 110] showed successful results using a chemically killed non-encapsulated pneumococcal vaccine in an animal model and in a clinical phase I trial (ClinicalTrials.gov Identifier: NCT01537185) [93, 111]. No serious adverse events were reported in these clinical trials, and currently, participants are being recruited to test its safety and tolerability with intramuscular administration in healthy Kenyan adults and toddlers who have been primed with PCV vaccines. We also investigated the possibility of creating an irradiated pneumococcal vaccine using non-encapsulated pneumococci. Irradiated killed pneumococci showed non-toxic effects in vitro, whereas chemically killed pneumonia did (unpublished data). In addition, irradiated WCVs elicited a significantly higher level of antibody responses in mice with intranasal and intramuscular vaccinations (Fig. 1). This shows that irradiation is one possible method to manufacture a whole killed pneumococcal vaccine.

Fig. 1
Irradiated pneumococcal vaccine induced antibody titers. Irradiated or formalin-treated vaccines were administered intranasally to CD-1 mice (n = 5) with phosphate buffered saline (PBS), cholera toxin (CT), or lipid A at days 0 and 14. Pneumococcal specific immunoglobulin A in bronchoalveolar lavage fluid (BALF) (A) and total immunoglobulin (B) were measured at 5 days after final immunization.

Salmonella enterica causes a variety of infectious diseases in animals and humans [112, 113]. Live attenuated vaccines generally confer better protection than killed vaccines; however, they are limited by their toxicity and cause mild diarrhea. Previte [114] first described the use of radiation to increase immunogenicity and decrease the toxicity of Salmonella compared to acetone-, alcohol-, and heat-killed vaccines in mouse and bovine salmonellosis models [115, 116]. Brucella species (B. melitensis, B. abortus, B. suis) are the causative agents of brucellosis, a chronic bacterial infection in animals and humans. Some live attenuated vaccines (S19, RB51, Rev1) are licensed for use in animals, but not in humans. However, the live vaccine has been implicated in several accidental infections in humans and animals [117, 118]. Several inactivated Brucella vaccines have been tested as alternative, safer vaccines. In a mouse model, irradiated B. abortus RB51 and B. neotomae induced protection against systemic and mucosal challenge with Brucella spp. Oral vaccination in mice also elicited the activation of CD4 and CD8+ T lymphocytes specific to infectious Brucella species [40, 77, 119, 120, 121, 122, 123, 124].

Irradiated Malaria Vaccines

Human malaria is primarily due to infection with one of five Plasmodium species: Plasmodium falciparum, Plasmodium vivax, Plasmodium malariae, Plasmodium ovale, and Plasmodium knowlesi. Of these, P. falciparum is responsible for 92% of deaths that mostly occur in children living in sub-Saharan Africa [125]. In 2012 there was an estimated 220 million clinical cases and 0.63 million deaths worldwide from malaria infection [23]. An effective vaccine for P. falciparum is needed for use in malaria-endemic populations, but no licensed malaria vaccines and candidates have consistently produced a protective level of efficacy [126].

Based on the life cycle of the malaria parasite and the process of infection, malaria vaccines are divided into four potential target groups; interruption of human to mosquito transmission (parasite sexual and mosquito stages), inhibition of clinical consequences (asexual blood stage), prevention of mosquito to human transmission, and pre-erythrocytic infections (sporozoite [SPZ]/liver stages) [127]. Among them, only vaccines targeting pre-erythrocytic infections showed any significant levels of protection in human clinical trials [128]. The RTS,S/AS01 subunit vaccine consisting of the P. falciparum circumsporozoite protein (CSP) fused with the hepatitis B surface antigen (HBsAg) is one of most advanced anti-malaria vaccines at a clinical level. Although this vaccine did not appear to elicit a CD8+ T cell response, CSP-HBsAg induced a specific CD4+ T cell response targeting the whole SPZ. A large-scale clinical phase III trial (ClinicalTrials.gov Identifier: NCT00866619) in African infants aged 6-8 weeks showed that RTS,S/A01 vaccines provided modest protection (26.0%-36.6% of vaccine efficacy) with no serious adverse effects [129].

A recent landmark finding that set the standards for immunological protection against malaria infection was established by immunization with irradiated SPZ [130, 131]. Because the parasite undergoes morphological changes and displays antigenic variation at each stage of infection, whole parasite vaccines have an advantage [132, 133, 134]. In the early 1940s, Russell and Mohan [130] first demonstrated that inactivated P. gallinaceum SPZ provided protection against challenge with infectious P. gallinaceum. In 1967, Nussenzweig et al. [131] reported that a killed P. berghei SPZ vaccine was unsuccessful, but that an X-ray irradiated SPZ vaccine provided significant protection in an SPZ-challenge mouse model.

In the 1970s, researchers showed that immunizing human volunteers with bites from irradiated mosquitoes carrying P. falciparum SPZ (PfSPZ) or P. vivax SPZ (PvSPZ) provided protection against challenges with infectious SPZ [135, 136, 137, 138, 139]. Because infected mosquitoes cannot be used for immunizing large numbers of individuals, a team at the Vaccine Research Center (VRC) of the National Institutes of Health (NIH) and Sanaria Inc. (Rockville, MD, USA), developed an injectable and cryopreserved irradiated PfSPZ vaccine that met the vaccine regulatory standards [18]. Sanaria Inc. succeeded in raising mosquitoes on an industrial scale to good manufacturing practice (GMP) levels and harvested large amounts of PfSPZ from the mosquito salivary glands. In the phase 1 clinical trials of irradiated PfSPZ (ClinicalTrials.gov Identifier: NCT01001650), all of the subjects administered five or six doses intravenously showed complete resistance to challenges by bites from infected mosquitoes at 3 weeks after their final immunization, whereas five of six unvaccinated controls developed malaria [136, 139]. Additional clinical trials of intravenous administration of the PfSPZ vaccine are planned in multiple locations (ClinicalTrials.gov Identifier: NCT02132299, NCT02215707, NCT0215091, NCT02115 516, NCT02418962).

Irradiated Cancer Cell-Based Vaccine Therapy

Cancer, which is a major health concern worldwide, is a leading cause of morbidity and mortality in developed and developing countries. There were ~14 million new cases and 8.2 million cancer deaths in 2012 [140]. Currently, traditional therapeutic treatments for cancer control and cure include radiotherapy and chemotherapy, which are commonly used worldwide and are considered the most effective ways to prevent tumor growth [3, 141, 142]. Chemotherapy targets the cells that grow and divide quickly, one of the major properties of tumor cells, but causes serious adverse effects as it also targets healthy, fast-dividing cells, such as blood cells and those lining the mouth, stomach, intestines and hair follicles. Hence, myelosuppression (decreased production of blood cells), mucositis (inflammation of the lining of the digestive tract), and alopecia (hair loss) commonly occur after chemotherapy [143, 144, 145, 146, 147]. Radiation therapy using ionizing radiation works by damaging the DNA of tumor cells, leading to cell death. Compared to surgery, radiotherapy is less painful, but the severity and longevity of side effects depend on the dose and duration of radiation administered. The symptoms vary from patient to patient and depend on the concurrent treatment, such as chemotherapy. Common acute side effects include nausea, vomiting, epithelial damage, stomach sores, intestinal discomfort and even infertility [148, 149, 150]. The side effects are caused by damage to the blood vessels and connective tissues, which can lead to fibrosis, lymphedema, and heart disease, which represent hidden future threats. Thus, new and better prophylactic treatments are needed.

A recently developed therapeutic treatment in cancer prevention is a cancer vaccine. Numerous different kinds of cancer vaccines have been tested in clinical trials, but the clinical benefits for the majority of cancer patients still need to be evaluated and confirmed. Cancer vaccines are designed to prevent or cure cancers using the patient's own whole tumor cells or part of the tumor-specific cancer antigens as the source of the vaccines. Unlike chemotherapies and radiotherapies, these vaccines would not cause serious side effects, which would offer an alternative treatment for patients in cases where traditional treatments are not effective. Furthermore, the combination of a cancer vaccine with other cancer therapies could enhance the efficacy of any treatment.

Cancer treatment vaccines are made with the patient's own blood dendrite cells stimulated with cancer antigen(s). These can effectively inhibit or stop tumor cell growth and prevent re-occurrence of cancer after chemotherapy and radiotherapy [151]. In 2010, the Food and Drug Administration (FDA) approved the first cancer treatment vaccine, Provenge (Dendreon, Seattle, WA, USA), which is a patient-customized vaccine targeting metastatic prostate cancer [152]. This vaccine is made from cultured dendritic cells taken from the patient and stimulated with prostatic acid phosphatase (PAP) antigen with granulocyte-macrophage colony-stimulating factor (GM-CSF). Re-infusion of the stimulated dendritic cells into the patient effectively stimulates T-cell dependent immunity, which kills the tumor cells expressing PAP [153]. Results from a clinical trial showed that Provenge reduced the risk of death by 22% and increased survival by 4.1 months compared to the placebo group [154]. However, the cost of current patient-customized therapeutic cancer vaccines is extremely high and the time interval of treatment is long. Both these factors will prevent the vaccine from being widely used worldwide. Provenge treatment consists of three infusions at ~two-week intervals for one month, and the cost for a complete course of treatment is $93,000 [140]. Because of these limitations, new types of cell-based cancer vaccines are being developed.

One of most promising therapeutic cell-based cancer vaccines is GVAX (GM-CSF gene-transduced irradiated cancer vaccine cells) which is one of the vaccines furthest along the process in pre-clinical and clinical trials [155]. Unlike patient-specific cancer vaccines, this vaccine has been developed using patient-specific cancer cells genetically modified to secrete GM-CSF. This makes it easy to manufacture vaccines for various tumor types, such as melanoma, renal, lung, prostate, and pancreatic tumors [156]. Irradiated tumor cells can involve the apoptotic bodies, which would be accepted by the dendritic cells. When the dendritic cells interact with the antigens expressed by irradiated tumor cells, they become mature and present the antigens. In addition, allogenic tumor cell secreting recombinant GM-CSF chemotactically attract immature dendritic cells to induce maturation. The dendritic cells presenting the antigen expressed by the irradiated tumor cells then activate CD4 and CD8 lymphocytes directly [157]. To date, there are many types of GVAX vaccine in clinical trials, either alone or in combination with other therapies, to improve treatment options (Table 2). For example, a GVAX vaccine for prostate cancer (GVAX-PCa) with a co-treatment of Ipilimumab, which is a humanized monoclonal antibody and functions as a CTLA-4 blocker, was first approved by the FDA for the treatment of advanced melanoma in 2011 [158]. Clinically, GVAX immunotherapy combined with Ipilimumab leads to the development of specific antibodies against the tumor cells which prolonged patient survival in cases of pancreatic and prostate cancers in clinical trials (ClinicalTrials.gov Identifier: NCT00836407 and NCT01510288) [25].

Conclusion

Gamma radiation is not a new technique, and has been extensively utilized in the past to sterilize foods and create inactivated vaccines. Whether gamma radiation is superior to conventional inactivation methods, such as heat and chemical treatments, remains a controversial issue. However, due to its ability to effectively penetrate pathogens and cancer cells and specifically target nucleic acids whilst causing less damage to surface antigenic proteins, demands for the use of gamma radiation are increasing to develop safe and simple vaccines. In addition, gamma radiation has several advantages over the use of inactive forms of vaccines, such as the ability to inactivate large volumes, they can be stored in closed containers, and there is no requirement to remove chemical compounds after inactivation. Despite these advantages, no irradiated vaccines have been licensed to date. Here, we reviewed and summarized the current situation regarding irradiated vaccines in pre-clinical and clinical studies. Some irradiated vaccines showed no surprising results compared to live attenuated- or chemically inactivated vaccines, but most of the pre-clinical studies suggested that irradiated vaccines provide more potential immunogenicity than other inactivation methods. Moreover, the metabolically active form of irradiated vaccines were able to activate cytotoxic T cells, which are important immune cells for treating intracellular pathogens and cancers. Therefore, radiation inactivation might provide a feasible, broad-spectrum, simple, and effective technique for the development of novel vaccines.

Notes

No potential conflict of interest relevant to this article was reported.

References

    1. DeLaney TF, Trofimov AV, Engelsman M, Suit HD. Advanced-technology radiation therapy in the management of bone and soft tissue sarcomas. Cancer Control 2005;12:27–35.
    1. Barker CA, Postow MA. Combinations of radiation therapy and immunotherapy for melanoma: a review of clinical outcomes. Int J Radiat Oncol Biol Phys 2014;88:986–997.
    1. Amin NP, Sher DJ, Konski AA. Systematic review of the cost effectiveness of radiation therapy for prostate cancer from 2003 to 2013. Appl Health Econ Health Policy 2014;12:391–408.
    1. McDonald JC. Industrial radiation processing: working behind the scenes. Radiat Prot Dosimetry 2004;109:173–174.
    1. Musilek L. Applied Radiation and Isotopes. The 7th International Topical Meeting on Industrial Radiation and Radio isotope Measurement Application (IRRMA-7). Foreword. Appl Radiat Isot 2010;68:517.
    1. Awan MS, Tabbasam N, Ayub N, et al. Gamma radiation induced mutagenesis in Aspergillus niger to enhance its microbial fermentation activity for industrial enzyme production. Mol Biol Rep 2011;38:1367–1374.
    1. Galante AM, Campos LL. Mapping radiation fields in containers for industrial gamma-irradiation using polycarbonate dosimeters. Appl Radiat Isot 2012;70:1264–1266.
    1. Choi JI, Yoon M, Joe M, et al. Development of microalga Scenedesmus dimorphus mutant with higher lipid content by radiation breeding. Bioprocess Biosyst Eng 2014;37:2437–2444.
    1. Sugimoto T, Shinozaki T, Naruse T, Miyamoto Y. Who was concerned about radiation, food safety, and natural disasters after the great East Japan earthquake and Fukushima catastrophe? A nationwide cross-sectional survey in 2012. PLoS One 2014;9:e106377.
    1. Cho HW, Howard CR, Lee HW. Review of an inactivated vaccine against hantaviruses. Intervirology 2002;45:328–333.
    1. McKeage K. Inactivated quadrivalent split-virus seasonal influenza vaccine (Fluarix(R) quadrivalent): a review of its use in the prevention of disease caused by influenza A and B. Drugs 2013;73:1587–1594.
    1. Grassly NC. Immunogenicity and effectiveness of routine immunization with 1 or 2 doses of inactivated poliovirus vaccine: systematic review and meta-analysis. J Infect Dis 2014;210 Suppl 1:S439–S446.
    1. Gasparini R, Amicizia D, Lai PL, Panatto D. Live attenuated influenza vaccine: a review. J Prev Med Hyg 2011;52:95–101.
    1. Mayr A. Historical review of smallpox, the eradication of smallpox and the attenuated smallpox MVA vaccine. Berl Munch Tierarztl Wochenschr 1999;112:322–328.
    1. Thomas TN, Reef S, Neff L, Sniadack MM, Mootrey GT. A review of the smallpox vaccine adverse events active surveillance system. Clin Infect Dis 2008;46 Suppl 3:S212–S220.
    1. Brown CC. A review of three pathology-based techniques for retrospective diagnosis of rinderpest, with comparison to virus isolation. Res Vet Sci 1997;63:103–106.
    1. Njeumi F, Taylor W, Diallo A, et al. The long journey: a brief review of the eradication of rinderpest. Rev Sci Tech 2012;31:729–746.
    1. Luke TC, Hoffman SL. Rationale and plans for developing a non-replicating, metabolically active, radiation-attenuated Plasmodium falciparum sporozoite vaccine. J Exp Biol 2003;206:3803–3808.
    1. Hewitson JP, Hamblin PA, Mountford AP. Immunity induced by the radiation-attenuated schistosome vaccine. Parasite Immunol 2005;27:271–280.
    1. Plotkin SA. In: History of vaccine development. New York: Springer; 2011.
    1. Artenstein AW. In: Vaccines: a biography. New York: Springer; 2010.
    1. Arama C, Troye-Blomberg M. The path of malaria vaccine development: challenges and perspectives. J Intern Med 2014;275:456–466.
    1. Jindal H, Bhatt B, Malik JS, Sk S, Mehta B. Malaria vaccine: a step toward elimination. Hum Vaccin Immunother 2014;10:1752–1754.
    1. Qin L, Smith BD, Tsai HL, et al. Induction of high-titer IgG antibodies against multiple leukemia-associated antigens in CML patients with clinical responses to K562/GVAX immunotherapy. Blood Cancer J 2013;3:e145.
    1. De Remigis A, de Gruijl TD, Uram JN, et al. Development of thyroglobulin antibodies after GVAX immunotherapy is associated with prolonged survival. Int J Cancer 2015;136:127–137.
    1. Le DT, Wang-Gillam A, Picozzi V, et al. Safety and survival with GVAX pancreas prime and Listeria Monocytogenes-expressing mesothelin (CRS-207) boost vaccines for metastatic pancreatic cancer. J Clin Oncol 2015;33:1325–1333.
    1. Rappuoli R, Bagnoli F. In: Vaccine design: innovative approaches and novel strategies. Norfolk: Caister Academic Press; 2011.
    1. Plotkin SA, Orenstein WA, Offit PA. In: Vaccines. Philadelphia, PA: Elsevier; 2008.
    1. Madsen T. Vaccination against whooping cough. JAMA 1933;101:187–188.
    1. Brown F. Review of accidents caused by incomplete inactivation of viruses. Dev Biol Stand 1993;81:103–107.
    1. Perrin P, Morgeaux S. Inactivation of DNA by beta-propiolactone. Biologicals 1995;23:207–211.
    1. Uittenbogaard JP, Zomer B, Hoogerhout P, Metz B. Reactions of beta-propiolactone with nucleobase analogues, nucleosides, and peptides: implications for the inactivation of viruses. J Biol Chem 2011;286:36198–36214.
    1. Swanson MC, Rosanoff E, Gurwith M, Deitch M, Schnurrenberger P, Reed CE. IgE and IgG antibodies to beta-propiolactone and human serum albumin associated with urticarial reactions to rabies vaccine. J Infect Dis 1987;155:909–913.
    1. Stauffer AO, Barbosa VC. Dissemination strategy for immunizing scale-free networks. Phys Rev E Stat Nonlin Soft Matter Phys 2006;74:056105.
    1. Horl WH, Riegel W, Schollmeyer P. Effect of gamma radiation versus ethylene oxide sterilization of dialyzers and blood lines on plasma levels of granulocyte elastase in hemodialyzed patients. Clin Nephrol 1985;24:232–236.
    1. Dean EE, Howie DL. Safety of food sterilization by ionizing radiation. Bull Parenter Drug Assoc 1964;18:12–26.
    1. Goldblith SA. Radiation sterilization of food. Nature 1966;210:433–434.
    1. Datta M. The 5 component acellular pertussis vaccine combined with tetanus and diphtheria toxoids was efficacious in adolescents and adults. Evid Based Med 2006;11:51.
    1. Datta SK, Okamoto S, Hayashi T, et al. Vaccination with irradiated Listeria induces protective T cell immunity. Immunity 2006;25:143–152.
    1. Magnani DM, Harms JS, Durward MA, Splitter GA. Nondividing but metabolically active gamma-irradiated Brucella melitensis is protective against virulent B. melitensis challenge in mice. Infect Immun 2009;77:5181–5189.
    1. Quach C. Vaccinating high-risk children with the intranasal live-attenuated influenza vaccine: the Quebec experience. Paediatr Respir Rev 2014;15:340–347.
    1. Wutzler P, Hardt R, Knuf M, Wahle K. Targeted vaccine selection in influenza vaccination. Dtsch Arztebl Int 2013;110:793–798.
    1. Pica N, Palese P. Toward a universal influenza virus vaccine: prospects and challenges. Annu Rev Med 2013;64:189–202.
    1. Johnston JA, Tincher LB, Lowe DK. Booster and higher antigen doses of inactivated influenza vaccine in HIV-infected patients. Ann Pharmacother 2013;47:1712–1716.
    1. Haber P, Moro PL, McNeil MM, et al. Post-licensure surveillance of trivalent live attenuated influenza vaccine in adults, United States, Vaccine Adverse Event Reporting System (VAERS), July 2005-June 2013. Vaccine 2014;32:6499–6504.
    1. Prutsky GJ, Domecq JP, Elraiyah T, Prokop LJ, Murad MH. Assessing the evidence: live attenuated influenza vaccine in children younger than 2 years: a systematic review. Pediatr Infect Dis J 2014;33:e106–e115.
    1. Jefferson T, Di Pietrantonj C, Al-Ansary LA, Ferroni E, Thorning S, Thomas RE. Vaccines for preventing influenza in the elderly. Cochrane Database Syst Rev 2010;(2):CD004876.
    1. Jefferson T, Rivetti A, Di Pietrantonj C, Demicheli V, Ferroni E. Vaccines for preventing influenza in healthy children. Cochrane Database Syst Rev 2012;8:CD004879.
    1. Govaert TM, Thijs CT, Masurel N, Sprenger MJ, Dinant GJ, Knottnerus JA. The efficacy of influenza vaccination in elderly individuals: a randomized double-blind placebo-controlled trial. JAMA 1994;272:1661–1665.
    1. Thompson WW, Shay DK, Weintraub E, et al. Mortality associated with influenza and respiratory syncytial virus in the United States. JAMA 2003;289:179–186.
    1. Warfield KL, Swenson DL, Olinger GG, et al. Ebola virus inactivation with preservation of antigenic and structural integrity by a photoinducible alkylating agent. J Infect Dis 2007;196 Suppl 2:S276–S283.
    1. Sharma A, Gupta P, Maheshwari RK. Inactivation of Chikungunya virus by 1,5 iodonapthyl azide. Virol J 2012;9:301.
    1. Nagayama A. Inactivation of influenza A virus by gentian violet (GV) and GV-dyed cotton cloth, and bactericidal activities of these agents. J Infect Chemother 2006;12:73–79.
    1. Kap M, Arron GI, Loibner M, et al. Inactivation of influenza A virus, adenovirus, and cytomegalovirus with PAXgene tissue fixative and formalin. Biopreserv Biobank 2013;11:229–234.
    1. Fedorova AA, Azzami K, Ryabchikova EI, et al. Inactivation of a non-enveloped RNA virus by artificial ribonucleases: honey bees and acute bee paralysis virus as a new experimental model for in vivo antiviral activity assessment. Antiviral Res 2011;91:267–277.
    1. Dumard CH, Barroso SP, de Oliveira GA, et al. Full inactivation of human influenza virus by high hydrostatic pressure preserves virus structure and membrane fusion while conferring protection to mice against infection. PLoS One 2013;8:e80785.
    1. Dembinski JL, Hungnes O, Hauge AG, Kristoffersen AC, Haneberg B, Mjaaland S. Hydrogen peroxide inactivation of influenza virus preserves antigenic structure and immunogenicity. J Virol Methods 2014;207:232–237.
    1. Belanger JM, Raviv Y, Viard M, de la Cruz MJ, Nagashima K, Blumenthal R. Effects of UVA irradiation, aryl azides, and reactive oxygen species on the orthogonal inactivation of the human immunodeficiency virus (HIV-1). Virology 2011;417:221–228.
    1. Ada G. Vaccines and vaccination. N Engl J Med 2001;345:1042–1053.
    1. Mullbacher A, Ada GL, Hla RT. Gamma-irradiated influenza A virus can prime for a cross-reactive and cross-protective immune response against influenza A viruses. Immunol Cell Biol 1988;66(Pt 2):153–157.
    1. Mullbacher A, Marshall ID, Blanden RV. Cross-reactive cytotoxic T cells to alphavirus infection. Scand J Immunol 1979;10:291–296.
    1. Mullbacher A, Marshall ID, Ferris P. Classification of Barmah Forest virus as an alphavirus using cytotoxic T cell assays. J Gen Virol 1986;67(Pt 2):295–299.
    1. Alsharifi M, Mullbacher A. The gamma-irradiated influenza vaccine and the prospect of producing safe vaccines in general. Immunol Cell Biol 2010;88:103–104.
    1. Putkonen P, Thorstensson R, Cranage M, et al. A formalin inactivated whole SIVmac vaccine in Ribi adjuvant protects against homologous and heterologous SIV challenge. J Med Primatol 1992;21:108–112.
    1. Murphey-Corb M, Martin LN, Davison-Fairburn B, et al. A formalin-inactivated whole SIV vaccine confers protection in macaques. Science 1989;246:1293–1297.
    1. Trauger R. Remune response. Nat Biotechnol 1998;16:314.
    1. Churdboonchart V, Sakondhavat C, Kulpradist S, et al. A double-blind, adjuvant-controlled trial of human immunodeficiency virus type 1 (HIV-1) immunogen (Remune) monotherapy in asymptomatic, HIV-1-infected thai subjects with CD4-cell counts of >300. Clin Diagn Lab Immunol 2000;7:728–733.
    1. Moss RB, Giermakowska W, Lanza P, et al. Cross-clade immune responses after immunization with a whole-killed gp120-depleted human immunodeficiency virus type-1 immunogen in incomplete Freund’s adjuvant (HIV-1 immunogen, REMUNE) in human immunodeficiency virus type-1 seropositive subjects. Viral Immunol 1997;10:221–228.
    1. Kang CY, Michalski C. In: HIV combination vaccine and prime boost. Google Patents; 2011.
    1. Rossio JL, Esser MT, Suryanarayana K, et al. Inactivation of human immunodeficiency virus type 1 infectivity with preservation of conformational and functional integrity of virion surface proteins. J Virol 1998;72:7992–8001.
    1. Sheppard HW. Inactivated- or killed-virus HIV/AIDS vaccines. Curr Drug Targets Infect Disord 2005;5:131–141.
    1. Patel SS, Wagstaff AJ. A cellular pertussis vaccine (Infanrix-DTPa; SB-3): a review of its immunogenicity, protective efficacy and tolerability in the prevention of Bordetella pertussis infection. Drugs 1996;52:254–275.
    1. Cherry JD. Historical review of pertussis and the classical vaccine. J Infect Dis 1996;174 Suppl 3:S259–S263.
    1. Grabenstein JD. Anthrax vaccine: a review. Immunol Allergy Clin North Am 2003;23:713–730.
    1. Buddle BM, Aldwell FE, Skinner MA, et al. Effect of oral vaccination of cattle with lipid-formulated BCG on immune responses and protection against bovine tuberculosis. Vaccine 2005;23:3581–3589.
    1. Secanella-Fandos S, Noguera-Ortega E, Olivares F, Luquin M, Julian E. Killed but metabolically active Mycobacterium bovis bacillus Calmette-Guerin retains the antitumor ability of live bacillus Calmette-Guerin. J Urol 2014;191:1422–1428.
    1. Sanakkayala N, Sokolovska A, Gulani J, et al. Induction of antigen-specific Th1-type immune responses by gamma-irradiated recombinant Brucella abortus RB51. Clin Diagn Lab Immunol 2005;12:1429–1436.
    1. Yamamoto T. Current status of cholera and rise of novel mucosal vaccine. Jpn J Infect Dis 2000;53:181–188.
    1. Kabir S. Cholera vaccines: the current status and problems. Rev Med Microbiol 2005;16:101–116.
    1. MDS. Development of 60Co-irradiated cholera vaccine [Internet]. Niigata: Niigata University Graduate School of Medical and Dental Science; [cited 2015 Mar 2].
    1. Pasnik DJ, Evans JJ, Klesius PH. A microwave-irradiated Streptococcus agalactiae vaccine provides partial protection against experimental challenge in Nile Tilapia, Oreochromis niloticus. World J Vaccines 2014;4:184–189.
    1. Tuasikal BJ. In: Streptococcus agalactiae irradiated vaccine candidate for subclinical mastitis prevention in ruminants [dissertation]. Bogor: Bogor Agricultural University; 2012.
    1. Tuasikal BJ, Wibawan IW, Pasaribu FH, Estuningsih S. Bacterial protein characterization of Streptococcus agalactiae by SDS-PAGE method for subclinical mastitis irradiated vaccine materials in dairy cattle. Atom Indonesia 2012;38:66–70.
    1. Gaidamakova EK, Myles IA, McDaniel DP, et al. Preserving immunogenicity of lethally irradiated viral and bacterial vaccine epitopes using a radio-protective Mn2+-Peptide complex from Deinococcus. Cell Host Microbe 2012;12:117–124.
    1. van Diemen PM, Yamaguchi Y, Paterson GK, Rollier CS, Hill AV, Wyllie DH. Irradiated wild-type and Spa mutant Staphylococcus aureus induce anti-S. aureus immune responses in mice which do not protect against subsequent intravenous challenge. Pathog Dis 2013;68:20–26.
    1. Burnside K, Lembo A, Harrell MI, et al. Vaccination with a UV-irradiated genetically attenuated mutant of Staphylococcus aureus provides protection against subsequent systemic infection. J Infect Dis 2012;206:1734–1744.
    1. Malley R, Srivastava A, Lipsitch M, et al. Antibody-independent, interleukin-17A-mediated, cross-serotype immunity to pneumococci in mice immunized intranasally with the cell wall polysaccharide. Infect Immun 2006;74:2187–2195.
    1. Malley R, Lipsitch M, Stack A, et al. Intranasal immunization with killed unencapsulated whole cells prevents colonization and invasive disease by capsulated pneumococci. Infect Immun 2001;69:4870–4873.
    1. Kim SB, Kim SJ, Lee BM, et al. Oral administration of Salmonella enterica serovar Typhimurium expressing swine interleukin-18 induces Th1-biased protective immunity against inactivated vaccine of pseudorabies virus. Vet Microbiol 2012;155:172–182.
    1. Rahman MM, Uyangaa E, Han YW, et al. Enhancement of Th1-biased protective immunity against avian influenza H9N2 virus via oral co-administration of attenuated Salmonella enterica serovar Typhimurium expressing chicken interferon-alpha and interleukin-18 along with an inactivated vaccine. BMC Vet Res 2012;8:105.
    1. Eldar A, Shapiro O, Bejerano Y, Bercovier H. Vaccination with whole-cell vaccine and bacterial protein extract protects tilapia against Streptococcus difficile meningoencephalitis. Vaccine 1995;13:867–870.
    1. Liberman C, Takagi M, Cabrera-Crespo J, et al. Pneumococcal whole-cell vaccine: optimization of cell growth of unencapsulated Streptococcus pneumoniae in bioreactor using animal-free medium. J Ind Microbiol Biotechnol 2008;35:1441–1445.
    1. Chawla S, Garg D, Jain RB, et al. Tuberculosis vaccine: time to look into future. Hum Vaccin Immunother 2014;10:420–422.
    1. Kaufmann SH. Tuberculosis vaccine development at a divide. Curr Opin Pulm Med 2014;20:294–300.
    1. Kaufmann SH, Lange C, Rao M, et al. Progress in tuberculosis vaccine development and host-directed therapies: a state of the art review. Lancet Respir Med 2014;2:301–320.
    1. Cha SB, Shin SJ. Mycobacterium bovis Bacillus Calmette-Guerin (BCG) and BCG-based vaccines against tuberculosis. J Bacteriol Virol 2014;44:236–243.
    1. White AD, Sibley L, Dennis MJ, et al. Evaluation of the safety and immunogenicity of a candidate tuberculosis vaccine, MVA85A, delivered by aerosol to the lungs of macaques. Clin Vaccine Immunol 2013;20:663–672.
    1. Davids V, Hanekom W, Gelderbloem SJ, et al. Dose-dependent immune response to Mycobacterium bovis BCG vaccination in neonates. Clin Vaccine Immunol 2007;14:198–200.
    1. McShane H, Williams A. A review of preclinical animal models utilised for TB vaccine evaluation in the context of recent human efficacy data. Tuberculosis (Edinb) 2014;94:105–110.
    1. da Costa C, Walker B, Bonavia A. Tuberculosis vaccines: state of the art, and novel approaches to vaccine development. Int J Infect Dis 2015;32:5–12.
    1. Mayo RE, Stanford JL. Double-blind placebo-controlled trial of Mycobacterium vaccae immunotherapy for tuberculosis in KwaZulu, South Africa, 1991-97. Trans R Soc Trop Med Hyg 2000;94:563–568.
    1. von Reyn CF, Mtei L, Arbeit RD, et al. Prevention of tuberculosis in Bacille Calmette-Guerin-primed, HIV-infected adults boosted with an inactivated whole-cell mycobacterial vaccine. AIDS 2010;24:675–685.
    1. Johnson JL, Kamya RM, Okwera A, et al. The Uganda-Case Western Reserve University Research Collaboration. Randomized controlled trial of Mycobacterium vaccae immunotherapy in non-human immunodeficiency virus-infected ugandan adults with newly diagnosed pulmonary tuberculosis. J Infect Dis 2000;181:1304–1312.
    1. Olson BJ, Habel K, Piggott WR. A comparative study of live and killed vaccines in experimental tuberculosis. Public Health Rep 1947;62:293–296.
    1. Paterson JC, Crombie DW, Coles JC. Protection by killed vole bacillus vaccine against experimental tuberculosis in guinea pigs. Can J Res E Med Sci 1949;27:37–42.
    1. O'Brien KL. PCV13 impact evaluations: the obvious and the unpredicted. Pediatr Infect Dis J 2013;32:264–265.
    1. Dagan R, Juergens C, Trammel J, et al. Efficacy of 13-valent pneumococcal conjugate vaccine (PCV13) versus that of 7-valent PCV (PCV7) against nasopharyngeal colonization of antibiotic-nonsusceptible Streptococcus pneumoniae. J Infect Dis 2015;211:1144–1153.
    1. Chapman KE, Wilson D, Gorton R. Serotype dynamics of invasive pneumococcal disease post-PCV7 and pre-PCV13 introduction in North East England. Epidemiol Infect 2013;141:344–352.
    1. Goncalves VM, Dias WO, Campos IB, et al. Development of a whole cell pneumococcal vaccine: BPL inactivation, cGMP production, and stability. Vaccine 2014;32:1113–1120.
    1. Moffitt KL, Yadav P, Weinberger DM, Anderson PW, Malley R. Broad antibody and T cell reactivity induced by a pneumococcal whole-cell vaccine. Vaccine 2012;30:4316–4322.
    1. Lu YJ, Leite L, Goncalves VM, et al. GMP-grade pneumococcal whole-cell vaccine injected subcutaneously protects mice from nasopharyngeal colonization and fatal aspiration-sepsis. Vaccine 2010;28:7468–7475.
    1. Coburn B, Grassl GA, Finlay BB. Salmonella, the host and disease: a brief review. Immunol Cell Biol 2007;85:112–118.
    1. Kim K, Shim J, Park S, et al. Evaluation of immune response for Vi-CRM(197) conjugated vaccine against Salmonella enterica serovar Typhi in mice. J Bacteriol Virol 2014;44:52–58.
    1. Previte JJ. Immunogenicity of irradiated Salmonella typhimurium cells and endotoxin. J Bacteriol 1968;95:2165–2170.
    1. Dima FV, Ivanov D, Dima SV. Active immunization against Salmonella typhi by oral administration of fast neutron irradiated cells. Ann N Y Acad Sci 1994;730:348–349.
    1. Begum RH, Rahman H, Ahmed G. Development and evaluation of gamma irradiated toxoid vaccine of Salmonella enterica var Typhimurium. Vet Microbiol 2011;153:191–197.
    1. Elzer PH, Edmonds MD, Hagius SD, Walker JV, Gilsdorf MJ, Davis DS. Safety of Brucella abortus strain RB51 in Bison. J Wildl Dis 1998;34:825–829.
    1. Olsen SC, Holland SD. Safety of revaccination of pregnant bison with Brucella abortus strain RB51. J Wildl Dis 2003;39:824–829.
    1. Ahn TH, Nishihara H, Carpenter CM, Taplin GV. Respiration of gamma irradiated Brucella abortus and Mycobacterium tuberculosis. Proc Soc Exp Biol Med 1962;111:771–773.
    1. Nishihara H, Ahn TH, Taplin GV, Carpenter CM. Immunogenicity of a gamma-irradiated Brucella melitensis vaccine in mice. Cornell Vet 1964;54:573–583.
    1. Oliveira SC, Zhu Y, Splitter GA. Recombinant L7/L12 ribosomal protein and gamma-irradiated Brucella abortus induce a T-helper 1 subset response from murine CD4+ T cells. Immunology 1994;83:659–664.
    1. Surendran N, Hiltbold EM, Heid B, et al. Heat-killed and gamma-irradiated Brucella strain RB51 stimulates enhanced dendritic cell activation, but not function compared with the virulent smooth strain 2308. FEMS Immunol Med Microbiol 2010;60:147–155.
    1. Moustafa D, Garg VK, Jain N, Sriranganathan N, Vemulapalli R. Immunization of mice with gamma-irradiated Brucella neotomae and its recombinant strains induces protection against virulent B. abortus, B. melitensis, and B. suis challenge. Vaccine 2011;29:784–794.
    1. Dabral N, Martha Moreno L, Sriranganathan N, Vemulapalli R. Oral immunization of mice with gamma-irradiated Brucella neotomae induces protection against intraperitoneal and intranasal challenge with virulent B. abortus 2308. PLoS One 2014;9:e107180.
    1. Gething PW, Patil AP, Smith DL, et al. A new world malaria map: Plasmodium falciparum endemicity in 2010. Malar J 2011;10:378.
    1. Schwartz L, Brown GV, Genton B, Moorthy VS. A review of malaria vaccine clinical projects based on the WHO rainbow table. Malar J 2012;11:11.
    1. In brief: PCV13 for adults 65 years and older. Med Lett Drugs Ther 2014;56:102.
    1. Okie S. Betting on a malaria vaccine. N Engl J Med 2005;353:1877–1881.
    1. RTS,S Clinical Trials Partnership. Agnandji ST, Lell B, et al. A phase 3 trial of RTS,S/AS01 malaria vaccine in African infants. N Engl J Med 2012;367:2284–2295.
    1. Russell PF, Mohan BN. The immunization of fowls against mosquito-borne Plasmodium gallinaceum by injections of serum and of inactivated homologous sporozoites. J Exp Med 1942;76:477–495.
    1. Nussenzweig RS, Vanderberg J, Most H, Orton C. Protective immunity produced by the injection of x-irradiated sporozoites of plasmodium berghei. Nature 1967;216:160–162.
    1. Hoffman SL, Goh LM, Luke TC, et al. Protection of humans against malaria by immunization with radiation-attenuated Plasmodium falciparum sporozoites. J Infect Dis 2002;185:1155–1164.
    1. Roestenberg M, McCall M, Hopman J, et al. Protection against a malaria challenge by sporozoite inoculation. N Engl J Med 2009;361:468–477.
    1. Roestenberg M, Teirlinck AC, McCall MB, et al. Long-term protection against malaria after experimental sporozoite inoculation: an open-label follow-up study. Lancet 2011;377:1770–1776.
    1. Laurens MB, Billingsley P, Richman A, et al. Successful human infection with P. falciparum using three aseptic Anopheles stephensi mosquitoes: a new model for controlled human malaria infection. PLoS One 2013;8:e68969.
    1. Seder RA, Chang LJ, Enama ME, et al. Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science 2013;341:1359–1365.
    1. Epstein JE, Richie TL. The whole parasite, pre-erythrocytic stage approach to malaria vaccine development: a review. Curr Opin Infect Dis 2013;26:420–428.
    1. Hoffman SL, Billingsley PF, James E, et al. Development of a metabolically active, non-replicating sporozoite vaccine to prevent Plasmodium falciparum malaria. Hum Vaccin 2010;6:97–106.
    1. Epstein JE, Tewari K, Lyke KE, et al. Live attenuated malaria vaccine designed to protect through hepatic CD8(+) T cell immunity. Science 2011;334:475–480.
    1. Anassi E, Ndefo UA. Sipuleucel-T (provenge) injection: the first immunotherapy agent (vaccine) for hormone-refractory prostate cancer. P T 2011;36:197–202.
    1. Walls B, Jordan L, Diaz L, Miller R. Targeted therapy for cutaneous oncology: a review of novel treatment options for non-melanoma skin cancer: part II. J Drugs Dermatol 2014;13:955–958.
    1. Iacovelli R, Pietrantonio F, Farcomeni A, et al. Chemotherapy or targeted therapy as second-line treatment of advanced gastric cancer: a systematic review and meta-analysis of published studies. PLoS One 2014;9:e108940.
    1. Jansman FG, Sleijfer DT, de Graaf JC, Coenen JL, Brouwers JR. Management of chemotherapy-induced adverse effects in the treatment of colorectal cancer. Drug Saf 2001;24:353–367.
    1. Cortes J, Calvo V, Ramirez-Merino N, et al. Adverse events risk associated with bevacizumab addition to breast cancer chemotherapy: a meta-analysis. Ann Oncol 2012;23:1130–1137.
    1. Cartwright TH. Adverse events associated with antiangiogenic agents in combination with cytotoxic chemotherapy in metastatic colorectal cancer and their management. Clin Colorectal Cancer 2013;12:86–94.
    1. Thomay AA, Nagorney DM, Cohen SJ, et al. Modern chemotherapy mitigates adverse prognostic effect of regional nodal metastases in stage IV colorectal cancer. J Gastrointest Surg 2014;18:69–74.
    1. Peng SL, Thomas M, Ruszkiewicz A, Hunter A, Lawrence M, Moore J. Conventional adverse features do not predict response to adjuvant chemotherapy in stage II colon cancer. ANZ J Surg 2014;84:837–841.
    1. Duncan W, MacDougall RH, Kerr GR, Downing D. Adverse effect of treatment gaps in the outcome of radiotherapy for laryngeal cancer. Radiother Oncol 1996;41:203–207.
    1. Rades D, Fehlauer F, Bajrovic A, Mahlmann B, Richter E, Alberti W. Serious adverse effects of amifostine during radiotherapy in head and neck cancer patients. Radiother Oncol 2004;70:261–264.
    1. Kerns SL, Ostrer H, Rosenstein BS. Radiogenomics: using genetics to identify cancer patients at risk for development of adverse effects following radiotherapy. Cancer Discov 2014;4:155–165.
    1. Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer 2012;12:265–277.
    1. Patel PH, Kockler DR. Sipuleucel-T: a vaccine for metastatic, asymptomatic, androgen-independent prostate cancer. Ann Pharmacother 2008;42:91–98.
    1. Small EJ, Schellhammer PF, Higano CS, et al. Placebo-controlled phase III trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J Clin Oncol 2006;24:3089–3094.
    1. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411–422.
    1. Nemunaitis J. Vaccines in cancer: GVAX, a GM-CSF gene vaccine. Expert Rev Vaccines 2005;4:259–274.
    1. Dummer R. GVAX (Cell Genesys). Curr Opin Investig Drugs 2001;2:844–848.
    1. Geary SM, Lemke CD, Lubaroff DM, Salem AK. Proposed mechanisms of action for prostate cancer vaccines. Nat Rev Urol 2013;10:149–160.
    1. Callahan MK, Wolchok JD. At the bedside: CTLA-4- and PD-1-blocking antibodies in cancer immunotherapy. J Leukoc Biol 2013;94:41–53.

Metrics
Share
Figures

1 / 1

Tables

1 / 2

ORCID IDs
PERMALINK