National Academies Press: OpenBook

Global Health Impacts of Vector-Borne Diseases: Workshop Summary (2016)

Chapter: A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech

« Previous: A1 - Emerging Insect-Transmitted Plant Diseases: The Bacterium Xylella fastidiosa as a Case Study - Rodrigo P. P. Almeida and L. Nunney
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

Schaad, N. W., E. Postnikova, G. Lacy, M. Fatmi, and C.-J. Chang. 2004. Xylella fastidiosa subspecies: X. fastidiosa subsp. piercei, subsp. nov., X. fastidiosa subsp. multiplex subsp. nov., and X. fastidiosa subsp. pauca subsp. nov. Systematic and Applied Microbiology 27:290–300.

Sorensen, J. T., and R. G. Gill. 1996. A range extension of Homalodisca vitripennis (Say) (Hemiptera: Clypeorrhyncha: Cicadellidae) to Southern California. Pan Pacific Entomologist 72:160–161.

Su, C.-C., C.-J. Chang, C.-M. Chang, H.-T. Shih, K.-C. Tzeng, F.-J. Jan, C.-W. Kao, and W.-L. Deng. 2013. Pierce’s disease of grapevines in Taiwan: Isolation, cultivation and pathogenicity of Xylella fastidiosa. Journal of Phytopathology 161:389–396.

Su, C.-C., W.-L. Deng, F.-J. Jan, C.-J. Chang, H. Huang, and J. Chen. 2014. Draft genome sequence of Xylella fastidiosa pear leaf scorch strain in Taiwan. Genome Announcements 2:e00166–14.

Sun, Q., Y. Sun, M. A. Walker, and J. M. Labavitch. 2013. Vascular occlusions in grapevines with Pierce’s disease make disease symptom development worse. Plant Physiology 161:1529–1541.

Turner, W. F., and H. N. Pollard. 1959. Life histories and behavior of five insect vectors of phony peach disease. U.S. Department of Agriculture Technical Bulletin 1188.

Van Sluys, M. A., M. C. de Oliveira, C. B. Monteiro-Vitorello, C. Y. Miyaki, L. R. Furlan, L. E. A. Camargo, A. C. R. da Silva, et al., 2003. Comparative analyses of the complete genome sequences of Pierce’s disease and citrus variegated chlorosis strains of Xylella fastidiosa. Journal of Bacteriology 185:1018–1026.

Young, D. A. 1958. A synopsis of the species of Homalodisca in the United States (Homoptera: Cicadellidae). Bulletin of the Brooklyn Entomology Society 53:7–13.

Yuan, X., L. Morano, R. Bromley, S. Spring-Pearson, R. Stouthamer, and L. Nunney. 2010. Multilocus sequence typing of Xylella fastidiosa causing Pierce’s disease and oleander leaf scorch in the United States. Phytopathology 100:601–611.

Zeilinger, A. R., and M. P. Daugherty. 2014. Vector preference and host defense against infection interact to determine disease dynamics. Oikos 123:613–622.

A2

GENETIC CONTROL OF AEDES MOSQUITOES
1

Luke Alphey,2,3Andrew McKemey,2Derric Nimmo,2Marco Neira Oviedo,2,4Renaud Lacroix,2Kelly Matzen,2and Camilla Beech2

Abstract

Aedes mosquitoes include important vector species such as Aedes aegypti, the major vector of dengue. Genetic control methods are being developed for several of these species, stimulated by an urgent need owing to the poor

___________________

1 Reprinted with permission from Maney Publishing. Originally printed as Alphey et al., 2013. Genetic control of Aedes mosquitoes. Pathogens and Global Health 107(4):170−179.

2 Oxitec Limited, 71 Milton Park, Oxford OX14 4RX, UK.

3 Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, UK.

4 Center for Infectious Disease Research, Pontificia Universidad Católica del Ecuador, Quito, Ecuador. Keywords: Aedes, Genetic engineering, RIDL, SIT, IIT, Wolbachia, Dengue.

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

effectiveness of current methods combined with an increase in chemical pesticide resistance. In this review we discuss the various genetic strategies that have been proposed, their present status, and future prospects. We focus particularly on those methods that are already being tested in the field, including RIDL and Wolbachia-based approaches.

Introduction

Aedes mosquitoes transmit a range of pathogens that cause substantial human morbidity, mortality, and suffering. Dengue, the most important mosquito-borne viral disease with 50–400 million infections per year worldwide (Bhatt et al., 2013; WHO, 2012), is transmitted primarily by Ae. aegypti. Several other Aedes species are competent vectors for dengue in the laboratory and Ae. albopictus in particular has been responsible for some transmission in the field, though it appears much less epidemiologically significant than Ae. aegypti (Lambrechts et al., 2010). The common name of Ae. aegypti is the yellow fever mosquito, indicating another major arbovirus transmitted by mosquitoes of this genus, and there are many more; chikungunya has come to prominence more recently with a major outbreak in the Indian Ocean in 2005–6 (Gérardin et al., 2008; Delatte et al., 2008) and some transmission in Italy in 2006 (Bonilauri et al., 2008). Pathogen transmission is not confined to viruses—lymphatic filariasis in the South Pacific is vectored by Ae. polynesiensis; specific characteristics of this vector may have contributed to the failure of drug-based control programmes in the region (Chambers et al., 2011; O’Connor et al., 2012).

A vaccine has long been available for yellow fever, but remains some way off for dengue, following disappointing results from a recent large trial of the leading candidate (Halstead, 2012; Sabchareon et al., 2012). With no licensed vaccine or specific drug (whether prophylactic or therapeutic), dengue control focuses on the major mosquito vector, Ae. aegypti—and vector control is expected to remain essential even when drugs or vaccines eventually become available. However, current mosquito control methods have limited effectiveness against some key species which breed in small dispersed bodies of water. For Ae. aegypti, these might be water storage containers or rain-water filled artificial containers such as buckets, vases, general refuse, or blocked rainwater gutters. Both private properties and public spaces will have large numbers of such potential breeding sites. Each one may be treated easily by tipping out the water or treating with a chemical or biological toxin, however finding and treating a high enough proportion for effective control is extremely difficult and impractical in most settings. Adulticides are also of limited effectiveness, compounded by increased resistance and the relative ineffectiveness of bednets against day-biting mosquitoes. The inadequacy of current technology is clear: for example, the efficient and well-resourced programme in Singapore, working with a cooperative citizenry, has not been able to prevent epidemic dengue (Egger et al., 2008; Ooi et al., 2006;

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

National Environment Agency, 2012). This, combined with recent enabling technical advances in mosquito genetics, provides the underlying motivation for the development of new genetics-based approaches.

Genetics-based approaches have several features in common.5 Since they depend on vertical (mating-based) transmission of heritable elements,6 they are extremely species-specific. Populations can only be affected by the genetic system if they can interbreed with carriers of that genetic system; other populations will not be directly affected. This species-specific aspect is very attractive from an environmental perspective, as it means that these approaches are exquisitely targeted to the pest or vector species of interest. On the other hand, this feature may be a limitation where multiple pest species are transmitting the same pathogen, in which case a more broad-spectrum approach may be preferred. An additional advantage of genetic control methods is that the control agent, modified insects, will actively disperse and seek mates, so the methods are “homing” or actively target-seeking, as well as specific.

Though some genetic strategies have been developed using classical genetics, such as the Sterile Insect Technique (SIT) (see section below: Population Suppression Strategies—Sterile Males Section), recombinant DNA methods provide a step change in our ability to design and build highly specified, versatile, powerful genetic systems. Several key Aedes species have now been transformed, either by recombinant DNA methods using transposon vectors (Labbé et al., 2010; Coates et al., 1998; Jasinskiene et al., 1998; Rodrigues et al., 2006; Fraser, 2012), or by artificial infection with various Wolbachia, a diverse group of intracellular bacteria (Chambers et al., 2011; Xi et al., 2005, 2006). This opens the door to the development of powerful new genetics-based tools with which to control major vector-borne diseases.

Classifying Genetic Control Strategies

A bewildering variety of genetic control strategies have been proposed; these can be categorised according to the intended outcome, or according to the expected dynamics of the genetic element in the target population. Regarding intended outcome, this may be to reduce the number of individual vector mosquitoes—population suppression—or to reduce the ability of individual mosquitoes within the population to transmit the pathogen. This latter approach is

___________________

5 Genetic control may be defined as “Dissemination, by mating or inheritance, of factors that reduce pest damage” and area-wide control as “Reducing pest damage using measures whose effectiveness depends on application over large expanses” (Mark Benedict, pers. comm.). All proposed genetic strategies are intended for area-wide use, though the minimum useful area varies by species and strategy.

6 One exception might be “paratransgenesis,” the use of modified microbes to change the phenotype of insects with which the microbes associate. Depending on the microbe, horizontal transfer of the modified microbe between insects might be possible. Paratransgenesis is not discussed further in this review.

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

known as “population replacement” or, because the mosquitoes are made refractory to transmission of the pathogen, “refractory insect strategy” (Braig and Yan, 2001; James, 2000; Alphey et al., 2002; Alphey, 2009). However, the target population is not really replaced; rather a genetic element is introduced into it through breeding of released modified mosquitoes with wild individuals, thereby changing the phenotype of some or all individuals in that population—those that carry the new genetic element.

Regarding the expected dynamics of the genetic element in the target population, the element may be intended to persist indefinitely in the target population, potentially also increasing in frequency within the target population and spreading to invade additional populations. These are termed “self-sustaining” genetic systems. The alternative is systems which will not spread or persist, rather they will decrease in prevalence over time and can be maintained in the target population only by periodic release of additional carriers. These are known as “self-limiting” genetic systems.

Population Suppression Strategies—Sterile Males

The most familiar genetics-based population suppression strategy is SIT. This relies on the release of large numbers of sterile males to seek, court, and mate wild females, thereby reducing the reproductive potential of the target wild population. If enough of the wild females mate sterile males then the target population will decline and collapse. SIT has been used successfully for more than 50 years against several major agricultural pests, using radiation-sterilised insects (Dyck et al., 2005; Knipling, 1955). However, the use of radiation imposes several undesirable limitations, including logistical issues, and the somatic damage unavoidably caused by the sterilising dose of radiation used (Andreasen and Curtis, 2005; Helinski et al., 2006, 2009; Helinski and Knols, 2008). Several field trials using radiation- or chemo-sterilised mosquitoes have been conducted, with some success, but there are also problems including poor performance of irradiated mosquitoes (Dame et al., 2009). Though classical methods have recently been revisited for Ae. albopictus (Bellini et al., 2007; Boyer et al., 2011) and An. arabiensis (Helsinki et al., 2008), several alternatives have been explored to avoid the need for irradiation, and to provide additional enhancements, while retaining the many attractive aspects of classical SIT (Alphey et al., 2010). Though “sterile” may strictly indicate agametic sterility, meaning that no gametes are produced, for SIT agametic sterility is not intended or used as it is important that spermatozoa are in fact produced. If aspermic males were used, sperm competition in remating females would likely lead to fertile sperm winning over (non-existent) sperm from sterile males. This would lead to most or all of the eggs from females that mate more than once being fertilised by unmodified sperm and therefore being viable, unless all of their mates are sterile. Increased remating might therefore represent a form of selectable behavioural resistance. However, the barriers to remating

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

vary; where physical barriers such as mating plugs occur selection for increased remating may be less likely. Instead of “agametic,” in the context of SIT and this review “sterile” simply means that some or all of the offspring die. For instance, Wolbachia can induce a form of sterility known as Cytoplasmic Incompatibility (CI), in which embryos from uninfected females fertilised by sperm from infected males fail to develop. Infected males are therefore sterile when mated with uninfected females, though fertile when mating with infected females. This can potentially be used as a sterilising principle for SIT, this variant being called the Incompatible Insect Technique, IIT (Brelsfoard et al., 2008). In classical SIT, the radiation doses used induce dominant lethal mutations in the irradiated sperm such that most eggs die after being fertilised by such sperm. About 95–99% sterility is typical for Mediterranean fruit fly SIT programmes (Bakri et al., 2005; Mumford, 2012); higher sterility can be achieved with more radiation, but at the cost of further damaging the insects. Wolbachia achieve a similar effect—death of offspring of incompatible crosses—in IIT, though the biochemical and genetic mechanism is unknown.

Sterility—death of most or all offspring—can also be achieved by using dominant lethal alleles introduced into the genome by recombinant DNA methods, rather than by irradiation. In the most direct analogous system, so far described only in Anopheles, a nuclease is expressed in the male germline (Windbichler et al., 2008). This gives a sterilising effect much like radiation—and presumably by a similar mechanism, induction of double-stranded breaks in the insect’s chromosomes. Interestingly, the system was designed to cut the X chromosome exclusively, and thereby selectively kill female offspring, though this was not achieved and would in any case be difficult in Aedes mosquitoes that lack a Y chromosome. The underlying molecular system, using sequence-specific nucleases called homing endonucleases (HEGs), is remarkably flexible depending on the precise design. In theory, both self-limiting systems like this SIT example and invasive, self-sustaining genetic systems can be developed with these tools (Burt, 2003; Deredec et al., 2008). Furthermore, although the SIT-like systems described here are clearly self-limiting, self-sustaining population suppression strategies using HEGs have been described, in which reduced-fitness traits are driven into the target population using the super-Mendelian inheritance property of HEGs; in principle this could drive a population or even a species to extinction (Burt, 2003; Deredec et al., 2008).

We have developed a SIT-like system called RIDL (Release of Insects carrying a Dominant Lethal) (Thomas et al., 2000). Here, rather than inducing dominant lethals when required, as with radiation, a dominant lethal transgene is inserted, but its expression is artificially repressed to allow the insects to be reared. One advantage of this approach over the use of DNA damage or CI is the ability to select the time of death of the offspring. Radiation and CI kill affected individuals as embryos, but where there is significant larval density-dependence, a later lethal period can be considerably preferable (Phuc et al., 2007; Atkinson

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

et al., 2007; Yakob and Alphey, 2008; White et al., 2010; Alphey et al., 2011; Barclay, 2005; Bax and Thresher, 2009).

All control interventions place pressures on the target population that may select for various forms of resistance, and genetic control methods are no exception. As mating-based systems, one obvious potential mode of resistance is assortative mating, whereby females are selected to avoid the engineered males. In practice, in decades of use of radiation-based SIT there have been few examples of this, a melon fly control programme in Okinawa being perhaps the only well-documented example (Koyama et al., 2004). Even then, control was successfully achieved simply by releasing more sterile males. Other genetic strategies may have additional potential resistance modes. The use of zygotically active lethal genes in RIDL provides flexibility in terms of engineering the time—and/or sex, see in the following section—of death. In principle, it also allows the possibility of resistance to the zygotic killing mechanism (Alphey et al., 2011b) though this has not yet been observed. Given the large number of effector molecules available, one might expect that new strains could be developed faster than such resistance would emerge; other approaches such as stacking traits may also be useful should this type of resistance prove an issue in practice.

Large-Scale Separation of Males and Females—Genetic Sexing Strains

A further issue is that of sex separation. This is not essential for efficacy—the New World screw-worm was eliminated from a continent by a classical SIT programme releasing both males and females—but it is highly desirable (Dyck et al., 2005). Female mosquitoes will bite and potentially transmit disease even if sterilised. The lifespan of released mosquitoes will likely be reduced by laboratory rearing and handling, significantly reducing their capacity to transmit disease in addition to any effect of the modification itself, nonetheless the possibility of deliberate or accidental release of females may adversely affect public acceptance. Sterile-male methods (e.g., SIT, IIT, RIDL) do not require the release of females, however self-sustaining releases of Wolbachia do require the release of some females because Wolbachia is maternally inherited. Therefore it has been proposed that special strategies using male-biased release should be used to minimise the number of females released (Hancock et al., 2011), though in fact no sex separation was used for the first such release trial (Hoffman et al., 2011).

For some strategies there are additional reasons to remove females beyond their potential to bite. For SIT, large-scale field experiments with Mediterranean fruit flies showed that male-only releases were 3–5 times more effective per male than mixed-sex releases; the sterile females are thought to “distract” the sterile males from seeking out wild females (Rendón et al., 2004). For the Wolbachia-based IIT specifically there is an additional requirement for sex separation—the infected females are fully fertile with both infected and uninfected males, furthermore all their progeny inherit the infection. This means that release of even a

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

single infected female could potentially lead to the alien Wolbachia spreading in the target population. Where the target species is naturally infected with a different, incompatible, strain of Wolbachia, the resulting bidirectional incompatibility will likely limit the spread of the new infection beyond the target area, at least for small target areas. However, if the target species is naturally uninfected, this could lead to the spread of the infection throughout the species. The natural history of Wolbachia, which indicates many independent invasion events, shows this is possible, but not the likelihood, which may be very low per female. This is likely to be seen as an undesirable outcome and therefore a significant risk, unless species-wide invasion is the intent of the release.

Sex separation can be efficiently achieved for some species of mosquitoes, including Ae. aegypti, using physical methods based on the size difference between male and female pupae (Ansari et al., 1977; Focks, 1980; Harris et al., 2011, 2012). Strains that allow genetics-based automated separation of males and females are known as “genetic sexing strains.” Several have been developed using classical genetics, notably the “MACHO” strain which contributed greatly to the success of an SIT programme against An. arabiensis in El Salvador (Dame et al., 2009; Kaiser et al., 1978). However, modern genetics provides more options and also allows such systems to be transferred more readily from one species to another. Several have been developed (Papathanos et al., 2009; Catteruccia et al., 2005; Fu et al., 2007, 2010; Ant et al., 2012; Jin et al., 2013). In principle, any selectable induced sexual dimorphism could be used, but in practice two approaches have been followed, either sex-specific expression of a fluorescent marker allowing automated sorting (Catteruccia et al., 2005; Marois et al., 2012), or sex-specific conditional lethality allowing facile elimination of one sex from a cohort during rearing (Fu et al., 2007). It is possible to use a repressible female-killing system both for sex separation and also for field control (Thomas et al., 2000; Alphey, 2002; Alphey and Andreasen, 2002; Alphey et al., 2008). Insects are reared with the lethal system repressed to provide a colony. Cohorts for release are then reared without the repressor, so that females are eliminated. The resulting males, homozygous for a dominant female-specific lethal gene are released to mate with wild females. All offspring from such a mating inherit one copy of the female-lethal transgene, so daughters die. These are both the vectors and the reproductive potential of the population. Heterozygous sons will pass the transgene on to half of their offspring, resulting in some additional control, though the high fitness cost of a female-lethal trait means that the transgene will be rapidly eliminated from the target population unless maintained by periodic release of additional homozygous males. This is female-specific RIDL, fsRIDL, which has some similarities to the classical field female-killing (FK) systems developed in Lucilia cuprina (Black and Alphey, 2011) and is in principle more efficient than SIT (Schliekelman and Gould, 2000). Furthermore, the use of female-lethal systems may provide additional benefits in terms of resistance management for other approaches used in an integrated vector management

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

programme (Alphey et al., 2007, 2009). fsRIDL strains have been developed for Ae. aegypti (Fu et al., 2010; Wise de Valdez et al., 2011), and Ae. albopictus (Labbé et al., 2012), using flightlessness as a lethal trait.

Refractory Insects

Several approaches have been described for making mosquitoes refractory to malaria, including the expression of specific antibodies (Isaacs et al., 2012), peptides (Ito et al., 2002), or manipulating cell signaling (Corby-Harris et al., 2010). For the arboviruses transmitted by Aedes mosquitoes, RNAi seems an attractive mechanism for suppressing virus replication. Transgene-based expression of a hairpin RNA corresponding to part of the DEN2 virus in either the midgut (Franz et al., 2006) or salivary glands (Mathur et al., 2010) has been shown to provide a strong block to virus transmission. However, for the midgut-expressing line, expression of the anti-DEN2 hairpin and the associated refractory phenotype were lost after about 13 generations (Franz et al., 2009), suggesting that expression may impose a significant fitness cost, and also perhaps that the unusual inverted repeat structure involved may be subject to some form of epigenetic silencing.

Gene Drive Systems

A refractory gene will only have an epidemiologically useful effect if it is present in a significant fraction of the target population. It will probably also have to keep both prevalence and effectiveness high for many vector generations. How can this be achieved? Getting to a high prevalence by simple introgression is difficult in a numerically large population, though not necessarily impossible (Rasgon, 2009). However, since the refractory gene is likely to impose a fitness cost on the mosquitoes, it is likely that both be selected against in terms of prevalence, and also perhaps in terms of loss of function (Marrelli et al., 2006). A system is therefore required which will increase the prevalence within the population over time, despite a selective disadvantage. Such systems are termed “gene drive systems.” Selfish DNA systems (Burt and Trivers, 2006), which have this property of spreading despite not providing an individual fitness benefit, are the main source of inspiration for the design of gene drive systems. Several systems have been proposed (Sinkins and Gould, 2006), but none developed even to proof-of-principle stage in a mosquito. However, a Medea-like system has been demonstrated in Drosophila melanogaster (Chen et al., 2007), using a design which should in principle be transferable to mosquitoes (Hay et al., 2010).

One interesting proposal is the “killer–rescue” system (Gould et al., 2008). By using a lethal transgene and an unlinked repressor, this provides an initial increase in allele frequency of the repressor, but over time both the lethal transgene and the repressor decline in frequency. Though having some gene drive properties, this is therefore still a self-limiting system, which helps to illustrate that there

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

is a spectrum of invasiveness or persistence in genetic systems. At one extreme we have high-penetrance dominant lethal systems killing both males and females, where the transgene is not expected to persist beyond the immediate progeny of the released individuals. Then there are female-lethal systems, where the sons survive but the transgene will still disappear rapidly due to its high fitness cost. Refractory genes that are designed to be neutral will also decline in frequency, but much more slowly due to their much lower fitness cost (some fitness penalty seems inevitable). A transient gene drive system like killer–rescue can provide some boost beyond the initial allele frequency, but still eventually declines. Then on the other side of the self-sustaining/self-limiting divide—which is a very real and significant divide, notwithstanding the shades of persistence and invasiveness on either side of it—we have frequency-dependent systems like underdominance (Davis et al., 2001; Magori and Gould, 2006; Curtis, 1968). This has a high invasion threshold making it relatively unlikely to invade non-target populations well isolated from any target populations. Medea-like systems have a much lower invasion threshold and so are much more likely to spread aggressively into distant populations (Chen et al., 2007; Hay et al., 2010; Marshall et al., 2011), though modifications can in principle be made to reduce this (Marshall et al., 2011). Transposons, long proposed as the basis for gene drive systems though not yet demonstrated, are also potentially highly invasive (James, 2000).

While the relationship of IIT and RIDL with the well-known SIT is clear, there are not such obvious analogies with current methods to guide the testing, deployment, and use of gene drive systems. Some affinity may be found with classical biological control, where the intention is to introduce a parasitoid or predator to control a pest population, expecting that the biocontrol agent will establish and provide long-lasting control, albeit usually incomplete, for the indefinite future. As with classical biological control, there are concerns regarding the lack of control over the gene drive system once released, its unknown evolutionary trajectory post-release, and the essentially irreversible nature of a release, at least in the case of large-scale releases. For these reasons, self-sustaining systems are seen as higher-risk (FAO/IAEA, 2002; Beech et al., 2012; Alphey and Beech, 2012; Benedict et al., 2008, 2010). On the other hand, while sterile-male control looks economically attractive (Atkinson et al., 2007; Alphey et al., 2011a) self-sustaining systems in principle have an even lower cost to deploy as fewer mosquitoes are required, at least after the initial introduction. This theoretical cost advantage depends on being able to use the gene drive system as a “fire- and-forget” weapon; the more expensive the post-release monitoring required, for example to assure the ongoing prevalence, stability, and effectiveness of the modification, the lower the cost differential is likely to be.

A further issue is the possibility that success may lead to decreased vigilance or the loss of capacity to implement previously effective measures if such existed. While this applies to all control methods, whether genetic or not, it may be a significant concern in respect of the use of long-term self-sustaining systems. The

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

“forget” part of “fire-and-forget” should therefore not be taken literally—such methods would still require careful ongoing monitoring for field efficacy, and the development of replacement strains prior to breakdown. This is likely to require significant ongoing resource expenditure.

Can Wolbachia Provide Both Refractoriness and a Gene Drive System?

One striking exception to the slow progress with refractoriness and gene drive systems has come from work on Wolbachia in Ae. aegypti. Though originally developed for IIT and life-shortening strategies, it was observed that infection with certain strains of Wolbachia dramatically reduced susceptibility to a range of pathogens (Hedges et al., 2008; Kambris et al., 2009; Moreira et al., 2009), though potentially increasing susceptibility to others (Hughes et al., 2012). Wolbachia are capable of spreading through insect populations as a heritable modification by manipulating the host’s reproductive biology (Burt and Trivers, 2006; Hancock et al., 2011)—in other words, Wolbachia has the properties of a gene drive system. This raised the possibility that certain strains of Wolbachia might provide a complete gene-drive-plus-refractory-gene package. Attention has focused on wMel, a strain of Wolbachia from Drosophila melanogaster and a laboratory-isolated pathogenic derivative wMelPop. Interestingly—and highlighting the diversity of Wolbachia—wMel infection has a similar dengue-blocking effect in Ae. albopictus, even though Ae. albopictus is naturally infected with two further strains of Wolbachia that do not have this effect (Blagrove et al., 2012). As with cytoplasmic incompatibility, the molecular basis of this pathogen-blocking phenotype is not known, though various studies have implicated upregulation of immune genes or production of reactive oxygen species, or competition for a limited resource such as cholesterol (Kambris et al., 2009; Moreira et al., 2009; Pan et al., 2012; Brennan et al., 2008).

In principle, therefore, a suitable strain of Wolbachia could provide an invasive refractoriness phenotype. Though such invasive genetic systems are seen as relatively risky for reasons outlined above, Wolbachia is not especially invasive, particularly for a strain that has a significant fitness cost, as appears to be the case for wMelPop (Hancock et al., 2011). Introduction of a single infected female can still lead to Wolbachia invading that population, especially if the effective population size is low (Jansen et al., 2008).

Since Wolbachia are naturally occurring, albeit not in Ae. aegypti and the relevant strains are from rather distantly related insects, this use of Wolbachia escapes the regulatory structures and oversight put in place for recombinant DNA technology (De Barro et al., 2011). This may seem rather odd if one considers that addition of any single gene, or less, of DNA from Wolbachia would trigger such an oversight, but the addition of the whole genome does not. However, it is clear that here, as for conventional genetic engineering of mosquitoes, the relevant research groups have worked hard to clarify and then to comply with all

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

applicable regulations (O’Connor et al., 2012; Harris et al., 2012; De Barro et al., 2011; Subramaniam et al., 2012; Beech et al., 2009; Mumford et al., 2009).

For any self-sustaining genetic system, key questions relate to the initial ability to spread and confer the desired phenotype, and the possibility that evolutionary responses will compromise this, or have some other undesirable effect. Though in principle the large-scale use of such systems may be reversible by further genetic intervention, restoring the status quo ante is at best uncertain; this irreversibility has been a major discussion point in respect of gene drive systems. In the case of Wolbachia, one may predict that the introduced strain will co-adapt with Ae. aegypti, reducing the fitness cost of infection but perhaps correspondingly reducing the extent of refractoriness, as both may have the same underlying cause of overproliferation in somatic cells (Lu et al., 2012). However, while the direction seems clear, the rate of decay is very hard to predict, and many generations of protection may be provided. Lack of permanent effect is hardly a reason not to act, but might this tapering protection have some negative aspect? Consequences might include selection for resistant strains of virus. Though initial experiments suggested that wMel infection gave strong refractoriness (Walker et al., 2011), subsequent data using blood from human patients indicated titre-dependent breakthrough (O’Neill, 2011). This suggests that a Wolbachia strain with refractoriness that is incomplete—either as its initial phenotype or arising through co-adaptation with the mosquito—could select for virus strains with higher titre in humans, an undesirable trait. It is also striking that, unlike normal uninfected mosquitoes, Ae. aegypti infected with wMelPop require human blood to produce viable eggs (McMeniman et al., 2011). This would appear to provide strong selection for increased human biting preference, a trait which is central to the transmission of human-specific pathogens, as well as to biting nuisance. Unlike the more catholic Ae. albopictus, Ae. aegypti has a strong preference for anthropophagy, but this is far from absolute and could presumably be increased by such selection (Scott et al., 1993; Siriyasatien et al., 2010; Valerio et al., 2010; Barrera et al., 2012).

These issues illustrate the difficulty of predicting the consequences of releasing a self-sustaining genetic system relating to future evolutionary responses. The use of a “black box” system such as Wolbachia has advantages and disadvantages relative to genetic engineering using well-characterised components. On the one hand Wolbachia is arguably natural—though this may also be true of the elements of an engineered system; in both cases the association with Aedes aegypti is artificially induced, a product of modern biotechnology. To further blur the lines, gene transfer from Wolbachia to insect nuclear genomes is well known, and this can lead to stable transfer of expressed genes (Klasson et al., 2009). Nonetheless, this “natural” aspect is somewhat reassuring, in that Wolbachia strains are already widespread in the environment without known negative effects—though that many strains are harmless does not imply that all are; one could not sustain such an argument for E. coli, for example. On the other hand, a complex

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

uncharacterised system is by definition less well understood and correspondingly more likely to throw up surprises. wMel has an estimated 1,270 protein-coding genes in 1.3 Mb of DNA121—vastly more complex than the 1–4 genes in about 10–20 kb typical for current transgenic insertions. The refractoriness phenotype was a major, beneficial surprise; the human blood requirement was also entirely unexpected, and less welcome. The future evolutionary trajectory of such a complex system may reveal additional surprises—positive or negative.

However, it is a fallacy, sometimes called the nirvana fallacy, to compare actual things with idealised alternatives, for example the risks of future action with a hypothetical risk-free world. Both inaction and alternative actions have risks of their own. Nonetheless, it may be difficult both for regulatory authorities and the general public to compare the relatively well-known risks and hazards of inaction with the unknown aspects of a new technology, even when—as for genetic control—the technology seems likely to offer potentially large net benefits.

Not a “Magic Bullet”

The above discussion has focused on genetic control methods alone. However, current control methods have some strengths as well as weaknesses; an optimal programme is therefore likely to integrate the best of current methods with new technology to achieve the goal of improved control. For example, short-term suppression by conventional methods is likely to be a desirable prelude to either sterile-male or refractory-insect methods as it will reduce the number of modified insects required to achieve a given effect. As further tools become available, such as drugs and vaccines, this integrated vector management approach will naturally expand to integrated disease management—again using an optimal mix of available tools. While there may be a certain inclination simply to “wait for the vaccine,” in practice both vaccine and vector control experts anticipate an ongoing requirement for vector control even when a cheap, effective vaccine is generally available (WHO, 2012)—a hoped-for but perhaps rather distant prospect.

Progress to the Field

In fact, after due consideration, national regulators in several countries have approved small-scale field trials as the next step in an incremental testing and scale-up process. Several self-limiting and one self-sustaining genetic system have been tested in the field to date (Hoffman et al., 2011; Harris et al., 2011, 2012; Lacroix et al., 2012; Brelsfoard and Dobson, 2011). Public perception has generally been positive, though these are early days. The use of Wolbachia, presented as “natural,”7 has largely avoided public concerns relating to the use of recombinant DNA methods. Public response to genetic control, either in general

___________________

7http://www.eliminatedengue.com/, accessed October 18, 2012 and April 17, 2013.

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

or relating to specific applications, may vary considerably depending on a wide range of social, political, epidemiological, presentational, and cultural factors, of which the genetic element is only one; furthermore, this response may vary over time. Even for a well-established approach such as vaccination, participation rates are rarely as high as programme managers would wish, and scare stories such as that regarding MMR vaccine in the UK can still shake public confidence. However, regulatory and social factors, while crucial to the adoption of any new technology, are not the main focus of this review.

Field trials of genetic control methods known to the authors are:

  1. 2009–2010 Cayman Islands: males of a RIDL strain of Ae. aegypti, OX513A,43 were shown to be able to compete successfully for mates with wild mosquitoes; 58 sustained release of these “sterile” males led to strong suppression of the target wild population (Harris et al., 2012).
  2. 2010 Malaysia: OX513A males were shown to have similar longevity and maximum dispersal to an unmodified comparator (Lacroix et al., 2012).
  3. 2010 French Polynesia: sustained release of Ae. polynesiensis males infected with a Wolbachia strain from Ae. riversi for IIT trial (O’Connor et al., 2012; Brelsfoard and Dobson, 2011).
  4. 2011–present: Brazil: sustained release of OX513A males led to strong suppression of a target wild population.8
  5. 2011–present Australia: release of wMel-infected male and female Ae. aegypti led to the invasion and establishment of wMel Wolbachia in two target wild populations (Hoffman et al., 2011); releases underway in three further areas.
  6. Australia: release of wMelPop-infected male and female Ae. aegypti undertaken in two target areas; present status unknown.9
  7. 2013–present Vietnam: release of wMelPop-infected male and female Ae. aegypti on an island (Delatte et al., 2008).

To our knowledge, each of these trials has been successful in accomplishing its experimental objectives, and in no case have any negative consequences to human health or the environment been identified.

Prospects for the Future

One may anticipate that each of the programmes described above will develop further over the coming years, though there will doubtless be numerous technical, legal, and social challenges. In addition, one may anticipate that some

___________________

8http://www.moscamed.org.br/2012/index.php, http://www.oxitec.com, accessed October 18, 2012.

9http://www.eliminatedengue.com, accessed April 17, 2013.

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

of the many approaches at earlier stages of development will progress towards field trials and use. In this regard one may particularly look to synthetic biology approaches to engineered refractoriness and gene drive systems—an approach that has been long heralded and where the daunting technical obstacles are slowly being overcome.

A specific technical question relating to both genetic and conventional vector control is “how low do you have to go?” What is the relationship between the number and competence of vectors and disease transmission? Current dengue control methods rely on population suppression. Genetics-based population suppression has the same aim, so can reasonably be evaluated on the same terms, looking for mosquito suppression, i.e., entomological endpoints. But what about refractory-insect methods, or indeed novel non-genetic methods such as spatial repellents? One would need to show an ability to reduce dengue—an epidemiological endpoint. However, this is extremely difficult for an area-wide intervention, as dengue is highly variable in time and space. Consequently, a trial to show disease suppression would likely need to have many separate treatment and control sites, each of a significant size and with many inhabitants. This is problematic in terms of scale but also in terms of funding—despite the potential, and outstanding early results, funding for genetic control has been extremely low relative to the resources devoted to drugs, vaccines, and insecticides.

Given adequate resources, the future for genetic control looks bright. Numerous research groups are developing exciting approaches; the first of these have successfully completed their first field trials. Genetic control may soon be deployed on a large scale, delivering clean, affordable, sustainable, scalable solutions to major human vector-borne diseases.

Acknowledgments

We thank Stephen Dobson, Hervé Bossin, and Mark Benedict for helpful comments on the manuscript and Pam Gray for proof-reading. Final content remains the responsibility of the authors.

References

Alphey, L. 2002. Re-engineering the sterile insect technique. Insect Biochemistry and Molecular Biology 32:1243–7.

Alphey, L. 2009. Natural and engineered mosquito immunity. Journal of Biology 8:40.

Alphey, L., and M. Andreasen. 2002. Dominant lethality and insect population control. Molecular Biochemistry and Parasitology 121:173–8.

Alphey, L. and C. Beech. 2012. Regulation of agricultural biotechnology: The United States and Canada. In: Chris A Wozniak, Alan McHughen (eds.). The Netherlands: Springer. Ch. 13. p. 281–99.

Alphey L., C.B. Beard, P. Billingsley, M. Coetzee, A. Crisanti, C. Curtis, et al. 2002. Malaria control with genetically manipulated insect vectors. Science 298:119–21.

Alphey, N., P.G. Coleman, C.A. Donnelly, and L. Alphey. 2007. Managing insecticide resistance by mass release of engineered insects. Journal of Economic Entomology 100:1642–9.

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

Alphey L., D. Nimmo, S. O’Connell, N. Alphey. 2008. In: Aksoy S, (ed.), Transgenesis and the management of vector-borne disease. Austin, TX: Landes Bioscience. 627: 93–103.

Alphey, N., L. Alphey, and M. Bonsall. 2009. Combining pest control and resistance management: synergy of engineered insects with Bt crops. Journal of Economic Entomology 102:717–32.

Alphey, L., M. Benedict, R. Bellini, G.G. Clark, D.A. Dame, M.W. Service et al. 2010. Sterile-insect methods for control of mosquito-borne diseases: an analysis. Vector Borne Zoonotic Disease 10:295–311.

Alphey, N., L. Alphey, and M.B. Bonsall. 2011a. A model framework to estimate impact and cost of genetics-based sterile insect methods for dengue vector control. PLoS One 6:e25384.

Alphey, N., L. Alphey, and M.B. Bonsall. 2011b. Modeling resistance to genetic control of insects. Journal of Theoretical Biology 27042–55.55

Andreasen, M.H. and C.F. Curtis. 2005. Optimal life stage for radiation sterilization of Anopheles males and their fitness for release. Medical Veterinary Entomology 19:238–44.

Ansari, M.A., K.R. Singh, G.D. Brooks, P.R. Malhotra, and V. Vaidyanathan. 1977. The development of procedures and techniques for mass rearing of Aedes aegypti. Indian Journal of Medical Research 65(Suppl):91–9.

Ant, T., M. Koukidou, P. Rempoulakis, H.F. Gong, A. Economopoulos, J. Vontas, et al. 2012. Control of the olive fruit fly using genetics-enhanced sterile insect technique. BMC Biology 10:51.

Atkinson, M.P., Z. Su, N. Alphey, L.S. Alphey, P.G. Coleman, and L.M. Wein. 2007. Analyzing the control of mosquito-borne diseases by a dominant lethal genetic system. PNAS 104:22.

Bakri, A., K. Mehta, and D.R. Lance. 2005. Sterilizing insects with ionizing radiation. In Sterile insect technique. Principles and practice in area-wide integrated pest management, edited by V.A. Dyck, J. Hendrichs, and A.S. Robinson. The Netherlands: Springer. Pp. 233–68.

Barclay, H.J. 2005. Mathematical models for the use of sterile insects. In Sterile insect technique. Principles and practice in area-wide integrated pest management, edited by V.A. Dyck, J. Hendrichs, and A.S. Robinson. The Netherlands: Springer. Pp. 147–74.

Barrera, R., A.M. Bingham, H.K. Hassan, M. Amador, A.J. Mackay, and T.R. Unnasch. 2012. Vertebrate hosts of Aedes aegypti and Aedes mediovittatus (Diptera: Culicidae) in rural Puerto Rico. Journal of Medical Entomology 49:917–21.

Bax, N.J. and R.E. Thresher. 2009. Ecological, behavioral, and genetic factors influencing the recombinant control of invasive pests. Ecological Applications 19:873–88.

Beech, C., J. Nagaraju, S. Vasan, R. Rose, R. Othman, V. Pillai, and T. Saraswasthy. 2009. Risk analysis of a hypothetical open field release of a self-limiting transgenic Aedes aegypti mosquito strain to combat dengue. Asia Pacific Journal of Molecular Biology and Biotechnology 17:97–108.

Beech, C., M. Koukidou, N.I. Morrison, and L. Alphey. 2012. Genetically modified insects: Science, use, status and regulation. ICGEB Collection of Biosafety Reviews 6:66–124.

Bellini, R., M. Calvitti, A. Medici, M. Carrieri, G. Celli, and S. Maini. 2007. In Area-wide control of insect pests, edited by M.B. Vreysen, A.S. Robinson, and J. Hendrichs. The Netherlands: Springer. Pp. 505–15.

Benedict, M., P. D’Abbs, S. Dobson, M. Gottlieb, L. Harrington, S. Higgs, et al. 2008. Guidance for contained field trials of vector mosquitoes engineered to contain a gene drive system: Recommendations of a scientific working group. Vector Borne and Zoonotic Diseases 8:127–66.

Benedict, M., M. Eckerstorfer, G. Franz, H. Gaugitsch, A. Greiter, A. Heissenberger, B. Knols, S. Kumschick, W. Nentwig, and W. Rabitsch. 2010. Defining environmental risk assessment criteria for genetically modified insects to be placed on the EU market. External report for European Food Safety Authority. http://www.efsa.europa.eu/en/scdocs/scdoc/71e.htm

Bhatt, S., P.W. Gething, O.J. Brady, J.P. Messina, A.W. Farlow, C.L. Moyes, et al. 2013. The global distribution and burden of dengue. Nature 496(7446): 504–7.

Black, W.C., 4th, L. Alphey, and A.A. James. 2011. Why RIDL is not SIT. Trends in Parasitology 27:362–70.

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

Blagrove, M.S., C. Arias-Goeta, A.B. Failloux, and S.P. Sinkins. 2012. Wolbachia strain wMel induces cytoplasmic incompatibility and blocks dengue transmission in Aedes albopictus. Proceedings of the National Academy of Sciences 109:255–60.

Bonilauri, P., R. Bellini, M. Calzolari, R. Angelini, L. Venturi, F. Fallacara et al. 2008. Chikungunya virus in Aedes albopictus, Italy. Emerging Infectious Diseases 14:852–4.

Boyer, S., J. Gilles, D. Merancienne, G. Lemperiere, and D. Fontenille. 2011. Sexual performance of male mosquito Aedes albopictus. Medical Veterinary Entomology 25:454–9.

Braig, H. and G. Yan. 2001. The spread of genetic constructs in natural insect populations. In Genetically engineered organisms: Assessing environmental and human health effects, edited by D. K. Letournaeu, B. E. Burrows. Boca Raton (Florida): CRC Press. Pp. 251–314.

Brelsfoard, C.L. and Dobson, S.L. 2011. Short note: An update on the utility of Wolbachia for controlling insect vectors and disease transmission. Asia Pacific Journal of Molecular Biology and Biotechnology 19:85–92.

Brelsfoard, C.L., Y. Sechan, and S.L. Dobson. 2008. Interspecific hybridization yields strategy for South Pacific filariasis vector elimination. PLoS Neglected Tropical Diseases 2:e129.

Brennan, L.J., B.A. Keddie, H.R. Braig, and H.L. Harris. 2008. The endosymbiont Wolbachia pipientis induces the expression of host antioxidant proteins in an Aedes albopictus cell line. PLoS One 3:e2083.

Burt, A. 2003. Site-specific selfish genes as tools for the control and genetic engineering of natural populations. Proceedings of the Royal Society: Biological Sciences 270:921–8.

Burt, A. and R. Trivers. 2006. Genes in conflict: The biology of selfish genetic elements. Cambridge, MA: Belknap Press, Harvard University Press.

Catteruccia, F., J.P. Benton, and A. Crisanti. 2005. An Anopheles transgenic sexing strain for vector control. Nature Biotechnology 23:1414–7.

Chambers, E.W., L. Hapairi, B.A. Peel, H. Bossin, and S.L. Dobson. 2011. Male mating competitiveness of a Wolbachia-introgressed Aedes polynesiensis strain under semi-field conditions. PLoS Neglected Tropical Disease 5(8):e1271.

Chen, C.H., H. Huang, C.M. Ward, J.T. Su, L.V. Schaeffer, M. Guo, et al. 2007. A synthetic maternal-effect selfish genetic element drives population replacement in Drosophila. Science 316:597–600.

Coates, C.J., N. Jasinskiene, L. Miyashiro, and A.A. James. 1998. Mariner transposition and transformation of the yellow fever mosquito, Aedes aegypti. Proceedings of the National Academy of Sciences 95:3748–51.

Corby-Harris, V., A. Drexler, L. Watkins de Jong, Y. Antonova, N. Pakpour, R. Ziegler, et al. 2010. Activation of Akt signaling reduces the prevalence and intensity of malaria parasite infection and lifespan in Anopheles stephensi mosquitoes. PLoS Pathogens 6:e1001003.

Curtis C.F. 1968. Possible use of translocations to fix desirable genes in insect pest populations. Nature 218:368–9.

Dame, D.A., C.F. Curtis, M.Q. Benedict, A.S. Robinson, and B.G. Knols. 2009. Historical applications of induced sterilisation in field populations of mosquitoes. Malaria Journal 8:S2.

Davis, S., N. Bax, and P. Grewe. 2001. Engineered underdominance allows efficient and economic introgression of traits into pest populations. Journal of Theoretical Biology 212:83–98.

De Barro, P., B. Murphy, C. Jansen, and J. Murray. 2011. The proposed release of the yellow fever mosquito, Aedes aegypti, containing a naturally occurring strain of Wolbachia pipientis, a question of regulatory responsibility. Journal für Verbraucherschutz und Lebensmittelsicherheit 6(Suppl 1):S33–40.

Delatte, H., C. Paupy, J.S. Dehecq, J. Thiria, A.B. Failloux, and D. Fontenille. 2008. Aedes albopictus, vector of chikungunya and dengue viruses in Reunion Island: Biology and control. Parasite 15:3–13.

Deredec, A., A. Burt, and H.C. Godfray. 2008. Population genetics of using homing endonuclease genes in vector and pest management. Genetics 179:2013–26.

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

Dyck, V.A., J. Hendrichs, and A.S. Robinson. 2005. Sterile insect technique: Principles and practice in area-wide integrated pest management. The Netherlands: Springer.

Egger, J.R., E.E. Ooi, D.W. Kelly, M.E. Woolhouse, C.R. Davies, and P.G. Coleman. 2008. Reconstructing historical changes in the force of infection of dengue fever in Singapore: Implications for surveillance and control. Bulletin of the World Health Organization 86:187–96.

FAO/IAEA. 2002. Status and risk assessment of the use of transgenic arthropods in plant protection. 48 Vienna: FAO/IAEA.

Focks, D.A. 1980. An improved separator for separating the developmental stages, sexes and species of mosquitoes. Mosquito News 19:144–47.

Franz, A.W., I. Sanchez-Vargas, Z.N. Adelman, C.D. Blair, B.J. Beaty, A.A. James, et al. 2006. Engineering RNA interference-based resistance to dengue virus type 2 in genetically modified Aedes aegypti. Proceedings of the National Academy of Sciences 103:4198–203.

Franz, A.W., I. Sanchez-Vargas, J. Piper, M.R. Smith, C.C. Khoo, A.A. James et al. 2009. Stability and loss of a virus resistance phenotype over time in transgenic mosquitoes harbouring an antiviral effector gene. Insect Molecular Biology 18:661–72.

Fraser, M.J., Jr. 2012. Insect transgenesis: Current applications and future prospects. Annual Review of Entomology 57:267–89.

Fu, G., K.C. Condon, M.J. Epton, P. Gong, L. Jin, G.C. Condon, et al. 2007. Female-specific insect lethality engineered using alternative splicing. Nature Biotechnology 25:353–7.

Fu, G., R.S. Lees, D. Nimmo, D. Aw, L. Jin. P. Gray, et al. 2010. Female-specific flightless phenotype for mosquito control. Proceedings of the National Academy of Sciences 107:4550–4.

Gérardin, P., V. Guernier, J. Perrau, A. Fianu, K. Le Roux, P. Grivard, et al. 2008. Estimating Chikungunya prevalence in La Reunion Island outbreak by serosurveys: Two methods for two critical times of the epidemic. BMC Infectious Diseases 8:99.

Gould, F., Y. Huang, M. Legros, and A.L. Lloyd. 2008. A killer–rescue system for self-limiting gene drive of anti-pathogen constructs. Proceedings of the Royal Society: Biological Sciences 275:2823–9.

Halstead, S.B. 2012. Dengue vaccine development: A 75% solution? Lancet 380(9853):1535–6.

Hancock, P.A., S.P. Sinkins, and H.C. Godfray. 2011a. Population dynamic models of the spread of Wolbachia. American Naturalist 177:323–33.

Hancock, P.A., S.P. Sinkins, and H.C. Godfray. 2011b. Strategies for introducing Wolbachia to reduce transmission of mosquito-borne diseases. PLoS Neglected Tropical Diseases 5:e1024.

Harris, A.F., D. Nimmo, A.R. McKemey, N. Kelly, S. Scaife, C.A. Donnelly, et al. 2011. Field performance of engineered male mosquitoes. Nature Biotechnology 29:1034–7.

Harris, A.F., A.R. McKemey, D. Nimmo, Z. Curtis, I. Black, S.A. Morgan, et al. 2012. Successful suppression of a field mosquito population by sustained release of engineered male mosquitoes. Nature Biotechnology 30:828–30.

Hay, B.A., C.H. Chen, C.M. Ward, H. Huang, J.T. Su, and M. Guo. 2010. Engineering the genomes of wild insect populations: Challenges, and opportunities provided by synthetic Medea selfish genetic elements. Journal of Insect Physiology 56:1402–13.

Hedges, L.M., J.C. Brownlie, S.L. O’Neill, and K.N. Johnson. 2008. Wolbachia and virus protection in insects. Science 322:702.

Helinski, M.E. and B.G. Knols. 2008. Mating competitiveness of male Anopheles arabiensis mosquitoes irradiated with a partially or fully sterilizing dose in small and large laboratory cages. Journal of Medical Entomology 45:698–705.

Helinski, M.E., A.G. Parker, and B.G. Knols. 2006. Radiation-induced sterility for pupal and adult stages of the malaria mosquito Anopheles arabiensis. Malaria Journal 5:41.

Helinski, M.E., M.M. Hassan, W.M. El-Motasim, C.A. Malcolm, B.G. Knols, and B. El-Sayed. 2008. Towards a sterile insect technique field release of Anopheles arabiensis mosquitoes in Sudan: Irradiation, transportation, and field cage experimentation. Malaria Journal 7:65.

Helinski, M.E., A.G. Parker, and B.G. Knols. 2009. Radiation biology of mosquitoes. Malaria Journal 8(Suppl 2):S6.

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

Hoffman, A.A., B.L. Montgomery, J. Popovici, I. Iturbe-Ormaetxe, P.H. Johnson, F. Muzzi, et al. 2011. Successful establishment of Wolbachia in Aedes populations to suppress dengue transmission. Nature 476:454–6.

Hughes, G.L., J. Vega-Rodriguez, P. Xue, and J.L. Rasgon. 2012. Wolbachia strain wAlbB enhances infection by the rodent malaria parasite Plasmodium berghei in Anopheles gambiae mosquitoes. Applied Environmental Microbiology 78:1491–95.

Isaacs, A.T., N. Jasinskiene, M. Tretiakov, I. Thiery, A. Zettor, C. Bourgouin, et al. 2012. Transgenic Anopheles stephensi coexpressing single-chain antibodies resist Plasmodium falciparum development. Proceedings of the National Academy of Sciences 109:E1922–30.

Ito, J., A. Ghosh, L.A. Moreira, E.A. Wimmer, and M. Jacobs-Lorena. 2002. Transgenic anopheline mosquitoes impaired in transmission of a malaria parasite. Nature 417:452–5.

James, A. 2000. In: Handler AM, James A.A., (eds.) Insect transgenesis. CRC Press.

Jansen, V.A., M. Turelli, and H.C. Godfray. 2008. Stochastic spread of Wolbachia. Proceedings of the Royal Society: Biological Sciences 275:2769–76.

Jasinskiene, N., C.J. Coates, M.Q. Benedict, A.J. Cornel, C.S. Rafferty, A.A. James et al. 1998. Stable transformation of the yellow fever mosquito, Aedes aegypti, with the Hermes element from the housefly. Proceedings of the National Academy of Sciences 95:3743–7.

Jin, L., A.S. Walker, G. Fu, T. Harvey-Samuel, T. Dafa’alla, A. Miles, T. Marubbi, D. Granville, N. Humphrey-Jones, S. O’Connell, N.I. Morrison, and L. Alphey. 2013. Engineered female-specific lethality for control of pest Lepidoptera. ACS Synthetic Biology 2(3):160-6

Kaiser, P.E., J.A. Seawright, D.A. Dame, and D.J. Joslyn. 1978. Development of a genetic sexing for Anopheles albimanus. Journal of Economic Entomology 71:766–71.

Kambris, Z., P.E. Cook, H.K. Phuc, and S.P. Sinkins. 2009. Immune activation by life-shortening Wolbachia and reduced filarial competence in mosquitoes. Science 326:134–6.

Klasson, L., Z. Kambris, P.E. Cook, T. Walker, and S.P. Sinkins. 2009. Horizontal gene transfer between Wolbachia and the mosquito Aedes aegypti. BMC Genomics 10:33.

Knipling E. 1955. Possibilities of insect control or eradication through the use of sexually sterile males. Journal of Economic Entomology 48:459–69.

Koyama, J., H. Kakinohana, and T. Miyatake. 2004. Eradication of the melon fly Bactrocera cucurbitae, in Japan: Importance of behavior, ecology, genetics, and evolution. Annual Review of Entomology 49:331–49.

Labbé, G.M., D.D. Nimmo, and L. Alphey. 2010. piggyBac- and PhiC31-mediated genetic transformation of the Asian tiger mosquito, Aedes albopictus (Skuse). PLoS Neglected Tropical Disease 4:e788.

Labbé, G.M., S. Scaife, S.A. Morgan, Z.H. Curtis, and L. Alphey. 2012. Female-specific flightless (fsRIDL) phenotype for control of Aedes albopictus. PLoS Neglected Tropical Diseases 6:e1724.

Lacroix, R., A.R. McKemey, N. Raduan, L. Kwee Wee, W. Hong Ming, T. Guat Ney, et al. 2012. Open field release of genetically engineered sterile male Aedes aegypti in Malaysia. PLoS One 7:e42771.

Lambrechts, L., T.W. Scott, and D.J Gubler. 2010. Consequences of the expanding global distribution of Aedes albopictus for dengue virus transmission. PLoS Neglected Tropical Diseases 4:e646.

Lu, P., G. Bian, X. Pan, and Z. Xi. 2012. Wolbachia induces density-dependent inhibition to dengue virus in mosquito cells. PLoS Neglected Tropical Diseases 6:e1754.

Magori, K., and F. Gould. 2006. Genetically engineered underdominance for manipulation of pest populations: A deterministic model. Genetics 172:2613–20.

Marois, E., C. Scali, J. Soichot, C. Kappler, E.A. Levashina, and F. Catteruccia. 2012. High-throughput sorting of mosquito larvae for laboratory studies and for future vector control interventions. Malaria Journal 11:302.

Marrelli, M.T., C.K. Moreira, D. Kelly, L. Alphey, and M. Jacobs-Lorena. 2006. Mosquito transgenesis: What is the fitness cost? Trends in Parasitology 22:197–202.

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

Marshall, J.M., G.W. Pittman, A.B. Buchman, and B.A. Hay. 2011. Semele: A killer-male, rescue-female system for suppression and replacement of insect disease vector populations. Genetics 187:535–51.

Mathur, G., I. Sanchez-Vargas, D. Alvarez, K.E. Olson, O. Marinotti, and A.A. James. 2010. Trans-gene-mediated suppression of dengue viruses in the salivary glands of the yellow fever mosquito, Aedes aegypti. Insect Molecular Biology 19:753–63.

McMeniman, C.J., G.L. Hughes, and S.L. O’Neill. 2011. A Wolbachia symbiont in Aedes aegypti disrupts mosquito egg development to a greater extent when mosquitoes feed on nonhuman versus human blood. Journal of Medical Entomology 48:76–84.

Moreira, L.A., I. Iturbe-Ormaetxe, J.A. Jeffery, G. Lu, A.T. Pyke, L.M. Hedges, et al. 2009. A Wolbachia symbiont in Aedes aegypti limits infection with dengue, chikungunya, and Plasmodium. Cell 139:1268–78.

Mumford, J.D. 2012. Science, regulation, and precedent for genetically modified insects. PLoS Neglected Tropical Diseases 6:e1504.

Mumford, J., M.M. Quinlan, C.J. Beech, L. Alphey, V. Bayard, M.L. Capurro, P. Kittayapong, J.D. Knight, M.T. Marrelli, K. Ombongi, J.M. Ramsey, and R. Reuben. 2009. MosqGuide: A project to develop best practice guidance for the deployment of innovative genetic vector control strategies for malaria and dengue. Asia Pacific Journal of Molecular Biology and Biotechnology 17:91–3.

National Environment Agency. 2012. Campaign against dengue. Available at http://www.dengue.gov.sg/

O’Connor, L., C. Plichart, A.C. Sang, C.L. Brelsfoard, H.C. Bossin, and S.L. Dobson. 2012. Open release of male mosquitoes infected with a Wolbachia biopesticide: Field performance and infection containment. PLoS Neglected Tropical Diseases 6:e1797.

O’Neill, S.L. 2011. Wolbachia infections of Aedes aegypti and their potential to control dengue transmission. American Journal of Tropical Medicine and Hygiene 85:169.

Ooi, E.E., K.T. Goh, and D.J. Gubler. 2006. Dengue prevention and 35 years of vector control in Singapore. Emerging Infectious Diseases 12:887–93.

Pan, X., G. Zhou, J. Wu, G. Bian, P. Lu, A.S. Raikhel, et al. 2012. Wolbachia induces reactive oxygen species (ROS)-dependent activation of the Toll pathway to control dengue virus in the mosquito Aedes aegypti. Proceedings of the National Academy of Sciences 109:E23–31.

Papathanos, P.A., H.C. Bossin, M.Q. Benedict, F. Catteruccia, C.A. Malcolm, L. Alphey, et al. 2009. Sex separation strategies: Past experience and new approaches. Malaria Journal 8(Suppl 2):S5.

Phuc, H.K., M.H. Andreasen, R.S. Burton, C. Vass, M.J. Epton, G. Pape, et al. 2007. Late-acting dominant lethal genetic systems and mosquito control. BMC Biology 5:11.

Rasgon, J.L. 2009. Multi-locus assortment (MLA) for transgene dispersal and elimination in mosquito populations. PLoS One 4:e5833.

Rendón, P., D. McInnis, D. Lance, and J. Stewart. 2004. Medfly (Diptera:Tephritidae) genetic sexing: Large-scale field comparison of males-only and bisexual sterile fly releases in Guatemala. Journal of Economic Entomology 97:1547–53.

Rodrigues, F.G., S.B. Oliveira, B.C. Rocha, and L.A. Moreira. 2006. Germline transformation of Aedes fluviatilis (Diptera:Culicidae) with the piggyBac transposable element. Memórias do Instituto Oswaldo Cruz 101:755–7.

Sabchareon, A., D. Wallace, C. Sirivichayakul, K. Limkittikul, P. Chanthavanich, S. Suvannadabba, et al. 2012. Protective efficacy of the recombinant, live-attenuated, CYD tetravalent dengue vaccine in Thai schoolchildren: A randomised, controlled phase 2b trial. Lancet 380(9853):1559–67.

Schliekelman, P., and F. Gould. 2000. Pest control by the release of insects carrying a female-killing allele on multiple loci. Journal of Economic Entomology 93:1566–79.

Scott, T.W., E. Chow, D. Strickman, P. Kittayapong, R.A. Wirtz, L.H. Lorenz, et al. 1993. Blood-feeding patterns of Aedes aegypti (Diptera: Culicidae) collected in a rural Thai village. Journal of Medical Entomology 30:922–7.

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×

Sinkins, S.P. and F. Gould. 2006. Gene drive systems for insect disease vectors. National Review of Genetics 7:427–35.

Siriyasatien, P., T. Pengsakul, V. Kittichai, A. Phumee, S. Kaewsaitiam, U. Thavara, et al. 2010. Identification of blood meal of field caught Aedes aegypti (L.) by multiplex PCR. Southeast Asian Journal of Tropical Medicine and Public Health 41:43–7.

Subramaniam, T.S., H.L. Lee, W.A. Nazni, and S. Murad. 2012. Genetically modified mosquito: The Malaysian public engagement experience. Biotechnology Journal 7:1323–7.

Thomas, D.D., C.A. Donnelly, R.J. Wood, and L.S. Alphey. 2000. Insect population control using a dominant, repressible, lethal genetic system. Science 287:2474–6.

Valerio, L., F. Marini, G. Bongiorno, L. Facchinelli, M. Pombi, B. Caputo, et al. 2010. Host-feeding patterns of Aedes albopictus (Diptera: Culicidae) in urban and rural contexts within Rome province, Italy. Vector Borne and Zoonotic Diseases 10:291–4.

Walker, T., P.H. Johnson, L.A. Moreira, I. Iturbe-Ormaetxe, F.D. Frentiu, C.J. McMeniman, et al. 2011. The wMel Wolbachia strain blocks dengue and invades caged Aedes aegypti populations. Nature 476:450–3.

White, S.M., P. Rohani, and S.M. Sait. 2010. Modelling pulsed releases for sterile insect techniques: Fitness costs of sterile and transgenic males and the effects on mosquito dynamics. Journal Applied Ecology 47:1329–39.

WHO. 2012. Global strategy for dengue prevention and control 2012–2020. Geneva: WHO.

Windbichler, N., P.A. Papathanos, and A. Crisanti. 2008. Targeting the X chromosome during spermatogenesis induces Y chromosome transmission ratio distortion and early dominant embryo lethality in Anopheles gambiae. PLoS Genetics 4:e1000291.

Wise de Valdez, M.R, D. Nimmo, J. Betz, H.F. Gong, A.A. James, L. Alphey, et al. 2011. Genetic elimination of dengue vector mosquitoes. Proceedings of the National Academy of Sciences 108:4772–5.

Wu, M., L.V. Sun, J. Vamathevan, M. Riegler, R. Deboy, J.C. Brownlie, et al. 2004. Phylogenomics of the reproductive parasite Wolbachia pipientis wMel: A streamlined genome overrun by mobile genetic elements. PLoS Biology 2:e69.

Xi, Z., C.C. Khoo, and S.L. Dobson. 2005. Wolbachia establishment and invasion in an Aedes aegypti laboratory population. Science. 310:326–8.

Xi, Z., C.C. Khoo, and S.L. Dobson. 2006. Interspecific transfer of Wolbachia into the mosquito disease vector Aedes albopictus. Proceedings of Biological Sciences 273:1317–22.

Yakob, L., L. Alphey, and M. Bonsall. 2008. Aedes aegypti control: The concomitant role of competition, space and transgenic technologies. Journal of Applied Ecology 45:1258–65.

Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 106
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 107
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 108
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 109
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 110
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 111
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 112
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 113
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 114
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 115
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 116
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 117
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 118
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 119
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 120
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 121
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 122
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 123
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 124
Suggested Citation:"A2 - Genetic Control of Aedes Mosquitoes - Luke Alphey, Andrew McKemey, Derric Nimmo, Marco Neira Oviedo, Renaud Lacroix, Kelly Matzen, and Camilla Beech ." National Academies of Sciences, Engineering, and Medicine. 2016. Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: The National Academies Press. doi: 10.17226/21792.
×
Page 125
Next: A3 - The Intensifying Storm: Domestication of Aedes aegypti, Urbanization of Arboviruses, and Emerging Insecticide Resistance - Barry J. Beaty, William C. Black IV, Lars Eisen, Adriana E. Flores, Julin E. Garca-Rejn, Mara Loroo-Pino, and Karla Saavedra-Rodriguez »
Global Health Impacts of Vector-Borne Diseases: Workshop Summary Get This Book
×
 Global Health Impacts of Vector-Borne Diseases: Workshop Summary
Buy Paperback | $80.00 Buy Ebook | $64.99
MyNAP members save 10% online.
Login or Register to save!
Download Free PDF

Pathogens transmitted among humans, animals, or plants by insects and arthropod vectors have been responsible for significant morbidity and mortality throughout recorded history. Such vector-borne diseases – including malaria, dengue, yellow fever, and plague – together accounted for more human disease and death in the 17th through early 20th centuries than all other causes combined. Over the past three decades, previously controlled vector-borne diseases have resurged or reemerged in new geographic locations, and several newly identified pathogens and vectors have triggered disease outbreaks in plants and animals, including humans.

Domestic and international capabilities to detect, identify, and effectively respond to vector-borne diseases are limited. Few vaccines have been developed against vector-borne pathogens. At the same time, drug resistance has developed in vector-borne pathogens while their vectors are increasingly resistant to insecticide controls. Furthermore, the ranks of scientists trained to conduct research in key fields including medical entomology, vector ecology, and tropical medicine have dwindled, threatening prospects for addressing vector-borne diseases now and in the future.

In June 2007, as these circumstances became alarmingly apparent, the Forum on Microbial Threats hosted a workshop to explore the dynamic relationships among host, pathogen(s), vector(s), and ecosystems that characterize vector-borne diseases. Revisiting this topic in September 2014, the Forum organized a workshop to examine trends and patterns in the incidence and prevalence of vector-borne diseases in an increasingly interconnected and ecologically disturbed world, as well as recent developments to meet these dynamic threats. Participants examined the emergence and global movement of vector-borne diseases, research priorities for understanding their biology and ecology, and global preparedness for and progress toward their prevention, control, and mitigation. This report summarizes the presentations and discussions from the workshop.

READ FREE ONLINE

  1. ×

    Welcome to OpenBook!

    You're looking at OpenBook, NAP.edu's online reading room since 1999. Based on feedback from you, our users, we've made some improvements that make it easier than ever to read thousands of publications on our website.

    Do you want to take a quick tour of the OpenBook's features?

    No Thanks Take a Tour »
  2. ×

    Show this book's table of contents, where you can jump to any chapter by name.

    « Back Next »
  3. ×

    ...or use these buttons to go back to the previous chapter or skip to the next one.

    « Back Next »
  4. ×

    Jump up to the previous page or down to the next one. Also, you can type in a page number and press Enter to go directly to that page in the book.

    « Back Next »
  5. ×

    Switch between the Original Pages, where you can read the report as it appeared in print, and Text Pages for the web version, where you can highlight and search the text.

    « Back Next »
  6. ×

    To search the entire text of this book, type in your search term here and press Enter.

    « Back Next »
  7. ×

    Share a link to this book page on your preferred social network or via email.

    « Back Next »
  8. ×

    View our suggested citation for this chapter.

    « Back Next »
  9. ×

    Ready to take your reading offline? Click here to buy this book in print or download it as a free PDF, if available.

    « Back Next »
Stay Connected!