2,451
Views
3
CrossRef citations to date
0
Altmetric
Review Article

Perceptions of plant breeding methods–from ‘phenotypic selection’ to ‘genetic modification’ and ‘new breeding technologies’

ORCID Icon
Received 20 Jan 2023, Accepted 02 Mar 2023, Published online: 22 Mar 2023

ABSTRACT

Plant breeding has been closely aligned with the development of civilisations and continues to be important for the supply of nutritious food and a key factor in reducing poverty and hunger. Plant breeding uses a range of techniques for both expanding and exploiting the genetic potential of plants. However, some techniques are deemed higher risk than others despite the end products of both processes at times being indistinguishable. While it is considered that the domestication of some plant species began over 10,000 years ago, it is only in the last 100 years or so that modern plant breeding has been used to develop thousands of cultivars in a range of plant species for food, feed, and recreation. In the last 25 years, genetic modification and, more recently, New Breeding Technologies have been used to introduce new variations into important plant species. This has resulted in mistrust and suspicion, and a range of regulatory systems. Product-based and process-based regulatory systems differ in the information required for decision-making. Methods used for the development and manipulation of plant traits are reviewed in an attempt to understand the reasons why some are deemed more acceptable than others.

Introduction

Plants through their ability to capture sunlight energy and store it have been and will continue to be the foundation of food–whether consumed directly or indirectly. Plant breeding which has contributed towards higher yielding and more nutritious crops and forages is one of the factors underpinning the development of civilisations (Hallauer Citation2011; Breseghello and Coelho Citation2013) and has alleviated poverty through initially increasing food supply locally but more recently globally (FAO Citation2022). Plant breeding brings together both the skill of the breeder to recognise value across a range of traits and understanding through science to engineer plants for the benefit of humanity (Hallauer Citation2011; Poehlman Citation2013). It is the application of techniques for both expanding and exploiting the genetic potential of plants (Stoskopf et al. Citation2019; Bowerman et al. Citation2023) that has been the key to continued improvements in crops and forages. The aim here is to review methods used for both the development and manipulation of plant traits, investigate their effectiveness, and attempt to understand the reasons why some methods are deemed more acceptable than others.

The importance of plant breeding

The history of plant breeding

Plant breeding is considered to have begun over 10,000 years ago (Hallauer Citation2011) through the gradual domestication of wild plant species. For example, modern domesticated maize (Zea mays ssp, mays) is derived from teosinte (Z. mays ssp. parviglumis), a wild species native to Mexico and Central America (Matsuoka et al. Citation2002; van Heerwaarden et al. Citation2011). Teosinte seed, when ripe fall from the cob, which is only made up of two rows of ‘kernels’, and the seed coat is full of silica and lignin making it indigestible and therefore easily distributed in faecal matter (Hake and Ross-Ibarra Citation2015). In contrast, modern maize kernels are large, soft coated, easily digestible, and held tightly into cobs up to 20 or more rows.

Similarly, domesticated wheat (Triticum aestivum) originated in the Middle East (Iraq, Syria, Lebanon, Israel, and Palestine) as a result of two successive cycles of natural interspecific hybridisation (Salamini et al. Citation2002). It was spread by humanity both west into Europe and east into Asia where adaptation to local environments occurred resulting in the development of landraces (Balfourier et al. Citation2019). Landraces are defined as ‘a cultivated, genetically heterogeneous variety that has evolved in a certain ecogeographical area and is therefore adapted to the edaphic and climatic conditions and to its traditional management and uses’ (Casañas et al. Citation2017). Landraces differ from ecotypes in that landraces result from being cultivated by farmers and becoming domesticated (Zeven Citation1998; Ortiz Citation2020). The more recent genetic modification to wheat occurred 70 years ago with the introduction of dwarfing genes and genes for photoperiod insensitivity during the Green Revolution which improved the harvest index and reduced lodging (Trethowan et al. Citation2007).

The vegetable brassicas cauliflower, brussels sprouts, broccoli, kale, kohlrabi, and cabbage have all been domesticated over many generations from the same precursor germplasm now known as Brassica oleracea (European Commission Citation2021). Determining the origin and evolutionary history of B. oleracea has been controversial. However, a recent study using newly generated RNA-seq data (which accurately maps short sequence read to a reference genome allowing quantification of gene(s) expression levels) for a diversity panel of 224 accessions, which represents 14 different B. oleracea crop types, and nine potential wild progenitor species have concluded that the Aegean endemic B. cretica is the closest living relative of cultivated B. oleracea, supporting an origin of cultivation in the Eastern Mediterranean region (Mabry et al. Citation2021). This study also considered that cultivated plants of this species can revert to a wild-like state with relative ease. The different sub-groups of B. oleracea developed through mutation and introgression from wild species during evolution or human selection (Ramchiary et al. Citation2011). Genetic analysis of these sub-groups has shown they are closely related despite the large morphological differences (Sadowski and Kole Citation2016).

Modern plant breeding has however a short history of about 100 years, resulting largely from the connection of Mendel’s principles of inheritance (Laird and Lange Citation2011; Bateson and Mendel Citation2013; Stenseth et al. Citation2022), Darwin’s theory of natural selection (Ruse Citation1975; Ospovat Citation1995), Vilmorin’s demonstration that parental selection is more effective if based on progeny testing (Gayon and Zallen Citation1998), and the use of appropriate experimental design and statistical analysis (Fisher Citation1925; Provine Citation1971; Hallauer Citation2011). This has led to the development of thousands of varieties or cultivars in major food crops and forage plants and continues to do so. Even with the advent of modern biotechnologies for manipulating the genome and linking genes to traits of value (Moose and Mumm Citation2008; Bowerman et al. Citation2023), the role of the plant breeders remains central to the delivery of new and innovative cultivars.

Plant traits

Phenotype is the expression of both the genetics of the plant and the environment in which the plant is growing. Despite an increase in knowledge of genomics and methods of manipulating the genome, it is the resulting phenotype that is important in delivering the value of the plant breeding or genetic improvement method used (Lee Citation2006). Phenotypic plasticity is defined as the degree to which a trait can change with changes in environment and management is itself under genetic control (West-Eberhard Citation1989). Trait expression can be controlled by either single or multiple genes and these themselves may interact with environmental variables. Single or major genes will follow the principles of Mendel in the way they segregate and express in progeny, while complex traits (such as yield or persistence) which are under multigene control are better understood by determining the breeding value of parental genotypes based on the performance of progeny (Falconer Citation1960).

Plant traits identified by breeders as important can be many and varied, but success in selecting them depends on their heritability (Nyquist and Baker Citation1991) which is a measure of the extent to which they can be manipulated from a genetic perspective. Two broad categories of heritability are often distinguished. Broad sense heritability is the ratio of total genetic variance (the sum of additive genetic variance, dominance genetic variance, and epistatic genetic variance) to phenotypic variance; while narrow sense heritability is the ratio of additive genetic variance (which is determined by gene frequency and by the average effect of substituting one allele for another) to phenotypic variance (Dudley and Moll Citation1969).

Methods for developing and manipulating plant traits

The ultimate success of plant breeding is reliant on both the ability of the breeder and the extent of variation available for the traits being sought or selected. Increasing trait variability and then capturing the desired trait expression is achieved through a number of methods, some of which can be regulated.

Phenotypic selection

The ability to accurately identify, define and measure important plant traits is the crucial factor underlying a successful plant breeding programme. This requires access to a wide range of germplasm within the species of interest, which might include wild relatives and landraces, the relevant environment in which to grow the plant material and the perceptive eye of the breeder.

Selection of the correct phenotype is the basic activity of plant breeding closely followed by using the appropriately controlled mating of plants that have been selected with desirable trait expression. The power of phenotypic selection has led to the development of modern crops and forages. Plant breeding methods have been extensively reviewed previously (Allard Citation1960; Mayo Citation1987; Poehlman Citation2013; Acquaah Citation2015; Stoskopf et al. Citation2019).

Interspecific hybridisation

While numerous books and articles have been written to examine and defend a variety of definitions of species (George and Mayden Citation2005; Zachos Citation2016), here species, as the primary natural taxonomic unit, is defined by being genetically isolated, able to interbreed within the species but unlikely to interbreed with another species (Wilkins Citation2007). However, using embryo rescue interspecific hybrids can be produced. Embryo rescue is an in vitro technique used when crossing between plants results in embryo abortion or degeneration (Pramanik et al. Citation2021). In plants, this along with polyploidy has occurred most frequently in ornamentals such as Rosa, Chrysanthemum, Gladiolus, Alstroemeria, Lilium, and orchids (Van Tuyl and Lim Citation2003). Plant biologists have long considered hybridisation as an important force in adaptation to environmental effects and to further speciation (Grant Citation1972; Bowley and Taylor Citation1987; Rieseberg et al. Citation2004). In crop and forage plants successful artificially-made interspecific hybrids include Triticale–a hybrid between wheat and rye (Secale cereale) (Randhawa et al. Citation2015; Losert et al. Citation2017); raphanobrassica–a hybrid between kale (Brassica oleracea) and radish (Raphanus raphanistrum subsp. sativus) (Dumbleton et al. Citation2022) (). In horticulture, interspecific hybrids have been widely used in tomato (Solanum lycopersicum) (Vanlay et al. Citation2022), sugarcane (Saccharum) (Irvine Citation1999), cucumber (Cucumis) (Tak et al. Citation2016), Prunus species (Layne and Sherman Citation1986; Szymajda et al. Citation2015), apple (Malus) (Korban Citation1986; Gross et al. Citation2012), chestnut (Castanea) (Pereira-Lorenzo et al. Citation2016), papaya (Carica species) (Drew et al. Citation1998), and banana (Musa species) (De Langhe et al. Citation2010), (). For some horticultural species that are propagated through grafting, such as apple (Malus x domestica), the use of rootstocks from wild relatives has been used to improve drought tolerance (Liu et al. Citation2012). Some rootstocks are themselves interspecific hybrids (Niu et al. Citation2019).

Table 1. Artificially-made interspecific hybrids used in agriculture and horticulture.

Polyploidy

Genome doubling or polyploidy can occur in two ways –allopolyploidy, resulting from hybridisation between two different species, or autopolyploidy, resulting from the duplication of a single genome. Greater new genetic diversity and hybrid vigour is most likely to result from allopolyploidy (Bell et al. Citation2013). Many crop species are natural polyploids (Mason and Batley Citation2015)–including potato (Solanum), sweet potato (Ipomoea), cassava (Manihot), taro (Colocassia), yams (Dioscoria), wheat (Triticum), sugarcane (Saccharum), cotton (Gossypium), canola (Brassica), strawberry (Fragaria), and kiwifruit (Actinidia) (Sattler et al. Citation2016).

Polyploidy has also been generated in some plant species to improve yield and nutritive quality (Sattler et al. Citation2016). Polyploids can be induced by sexual polyploidisation using a range of chemicals including colchicine, nitrous oxide, oryzalin, caffeine, trifluraline, puromycin, and benzobenil (Younis et al. Citation2014), resulting in the union of unreduced male and female gametes that have not undergone normal meiosis and still have a 2n constitution (Ramanna and Jacobsen Citation2003). The anti-mitotic agent colchicine can also be used to disrupt mitosis resulting in chromosome doubling in meristematic cells giving rise to a polyploid shoot (Planchais et al. Citation2000; Nair Citation2004; Ranney Citation2006). While not all polyploids deliver advantages, the opportunities they might provide are overcoming barriers to hybridisation, development of sterile cultivars, restoration of fertility in wide hybrids, improved pest resistance and stress tolerance, and enhanced vigour (Ranney Citation2006).

Doubling the chromosome number to create tetraploids has been used in forages such as perennial ryegrass (Lolium perenne) primarily to increase nutritive value and digestibility (Balocchi and López Citation2009). Further, it has been demonstrated that tetraploids of ryegrass are more likely to have higher yields under severe drought conditions and cold than diploids (Kemesyte et al. Citation2017; Lee et al. Citation2019a), but they are potentially more susceptible to damage from root feeding invertebrates (Tozer et al. Citation2017).

Heterotic breeding systems

Heterosis or hybrid vigour can be created by crossing genetically diverse and often inbred populations (Frankel Citation1983; Shen et al. Citation2006; Stupar et al. Citation2008). Mendel observed heterosis in his pea populations, as did early horticulturists in the eighteenth century (Ashby Citation1937), but the first designed experiments to understand this phenomenon were carried out by Darwin and documented in ‘The effects of cross- and self-fertilization in the vegetable kingdom’ published in 1876 (Mather Citation1955), Hybrid vigour has been effectively used in crop and vegetable species resulting in superior performance relative to the parental lines (Fujimoto et al. Citation2018). Production of hybrid seed relies on the prevention of self-fertilisation which can be achieved using a male-sterile female parent. Male sterility can be induced by cytoplasmic male sterility (CMS), nuclear-controlled environment-sensitive genic male sterility (EGMS), or genic male sterility (Chen and Liu Citation2014; Kim and Zhang Citation2018). CMS is caused by mitochondrial genes together with nuclear genes, while genic male sterility is caused by nuclear genes alone, and EGMS depends on environmental condition such as photoperiod or temperature. CMS-induced male-sterile female lines can only be maintained by crossing with a maintainer male fertile line (due to mitochondrial DNA only being maternally inherited) while EGMS-induced male-sterile female lines can be self-propagated without a maintainer line. CMS resulting from either spontaneous or artificial mutations has been used in maize, rice (Oryza sativa), sunflower (Helianthus annuus), a range of brassica species (Yamagishi and Bhat Citation2014), sorghum (Sorghum bicolor), wheat, common bean (Proteus vulgaris), pepper (Capsicum annuum), carrot (Daucus carota), and sugar beet (Beta vulgaris) (Chen and Liu Citation2014). In species such as maize with a natural separation of male and female reproductive organs, removing the male structure through detasseling is another method of creating hybrids. However, it is a laborious method and the use of cytoplasmic male sterility which is maternally inherited is preferred (Laughnan and Gabay-Laughnan Citation1983; Levings Citation1993). Heterosis can result in significant yield increases over open-pollinated cultivars of up to 120% in oilseed brassica crops (Brandle and McVetty Citation1989), 113% in maize (Zanoni and Dudley Citation1989), 125% for 10-ear weight in sweet corn (Dickert and Tracy Citation2002), 134% in rice (Virmani et al. Citation1982), and 130% in wheat (Duvick Citation1999). Prior to the development of hybrid maize in the 1930s, open-pollinated cultivars were yielding about 2 tons per ha of grain in the USA while hybrid cultivars in 2018 were yielding close to 12 tons per ha of grain (Hochholdinger and Baldauf Citation2018).

Chemical and radiation-induced mutagenesis

Mutations are heritable changes to the genetic material of an organism (Mba et al. Citation2010; Mba Citation2013). Spontaneous mutations are the primary source of genetic variation in organisms (Kharkwal Citation2012). Mutagenesis can be induced to increase trait variability by exposing the seed to certain chemicals, x-rays, or radiation (Kodym and Afza Citation2003; Mba Citation2013; Oladosu et al. Citation2016; Ahmar et al. Citation2020). Mutations either spontaneous or induced are random events, and it is the breeders’ ability to recognise useful and beneficial variations that remains the key to a successful outcome. The International Atomic Energy Agency (IAEA) Mutant Variety Database (IAEA Citation2022) is a repository of voluntarily contributed information on officially released mutant crop varieties of both seed and vegetative crops. This database indicates the largest number of cultivars released that involved induced mutation breeding was rice (with 873 cultivars), followed by barley (Hordeum vulgare) (307), chrysanthemum (285), wheat (265), soybean (Glycine max) (182), maize (89), and horticultural fruit species (81). The highest number of mutation-induced cultivars are used in Asia (with 2087 cultivars), followed by Europe (960), North America (211), Africa (82), Latin America (53), and Australia/Pacific (9).

Reverse genetics and breeding

Reverse genetic approaches identify gene function through three methods (Kumar et al. Citation2022): (a) phenotypic changes due to knocking out/altering/silencing the target gene(s) using molecular techniques such as RNAi (interferes with the translation of target mRNA transcript eventually suppressing the gene expression (Saurabh et al. Citation2014)), virus induced silencing of gene (VIGs) (Senthil-Kumar and Mysore Citation2011), and homologous recombination which creates covalent linkages between DNA in regions of highly similar or identical sequence (Schuermann et al. Citation2005), (b) through expression analysis when a target candidate gene is inserted into the species of interest, and (c) screening the target gene(s) in the mutant populations developed by random disruption of genes through mutagens and then determining the phenotypic result (Jankowicz-Cieslak and Till Citation2015). The latter uses a technique called Targeting Induced Local Lesions IN Genomes (TILLING) (McCallum et al. Citation2000). TILLING does not involve transgenic modifications but uses gene-specific primers to amplify DNA and then specific nucleases to identify mismatches between wild-type and mutant DNA (Henikoff et al. Citation2004). This process has the potential for removing the need to screen large putative mutant populations for the desired phenotype and has been used in a large number of crops, including rice, maize, wheat, sugar beet, barley, soybean, pea (Pisum sativum), bean (Phaseolus vulgaris), tomato (Solanum lycopersicum), and the vegetatively propagated banana (Musa spp.) (McCallum et al. Citation2000).

Single seed descent

Used in self-fertilising crops single seed descent is used to identify phenotypic traits or characteristics that exceed those of the parents for economically important traits such as seed yield (Chahota et al. Citation2007) and fix these traits for expression in succeeding generations (Lenaerts et al. Citation2019) through advancing lines to homozygosity. It has the advantages over the normal pedigree breeding method of requiring less time and labour, and transgressive segregation (i.e. producing individuals in subsequent generations that are superior to both the parents) can be fixed and genetic advance obtained (Snape and Riggs Citation1975; Chahota et al. Citation2007; Jumbo et al. Citation2011). Single seed descent genotypes have been effectively used to study the genetic variation within a species, as demonstrated in a large collection of Durum wheat (Triticum durum) (Pignone et al. Citation2015).

Doubled haploidy

Haploids are sporophytes that have the gametic chromosome number (n). Double haploids can be spontaneous at low frequency but can also be induced to generate homozygous lines without the need for using several generations of selfing (Germanà Citation2011) and then use these for obtaining hybrid cultivars and/or provide access to recessive genes and for biotechnological manipulations (Lübberstedt and Frei Citation2012; Prigge and Melchinger Citation2012). Anther culture is often the preferred method for double haploid production in many crops (Sopory and Munshi Citation1996). It is only achievable in species able to be tissue cultured, but this does include rice for which this breeding method has led to the release of many cultivars (Grewal et al. Citation2011; Mishra and Rao Citation2016).

Apomixis

Apomictic plants produce seeds with embryos derived directly from maternal tissue, not through sexual fertilisation. It has been proposed that apomixis could be used to develop self-reproducing hybrids (Bashaw Citation1980; Duvick Citation1999). Apomixis has been observed in over 300 species (Hann and Bashaw Citation1987). Using apomixis to develop new hybrids would bypass multi-generation selfing for inbred development and eliminate the need for large-scale and expensive cross-pollination blocks to produce hybrid seed (Duvick Citation1999; Hanna and Bashaw Citation1987). Several apomictic mechanisms have been shown to be genetically controlled however, it would appear that to date this development has only been used commercially in the breeding of tropical grasses, such as Brachiaria (Miles Citation2007).

Selection of genotypes with desired traits using genetic and molecular markers

The advantages and disadvantages of different genetic markers used in identifying desired genotypes for further breeding have been recently summarised by Nadeem et al. (Citation2018).

Marker assisted selection

Molecular DNA markers can increase the efficiency of breeding programmes. This has been used effectively for manipulating simple traits but has been slower for complex traits, such as salt tolerance (Ashraf and Foolad Citation2013). DNA markers that have been used in Marker Assisted Selection include Restriction Fragment Length Polymorphism (RFLP), Random Amplified Polymorphic DNA (RAPD), Amplified Fragment Length Polymorphism (AFLP), Simple Sequence Repeats (SSRs) or microsatellites, and Single Nucleotide Polymorphisms (SNPs) (Collard et al. Citation2005; Jiang Citation2013; Nadeem et al. Citation2018; Lamichhane and Thapa Citation2022). Marker Assisted Selection has played a prominent role in cereal crop breeding for resistance to obligate biotrophic pathogens such as leaf rust in wheat (Vida et al. Citation2009), However, while used in the commercial breeding of several crops, including wheat, rice, barley, and maize (Hasan et al. Citation2021), in general, it has not been fully integrated into all conventional plant breeding programmes (Young Citation1999; Gupta et al. Citation2010). Reasons for this low impact of Marker Assisted Selection include reliability and accuracy of quantitative trait loci mapping, association between marker and trait is poorly correlated, impacts of genetic background used to identify markers and a lack of transferability to other germplasm, and environmental interactions (Collard and Mackill Citation2008).

Genomic selection

Genomic selection (Lenaerts et al. Citation2019) is based on making genomic predictions from very large numbers of DNA markers rather than focusing on specific genes or quantitative trait loci defined as the section of DNA that correlates with a variation of a quantitative trait in the phenotype of a population (Desta and Ortiz Citation2014; Heslot et al. Citation2015). Molecular tools are therefore used to identify superior genotypes and track trait variation. Using a large amount of marker information genomic selection can be used to calculate genomic estimated breeding values (GEBVs) for complex traits that older Marker Assisted Selection systems were ineffective in achieving and were therefore less efficient in accelerating genetic gain (Heffner et al. Citation2010; Merrick et al. Citation2022a and Citation2022b; Sandhu et al. Citation2022a). Genomic tools have also enabled a better understanding of the complex interactions between plants and pathogens at a molecular level to better identify host defences in resistant and susceptible interactions to provide consideration for the introgression of these traits into breeding programmes (Kankanala et al. Citation2019). Genomic selection has been heralded as a means to accelerating the efficiency of molecular selection for complex traits associated with climate-resilient crops (Budhlakoti et al. Citation2022), seed yield (Hu et al. Citation2022), growth and wood property traits of forest trees (Grattapaglia Citation2022), and in plants such as sugarcane with the large complex genome, high levels of polyploidy and heterozygosity (Sandhu et al. Citation2022b). A recent review concluded that plant breeders can improve genetic gains through the use of genome selection by utilising multi-trait and, multi-environment models, high-throughput phenotyping, and machine learning (Merrick et al. Citation2022a). Another recent review likens genomic selection to ‘instead of trying to find the needle in a haystack, i.e. the “magic” gene in the complex and fluid genome, genomic selection more efficiently and humbly ‘buys the whole haystack’ of genomic effects to predict complex phenotypes’ (Grattapaglia Citation2022).

Genotyping-by-sequencing

Genotyping-by-sequencing has enabled the development of genome-wide markers in non-model polyploid plants (Baral et al. Citation2020). Sequence-based markers that have been used are Single-Nucleotide Polymorphism (SNP) and Diversity Arrays Technology (DArT) markers (Nadeem et al. Citation2018). The ability to use markers for traits of interest is enhanced through using genome-wide SNP markers which provide more accurate selection of parental genotypes (Meuwissen et al. Citation2001; Heffner et al. Citation2009; Crossa et al. Citation2011, Citation2017).

Genetic engineering and genetic modification (GM)

Genetic engineering and modification are here defined as the manipulation of an organism’s genes by introducing, eliminating or rearranging specific genes using the methods of molecular biology (USDA Citation2022). This process can allow the introduction of new traits and variation otherwise not available in the unmodified genome, such that ‘genetic material has been altered in a way that does not occur naturally by mating and/or natural recombination’ (EU Directive Citation2001). This broad all-encompassing definition from the European Union has almost certainly influenced decisions in parts of the world where the options are fewer and less attractive to alleviate human suffering wherever possible (Herring Citation2008). The benefits, risks, and intended and unintended consequences of this technology have been recently reviewed (Caradus Citation2022a) along with opportunities they present for the future (Bowerman et al. Citation2023). There is a range of methods used to modify plant genomes some or all of which are regulated depending on the country. In New Zealand, all are currently regulated under the Hazardous Substances and New Organisms (HSNO) Act 1996 administered by the Ministry for the Environment (New Zealand Legislation Citation1996) (Caradus Citation2022b). Adapting regulatory frameworks to accommodate rapidly changing genome editing technologies to balance risk against benefits is a challenge which some jurisdictions are adjusting to better than others (Rozas et al. Citation2022).

Transgenesis

Transgenesis involves using recombinant DNA technologies to introduce genes from other organisms to another. It may also involve genes artificially synthesised in the laboratory(Herrera-Estrella et al. Citation2004). This methodology has been extensively used over the last 25 years to deliver 525 different transgenic events, in 32 crops and flower species across 26 countries (Caradus Citation2022a). Transgenesis has predominantly used either an Agrobacterium-mediated process (Gelvin Citation2000), or biolistics (Sanford Citation1990) to introduce the transgene(s), along with a selectable marker (for ease of differentiating between genotypes with the transgene and those without) and a promoter. Less frequently used methods include the use of electroporation, viral vectors, polymers, and nanoparticles (Cunningham et al. Citation2018; Ahmar et al. Citation2021). Selectable markers might be either herbicide resistant gene, antibiotic resistant gene or inducible growth, or differentiation of transformed tissues (Miki and McHigh Citation2004). The promoters used might be one that is constitutive (drives expression of a gene in all plant tissues and throughout all developmental stages), tissue-specific (expresses the gene in only a specific type of tissue), and inducible (initiates gene expression under certain external conditions) (Liu Citation2009; Smirnova et al. Citation2012).

Using plant breeding to combine stacked genes into a single breeding line can be unwieldy with the population size needed to isolate a stacked F2 plant increasing exponentially with the increasing number of unlinked genes (Pathak and Srivastava Citation2020). Repeated recombinase-mediated DNA cassette exchanges linking the introduced genes and stacking them into a locus improve efficiency (Petolino and Kumar Citation2016; Que et al. Citation2010). For example, multi-gene integration in crops for agronomically relevant traits has been achieved in soybean were genes fatty acid ω−6 desaturase 2 and acyl-acyl carrier protein thioesterase 2 were silenced to improve oleic acid content (Li et al. Citation2010). Similarly, site-specific integration of a five-gene cassette has been demonstrated successfully in rice (Pathak and Srivastava Citation2020).

Cisgenesis and intragenesis

Cisgenesis is defined as transferring a gene from the same or a closely related species (Holme et al. Citation2013; Freddy et al. Citation2022; Hefferon Citation2022; Koul Citation2022) along with its own regulatory promoter and terminator (Kumar et al. Citation2020). This can lead to a change in the expression pattern of the desired gene driven by regulatory elements from other genes (Kumar et al. Citation2020). Similarly, intragenesis involves the construction of the entire vector region destined for transfer to a species from the genome of that species (Rommens Citation2004; Rommens et al. Citation2004; Conner et al. Citation2007; Rommens et al. Citation2007; Barrell et al. Citation2010; Barman et al. Citation2020). For well over a decade, cisgenesis and intragenesis have promised a ‘clean’ DNA delivery system for gene transfer. There is no published evidence of cisgenic or intragenic events having ever been commercialised and their promise remains to be fulfilled.

Targeted gene editing

Gene editing or sequence-specific nuclease technology (Schaart et al. Citation2016; Mohanta et al. Citation2017; Songstad et al. Citation2017) differs from previous techniques in manipulating the plant genome by being more precise and more versatile, and relatively rapid to undertake. Gene editing methods rely on specific DNA binding proteins which are found in all cells to ensure that genetic material is appropriately expressed, replicated, and transmitted from one generation to the next. These DNA binding sites are short DNA segments referred to as motifs or signatures that can be used to predict protein function (Das and Dai Citation2007). Early approaches to introduce site-specific modifications to plant genomes in an attempt to control gene expression used oligonucleotides, small molecules (Gottesfeld et al. Citation1997), or self-splicing (Yang et al. Citation1996). However, more recent technologies using principles of DNA–protein recognition resulted in the development of site-directed zinc finger nucleases (ZFNs) and TAL effector nucleases (TALENs) and then most recently clustered regularly interspaced palindromic repeats (CRISPR) and their associated (Cas) nucleases (Doudna and Charpentier Citation2014).

Oligonucleotide-directed mutagenesis (ODM) is a precise genome editing technology that uses oligonucleotides (short single strands of synthetic DNA or RNA that serve as the starting point for molecular biology applications) to make targeted edits in plasmid, episomal, and chromosomal DNA (Oh and May Citation2001; Dong et al. Citation2006; Gocal Citation2015; Gocal et al. Citation2015; Sauer et al. Citation2016). While the use of oligonucleotides to precisely direct, mutagenesis in preselected sequences has been demonstrated to be effective it has not been widely deployed in crops (Cardi and Neal Stewart Citation2016).

Chimeric zinc finger nucleases allow direct targeting of chromosome sites for gene insertion (Bibikova et al. Citation2003) by exploiting the natural recognition mechanism of cellular DNA repair machinery, to make sequence-specific double-stranded DNA breaks at a target locus (Townsend et al. Citation2009; Shukla et al. Citation2009; Kumar et al. Citation2020).

Transcription activator-like (TAL) effector proteins (TALENS) are an effective technology for genome editing that use targetable nucleases to induce double-strand breaks into specific DNA sites, which are then repaired by mechanisms that can be exploited to create sequence alterations at the cleavage site (Joung and Sander Citation2013). Transcription activator-like effectors (TALEs) are major virulence factors secreted by the plant pathogenic bacterial genus Xanthomonas, which causes disease in plants such as rice and cotton. TALEs are injected into host cells and interfere with cellular activities by activating the transcription of specific target genes (Bogdanove et al. Citation2010; Kumar et al. Citation2020). TALENS has been most effectively used to knock out genes expressing unwanted traits (Bezie et al. Citation2021).

Clustered regularly interspaced palindromic repeats (CRISPR) and their associated (Cas) nucleases allow a process of rapidly and efficiently targeting protein domains in areas of interest in a genome (Zhang et al. Citation2014; Ma et al. Citation2015; Doudna and Sternberg Citation2017; Eş et al. Citation2019). The CRISPR/CasX (i.e. Cas9 and Cas12 and its variants) system consists of two essential components–Cas nucleases which allow a process of rapidly and efficiently targeting areas of interest in a genome, and a single-guide RNA which targets the section of DNA to be edited (Hsu et al. Citation2014; Lee et al. Citation2019b). An extensive review has shown that CRISPR/Cas9 technology has been effectively used by researchers to bring efficient targeted transformation to a range of crop species (Bezie et al. Citation2021). Gene editing using CRISPR deliver three types of outcomes. CRISPR gene editing uses Site-Directed Nucleases (SDN) which uses different DNA-cutting enzymes (nucleases) that are directed to cut the DNA at a predetermined location by a range of different DNA binding systems (Hsu et al. Citation2014; Lee et al. Citation2019b). After the double strand, the cut is made, the cell’s own DNA repair mechanism recognises the break and repairs the damage, using one of two pathways that are naturally present in cells (1) non-homologous end-joining (NHEJ): The cut DNA is re-joined, but while doing this errors are made resulting in random small deletions (up to 20) or additions (a few base pairs) of nucleotides at the cut site or (2) homology-directed repair (HDR): a donor DNA that carries the desired change and has homology with the target site is used to introduce this change at the cut site (Bibikova et al. Citation2002; Cong et al. Citation2013). This leads to one of three outcomes–gene disruption or deletion (SDN1), gene correction or modification (SDN2), or DNA insertion (SDN3) (Doudna and Charpentier Citation2014; FAO Citation2022).

The European Food Safety Authority has recently declared that the documents for ‘Guidance for risk assessment of food and feed from genetically modified plants’ and the ‘Guidance on the environmental risk assessment of genetically modified plants’ are ‘not relevant for the risk assessment of plants developed via SDN-1, SDN-2 or oligonucleotide-directed mutagenesis approaches if the genome of the final product does not contain exogenous DNA’ (Naegeli et al. Citation2020). In reality, SDN-1 and SDN-2 type gene edits are cisgenic in nature (Cabrera-Ponce et al. Citation2022; Ghose et al. Citation2022), and SDN-1 type knockout gene edits have been confusingly referred to as null lines (Kim et al. Citation2023).

Alternatives to the commonly used Cas9 single-component effector protein have been identified and include Cas12/Cpf1 which is a single RNA-guided endonuclease which cleaves DNA via a staggered DNA double-stranded break motif (Zetsche et al. Citation2015; Yin et al. Citation2017; Bandyopadhyay et al. Citation2020; Kumar et al. Citation2020). Different Cas effector proteins can result in a range of on-target mutations in plants, ranging from 90 to 100% for Cas9 and 0%–60% for Cas12a (Lee et al. Citation2019b). Others have used CRISPR/Cas9 ribonucleoprotein complexes to achieve consistently no off-target mutations (Liang et al. Citation2017). Compared with other forms of mutagenesis (e.g. chemical mutagenesis using ethyl methanesulfonate or X-ray radiation, etc.) off-target effects of CRISPR-Cas9 are trivial. Even with low levels of off-target mutations, sequence analysis can be used to identify genotypes with off-target events to ensure they are not selected.

A comparison of the four main genome editing methods oligonucleotide-directed mutagenesis, zinc finger, TALEN, and CRISPR is provided by Gao et al. (Citation2020). They conclude that compared with CRISPR, both zinc finger and TALEN-mediated gene editing techniques have some cumbersome protein engineering steps, high costs, and are difficult to multiplex.

RNA-directed DNA methylation and epigenesis

RNA-directed DNA methylation (i.e. where a methyl (CH3) group molecule gets added to DNA) induces transcriptional gene silencing via methylation of the target gene promoter sequence by RNA interference (RNAi) (Schaart et al. Citation2016), is unique to plants (Erdmann and Picard Citation2020). Epigenetic engineering technologies make it experimentally possible to modify individual chromatin marks (sites where DNA methylation, small interfering RNA, or histone variants are associated with an epigenetic event) at specific user-defined sites (Margueron and Reinberg Citation2010; Köferle et al. Citation2015). Epigenetic processes change the activity of genes without changing a DNA sequence, but still leads to modifications that can be transmitted to daughter cells (although some epigenetic changes can be reversed) (Weinhold Citation2006). Epigenetic processes can occur through DNA methylation, acetylation, phosphorylation, ubiquitylation, and sumolyation. Epigenetic changes occur naturally through environmental impacts, nutrition, and ageing (Huidobro et al. Citation2013). Initially, understanding the epigenetic regulation of gene expression in plants was undertaken in the lab-species Arabidopsis. Evidence would suggest heritable epiallelic variations associated with a trait of interest could be utilised for crop improvement, particularly in combination with targeted gene editing (Puchta Citation2016; Adli Citation2018; Kumar Citation2019). This includes the understanding of epigenetic regulation in crop plants in response to both biotic (Huang and Jin Citation2022) and abiotic stresses (Perrella et al. Citation2022; Singh and Prasad Citation2022; Verma et al. Citation2022).

Grafting with GM rootstocks

Grafting onto GM rootstock occurs when the top of a non-GM plant (scion) is grafted on a GM rootstock such that the scion may benefit from traits conferred by transgenes in the rootstock, but its products do not contain the transgene itself. (Schaart et al. Citation2016; Hu and Gao Citation2023). Using transgenic rootstock to protect the grafted non-transgenic scion of grapevine from Xylella fastidiosa (which causes Pierce’s disease) is an example of this technology (Dandekar et al. Citation2019). In this case, a plant polygalacturonase inhibitory protein (PGIP) that prevents Pierce’s disease by inhibiting the breakdown of pectin present in primary cell walls is expressed in the transgenic rootstock transiting into the xylem of the grafted scion.

Agroinfiltration

Agroinfiltration is a technique using Agrobacterium transformation as a tool to achieve temporary and local expression of genes in plant tissue. Agroinfiltration is applied for testing the reaction of target plants to transgenic proteins, or for functional gene analysis in plants (Schaart et al. Citation2016).

Synthetic biology

Synthetic biology in plants refers to the introduction of new synthetic genomic pathways that deliver natural products and is expected to provide significant opportunities in agriculture (Zhu et al. Citation2021). ‘All definitions encompass the notion that applications of synthetic biology involve the creation of novel living systems through synthesising and assembling artificial and/or natural components’ (Jin et al. Citation2019). In terms of crop improvement outcomes, all have been at laboratory scale with most commercial-scale applications occurring in microbes that can be fermented (Wehrs et al. Citation2019; Yilmaz et al. Citation2022; Kwan et al. Citation2023).

Null segregants

Null segregants are non-GM progeny derived from crossing a GM plant with a wild-type non-GM plant. The value of null segregants is primarily with horticultural plants where it can take several years for plants to reach maturity and the introduction of new traits by crossing plants over multiple generations can take many years. By using genetically modifying germplasm with a shortened flowering time in the breeding programme (Flachowsky et al. Citation2009), so that they mature more rapidly (e.g. mature in 1 year instead of 4 years), means that new desirable characteristic(s) can be incorporated into elite commercial cultivars in significantly less time. The genetic modification is simply used to speed up the breeding programme and the new trait is the result of a non-GM change in the genome. To produce the commercial cultivar the line with the now-stably incorporated new trait is crossed one or two times with a non-GM line. The genetic modification for speed breeding is therefore selected out and the new cultivar now has the desired trait(s), matures at the normal rate, and is not genetically modified (Royal Society Te Apārangi Citation2019).

Advantages of new breeding technologies (NBT)

Some of the methods used to provide increased variation and opportunities for trait selection have been termed New Breeding Technologies (NBT) or New Genomic Techniques–namely, genome or gene editing to modify DNA at one or more specific sites using CRISPR, Zinc Finger Nucleases, or TALENs; introducing targeted changes to a small number of bases of DNA using oligonucleotide-directed mutagenesis; cisgenesis; intragenesis (inserting a reorganised regulatory coding region of a gene from the same species); and using epigenetic processes to change the activity of genes without changing a DNA sequence (Parisi and Rodríguez-Cerezo Citation2021).

Many have stressed that differences between older transgenic technologies and New Breeding Technologies challenge the appropriateness of many existing regulatory systems. The benefits of New Breeding Technologies compared with transgenesis are described in .

Table 2. Comparison of benefits and challenges of New Breeding Technologies compared with transgenesis

Precisely targeted gene editing is becoming an increasingly important and dominating method which is being promoted as ‘an advance in technology that has the potential to transform some aspects of the world’s agrifood systems for the better’ (FAO Citation2022). The potential influence and power of genome editing have led to claims that science is now ‘not only able to “read” the “Book of Life”, but also to “write” it and “edit” it’ (European Commission Citation2021). This has led to the question of ethics and how metaphors used to describe new technologies can influence public understanding (O'Keefe et al. Citation2015).

As a market, genome editing across pharmacology, biotechnology, and academia is estimated to be worth US$5.1 billion and with a CAGR of 18.2% expected to grow to US$11.7 billion by 2021 (Markets and Markets Citation2022). Other estimates vary between $7.4 billion by 2031 (Allied Market Research Citation2022) and $18.5 billion by 2028 (Biospace Citation2023). This is being driven by growth in funding by governments, increased application areas, and the introduction of CRISPR CasX. The potential benefits of integrating gene editing into plant breeding programmes using targets and traits with application in New Zealand has been reviewed and concludes that New Zealand is torn between the cautious ‘wait-and-see-approach’ with regard to the regulation of gene editing and implementing innovative solutions which are essential for the industry to maintain its global competitiveness (Fritsche et al. Citation2018).

Research with New Breeding Technologies has been used in the most part to demonstrate proof of concept. Most of these have been aimed at improved disease resistance, improved crop quality, flowering traits, and herbicide resistance. However, several finished cultivars have been submitted for de-regulation and a few have been commercialised (). There is also the promise of other traits of value that New Breeding Technologies could provide (Jones et al. Citation2022), including abiotic stress tolerances (Zafar et al. Citation2020); environmental stress resilience (Kouhen et al. Citation2022); and improved yield (Gao et al. Citation2020). An extensive summary of the use of gene editing by plant species, traits, techniques, and applications is available through an interactive internet database (eusage Citation2023).

Table 3. Examples of cultivars or germplasm developed using different methods of genetic engineering and genetic modification–those in bold have been submitted for de-regulation or commercialised. Refer also to Penna and Jain Citation2023) for a summary of successful examples of genome editing in fruit crops.

Perceptions–why are some methods causing concerns?

Of the wide range of methods used for creating and manipulating genetic variation of plants only those defined as genetic engineering, which in some countries includes New Breeding Technologies, are likely to be regulated (). The extent of this regulation does vary from country to country, and currently there is no internationally recognised or agreed standard for what should and should not be regulated. It has been argued that ‘current uncertain and complex global regulatory situation is stifling innovation and the realisation of the full potential of these technologies’ (Tagliabue Citation2016; Atanassova and Keiper Citation2018). Many countries, including New Zealand, regulate plant breeding outcomes based on the process used to produce them while others regulate the risk of the product with less concern being given to the process used to produce them (). Reasons for regulation are sound in that they are ensuring that products produced result in low levels of risk to human health and the environment. However, achieving this through regulating the process makes no logical sense when it is the end product that is released of which the process used to produce it is only the means to an end. Indeed the process-based regulatory systems are an overly precautionary approach which has the unintended consequence of stigmatising the use of some biotechnology methods (Tait Citation2001; Marchant and Stevens Citation2015; Aerni Citation2019; Smyth and Lassoued Citation2019; Anyshchenko and Yarnold Citation2021; Caradus Citation2022). Process-based regulatory systems also suffer from becoming increasingly obsolete as the scope of risks associated with processes become better understood (Atanassova and Keiper Citation2018). However, changing regulatory laws can be slow and contentious with polarised views reducing arguments to differences of opinion rather than pragmatically seeking a fact-based solution.

Table 4. Review of some country differences in regulation of genetic engineering techniques and in particular New Breeding Technologies (NBTs) (also refer to Atanassova and Keiper Citation2018; Menz et al. Citation2020; and Turnbull et al. Citation2021; Jones et al. Citation2022).

The driving forces behind these concerns about the processes used to deliver new and unique variations are general concerns by some consumers resulting from suspicions of corporate multinationals, that GM crops are deemed to be un-natural and ‘playing with nature’, the so-called ‘precautionary approach’, absence of economic or environmental benefit, a significant level of misinformation, controversy surrounding negative results, and distrust of science.

Consumer concerns

Consumer acceptance is crucial for food products to be successfully marketed and recognised (MacFie Citation2007; Krishna and Qaim Citation2008; Maghari and Ardekani Citation2011). Concerns include impacts on human health, the environment through the creation of super weeds, and adverse effects of beneficial species and biodiversity (Gaharwar et al. Citation2021). Consumer attitudes are influenced by the perception of risks and benefits, knowledge and trust, and personal values (Lucht Citation2015). Some anxiety about the use of GM crops is driven by scare stories and pseudo-science provided by anti-GM opinion (McHughen Citation2013). The New Zealand economy is heavily reliant on export income from primary produce where the countrywide market perspective is to portray and clean green image (Tourism NZ Citation2009). Using GM crops and forages has been construed as the antithesis of a ‘clean green image’ (Edwards Citation2017). A recent review on consumer attitudes towards the use of GM crops and forages in New Zealand concluded that

while there will always be a proportion of consumers against the use of GM in food production, the published evidence would suggest that the use of GM plants in New Zealand for food production will have no long-term deleterious effects in overseas markets. (Caradus Citation2022b)

Indeed the consequences of GM crops grown for food and feed over the last 25 years have shown that ‘GM technologies like many non-GM technologies can bring risks, but these can and have been monitored and quantified, allowing decisions balancing commercial, societal and environmental benefits against measurable risks’ (Caradus Citation2022a). Another recent review (Bowerman et al. Citation2023) concluded that

the limited data available suggests that there may be some willingness to consider novel crops modified to be more resilient if they indeed benefit farmers (particularly those seen as family farmers as opposed to multinationals or corporates), and if the crop fulfils other beneficial desiderata such as reduction of agrochemical use or documentable environmental benefits, and if it is produced using gene editing rather than conventional GM.

This review also confirms that often in promoting the value of new gene manipulation technologies the social aspects and impacts are overlooked. Involving public engagement more in the process of delivering new technologies rather than simply seeking consent is a priority.

Suspicion of corporate multinationals

The majority of commercialised GM crops have been developed by a few multinational companies (Kumar et al. Citation2020). The attitude, real or perceived, of some of these large corporate multinationals in commercialising the first GM crops in the mid-1990s has tarnished the technology and has been a major driver of antagonism towards the use of GM crops for food and feed (Lewontin Citation2000; Patel et al. Citation2005; Amann et al. Citation2007; Clancy Citation2016). Those first GM crops offered either herbicide resistance or resistance to some economically important insect pests (Qaim Citation2015). Criticisms included:

  • GM crops with herbicide resistance were developed so that corporates could continue to make a return on sales of the herbicides (Bonny Citation2008, Citation2011).

  • The traits were all input traits that benefited the farmer with little concern for providing benefits to the consumer. Indeed ‘a new food technology may become an issue when consumers are convinced that this technology provides no additional value to them or to society and may only have advantages for producers and the industry’ (Siegrist Citation2008).

  • Intellectual property protection on the technology would make it more expensive than conventional cultivars, and this cost would be passed onto the consumer. However, a European Parliament study has concluded that there is no monopolistic pricing power present in the delivery of GM crop technologies (Wesseler et al. Citation2015).

  • Corporations developing and marketing GM technologies acted with arrogance and over-confidence (Sandler Citation2004).

  • GM crop seed would be too expensive for poorer farmers (ETC Group Citation2013).

  • Farmers were held hostage to the ‘agro-industrial machine’ due to their control over the use of GM technologies (Tirado and Johnston Citation2010).

  • The argument that GM technologies are needed to secure future food production was viewed simply as a reflection of corporate interests (Jacobsen et al. Citation2013).

  • Concern about corporate ownership of seed and threats to the purity of indigenous crops (Schmidt Citation2005).

  • Poor communication, both in substance and means, on the balance of benefits and risks of this new technology was poor (Cook et al. Citation2006;Nicolia et al. Citation2014; Quemada Citation2022).

It has been determined that many of these concerns are still real with control of New Breeding Technologies being largely with a small number of influential companies (Clapp and Ruder Citation2020).

‘Playing with nature’

Risk perception can be partly driven by notions of what is seen as unnatural and immoral activities of a modern technology (Sjöberg Citation2000). This is largely a moral position related to the level of understanding of consequences and whether GM crops in agriculture are beneficial or harmful to those consuming the food or to the environment and therefore deemed acceptable (Gregorowius et al. Citation2012). GM crops and food are also viewed by some as not being ‘natural’ (Rozin Citation2005, Citation2006; Siegrist Citation2008). However, when considering the visibly significant changes made through phenotype selection breeding in some domesticated crops, for example in the development of modern maize non-GM cultivars, why is that now considered more natural (or certainly not criticised as unnatural) than comparatively small changes in genes and gene function made by genetic modification technologies? While that is the case now for hybrid maize, it was not always the case. Despite the superior performance of maize hybrids and their rapid acceptance by farmers (Sprague Citation1946) in the 1970s were blamed for social and environmental problems arising from industrialised agriculture (Curry Citation2022).

The precautionary principle and precautionary approaches

An early accepted definition of the Precautionary Principle was that it ‘seeks to impose early preventive measures to ward off even those risks for which we have little or no basis on which to predict the future probability of harm’ (Wiener Citation2001). It is based on the adage of ‘better safe than sorry’ (Margolis Citation1997). Alternatively, the precautionary approach is more about how a country, organisation or individual determines how they may evaluate the likelihood of specific risks in relation to the Precautionary Principle. As pointed out by Conko (Citation2003), often the two terms are not differentiated as has occurred in the European Commission’s communication on the precautionary principle (European Commission Citation2000), and the Canadian governments environmental regulatory agency (Vanderzwaag et al. Citation2002). There is a view that precautionary principles should be motivated by ‘decision making under conditions of uncertainty (which) necessitate taking a precautionary approach to decision making’ on a case-by-case basis (Hartzell-Nichols Citation2013). Either way ‘the best elements of a precautionary approach demand good science and challenge the scientific community to improve methods used for risk assessment’ (Hayes Citation2005).

The Cartagena Protocol on Biosafety (Cartagena Protocol Citation2000) outlines the risk assessments required for the trans-boundary movement of modified organisms, and specifically places the precautionary approach (note the term Precautionary Principle is not used) as a fundamental concept when using genetically modified organisms. The precautionary approach in environmental law is outlined in Principle 15 of the Rio Declaration on Environment and Development152 (Citation1992):

In order to protect the environment, the precautionary approach shall be widely applied by States according to their capabilities. Where there are threats of serious or irreversible damage, lack of full scientific certainty shall not be used as a reason for postponing cost-effective measures to prevent environmental degradation. (Rio Declaration Citation1992)

This ‘precautionary approach’ has had a significant impact on how genetically modified plants are regulated in a number of countries including the European Union and New Zealand, amongst others, ‘even though there is little evidence of serious or irreversible damage to the widespread use of these crops in the rest of the world’ (Conko Citation2003; NASEM Citation2016; The Royal Society Citation2016; Adenle Citation2017; European Commission Citation2021). Indeed the Royal Commission on Genetic Modification (Eichelbaum et al. Citation2001) concluded that New Zealand should keep it options open and ‘it would be unwise to turn our back on the potential advantages on offer, but we should proceed carefully, minimising and managing risks’. This is simply the precautionary principle explained. However, 12 years after the Royal Commission, ‘little progress has been made towards developing the public policy capacity necessary to make effective strategic decisions on GM crops in New Zealand’ (McGuinness Institute Citation2013), and even since then nothing has changed. The McGuinness Institute concluded that ‘politicians and public policy analysts are faced with an uneven and incomplete policy landscape, leaving them ill equipped to make sound decisions on New Zealand’s position regarding the release of a GM organism’.

In Europe, where the precautionary principle is also adhered to for all GM technologies (European Commission Citation2000), there has been a plea to use the ‘innovation principle’ where relevant risk assessment would be designed on a case-per-case base, to enable benefiting of gene-edited products while complying with relevant risks management (Jouanin et al. Citation2018). An innovation principle means ensuring that whenever policy is developed, the impact on innovation is fully assessed. The principle should provide guidance to ensure that the choice, design, and regulatory tools foster innovation, rather than hamper it (European Political Strategy Centre Citation2016).

Perceived absence of social, environmental, and economic benefits

While the economic benefits of GM crops have demonstrated considerable benefits to farmers, consumers, and the environment where they have been used (Caradus Citation2022a), in New Zealand, it has been stated that ‘there has been a clear absence of any commercial or other benefit to the New Zealand public’ of the investment to date, largely by the government, in developing genetically modified crops and forages (McGuinness Institute Citation2013). They went further on the basis that

New Zealand is not equipped to make a decision on the release of a GMO in the outdoors; however, we do consider there is sufficient evidence to make a decision on New Zealand becoming a dedicated GM-free food and fibre producer in the short to medium term.

The likely positive consideration of any application to EPA for a conditional release of a GM or gene-edited technology in New Zealand requires a convincing cost–benefit analysis. The onus is on science here to deliver that convincing analysis.

The same applies to ensuring that environmental and societal benefits are also espoused. Societal benefits will ensure that both the quantity and quality of food produced are sufficient for a growing world population (Hickey et al. Citation2019; Zaidi et al. Citation2019), while environmental benefits will ensure that quality of air, land, and water is improved. Many of the benefits resulting from the use of GM crops (Qaim Citation2020; Caradus Citation2022a) can also be attributed to future opportunities provided by New Breeding Technologies.

In two surveys, one of 555 articles from journals, company web pages, and web pages of governmental agencies which summarised 1328 studies/applications of genome-editing in model plants and agricultural crops in the period January 1996 to May 2018 (Modrzejewski et al. Citation2019), and the second of 62 plant breeding companies taken between January and May 2020 (Jorasch Citation2020) identified the types of traits being sought through using New Breeding Technologies (). Proponents of New Breeding Technologies propose that environmental benefits will result from improving crop performance in more hostile climatic conditions, while also mitigating carbon emissions; reducing the use of toxic synthetic chemistry; and increasing efficiency and productivity on-farm and in so doing reducing pressures on natural resources and benefiting farmer incomes, supporting both environmental and economic sustainability goals (Clapp and Ruder Citation2020).

Table 5. Percentage of traits sought using New Breeding Technologies by researchers (Modrzejewski et al. Citation2019) and crop breeding companies (Jorasch Citation2020).

Misinformation

Misinformation defined as ‘false or inaccurate information, especially that which is deliberately intended to deceive’ abounds in society on a range of topics including GM crops and food. A review of mainstream and online news media on genetically modified crops and food, with a potential readership of 256 million, over a two-year period (2019–2021) found an overall falsehood rate of 9%, none of which was positive in sentiment (Lynas et al. Citation2022). They concluded ‘that misinformation about GMOs in the mainstream media is still a significant problem and outranks the proportion of misinformation in other comparable debates such as COVID-19 and vaccines’. The confusion created by misinformation not only impacts public attitudes but also affects government agencies, professional organisations, and the scientific community (Kabat Citation2017; Lelieveld and Andersen Citation2020). In a survey, it was found that ‘public attitudes to public confusion about food safety and health risks’ was the dominating factor (38% of respondents) related to the use of New Breeding Technologies in crops (Lassoued et al. Citation2020)

Misinformation in and about science is a real issue. This includes the predisposition to publish positive results and leave negative or non-significant results unpublished, to stray into unrealistic exaggeration, extrapolation and speculation, use citation bias (preferentially citing papers that support a claim over those that undermine it) or simply using citations incorrectly, sometimes unwittingly, to justify claims they do not in fact support, and inappropriate statistical analysis and/or interpretation (West and Bergstrom Citation2021). A very popular strategy for misinforming society is to spread doubts by referring to the uncertainty of scientific conclusions about pertinent issues of the day such as the health impacts of tobacco and the impact of carbon emissions on climate change (Oreskes and Conway Citation2010). The same could apply to attitudes towards GM crops and food. For example, it has been argued that an altered plant DNA sequence that does not exist in nature could be a health issue due to ‘many GM foods having some common toxic effects’ and some GM food can compromise immune systems (Dona and Arvanitoyannis Citation2009). Yet, the overwhelming evidence based on over 25 years of reporting and debate would indicate that this conclusion is unfounded (Caradus Citation2022a). However, this will not stop this statement being re-stated elsewhere.

Negative results

A number of publications have shown negative effects of GM crops and food, notably the ‘Monarch butterfly controversy’ (Losey et al. Citation1999), the Pusztai study (Ewen and Pusztai Citation1999), Seralini study (Séralini et al. Citation2014), and the more recent Shen study (Shen et al. Citation2022). There has been vigorous debate on each of these (refer to Caradus Citation2022a) and in some cases has resulted in retraction (e.g. the case of the Séralini study) and then defence of the original study sighting collusion and corruption (Qaim Citation2016; Novotny Citation2018). For the lay person, these results are confusing and possibly alarming, and certainly are difficult to explain in the knowledge that for more than 25 years GM crops have been produced and marketed on a commercial basis and have been consumed by millions upon millions of people and livestock, having gone through pre-market regulatory approval, and yet have shown no evidence of adverse health or nutritional effects in the general population (Kour et al. Citation2022).

Distrust of science

In an online survey, conducted in 2018, of just under 20,000 adults across 23 countries about the level of trust by the public in 18 professions, scientists were seen as the most trustworthy profession globally, followed by doctors and teachers, while politicians and government ministers were the least trustworthy (Ipsos Citation2019). However, despite that encouraging statistic scientists cannot be complacent. While scientists had the lowest level of untrustworthiness at 11%, that still indicates that about 10% of the population does not trust scientists or believe in science. But clearly, ‘science relies on public trust for its funding and opportunities to interface with the world’ (West and Bergstrom Citation2021). The widespread confusion about issues such as GM crops and food ‘contributes to an increasing public distrust of science since scientists are seemingly incapable of resolving these controversies’ (Kabat Citation2017). Distrust is driven by a number of factors including religious belief, level of education, political affiliation, socioeconomic status, and simply by the large number of controversies being publicly debated. But not all scientific controversies are of the same type. ‘On some issues, such as vaccines and GM crops, we have solid experimental and epidemiological data based on rigorous research, which enables us to make strong inferences about causality and the rate of adverse effects’ (Kabat Citation2017).

Some, but not all, of this misinformation and distrust of scientists and science can be corrected by reassessing career-related incentives associated with publishing, ensuring productivity metrics are a measure and not a target, addressing better ways of undertaking peer review and evaluating reference lists to reduce citation errors, and increasing the number of science reporters both inside and outside of science institutions to improve public engagement and understanding of science (West and Bergstrom Citation2021). Scientists have an important role in addressing misinformation by not ‘pretending that there is a scientific consensus on controversial issues when there is not’ (Kabat Citation2017). This requires those representing science to engage in transparent, open, and honest debate, examining without bias all available information, resisting exaggeration, and speculation so that a balanced and fact-based case is presented.

Engagement with European consumer experts and societal stakeholders to consider their perceptions, expectations, and acceptability of improving crops and New Plant Breeding Technologies for future-proofing the agri-food systems indicated that ‘governments to take a proactive role in regulating them, ensure openness and transparency in breeding new crop cultivars, and inform consumers about the effects of these breeding programmes and the risks and benefits of the new crop varieties developed’ (Nair et al. Citation2023).

GM crops and food concerns–real and justified?

Systematic reviews of intended and unintended consequences of 25 years of GM crops have concluded that perceived risks associated with GM crops are low to non-existent and that GM crops provide considerable benefits and will provide tangible solutions for many of the current challenges facing humanity, improving societal, economic, and environmental outcomes (Herman and Price Citation2013; Qaim Citation2016; Carzoli et al. Citation2018; Delaney et al. Citation2018; Ladics Citation2019; Louwaars Citation2019; Herman et al. Citation2020; Caradus Citation2022a; Vega Rodríguez et al. Citation2022). In addition, GM crops that provide food and feed are the most highly regulated biological technology in the world (DeFrancesco Citation2013; Baulcombe et al. Citation2014; Brune et al. Citation2021), which would indicate that risks associated with them are low and are significantly outweighed by the benefits.

Many of the New Breeding Technologies used to provide new trait opportunities for crop and forage plants result in products that are indistinguishable from those developed using alternative non-regulated techniques (Glenn et al. Citation2017) and, as a result, some will argue that are safe for use (Schouten et al. Citation2006; Schaart and Visser Citation2009), although others argue ‘that precision, cannot be considered an indication of safety per se, especially in relation to novel traits created by such modifications’ (Eckerstorfer et al. Citation2019). Criteria for categorising and evaluating cultivars produced from New Breeding Technologies have been proposed and include the rationale for plant breeding including the effect achieved, criteria concerning the method of application, the process, and the product (Lusser and Davies Citation2013). A multi-country (USA, Canada, Belgium, France, and Australia) assessment of consumers’ willingness-to-consume CRISPR-produced food compared to GM and non-GM derived foods, indicated that 56, 47, 46, 30, and 51% of respondents, respectively would consume both GM and CRISPR derived foods (Shew et al. Citation2018). However, an additional 15, 13, 10, 30, and 9% respectively would consume CRISPR-produced foods, compared with 8, 5, 6, 3, and 7% for GM-produced foods.

The question to debate is whether using a biotechnology tool is more unnatural or artificial and therefore riskier than earlier methods of manipulating the plant genome? Ethically for some, it is considered wrong to mix genetic material ‘unnaturally’ in a way that cannot occur in nature. But is it also wrong to create ‘unnatural’ genetic mixes not known to exist in nature but by a process where the genetic mix is within the same organism (Bruce Citation2017)? Non-GM breeding, where there is complete freedom to operate, results in this outcome as does some gene editing technologies, which in some countries is tightly regulated (). As others have stated ‘scientifically, it is illogical to regulate systems that produce single, very precise genomic changes, while others, such as chemical and radiation mutagenesis, which produces thousands of random mutations, remain unregulated’ (Mao et al. Citation2019). However, this is not an encouragement for regulators to now include chemical and radiation mutagenesis within their purview. Over the many decades that these forms of mutagenesis have been used, there have been no recorded issues of concern in commercialised cultivars (Oladosu et al. Citation2016). Further, the plant breeding process irrespective of whether it involves conventionally bred, genetically modified, or gene-edited crops provides multiple opportunities to eliminate adverse unintended effects resulting from any of those processes used to develop and select new or improved traits (Glenn et al. Citation2017; Louwaars Citation2019; Brune et al. Citation2021).

Table 6. Comparison of using chemical or radiation mutagenesis with CRISPR site-directed nucleases gene editing (derived from Lema Citation2021).

Concluding comment

Some have argued that with world population projected to be 10 billion by 2050 ‘that postponing technologies that can accelerate breeding makes no economic sense and should justify immediate adoption of accelerated breeding practices’ (Lenaerts et al. Citation2019). However, there are still misgivings due to an unwillingness to differentiate GM crops (generated by transgenesis) from genome-edited crops. Nine out of the top 10 countries by population (i.e. except Pakistan) having either paved a way or stated intentions to open up for easy use of gene-edited plants in commercial agriculture (Sprink et al. Citation2022). Many countries are now either not regulating or seriously considering not regulating SDN1 and, in some cases, SDN2 gene edits as genetically modified organisms except for the European Community and New Zealand (Buchholzer and Frommer Citation2023). A willingness by political decision and lawmakers is an essential element for any progress to be achieved in using the New Breeding Technologies for solving problems (causes and impacts of changing climate, malnourishment, and environmental degradation) currently facing society.

For millennia, human beings have manipulated the genetics of crops and forages, mostly without precision or knowledge of genetic systems, and without legal regulation. However, over the last 25 years with the advent of capability to introduce genes of known effect and with genetic methods that are increasingly precise and targeted regulatory systems has slowed delivery in many territories. Having said that balancing the risk and benefit of any new technological advance is important and required. But regulators must dispense with process-driven regulatory systems for more informed and nuanced product-based regulatory processes. The disconnect between the permitted consumption of foods derived from genetically modified crops but the inability to use genetically modified crops for animal feed (as occurs in New Zealand) is illogical and needs resolution. Similar inconsistencies exist in some territories (e.g. Europe) where importing genetically modified crops for animal feed is permitted but farmers are unable to grow these crops. Societal perspectives and public engagement about emerging technologies are crucial for their successful implementation (Bowerman et al. Citation2023). The science community has a responsibility to work with government and industry decision-makers to counter misinformation, and outline the social, environmental, and economic benefits of New Breeding Technologies through both listening and engaging in a constructive and empathetic manner.

Disclosure statement

The author is employed by Grasslanz Technology Ltd. which has a R&D investment portfolio that includes both genetic modification and gene editing of forages and microbes to provide mitigating solutions to current environmental and animal welfare issues facing both New Zealand and other pastoral economies.

References

  • Acquaah G. 2015. Conventional plant breeding principles and techniques. In: Al-Khayri J, Jain S, Johnson D, editor. Advances in plant breeding strategies: breeding, biotechnology and molecular tools. Cham: Springer; p. 115–158. doi:10.1007/978-3-319-22521-0_5
  • Adenle AA. 2017. Chapter 14. The precautionary principle as a barrier to GMO risk analysis: elicitation of experts’ viewpoints. In: Adenle AA, Murphy DJ, Morris EJ, editors. Genetically modified organisms in developing countries: risk analysis and governance. Cambridge: Cambridge University Press; p. 162–172.
  • Adli M. 2018. The CRISPR tool kit for genome editing and beyond. Nature Communications. 9:Article 1911. doi:10.1038/s41467-018-04252-2.
  • Aerni P. 2019. Politicizing the precautionary principle: why disregarding facts should not pass for farsightedness. Frontiers in Plant Science. 10:Article 1053. doi:10.3389/fpls.2019.01053.
  • Ahmar S, Gill RA, Jung KH, Faheem A, Qasim MU, Mubeen M, Zhou W. 2020. Conventional and molecular techniques from simple breeding to speed breeding in crop plants: recent advances and future outlook. International Journal of Molecular Sciences. 21:Article 2590. doi:10.3390/ijms21072590.
  • Ahmar S, Mahmood T, Fiaz S, Mora-Poblete F, Shafique MS, Chattha MS, Jung K-H. 2021. Advantage of nanotechnology-based genome editing system and its application in crop improvement. Frontiers in Plant Science. 12:Article 663849. doi:10.3389/fpls.2021.663849.
  • Allard RW. 1960. Principles of plant breeding. New York: John Wiley and Sons Inc.
  • Allied Market Research. 2022. Gene editing market by technology: Global opportunity analysis and industry forecast 2021-2031; [accessed 25 February 2023] https://www.alliedmarketresearch.com/gene-editing-market-A10973.
  • Amann W, Khan S, Salzmann O, Steger U, Lonescu-Somer A. 2007. Managing external pressures through corporate diplomacy. Journal of General Management. 33:33–50. doi:10.1177/030630700703300103.
  • Andersson M, Turesson H, Nicolia A, Fält A-S, Samuelsson M, Hofvander P. 2017. Efficient targeted multiallelic mutagenesis in tetraploid potato (solanum tuberosum) by transient CRISPR-Cas9 expression in protoplasts. Plant Cell Reports. 36:117–128. doi:10.1007/s00299-016-2062-3.
  • Anyshchenko A, Yarnold J. 2021. From ‘mad cow’ crisis to synthetic biology: challenges to EU regulation of GMOs beyond the European context. International Environmental Agreements: Politics, Law and Economics. 21:391–404. doi:10.1007/s10784-020-09516-1.
  • Ashby E. 1937. Studies in the inheritance of physiological characters: III. Hybrid vigour in the tomato: part. 1. Manifestations of hybrid vigour from germination to the onset of flowering. Annals of Botany. 1:11–41. http://www.jstor.org/stable/42908405.
  • Ashraf M, Foolad MR. 2013. Crop breeding for salt tolerance in the era of molecular markers and marker-assisted selection. Plant Breeding. 132:10–20. doi:10.1111/pbr.12000.
  • Atanassova A, Keiper F. 2018. Plant breeding innovation: a global regulatory perspective. Cereal Chemistry. 95:8–16. doi:10.1002/cche.10021.
  • Balfourier F, Bouchet S, Robert S, De Oliveira R, Rimbert H, Kitt J, Choulet F, International Wheat Genome Sequencing Consortium, Breed Wheat Consortium, Paux E. 2019. Worldwide phylogeography and history of wheat genetic diversity. Science Advances. 5:Article eaav0536. doi:10.1126/sciadv.aav0536.
  • Balocchi OA, López IF. 2009. Herbage production, nutritive value and grazing preference of diploid and tetraploid perennial ryegrass cultivars (lolium perenne L.). Chilean Journal of Agricultural Research. 69:331–339.
  • Bandyopadhyay A, Kancharla N, Javalkote VS, Dasgupta S, Brutnell TP. 2020. CRISPR-Cas12a (Cpf1): A versatile tool in the plant genome editing tool box for agricultural advancement. Frontiers in Plant Science. 11:Article 584151. doi:10.3389/fpls.2020.584151.
  • Baral K, Coulman B, Biligetu B, Fu Y-B. 2020. Advancing crested wheatgrass [agropyron cristatum (L.) Gaertn.] breeding through genotyping-by-sequencing and genomic selection. PLOS ONE. 15: Article 20239609. doi:10.1371/journal.pone.0239609.
  • Barman M, Ghosh S, Das K, Mondol SA. 2020. Intragenesis as a sustainable crop improvement method: a review. International Journal of Current Microbiology and Applied Sciences. 9:773–782. doi:10.20546/ijcmas.2020.904.092.
  • Barrell PJ, Jacobs JME, Baldwin SJ, Conner AJ. 2010. Intragenic vectors for plant transformation within gene pools. CABI Reviews: Perspectives in Agriculture, Veterinary Science, Nutrition and Natural Resources. 5(010):18pp. doi:10.1079/PAVSNNR20205010.
  • Bashaw EC. 1980. Hybridization of crop plants. In: Fehr WR, Hadley HH, editors. Hybridization of crop plants. Chapter 3. ASA, CSSA and SSSA Books; p. 45–63. doi:10.2135/1980.hybridizationofcrops.c3.
  • Basso MF, Arraes FB, Grossi-de-Sa M, Moreira VJ, Alves-Ferreira M, Grossi-de-Sa MF. 2020. Insights into genetic and molecular elements for transgenic crop development. Frontiers in Plant Science. 11:Article 509. doi:10.3389/fpls.2020.00509.
  • Bateson W, Mendel G. 2013. Mendel's principles of heredity. Courier Corporation.
  • Baulcombe D, Dunwell J, Jones J, Pickett J, Puigdomenech P. 2014. GM Science update: a report to the Council for Science and Technology; [accessed 12 January 2023]. https://centaur.reading.ac.uk/36228/1/GM%20Science%20Update%20-%20Report%20to%20CST%20110314.pdf.
  • Bell GD, Kane NC, Rieseberg LH, Adams KL. 2013. RNA-seq analysis of allele-specific expression, hybrid effects, and regulatory divergence in hybrids compared with their parents from natural populations. Genome Biology and Evolution. 5:1309–1323. doi:10.1093/gbe/evt072.
  • Bezie Y, Tilahun T, Atnaf M, Taye M. 2021. The potential applications of site-directed mutagenesis for crop improvement: a review. Journal of Crop Science and Biotechnology. 24:229–244. doi:10.1007/s12892-020-00080-3.
  • Bibikova M, Beumer K, Trautman JK, Carroll D. 2003. Enhancing gene targeting with designed zinc finger nucleases. Science. 300:764–764. doi:10.1126/science.1079512.
  • Bibikova M, Golic M, Golic KG, Carroll D. 2002. Targeted chromosomal cleavage and mutagenesis in drosophila using zinc-finger nucleases. Genetics. 161:1169–1175. doi:10.1093/genetics/161.3.1169.
  • Biospace. 2023. Gene editing market size to reach USD 18.50 billion in 2028 | Industry trend – extensive use of gene editing in development of personalized medicine; [accessed 25 February 2023]. https://www.biospace.com/article/gene-editing-market-size-to-reach-usd-18-50-billion-in-2028-industry-trend-extensive-use-of-gene-editing-in-development-of-personalized-medicine/.
  • Bogdanove AJ, Schornack S, Lahaye T. 2010. TAL effectors: finding plant genes for disease and defense. Current Opinion in Plant Biology. 13:394–401. doi:10.1016/j.pbi.2010.04.010.
  • Bonny S. 2008. Genetically modified glyphosate-tolerant soybean in the USA: adoption factors, impacts and prospects. A review. Agronomy for Sustainable Development. 28:21–32. doi:10.1051/agro:2007044.
  • Bonny S. 2011. Herbicide-tolerant transgenic soybean over 15 years of cultivation: pesticide use, weed resistance, and some economic issues. The case of the USA. Sustainability. 3:1302–1322. doi:10.3390/su3091302.
  • Bowerman AF, Byrt CS, Roy SJ, Whitney SM, Mortimer JC, Ankeny RA, Gilliham M, Zhang D, Millar AA, Rebetzke GJ, Pogson BJ. 2023. Potential abiotic stress targets for modern genetic manipulation. The Plant Cell. 35:139–161. doi:10.1093/plcell/koac327.
  • Bowley SR, Taylor NL. 1987. Introgressive hybridization. In: Christie BR, editor. CRC handbook of plant science in agriculture. Vol. 1. Boca Raton (FL): CRC Press; p. 23–59.
  • Brandle JE, McVetty PBE. 1989. Heterosis and combining ability in hybrids derived from oilseed rape cultivars and inbred lines. Crop Science. 29:1191–1194. doi:10.2135/cropsci1989.0011183X002900050020x.
  • Breseghello F, Coelho ASG. 2013. Traditional and modern plant breeding methods with examples in rice (Oryza sativa L.). Journal of Agricultural and Food Chemistry. 61:8277–8286. doi:10.1021/jf305531j.
  • Bruce DM. 2017. Genome editing: does it move the goalposts on the GM playing field? In: San-Epifanio LE, editor. Towards a new regulatory framework for GM crops in the European union: scientific, ethical, social and legal issues and the challenges ahead. Chapter 7. Wageningen, The Netherlands: Wageningen Academic Publishers; p. 123–129.
  • Brune P, Chakravarthy S, Graser G, Mathesius CA, McClain S, Petrick JS, Sauve-Ciencewicki A, Schafer B, Silvanovich A, Brink K, Burgin K, et al. 2021. Core and supplementary studies to assess the safety of genetically modified (GM) plants used for food and feed. Journal of Regulatory Science. 9:45–60. doi:10.21423/jrs-v09i1brune.
  • Buchholzer M, Frommer WB. 2023. An increasing number of countries regulate genome editing in crops. New Phytologist. 237:12–15. doi:10.1111/nph.18333.
  • Budhlakoti N, Kushwaha AK, Rai A, Chaturvedi KK, Kumar A, Pradhan AK, Kumar U, Kumar RR, Juliana P, Mishra DC, Kumar S. 2022. Genomic selection: a tool for accelerating the efficiency of molecular breeding for development of climate-resilient crops. Frontiers in Genetics. 13:Article 832153. doi:10.3389/fgene.2022.832153
  • Cabrera-Ponce JL, Barraza A, Alvarez-Venegas R. 2022. Cisgenic crops: major strategies to create cisgenic plants based on genome editing. In: Chaurasia A, Kole C, editors. Cisgenic crops: potential and prospects. concepts and strategies in plant sciences. Cham: Springer; p. 213–235. doi:10.1007/978-3-031-06628-3_11
  • Calyxt. 2019. First commercial sale of Calyxt high oleic soybean oil on the U.S. market. Press Release; [accessed 19 January 2023]. https://calyxt.com/wp-content/uploads/2019/02/20190226_PR-Calyno-Commercialization.pdf.
  • Caradus JR. 2022. Ergot alkaloids in New Zealand pastures and their impact. New Zealand Journal of Agricultural Research. 65:1–41. doi:10.1080/00288233.2020.1785514
  • Cardi T, Neal Stewart C. 2016. Progress of targeted genome modification approaches in higher plants. Plant Cell Reports. 35:1401–1416. doi:10.1007/s00299-016-1975-1.
  • Cartagena Protocol. 2000. Cartagena protocol on biosafety to the convention on biological diversity; [accessed 1 January 2023]. https://www.cbd.int/doc/legal/cartagena-protocol-en.pdf.
  • Carzoli AK, Aboobucker SI, Sandall LL, Lübberstedt TT, Suza WP. 2018. Risks and opportunities of GM crops: Bt maize example. Global Food Security. 19:84–91. doi:10.1016/j.gfs.2018.10.004.
  • Casañas F, Simó J, Casals J, Prohens J. 2017. Toward an evolved concept of landrace. Frontiers in Plant Science. 08:Article 145. doi:10.3389/fpls.2017.00145.
  • CFIA. 2016. Canadian Food Inspection Agency. Regulating agricultural biotechnology in Canada: an overview; [accessed 28 December 2022]. https://inspection.canada.ca/plant-varieties/plants-with-novel-traits/general-public/regulating-agricultural-biotechnology/eng/1338187581090/1338188593891.
  • Chahota RK, Kishore N, Dhiman KC, Sharma TR, Sharma SK. 2007. Predicting transgressive segregants in early generation using single seed descent method-derived micro-macrosperma genepool of lentil (lens culinaris medikus). Euphytica. 156:305–310. doi:10.1007/s10681-007-9359-9.
  • Chakwizira E, Maley S, Dumbleton A. 2021. Water use efficiency of raphanobrassica and forage rape. Agronomy Journal of New Zealand. 51:35–46.
  • Chandrasekaran J, Brumin M, Wolf D, Leibman D, Klap C, Pearlsman M, Sherman A, Arazi T, Gal-On A. 2016. Development of broad virus resistance in non-transgenic cucumber using CRISPR/Cas9 technology. Molecular Plant Pathology. 17:1140–1153. doi:10.1111/mpp.12375.
  • Char SN, Unger-Wallace E, Frame B, Briggs SA, Main M, Spalding MH, Vollbrecht E, Wang K, Yang B. 2015. Heritable site-specific mutagenesis using TALENs in maize. Plant Biotechnology Journal. 13:1002–1010. doi:10.1111/pbi.12344.
  • Chawla R, Shakya R, Rommens CM. 2012. Tuber-specific silencing of asparagine synthetase-1 reduces the acrylamide-forming potential of potatoes grown in the field without affecting tuber shape and yield. Plant Biotechnology Journal. 10:913–924. doi:10.1111/j.1467-7652.2012.00720.x.
  • Chen L, Liu Y-G. 2014. Male sterility and fertility restoration in crops. Annual Review of Plant Biology. 65:579–606. doi:10.1146/annurev-arplant-050213-040119.
  • Clancy KA. 2016. The politics of genetically modified organisms in the United States and Europe. Basingstoke, UK and New York: Palgrave Macmillan published by Springer International, Switzerland. doi: 10.1007/978-3-319-33984-9
  • Clapp J, Ruder S-L. 2020. Precision technologies for agriculture: digital farming, gene-edited crops, and the politics of sustainability. Global Environmental Politics. 20:49–69. doi:10.1162/glep_a_00566.
  • Clasen BM, Stoddard TJ, Luo S, Demorest ZL, Li J, Cedrone F, Tibebu R, Davison S, Ray EE, Daulhac A, Coffman A. 2016. Improving cold storage and processing traits in potato through targeted gene knockout. Plant Biotechnology Journal. 14:169–176. doi:10.1111/pbi.12370.
  • Collard BCY, Jahufer MZZ, Brouwer JB, Pang ECK. 2005. An introduction to markers, quantitative trait loci (QTL) mapping and marker-assisted selection for crop improvement: The basic concepts. Euphytica. 142:169–196. doi:10.1007/s10681-005-1681-5.
  • Collard BCY, Mackill DJ. 2008. Marker-assisted selection: an approach for precision plant breeding in the twenty-first century. Philosophical Transactions of the Royal Society B: Biological Sciences. 363:557–572. doi:10.1098/rstb.2007.2170.
  • Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, Wu X, Jiang W, Marraffini LA, Zhang F. 2013. Multiplex genome engineering using CRISPR/Cas systems. Science. 339:819–823. https://www.science.org/doi/abs/10.1126science.1231143.
  • Conko G. 2003. Safety, risk and the precautionary principle: rethinking precautionary approaches to the regulation of transgenic plants. Transgenic Research. 12:639–647. doi:10.1023/B:TRAG.0000005157.45046.8e.
  • Conner AJ, Barrell PJ, Baldwin SJ, Lokerse AS, Cooper PA, Erasmuson AK, Nap JP, Jacobs JME. 2007. Intragenic vectors for gene transfer without foreign DNA. Euphytica. 154:341–353. doi:10.1007/s10681-006-9316-z.
  • Cook G, Robbins PT, Pieri E. 2006. “Words of mass destruction”: British newspaper coverage of the genetically modified food debate, expert and non-expert reactions. Public Understanding of Science. 15:5–29. doi:10.1177/0963662506058756.
  • Crossa J, Pérez P, de los Campos G, Mahuku G, Dreisigacker S, Magorokosho C. 2011. Genomic selection and prediction in plant breeding. Journal of Crop Improvement. 25:239–261. doi:10.1080/15427528.2011.558767.
  • Crossa J, Pérez-Rodríguez P, Cuevas J, Montesinos-López O, Jarquı´n D, de Los Campos G, Burgueño J, González-Camacho JM, Pérez-Elizalde S, Beyene Y, Dreisigacker S. 2017. Genomic selection in plant breeding: methods, models, and perspectives. Trends in Plant Science. 22:961–975. doi:10.1016/j.tplants.2017.08.011.
  • Cunningham FJ, Goh NS, Demirer GS, Matos JL. 2018. Nanoparticle-mediated delivery towards advancing plant genetic engineering. Trends in Biotechnology. 36:882–897. doi:10.1016/j.tibtech.2018.03.009.
  • Curry HA. 2022. Hybrid seeds in history and historiography. Isis. 113:610–617. doi:10.1086/721075.
  • Curtin SJ, Xiong Y, Michno JM, Campbell BW, Stec AO, Čermák T, Starker C, Voytas DF, Eamens AL, Stupar RM. 2018. CRISPR/cas9 and TALENs generate heritable mutations for genes involved in small RNA processing of glycine max and medicago truncatula. Plant Biotechnology Journal. 16:1125–1137. doi:10.1111/pbi.12857.
  • Dandekar AM, Jacobson A, Ibáñez AM, Gouran H, Dolan DL, Agüero CB, Sl U, Just R, Zaini PA. 2019. Trans-graft protection against Pierce’s disease mediated by transgenic grapevine rootstocks. Frontiers in Plant Science. 10:Article 84. https://www.frontiersin.org/articles/10.3389fpls.2019.00084.
  • Das MK, Dai HK. 2007. A survey of DNA motif finding algorithms. BMC Bioinformatics. 8(7):Article S21. doi:10.1186/1471-2105-8-S7-S21.
  • DeFrancesco L. 2013. How safe does transgenic food need to be? Nature Biotechnology. 31:794–802. doi:10.1038/nbt.2686.
  • Delaney B, Goodman RE, Ladics GS. 2018. Food and feed safety of genetically engineered food crops. Toxicological Sciences. 162:361–371. doi:10.1093/toxsci/kfx249.
  • De Langhe E, Hřibová E, Carpentier S, Doležel J, Swennen R. 2010. Did backcrossing contribute to the origin of hybrid edible bananas? Annals of Botany. 106:849–857. doi:10.1093/aob/mcq187.
  • Desta ZA, Ortiz R. 2014. Genomic selection: genome-wide prediction in plant improvement. Trends in Plant Science. 19:592–601. doi:10.1016/j.tplants.2014.05.006.
  • De Thomazella DPT, Seong K, Mackelprang R, Dahlbeck D, Geng Y, Gill US, Qia T, Pham J, Giuseppe P, Lee CT, et al. 2021. Loss of function of a DMR6 ortholog in tomato confers broad-spectrum disease resistance. Proceedings of the National Academy of Sciences. 118: Article e2026152118. doi:10.1073/pnas.2026152118.
  • de Vetten N, Wolters A, Raemakers K, van der Meer I, ter Stege R, Heeres E, Heeres P, Visser R. 2003. A transformation method for obtaining marker-free plants of a cross-pollinating and vegetatively propagated crop. Nature Biotechnology. 21:439–442.
  • Dhekney SA, Li ZT, Gray DJ. 2011. Grapevines engineered to express cisgenic vitis vinifera thaumatin-like protein exhibit fungal disease resistance. Vitro Cellular & Developmental Biology - Plant. 47:458–466. doi:10.1007/s11627-011-9358-3.
  • Dickert TE, Tracy WF. 2002. Heterosis for flowering time and agronomic traits among early open-pollinated sweet corn cultivars. Journal of the American Society for Horticultural Science. 127:793–797. https://journals.ashs.org/jashs/view/journals/jashs/127/5/article-p793.xml.
  • Dona A, Arvanitoyannis IS. 2009. Health risks of genetically modified foods. Critical Reviews in Food Science and Nutrition. 49:164–175. doi:10.1080/10408390701855993.
  • Dong C, Beetham P, Vincent K, Sharp P. 2006. Oligonucleotide-directed gene repair in wheat using a transient plasmid gene repair assay system. Plant Cell Reports. 25:457–465. doi:10.1007/s00299-005-0098-x.
  • Doudna JA, Charpentier E. 2014. The new frontier of genome engineering with CRISPR-Cas9. Science. 346:Article 1258096. https://www.science.org/doi/10.1126science.1258096
  • Doudna JA, Sternberg SH. 2017. A crack in creation: gene editing and the unthinkable power to control evolution. HarperCollins. https://books.google.com/books/about/A_Crack_In_Creation.html?hl = de&id = F8WlDAAAQBAJ.
  • Drew RA, Magdalita PM, O'Brien CM. 1998. Development of Carica interspecific hybrids. Acta Horticulturae. 2(461):285–292. doi:10.17660/ActaHortic.1998.461.31.
  • Duan YB, Li J, Qin RY, Xu RF, Li H, Yang YC, Ma H, Li L, Wei PC, Yang JB. 2016. Identification of a regulatory element responsible for salt induction of rice OsRAV2 through ex situ and in situ promoter analysis. Plant Molecular Biology. 90:49–62. doi:10.1007/s11103-015-0393-z.
  • Dudley JW, Moll RH. 1969. Interpretation and use of estimates of heritability and genetic variances in plant breeding. Crop Science. 9:257–262. doi:10.2135/cropsci1969.0011183X000900030001x.
  • Dumbleton A, Foley F, Westwood C, Box G. 2022. The development of Pallaton raphanobrassica for New Zealand farming systems. Journal of New Zealand Grasslands. 83:107–114. doi:10.33584/jnzg.2021.83.3505.
  • Duvick DN. 1999. ASA, CSSA, and SSSA books. In: Coors JG, Pandey S, editors. Genetics and exploitation of heterosis in crops. Chapter 27. p. 295–304. doi:10.2134/1999.geneticsandexploitation.c27
  • Eckerstorfer MF, Dolezel M, Heissenberger A, Miklau M, Reichenbecher W, Steinbrecher RA, Waßmann F. 2019. An EU perspective on biosafety considerations for plants developed by genome editing and other new genetic modification techniques (nGMs). Frontiers in Bioengineering and Biotechnology. 7:Article 31.10.3389/fbioe.2019.00031.
  • Edwards S. 2017. Research into genetically modified organisms in New Zealand: an examination of a sociotechnical controversy. Case Studies in the Environment. 1:1–8.
  • EFSA GMO Panel (EFSA Panel on Genetically Modified Organisms), Naegeli H, Bresson J-L, Dalmay T, Dewhurst IC, Epstein MM, Firbank LG, Guerche P, Hejatko J, Moreno FJ, Mullins E, et al. 2020. Applicability of the EFSA opinion on site-directed nucleases type 3 for the safety assessment of plants developed using site-directed nucleases type 1 and 2 and oligonucleotide-directed mutagenesis. EFSA Journal. 18(6299):14. doi:10.2903/j.efsa.2020.6299.
  • Eichelbaum T, Allan J, Fleming J, Randerson R. 2001. Report of the Royal Commission on Genetic Modification. Wellington, New Zealand. [accessed 1 January 2023]. https://environment.govt.nz/assets/Publications/Files/Royal-Commission-on-GM-in-NZ-Final.pdf.
  • Erdmann RM, Picard CL. 2020. RNA-directed DNA methylation. PLOS Genetics. 16:Article e1009034. doi:10.1371/journal.pgen.1009034.
  • Eş I, Gavahian M, Marti-Quijal FJ, Lorenzo JM, Khaneghah AM, Tsatsanis C, Kampranis SC, Barba FJ. 2019. The application of the CRISPR-Cas9 genome editing machinery in food and agricultural science: current status, future perspectives, and associated challenges. Biotechnology Advances. 37:410–421. doi:10.1016/j.biotechadv.2019.02.006.
  • ETC Group. 2013. Putting the cartel before the horse … and farm, seeds, soil, peasants, etc. who will control agricultural inputs, 2013? Communiqué No. 111. p. 40. https://www.etcgroup.org/sites/www.etcgroup.org/files/CartelBeforeHorse11Sep2013.pdf.
  • EU Directive. 2001. Directive 2001/18/EC of the European Parliament and of the Council of 12 March 2001 on the deliberate release into the environment of genetically modified organisms and repealing Council Directive 90/220/EEC; [accessed 1 January 2023]. https://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri = CONSLEG:2001L0018:20080321:EN:PDF.
  • European Commission. 2000. Communication from the Commission on the Pre-Cautionary Principle: COM (2000) 1 final; [accessed 2 January 2023]. http://eur-lex.europa.eu/legal-content/EN/TXT/?uri = CELEX%3A52000DC0001.
  • European Commission. 2021. Directorate-General for research and innovation. Ethics of genome editing. European group on ethics in science and new technologies. Opinion. 32:112. https://data.europa.eu/doi/10.2777659034.
  • European Political Strategy Centre. 2016. Towards an innovation principle endorsed by better regulation (EPSC Strategic Notes No. Issue 14); [accessed 2 January 2023]. https://wayback.archive-it.org/12090/20191129102319/https://ec.europa.eu/epsc/publications/strategic-notes/towards-innovation-principle-endorsed-better-regulation_en.
  • eusage. 2023. European sustainable agriculture through genome editing. The European Community study on new genomic techniques; [accessed 11 January 2023]. https://www.eu-sage.eu/.
  • Ewen SW, Pusztai A. 1999. Effect of diets containing genetically modified potatoes expressing Galanthus nivalis lectin on rat small intestine. The Lancet. 354:1353–1354. doi:10.1016/S0140-6736(98)05860-7.
  • Falconer D. 1960. Introduction to quantitative genetics. Edinburgh: Oliver and Boyd.
  • FAO. 2022. Gene editing and agrifood systems. Food and Agriculture Organization of the United Nations, Rome. doi:10.4060/cc3579en
  • Farhat S, Jain N, Singh N, Sreevathsa R, Das PK, Rai R, Yadav S, Kumar P, Sarkar A, Jain A. 2019. CRISPR-Cas9 directed genome engineering for enhancing salt stress tolerance in rice. Seminars in Cell & Developmental Biology. 96:91–99. doi:10.1016/j.semcdb.2019.05.003.
  • FDA. 2020. US Food and Drug Administration. Food from new plant varieties; [accessed 28 December 2022]. https://www.fda.gov/food/food-ingredients-packaging/food-new-plant-varieties.
  • Fisher RA. 1925. Statistical methods for research workers. Edinburgh: Oliver and Boyd. 239p.
  • Fister AS, Landherr L, Maximova SN, Guiltinan MJ. 2018. Transient expression of CRISPR/Cas9 machinery targeting TcNPR3 enhances defense response in theobroma cacao. Frontiers in Plant Science. 9:Article 268. doi:10.3389/fpls.2018.00268.
  • Flachowsky H, Hanke M-V, Piel A, Strauss SH, Fladung M. 2009. A review on transgenic approaches to accelerate breeding of woody plants. Plant Breeding. 128:217–226. http://onlinelibrary.wiley.com/doi/10.1111j.1439-0523.2008.01591.x/pdf.
  • Frankel R. 1983. Heterosis. In: Reappraisal of theory and practice. Berlin: Springer-Verlag.
  • Fraser PD, Enfissi EMA, Bramley PM. 2009. Genetic engineering of carotenoid formation in tomato fruit and the potential application of systems and synthetic biology approaches. Archives of Biochemistry and Biophysics. 483:196–204. doi:10.1016/j.abb.2008.10.009.
  • Freddy BO, Aimé DN, Jacques LNB, Sébastien LN. 2022. Cisgenesis and plant breeding: a review. In: Chaurasia A, Kole C, editors. Cisgenic crops: potential and prospects. Concepts and strategies in plant sciences. Cham: Springer; p. 79–87. doi:10.1007/978-3-031-06628-3_5.
  • Fritsche S, Poovaiah C, MacRae E, Thorlby G. 2018. A New Zealand perspective on the application and regulation of gene editing. Frontiers in Plant Science. 9:Article 1323. https://www.frontiersin.org/articles/10.3389fpls.2018.01323.
  • FSANZ. 2021. Food Standards Australia New Zealand. Genetically modified foods; [accessed 28 December 2022]. https://www.foodstandards.govt.nz/consumer/gmfood/Pages/default.aspx.
  • FSANZ. 2022. Food Standards Australia New Zealand. GM Foods in Australia and New Zealand; [accessed 28 December 2022]. https://www.foodstandards.gov.au/consumer/gmfood/Documents/GM%20Foods%20in%20Australia%20and%20New%20Zealand.pdf.
  • Fujimoto R, Uezono K, Ishikura S, Osabe K, Peacock WJ, Dennis ES. 2018. Recent research on the mechanism of heterosis is important for crop and vegetable breeding systems. Breeding Science. 68:145–158. doi:10.1270/jsbbs.17155.
  • Gadaleta A, Giancaspro A, Blechl AE, Blanco A. 2008. A transgenic durum wheat line that is free of marker genes and expresses 1DY10. Journal of Cereal Science. 48:439–445. doi:10.1016/j.jcs.2007.11.005.
  • Gaharwar US, Verma A, Singh R, Prasad T, Rajamani P. 2021. Health risks and environmental concerns of GM crop adoption. In: Singh P, Borthakur A, Singh AA, Kumar A, Singh KK, editors. Policy issues in genetically modified crops. Academic Press; p. 371–400. doi:10.1016/B978-0-12-820780-2.00017-0
  • Gao H, Gadlage MJ, Lafitte HR, Lenderts B, Yang M, Schroder M, Farrell J, Snopek K, Peterson D, Feigenbutz L. 2020. Superior field performance of waxy corn engineered using CRISPR–Cas9. Nature Biotechnology. 38:579–581. doi:10.1038/s41587-020-0444-0.
  • Gayon J, Zallen DT. 1998. The role of the vilmorin company in the promotion and diffusion of the experimental science of heredity in France, 1840-1920. Journal of the History of Biology. 31:241–262. http://www.jstor.org/stable/4331479.
  • GDM Seeds. 2022. The Colombian government approves soy edited by GDM with low presence of sugars; [accessed 19 January 2023]. https://www.gdmseeds.com/wp-content/uploads/2023/01/ING-Soja-Colombia.pdf.
  • Gelvin SB. 2000. Agrobacterium and plant genes involved in T-DNA transfer and integration. Annual Review of Plant Physiology and Plant Molecular Biology. 51:223–256.
  • George AL, Mayden RL. 2005. Species concepts and the Endangered Species Act: how a valid biological definition of species enhances the legal protection of biodiversity. Natural Resources Journal. 45:369–407. http://www.jstor.org/stable/24888981.
  • Germanà MA. 2011. Anther culture for haploid and doubled haploid production. Plant Cell, Tissue and Organ Culture (PCTOC). 104:283–300. doi:10.1007/s11240-010-9852-z.
  • Ghose K, Yuan N, Dampanaboina L, Mendu V. 2022. Cisgenesis in the era of genome editing and modern plant biotechnology. In: Chaurasia A, Kole C, editors. Cisgenic crops: potential and prospects. concepts and strategies in plant sciences. Cham: Springer; p. 257–279. doi:10.1007/978-3-031-06628-3_13
  • Glenn KC, Alsop B, Bell E, Goley M, Jenkinson J, Liu B, Martin C, Parrott W, Souder C, Sparks O, et al. 2017. Bringing new plant varieties to market: plant breeding and selection practices advance beneficial characteristics while minimizing unintended changes. Crop Science. 57:2906–2921. doi:10.2135/cropsci2017.03.0199.
  • Gocal G. 2015. Non-transgenic trait development in crop plants using oligo-directed mutagenesis: cibus’ rapid trait development system. In: Eaglesham A, Hardy RW, editors. NABC report 26. New DNA-editing approaches: methods, applications and policy for agriculture. Ithaca (NY): North American Agricultural Biotechnology Council; p. 97–105.
  • Gocal GFW, Schopke C, Beetham PR. 2015. Oligo-mediated targeting gene editing. In: Zhang F, Puchta H, Thompson JG, editors. Advances in new technology for targeted modification of plant genomes. New York, NY: Springer; p. 73–89.
  • Gomez MA, Lin DZ, Moll T, Chauhan RD, Hayden L, Renninger K, Beyene G, Taylor NJ, Carrington J, Staskawicz B, Bart R. 2019. Simultaneous CRISPR/Cas9-mediated editing of cassava eIF4E isoforms nCBP-1 and nCBP-2 reduces cassava brown streak disease symptom severity and incidence. Plant Biotechnology Journal. 17:421–434. doi:10.1111/pbi.12987.
  • Gottesfeld JM, Neely L, Trauger JW, Baird EE, Dervan PB. 1997. Regulation of gene expression by small molecules. Nature. 387:202–205. doi:10.1038/387202a0.
  • Grant PR. 1972. Convergent and divergent character displacement. Biological Journal of the Linnean Society. 4:39–68. doi:10.1111/j.1095-8312.1972.tb00690.x.
  • Grattapaglia D. 2022. Twelve years into genomic selection in forest trees: climbing the slope of enlightenment of marker assisted tree breeding. Forests. 13:Article 1554. doi:10.3390/f13101554.
  • Gregorowius D, Lindemann-Matthies P, Huppenbauer M. 2012. Ethical discourse on the use of genetically modified crops: a review of academic publications in the fields of ecology and environmental ethics. Journal of Agricultural and Environmental Ethics. 25:265–293. doi:10.1007/s10806-011-9330-6.
  • Grewal D, Manito C, Bartolome V. 2011. Doubled haploids generated through anther culture from crosses of elite indica and japonica cultivars and/or lines of rice: large-scale production, agronomic performance, and molecular characterization. Crop Science. 51:2544. doi:10.2135/cropsci2011.04.0236.
  • Gross BL, Henk AD, Forsline PL, Richards CM, Volk GM. 2012. Identification of interspecific hybrids among domesticated apple and its wild relatives. Tree Genetics & Genomes. 8:1223–1235. doi:10.1007/s11295-012-0509-4.
  • Gupta PK, Kumar J, Mir RR, Kumar A. 2010. Marker-assisted selection as a component of conventional plant breeding. Plant Breeding Reviews. 33:145–217.
  • Hake S, Ross-Ibarra J. 2015. The natural history of model organisms: genetic, evolutionary and plant breeding insights from the domestication of maize. eLife. 4:Article e05861. doi:10.7554/eLife.05861.
  • Hallauer AR. 2011. Evolution of plant breeding. Crop Breeding and Applied Biotechnology. 11:197–206. doi:10.1590/S1984-70332011000300001.
  • Han J, Guo B, Guo Y, Zhang B, Wang X, Qiu LJ. 2019. Creation of early flowering germplasm of soybean by CRISPR/Cas9 technology. Frontiers in Plant Science. 10:Article 1446. doi:10.3389/fpls.2019.01446.
  • Hanna WW, Bashaw EC. 1987. Apomixis: its identification and use in plant breeding. Crop Science. 27:1136–1139. doi:10.2135/cropsci1987.0011183X002700060010x.
  • Hartzell-Nichols L. 2013. From ‘the’ precautionary principle to precautionary principles. Ethics, Policy & Environment. 16:308–320. doi:10.1080/21550085.2013.844569.
  • Hasan N, Choudhary S, Naaz N, Sharma N, Laskar RA. 2021. Lipase from rhizopus oryzae R1: in-depth characterization, immobilization, and evaluation in biodiesel production. Journal of Genetic Engineering and Biotechnology. 19:1–26. doi:10.1186/s43141-020-00094-y.
  • Haun W, Coffman A, Clasen BM, Demorest ZL, Lowy A, Ray E, Retterath A, Stoddard T, Juillerat A, Cedrone F. 2014. Improved soybean oil quality by targeted mutagenesis of the fatty acid desaturase 2 gene family. Plant Biotechnology Journal. 12:934–940. doi:10.1111/pbi.12201.
  • Haverkort AJ, Struik PC, Visser RGF, Jacobsen E. 2009. Applied biotechnology to combat late blight in potato caused by phytophthora infestans. Potato Research. 52:249–264.
  • Hayes AW. 2005. The precautionary principle. Arhiv za Higijenu Rada i Toksikologiju. 56:161–166. [accessed 26 February 2023]. https://core.ac.uk/download/pdf/14375077.pdf.
  • Hefferon K. 2022. Cis genesis of crops. In: Chaurasia A, Kole C, editors. Cisgenic crops: potential and prospects. concepts and strategies in plant sciences. Cham: Springer; p. 67–78. doi:10.1007/978-3-031-06628-3_4
  • Heffner EL, Lorenz AJ, Jannink J-L, Sorrells ME. 2010. Plant breeding with genomic selection: gain per unit time and cost. Crop Science. 50:1681–1690. doi:10.2135/cropsci2009.11.0662.
  • Heffner EL, Sorrells ME, Jannink JL. 2009. Genomic selection for crop improvement. Crop Science. 49:1–12. doi:10.2135/cropsci2008.08.0512.
  • Henikoff S, Till BJ, Comai L. 2004. TILLING. traditional mutagenesis meets functional genomics. Plant Physiology. 135:630–636. doi:10.1104/pp.104.041061.
  • Herman RA, Gaffney J, Storer NP. 2020. Enlightened oversight of genetically engineered crops for the next generation. Agricultural & Environmental Letters. 5:e20004. doi:10.1002/ael2.20004.
  • Herman RA, Price WD. 2013. Unintended compositional changes in genetically modified (GM) crops: 20 years of research. Journal of Agricultural and Food Chemistry. 61:11695–11701. doi:10.1021/jf400135r.
  • Herrera-Estrella L, Simpson J, Martínez-Trujillo M. 2004. Transgenic plants. In: Peña L, editor. Transgenic plants: methods and protocols. Methods in molecular biology™. Humana Press; 286 p. 003–032. doi:10.1385/1-59259-827-7:003
  • Herring R. 2008. Opposition to transgenic technologies: ideology, interests and collective action frames. Nature Reviews Genetics. 9:458–463. doi:10.1038/nrg2338.
  • Heslot N, Jannink J-L, Sorrells ME. 2015. Perspectives for genomic selection applications and research in plants. Crop Science. 55:1–12. doi:10.2135/cropsci2014.03.0249.
  • Hickey LT, Hafeez N, Robinson H, Jackson SA, Leal-Bertioli S, Tester M, Gao C, Godwin ID, Hayes BJ, Wulff BB. 2019. Breeding crops to feed 10 billion. Nature Biotechnology. 37:744–754. doi:10.1038/s41587-019-0152-9.
  • Hochholdinger F, Baldauf JA. 2018. Heterosis in plants. Current Biology. 28:R1089–R1092.
  • Holme IB, Dionisio G, Brinch-Pedersen H, Wendt T, Madsen CK, Vincze E, Holm PB. 2012. Cisgenic barley with improved phytase activity. Plant Biotechnology Journal. 10:237–247.
  • Holme IB, Madsen CK, Wendt T, Brinch-Pedersen H. 2020. Horizontal stacking of PAPhy_a cisgenes in barley is a potent strategy for increasing mature grain phytase activity. Frontiers in Plant Science. 11:Article 592139. doi:10.3389/fpls.2020.592139.
  • Holme IB, Wendt T, Holm PB. 2013. Intragenesis and cisgenesis as alternatives to transgenic crop development. Plant Biotechnology Journal. 11:395–407. doi:10.1111/pbi.12055.
  • Hsu PD, Lander ES, Zhang F. 2014. Development and applications of CRISPR-Cas9 for genome engineering. Cell. 157:1262–1278. doi:10.1016/j.cell.2014.05.010.
  • Hu J, Chen B, Zhao J, Zhang F, Xie T, Xu K, Gao G, Yan G, Li H, Li L, Ji G, et al. 2022. Genomic selection and genetic architecture of agronomic traits during modern rapeseed breeding. Nature Genetics. 54:694–704. doi:10.1038/s41588-022-01055-6.
  • Hu J, Gao C. 2023. CRISPR-edited plants by grafting. Nature Biotechnology. [accessed 12 March 2023]. doi:10.1038/s41587-022-01516-7.
  • Huang C-Y, Jin H. 2022. Coordinated epigenetic regulation in plants: a potent managerial tool to conquer biotic stress. Frontiers in Plant Science. 12: Article 795274. doi:10.3389/fpls.2021.795274.
  • Huang H, Cui T, Zhang L, Yang Q, Yang Y, Xie K, Fan C, Zhou Y. 2020. Modifications of fatty acid profile through targeted mutation at BnaFAD2 gene with CRISPR/Cas9-mediated gene editing in brassica napus. Theoretical and Applied Genetics. 133:2401–2411. doi:10.1007/s00122-020-03607-y.
  • Huidobro C, Fernandez AF, Fraga MF. 2013. Aging epigenetics: causes and consequences. Molecular Aspects of Medicine. 34:765–781. doi:10.1016/j.mam.2012.06.006.
  • IAEA. 2022. The International Atomic Energy Agency (IAEA) Mutant Variety Database; [accessed 29 December 2022]. https://nucleus.iaea.org/sites/mvd/SitePages/Home.aspx#.
  • Ipsos. 2019. Global trust in professions – Who do global citizens trust?; [accessed 30 December 2022]. https://www.ipsos.com/sites/default/files/ct/news/documents/2019-09/global-trust-in-professions-trust-worthiness-index-2019.pdf.
  • Irvine JE. 1999. Saccharum species as horticultural classes. Theoretical and Applied Genetics. 98(2):186–194. doi:10.1007/s001220051057.
  • ISAAA database. 2023. GM Approval Database. https://www.isaaa.org/gmapprovaldatabase/.
  • Jacobsen SE, Sørensen M, Pedersen SM, Weiner J. 2013. Feeding the world: genetically modified crops versus agricultural biodiversity. Agronomy for Sustainable Development. 33:651–662. doi:10.1007/s13593-013-0138-9.
  • Jankowicz-Cieslak J, Till BJ. 2015. Forward and reverse genetics in crop breeding. In: Al-Khayri J, Jain S, Johnson D, editors. Advances in plant breeding strategies: breeding, biotechnology and molecular tools. Switzerland: Springer International Publishing; p. 215–240. doi:10.1007/978-3-319-22521-0_8.
  • Jiang GL. 2013. Molecular markers and marker-assisted breeding in plants. In: Andersen SB, editor. Plant breeding from laboratories to fields. Rijeka (Croatia): InTechOpen; Chapter 3, p. 45–83.
  • Jin S, Clark B, Kuznesof S, Lin X, Frewer LJ. 2019. Synthetic biology applied in the agrifood sector: public perceptions, attitudes and implications for future studies. Trends in Food Science & Technology. 91:454–466. doi:10.1016/j.tifs.2019.07.025.
  • Jo KR, Kim CJ, Kim SJ, Kim TY, Bergervoet M, Jongsma MA, Visser RG, Jacobsen E, Vossen JH. 2014. Development of late blight resistant potatoes by cisgene stacking. BMC Biotechnology. 14: Article 50. doi:10.1186/1472-6750-14-50.
  • Jones MGK, Fosu-Nyarko J, Iqbal S, Adeel M, Romero-Aldemita R, Arujanan M, Kasai M, Wei X, Prasetya B, Nugroho S, et al. 2022. Enabling trade in gene-edited produce in Asia and australasia: the developing regulatory landscape and future perspectives. Plants. 11. doi:10.3390/plants11192538
  • Jorasch P. 2020. Potential, challenges, and threats for the application of new breeding techniques by the private plant breeding sector in the EU. Frontiers in Plant Science. 11:Article 582011. doi:10.3389/fpls.2020.582011.
  • Jouanin A, Boyd L, Visser RG, Smulders MJ. 2018. Development of wheat with hypoimmunogenic gluten obstructed by the gene editing policy in Europe. Frontiers in Plant Science. 9:Article 1523. doi:10.3389/fpls.2018.01523.
  • Jouanin A, Gilissen LJ, Schaart JG, Leigh FJ, Cockram J, Wallington EJ, Boyd LA, van den Broeck HC, van der Meer IM, America AH, Visser RG. 2020. CRISPR/cas9 gene editing of gluten in wheat to reduce gluten content and exposure—reviewing methods to screen for coeliac safety. Frontiers in Nutrition. 7:Article 51. doi:10.3389/fnut.2020.00051.
  • Jouanin A, Schaart JG, Boyd LA, Cockram J, Leigh FJ, Bates R, Wallington EJ, Visser RG, Smulders MJ. 2019. Outlook for coeliac disease patients: towards bread wheat with hypoimmunogenic gluten by gene editing of α- and γ-gliadin gene families. BMC Plant Biology. 19:Article 333. doi:10.1186/s12870-019-1889-5.
  • Joung J, Sander J. 2013. TALENs: a widely applicable technology for targeted genome editing. Nature Reviews Molecular Cell Biology. 14:49–55. doi:10.1038/nrm3486.
  • Jumbo M, Weldekidan T, Holland JB, Hawk JA. 2011. Comparison of conventional, modified single seed descent, and doubled haploid breeding methods for maize inbred line development using germplasm enhancement of maize breeding crosses. Crop Science. 51:1534–1543. doi:10.2135/cropsci2010.10.0594.
  • Jung JH, Altpeter F. 2016. TALEN mediated targeted mutagenesis of the caffeic acid O-methyltransferase in highly polyploid sugarcane improves cell wall composition for production of bioethanol. Plant Molecular Biology. 92:131–142. doi:10.1007/s11103-016-0499-y.
  • Kabat GC. 2017. Taking distrust of science seriously. EMBO Reports. 18:1052–1055. doi:10.15252/embr.201744294.
  • Kanazawa A, Inaba JI, Shimura H, Otagaki S, Tsukahara S, Matsuzawa A, Bm K, Goto K, Masuta C. 2011. Virus-mediated efficient induction of epigenetic modifications of endogenous genes with phenotypic changes in plants. The Plant Journal. 65:156–168. doi:10.1111/j.1365-313X.2010.04401.x.
  • Kankanala P, Nandety RS, Mysore KS. 2019. Genomics of plant disease resistance in legumes. Frontiers in Plant Science. 10:Article 1345. doi:10.3389/fpls.2019.01345.
  • Kannan B, Jung JH, Moxley GW, Lee S-M, Altpeter F. 2018. TALEN-mediated targeted mutagenesis of more than 100 COMT copies/alleles in highly polyploid sugarcane improves saccharification efficiency without compromising biomass yield. Plant Biotechnology Journal. 16:856–866. doi:10.1111/pbi.12833.
  • Kemesyte V, Statkeviciute G, Brazauskas G. 2017. Perennial ryegrass yield performance under abiotic stress. Crop Science. 57:1935–1940. doi:10.2135/cropsci2016.10.0864.
  • Kharkwal MC. 2012. A brief history of plant mutagenesis. In: Shu QY, Forster BP, Nakagawa H, editors. Plant mutation breeding and biotechnology. Wallingford: CABI; p. 21–30.
  • Kim JY, Kim JH, Jang YH, Yu J, Bae S, Kim M-S, Cho Y-G, Jung YJ, Kang KK. 2023. Transcriptome and metabolite profiling of tomato SGR-knockout null lines using the CRISPR/Cas9 system. International Journal of Molecular Sciences. 24:Article 109. doi:10.3390/ijms24010109.
  • Kim Y-J, Zhang D. 2018. Molecular control of male fertility for crop hybrid breeding. Trends in Plant Science. 23:53–65. doi:10.1016/j.tplants.2017.10.001.
  • Kodym A, Afza R. 2003. Physical and chemical mutagenesis. In: Grotewol E, editor. Plant functional genomics. Methods in molecular biology™, vol 236. Totowa, NJ, USA: Humana Press; p. 189–204. doi:10.1385/1-59259-413-1:189.
  • Köferle A, Stricker SH, Beck S. 2015. Brave new epigenomes: the dawn of epigenetic engineering. Genome Medicine. 7:Article 59. doi:10.1186/s13073-015-0185-8.
  • Korban SS. 1986. Interspecific hybridization in Malus. HortScience. 21:41–48. https://journals.ashs.org/hortsci/view/journals/hortsci/21/1/article-p41_b.xml.
  • Kouhen M, García-Caparrós P, Twyman RM, Abdelly C, Mahmoudi H, Schillberg S, Debez A. 2022. Improving environmental stress resilience in crops by genome editing: insights from extremophile plants. Critical Reviews in Biotechnology. [accessed 12 March 2023]. doi:10.1080/07388551.2022.2042481.
  • Koul B. 2022. Cisgenics and crop improvement. In: Koul B, editor. Cisgenics and transgenics. Singapore: Springer; p. 107–129. doi:10.1007/978-981-19-2119-3_3
  • Kour J, Sangeeta DH, Tomar M, Bala K, Mishra RC. 2022. Genetically modified plant and animal foods. In: Kour J, Sharma V, Khanday I, editors. Genetically modified crops and food security: commercial, ethical and health considerations. Chapter1. Taylor & Francis. doi:10.4324/9781003278566-1.
  • Krishna VV, Qaim M. 2008. Consumer attitudes toward GM food and pesticide residues in India. Review of Agricultural Economics. 30:233–251. doi:10.1111/j.1467-9353.2008.00402.x.
  • Kuluev BR, Gumerova GR, Mikhaylova EV, Gerashchenkov GA, Rozhnova NA, Vershinina ZR, Khyazev AV, Matniyazov RT, Baymiev A, Baymiev A, Chemeris AV. 2019. Delivery of CRISPR/Cas components into higher plant cells for genome editing. Russian Journal of Plant Physiology. 66:694–706. doi:10.1134/S102144371905011X.
  • Kumar J, Kumar A, Gupta S, Kumar D, DePauw S. 2022. Reverse genetic approaches for breeding nutrient-rich and climate-resilient cereal and food legume crops. Heredity. 128:473–496. doi:10.1038/s41437-022-00513-5.
  • Kumar K, Gambhir G, Dass A, Tripathi AK, Singh A, Jha AK, Yadava P, Choudhary M, Rakshit S. 2020. Genetically modified crops: current status and future prospects. Planta. 251: Article 91. doi:10.1007/s00425-020-03372-8.
  • Kumar S. 2019. Epigenetics and epigenomics for crop improvement: current opinion. Advances in Biotechnology and Microbiology. 14:Article 555879. doi:10.19080/AIBM.2019.14.555879.
  • Kwan BD, Seligmann B, Nguyen TD, Franke J, Dang TT. 2023. Leveraging synthetic biology and metabolic engineering to overcome obstacles in plant pathway elucidation. Current Opinion in Plant Biology. 71: Article 102330. doi:10.1016/j.pbi.2022.102330.
  • Ladics GS. 2019. Assessment of the potential allergenicity of genetically-engineered food crops. Journal of Immunotoxicology. 16:43–53. doi:10.1080/1547691X.2018.1533904.
  • Laird NM, Lange C. 2011. Principles of inheritance: Mendel’s laws and genetic models. In: The fundamentals of modern statistical genetics. New York: Statistics for Biology and Health. Springer; p. 15–30. doi:10.1007/978-1-4419-7338-2_2
  • Lamichhane S, Thapa S. 2022. Advances from conventional to modern plant breeding methodologies. Plant Breeding and Biotechnology. 10:1–14. doi:10.9787/PBB.2022.10.1.1.
  • Lassoued R, Macall DM, Smyth SJ, Phillips PW, Hesseln H. 2020. How should we regulate products of new breeding techniques? Opinion of surveyed experts in plant biotechnology. Biotechnology Reports. 26: Article e00460. doi:10.1016/j.btre.2020.e00460.
  • Laughnan JR, Gabay-Laughnan S. 1983. Cytoplasmic male sterility in maize. Annual Review of Genetics. 17:27–48.
  • Layne RE, Sherman WB. 1986. Interspecific hybridization of Prunus. HortScience. 21:48–51. https://journals.ashs.org/hortsci/view/journals/hortsci/21/1/article-p48.xml.
  • Lee K, Zhang Y, Kleinstiver BP, Guo JA, Aryee MJ, Miller J, Malzahn A, Zarecor S, Lawrence-Dil CJ, Joung JK. 2019a. Activities and specificities of CRISPR/Cas9 and Cas12a nucleases for targeted mutagenesis in maize. Plant Biotechnology Journal. 17:362–372. doi:10.1111/pbi.12982.
  • Lee M. 2006. The phenotypic and genotypic eras of plant breeding. In: Lamkey KR, Lee M, editors. Plant breeding: the Arnel R. Hallauer International Symposium. Ames: Blackwell Publishing; p. 213–218.
  • Lee MA, Howard-Andrews V, Chester M. 2019b. Resistance of multiple diploid and tetraploid perennial ryegrass (lolium perenne L.) varieties to three projected drought scenarios for the UK in 2080. Agronomy. 9: Article 159. doi:10.3390/agronomy9030159.
  • Lelieveld H, Andersen V. 2020. Debunking misinformation about food. Scientific Bulletin. Series F. Biotechnologies. 23:237–247.
  • Lema M. 2021. Breaking barriers with breeding: a primer on new breeding innovations for food security. ISAAA Brief. 56:23–31.
  • Lenaerts B, Collard BCY, Demont M. 2019. Review: improving global food security through accelerated plant breeding. Plant Science. 278: Article 110207. doi:10.1016/j.plantsci.2019.110207.
  • Levings CS. 3rd. 1993. Thoughts on cytoplasmic male sterility in cms-T maize. The Plant Cell. 5:1285–1290. doi:10.2307/3869781.
  • Lewontin RC. 2000. The maturing of capitalist agriculture: farmer as proletarian. In: Magdoff F, Foster JB, Buttel FH, editors. Hungry for profit: the agribusiness threat to farmers, food, and the environment. New York: Publisher: Monthly Review Press; p. 93–106. https://books.google.co.nz/books?id = Iu78TKUEPv4C.
  • Li A, Jia S, Yobi A, Ge Z, Sato SJ, Zhang C, Angelovici R, Clemente TE, Holding DR. 2018. Editing of an alpha-kafirin gene family increases, digestibility and protein quality in sorghum. Plant Physiology. 177:1425–1438. doi:10.1104/pp.18.00200.
  • Li C, Unver T, Zhang B. 2017. A high-efficiency CRISPR/Cas9 system for targeted mutagenesis in cotton (gossypium hirsutum L.). Scientific Reports. 7: Article 43902. doi:10.1038/srep43902.
  • Li J, Scarano A, Mora Gonzalez N, D’Orso F, Yue Y, Nemeth K, Saalbach G, Hill L, Oliveira Martins C, Moran R, et al. 2022. Biofortified tomatoes provide a new route to vitamin D sufficiency. Nature Plants. 8:611–616. doi:10.1038/s41477-022-01154-6.
  • Li T, Liu B, Spalding M, Weeks DP, Yang B. 2012. High-efficiency TALEN-based gene editing produces disease-resistant rice. Nature Biotechnology. 30:390–392. doi:10.1038/nbt.2199.
  • Li Z, Liu Z-B, Xing A, Moon BP, Koellhoffer JP, Huang L, Ward RT, Falco CE, Cigan SC, M A. 2015. Cas9-guide RNA directed genome editing in soybean. Plant Physiology. 169:960–970. doi:10.1104/pp.15.00783.
  • Li Z, Moon BP, Xing A, Liu ZB, McCardell RP, Damude HG, Falco SC. 2010. Stacking multiple transgenes at a selected genomic site via repeated recombinase-mediated DNA cassette exchanges. Plant Physiology. 154:622–631. doi:10.1104/pp.110.160093.
  • Liang Z, Chen K, Li T, Zhang Y, Wang Y, Zhao Q, Liu J, Zhang H, Liu C, Ran Y, Gao C. 2017. Efficient DNA-free genome editing of bread wheat using CRISPR/Cas9 ribonucleoprotein complexes. Nature Communications. 8: Article 14261. doi:10.1038/ncomms14261.
  • Liu B, Li M, Cheng L, Liang D, Zou Y, Ma F. 2012. Influence of rootstock on antioxidant system in leaves and roots of young apple trees in response to drought stress. Plant Growth Regulation. 67:247–256. doi:10.1007/s10725-012-9683-5.
  • Liu D. 2009. Design of gene constructs for transgenic maize. In: Scott MP, editor. Transgenic maize. Methods in molecular biology™, vol 526. Totowa, NJ: Humana Press; p. 3–20. doi:10.1007/978-1-59745-494-0_1.
  • Losert D, Maurer HP, Marulanda JJ, Würschum T. 2017. Phenotypic and genotypic analyses of diversity and breeding progress in European triticale (× triticosecale wittmack). Plant Breeding. 136:18–27. doi:10.1111/pbr.12433.
  • Losey JE, Rayor LS, Carter ME. 1999. Transgenic pollen harms monarch larvae. Nature. 399:214. doi:10.1038/20338.
  • Louwaars N. 2019. Food safety and plant breeding - why are there no problems in practice? In: Urazbaeva A, Szajkowska A, Wernaart BFW, Franssens NT, Vaskoska RS, editors. The functional field of food law. Wageningen, The Netherlands: Wageningen Academic Press Publishers; p. 89–101.
  • Lübberstedt T, Frei UK. 2012. Application of doubled haploids for target gene fixation in backcross programmes of maize. Plant Breeding. 131:449–452. doi:10.1111/j.1439-0523.2011.01948.x.
  • Lucht JM. 2015. Public acceptance of plant biotechnology and GM crops. Viruses. 7:4254–4281. doi:10.3390/v7082819.
  • Lusser M, Davies HV. 2013. Comparative regulatory approaches for groups of new plant breeding techniques. New Biotechnology. 30:437–446. doi:10.1016/j.nbt.2013.02.004.
  • Lynas M, Adams J, Conrow J. 2022. Misinformation in the media: global coverage of GMOs 2019-2021. GM Crops & Food. [accessed 12 March 2023]. doi:10.1080/21645698.2022.2140568.
  • Ma X, Zhang Q, Zhu Q, Liu W, Chen Y, Qiu R, Wang B, Yang Z, Li H, Lin Y, et al. 2015. A robust CRISPR/Cas9 system for convenient, high-efficiency multiplex genome editing in monocot and dicot plants. Molecular Plant. 8:1274–1284. doi:10.1016/j.molp.2015.04.007.
  • Mabry ME, Turner-Hissong SD, Gallagher EY, McAlvay AC, An H, Edger PP, Moore JD, Pink DAC, Teakle GR, Stevens CJ, et al. 2021. The evolutionary history of wild, domesticated, and feral Brassica oleracea (Brassicaceae). Molecular Biology and Evolution. 38:4419–4434. doi:10.1093/molbev/msab183.
  • MacFie H. 2007. Consumer-led food product development. Woodhead: Woodhead Publishing Ltd, Cambridge.
  • Maghari BM, Ardekani AM. 2011. Genetically modified foods and social concerns. Avicenna Journal of Medical Biotechnology. 3:109–117.
  • Malnoy M, Viola R, Jung MH, Koo OJ, Kim S, Kim JS, Velasco R, Nagamangala Kanchiswamy C. 2016. DNA-free genetically edited grapevine and apple protoplast using CRISPR/Cas9 ribonucleoproteins. Frontiers in Plant Science. 7: Article 1904. doi:10.3389/fpls.2016.01904.
  • Mao Y, Botella JR, Liu Y, Zhu J-K. 2019. Gene editing in plants: progress and challenges. National Science Review. 6:421–437. doi:10.1093/nsr/nwz005.
  • Marchant GE, Stevens YA. 2015. A new window of opportunity to reject process-based biotechnology regulation. GM Crops & Food. 6:233–242.
  • Margolis H. 1997. Dealing with isk: why the public and the experts disagree on environmental issues. United Kingdom: University of Chicago Press.
  • Margueron R, Reinberg D. 2010. Chromatin structure and the inheritance of epigenetic information. Nature Reviews Genetics. 11:285–296. doi:10.1038/nrg2752.
  • Markets and Markets. 2022. Genome editing market size. Market Research Report Code BT3380, published November 2021; [accessed 1 January 2023]. https://www.marketsandmarkets.com/Market-Reports/genome-editing-engineering-market-231037000.html.
  • Mason AS, Batley J. 2015. Creating new interspecific hybrid and polyploid crops. Trends in Biotechnology. 33:436–441. doi:10.1016/j.tibtech.2015.06.004.
  • Massel K, Hintzsche J, Restall J, Kerr ED, Schulz BL, Godwin ID. 2023. CRISPR-knockout of β-kafirin in sorghum does not recapitulate the grain quality of natural mutants. Planta. 257:8. doi:10.1007/s00425-022-04038-3.
  • Mather K. 1955. The genetical basis of heterosis. Proceedings of the Royal Society of London. Series B-Biological Sciences. 144:143–150.
  • Matsuoka Y, Vigouroux Y, Goodman MM, Sanchez GJ, Buckler E, Doebley J. 2002. A single domestication for maize shown by multilocus microsatellite genotyping. Proceedings of the National Academy of Sciences. 99:6080–6084. doi:10.1073/pnas.052125199.
  • Mayo O. 1987. The theory of plant breeding. Oxford: Clarendon Press.
  • Mba C. 2013. Induced mutations unleash the potentials of plant genetic resources for food and agriculture. Agronomy. 3:200–231. doi:10.3390/agronomy3010200.
  • Mba C, Afza R, Bado S, Jain SM. 2010. Induced mutagenesis in plants using physical and chemical agents. In: Davey MR, Anthony P, editors. Plant cell culture: essential methods. Chichester, UK: Wiley-Blackwell; p. 111–130.
  • McCallum CM, Comai L, Greene EA, Henikoff S. 2000. Targeted screening for induced mutations. Nature Biotechnology. 18:455–457. doi:10.1038/74542.
  • McGuinness Institute. 2013. An overview of genetic modification in New Zealand 1973–2013: the first forty years. Project 2058; Report 16, September 2013, p. 140; [accessed 1 January 2023] http://www.mcguinnessinstitute.org/project-2058-reports/.
  • McHughen A. 2013. GM crops and foods. GM Crops & Food. 4:172–182. doi:10.4161/gmcr.26532.
  • Menz J, Modrzejewski D, Hartung F, Wilhelm R, Sprink T. 2020. Genome edited crops touch the market: a view on the global development and regulatory environment. Frontiers in Plant Science. 11: Article 586027. doi:10.3389/fpls.2020.586027.
  • Mergoum M, Sapkota S, ElDoliefy AEA, Naraghi SM, Pirseyedi S, Alamri MS, AbuHammad W. 2019. Triticale (x Triticosecale Wittmack) breeding. In: Al-Khayri J, Jain S, Johnson D, editors. Advances in plant breeding strategies: cereals. Cham: Springer; p. 405–451. doi:10.1007/978-3-030-23108-8_11
  • Merrick LF, Herr AW, Sandhu KS, Lozada DN, Carter AH. 2022a. Utilizing genomic selection for wheat population development and improvement. Agronomy. 12: Article 522. doi:10.3390/agronomy12020522.
  • Merrick LF, Herr AW, Sandhu KS, Lozada DN, Carter AH. 2022b. Optimizing plant breeding programs for genomic selection. Agronomy. 12: Article 714. doi:10.3390/agronomy12030714.
  • Meuwissen THE, Hayes BJ, Goddard ME. 2001. Prediction of total genetic value using genome-wide dense marker maps. Genetics. 157:1819–1829. doi:10.1093/genetics/157.4.1819.
  • Miki B, McHugh S. 2004. Selectable marker genes in transgenic plants: applications, alternatives and biosafety. Journal of Biotechnology. 107:193–232. doi:10.1016/j.jbiotec.2003.10.011.
  • Miles JW. 2007. Apomixis for cultivar development in tropical forage grasses. Crop Science. 47:S-238–S-249. doi:10.2135/cropsci2007.04.0016IPBS.
  • Miroshnichenko D, Timerbaev V, Okuneva A, Klementyeva A, Sidorova T, Pushin A, Dolgov S. 2020. Enhancement of resistance to PVY in intragenic marker-free potato plants by RNAi-mediated silencing of eIF4E translation initiation factors. Plant Cell, Tissue and Organ Culture (PCTOC). 140:691–705. doi:10.1007/s11240-019-01746-9.
  • Mishra R, Rao GJN. 2016. In-vitro androgenesis in rice: advantages, constraints and future prospects. Rice Science. 23:57–68. doi:10.1016/j.rsci.2016.02.001.
  • Modrzejewski D, Hartung F, Sprink T, Krause D, Kohl C, Wilhelm R. 2019. What is the available evidence for the range of applications of genome-editing as a new tool for plant trait modification and the potential occurrence of associated off-target effects: a systematic map. Environmental Evidence. 8: Article 27. doi:10.1186/s13750-019-0171-5.
  • Mohanta TK, Bashir T, Hashem A, Abd_Allah EF, Bae H. 2017. Genome editing tools in plants. Genes. 8: Article 27. doi:10.3390/genes8120399.
  • Moose SP, Mumm RH. 2008. Molecular plant breeding as the foundation for 21st century crop improvement. Plant Physiology. 147:969–977. doi:10.1104/pp.108.118232.
  • MPI. 2021. Ministry for Primary Industries. Labelling requirements for genetically modified food; [accessed 28 December 2022]. https://www.mpi.govt.nz/food-business/labelling-composition-food-drinks/specific-product-labelling/labelling-requirements-for-genetically-modified-food/.
  • Nadeem MA, Nawaz MA, Shahid MO, Doğan Y, Comertpay G, Yıldız M, Hatipoğlu R, Ahmad F, Alsaleh A, Labhane N. 2018. DNA molecular markers in plant breeding: current status and recent advancements in genomic selection and genome editing. Biotechnology & Biotechnological Equipment. 32:261–285. doi:10.1080/13102818.2017.1400401.
  • Nair A, Fischer ARH, Moscatelli S, Socaciu C, Kohl C, Stetkiewicz SS, Menary J, Baekelandt A, Nanda AK, Jorasch P, et al. 2023. European consumer and societal stakeholders’ response to crop improvements and new plant breeding techniques. Food and Energy Security. 12: Article e417. doi:10.1002/fes3.417.
  • Nair RM. 2004. Developing tetraploid perennial ryegrass (Lolium perenne L.) populations. New Zealand Journal of Agricultural Research. 47:45–49. doi:10.1080/00288233.2004.9513569.
  • NASEM. 2016. Genetically engineered crops: experiences and prospects. Washington, DC: National Academies Press. doi:10.17226/23395.
  • New Zealand Legislation. 1996. Hazardous Substances and New Organisms Act 1996; [accessed 2021 Dec 5]. https://legislation.govt.nz/act/public/1996/0030/latest/DLM381228.html.
  • Nicolia A, Manzo A, Veronesi F, Rosellini D. 2014. An overview of the last 10 years of genetically engineered crop safety research. Critical Reviews in Biotechnology. 34:77–88. doi:10.3109/07388551.2013.823595.
  • Niu C, Li H, Jiang L, Yan M, Li C, Geng D, Xie Y, Yan Y, Shen X, Chen P, Dong J. 2019. Genome-wide identification of drought-responsive microRNAs in two sets of Malus from interspecific hybrid progenies. Horticulture Research. 6: Article 75. doi:10.1038/s41438-019-0157-z.
  • Novotny E. 2018. Retraction by corruption: the 2012 Séralini paper. Journal of Biological Physics and Chemistry. 18:32–56. doi:10.4024/19NO17F.jbpc.18.01.
  • Nyquist WE, Baker RJ. 1991. Estimation of heritability and prediction of selection response in plant populations. Critical Reviews in Plant Sciences. 10:235–322. doi:10.1080/07352689109382313.
  • OGTR. 2021. Office of the Gene Technology Regulator. Technical review of the Gene Technology Regulations 2001 Decision Regulation Impact Statement; [accessed 28 December 2022]. https://www.ogtr.gov.au/sites/default/files/files/2021-07/decision_ris.pdf.
  • Oh TJ, May GD. 2001. Oligonucleotide-directed plant gene targeting. Current Opinion in Biotechnology. 12:169–172. doi:10.1016/S0958-1669(00)00194-4.
  • O'Keefe M, Perrault S, Halpern J, Ikemoto L, Yarborough M. 2015 “Editing” genes: a case study about how language matters in bioethics. The American Journal of Bioethics. 15:3–10. doi:10.1080/15265161.2015.1103804.
  • Okuzaki A, Toriyama K. 2004. Chimeric RNA/DNA oligonucleotide-directed gene targeting in rice. Plant Cell Reports. 22:509–512. doi:10.1007/s00299-003-0698-2.
  • Oladosu Y, Rafii MY, Abdullah N, Hussin G, Ramli A, Rahim HA, Miah G, Usman M. 2016. Principle and application of plant mutagenesis in crop improvement: a review. Biotechnology & Biotechnological Equipment. 30:1–16. doi:10.1080/13102818.2015.1087333.
  • Oreskes N, Conway EM. 2010. Merchants of doubt: how a handful of scientists obscured the truth on issues from tobacco smoke to global warming. London (UK): Bloomsbury.
  • Ortiz R. 2020. Göte Turesson’s research legacy to hereditas: from the ecotype concept in plants to the analysis of landraces’ diversity in crops. Hereditas. 157:44. doi:10.1186/s41065-020-00159-5.
  • Ospovat D. 1995. The development of Darwin's theory: natural history, natural theology, and natural selection, 1838-1859. Cambridge.: Cambridge University Press.
  • Pandey PK, Quilichini TD, Vaid N, Gao P, Xiang D, Datla R. 2019. Versatile and multifaceted CRISPR/Cas gene editing tool for plant research. Seminars in Cell & Developmental Biology. 96:107–114. Academic Press. doi:10.1016/j.semcdb.2019.04.012
  • Parisi C, Rodríguez-Cerezo E. 2021. Current and future market applications of new genomic techniques. EUR 30589 EN. Luxembourg: Publications Office of the European Union. 52. doi:10.2760/02472, JRC123830.
  • Patel R, Torres RJ, Rosset P. 2005. Genetic engineering in agriculture and corporate engineering in public debate: risk, public relations, and public debate over genetically modified crops. International Journal of Occupational and Environmental Health. 11:428–436. doi:10.1179/oeh.2005.11.4.428.
  • Pathak B, Srivastava V. 2020. Recombinase-mediated integration of a multigene cassette in rice leads to stable expression and inheritance of the stacked locus. Plant Direct. 4:1–10. doi:10.1002/pld3.236.
  • Peng A, Chen S, Lei T, Xu L, He Y, Wu L, Yao L, Zou X. 2017. Engineering canker-resistant plants through CRISPR/Cas9-targeted editing of the susceptibility gene CsLOB1 promoter in citrus. Plant Biotechnology Journal. 15:1509–1519. doi:10.1111/pbi.12733.
  • Penna S, Jain SM. 2023. Fruit crop improvement with genome editing, in vitro and transgenic approaches. Horticulturae. 9: Article 58. doi:10.3390/horticulturae9010058.
  • Pereira-Lorenzo S, Costa R, Anagnostakis S, Serdar U, Yamamoto T, Saito T, Ramos-Cabrer AM, Ling Q, Barreneche T, Robin C, Botta R. 2016. Interspecific hybridization of chestnut. Polyploidy and Hybridization for Crop Improvement. 15:377–408.
  • Perrella G, Bäurle I, van Zanten M. 2022. Epigenetic regulation of thermomorphogenesis and heat stress tolerance. New Phytologist. 234:1144–1160. doi:10.1111/nph.17970.
  • Petolino JF, Kumar S. 2016. Transgenic trait deployment using designed nucleases. Plant Biotechnology Journal. 14:503–509. doi:10.1111/pbi.12457.
  • Pignone D, De Paola D, Rapanà N, Janni M. 2015. Single seed descent: a tool to exploit durum wheat (Triticum durum desf.) genetic resources. Genetic Resources and Crop Evolution. 62:1029–1035. doi:10.1007/s10722-014-0206-2.
  • Planchais S, Glab N, Inzé D, Bergonioux C. 2000. Chemical inhibitors: a tool for plant cell cycle studies. FEBS Letters. 476:78–83. doi:10.1016/S0014-5793(00)01675-6.
  • Poehlman JM. 2013. Breeding field crops. The Netherlands: Springer Netherlands.
  • Pramanik KA, Sahoo JP, Mohapatra PP, Acharya LK, Jena CH. 2021. Insights into the embryo rescue-a modern in-vitro crop improvement approach in horticulture. Plant Cell Biotechnology Molecular Biology. 22:20–33.
  • Prigge V, Melchinger AE. 2012. Production of haploids and doubled haploids in maize. In: Loyola-Vargas V, Ochoa-Alejo N, editors. Plant cell culture protocols. Totowa, NJ: Humana Press; p. 161–172. doi:10.1007/978-1-61779-818-4_13
  • Provine WB. 1971. The origins of theoretical population genetics. Chicago: The University of Chicago Press.
  • Puchta H. 2016. Using CRISPR/Cas in three dimensions: towards synthetic plant genomes, transcriptomes and epigenomes. The Plant Journal. 87:5–15. doi:10.1111/tpj.13100.
  • Qaim M. 2015. Global impact of GM crops, 1996–2015. In: Clive James C, Teng P, Arujanan M, Aldemita RR, Flavell RB, Brookes G, Qaim M, editors. Invitational essays to celebrate the 20th anniversary of the commercialization of biotech crops (1996 to 2015): progress and promise. ISAAA brief 51. Ithaca, NY: ISAAA; p. 54–56.
  • Qaim M. 2016. Genetically modified crops and agricultural development. New York: Palgrave Macmillan. https://books.google.co.nz/books?id = s-kYDAAAQBAJ.
  • Qaim M. 2020. Role of new plant breeding technologies for food security and sustainable agricultural development. Applied Economic Perspectives and Policy. 42:129–150. doi:10.1002/aepp.13044.
  • Que Q, Chilton MD, de Fontes CM, He C, Nuccio M, Zhu T, Wu Y, Chen JS, Shi L. 2010. Trait stacking in transgenic crops: challenges and opportunities. GM Crops. 1:220–229. doi:10.4161/gmcr.1.4.13439.
  • Quemada H. 2022. Lessons learned from the introduction of genetically engineered crops: relevance to gene drive deployment in Africa. Transgenic Research. 31:285–311. doi:10.1007/s11248-022-00300-2.
  • Raju SKK, Shao M-R, Sanchez R, Xu Y-Z, Sandhu A, Graef G, Mackenzie S. 2018. An epigenetic breeding system in soybean for increased yield and stability. Plant Biotechnology Journal. 16:1836–1847. doi:10.1111/pbi.12919.
  • Ramanna MS, Jacobsen E. 2003. Relevance of sexual polyploidization for crop improvement: a review. Euphytica. 133:3–8. doi:10.1023/A:1025600824483.
  • Ramchiary N, Park S, Lim YP. 2011. Classical breeding and genetic analysis of vegetable brassicas. In: Sadowski J, Kole C, editors. Genetics, genomics and breeding of vegetable brassicas. Chapter 1. Boca Raton (FL): CRC Press; p. 34–61.
  • Randhawa HS, Bona L, Graf RJ. 2015. Triticale breeding—progress and prospect. In: Eudes F, editor. Triticale. Cham: Springer; p. 15–32. doi:10.1007/978-3-319-22551-7_2
  • Ranney TG. 2006. Polyploidy: from evolution to new plant development. Proceedings International Plant Propagators’ Society. 56:137–142.
  • Ren C, Liu X, Zhang Z, Wang Y, Duan W, Li S, Liang Z. 2016. CRISPR/Cas9-mediated efficient targeted mutagenesis in chardonnay (Vitis vinifera L.). Scientific Reports. 6: Article 32289. doi:10.1038/srep32289.
  • Rieseberg LH, Church SA, Morjan CL. 2004. Integration of populations and differentiation of species. New Phytologist. 161:59–69. doi:10.1046/j.1469-8137.2003.00933.x.
  • Rio Declaration. 1992. Rio declaration on environment and development. The United Nations Conference on Environment and Development, Rio de Janeiro from 3 to 14 June 1992; [accessed 1 January 2023]. https://www.cbd.int/doc/ref/rio-declaration.shtml.
  • Rogers C, Oldroyd GED. 2014. Synthetic biology approaches to engineering the nitrogen symbiosis in cereals. Journal of Experimental Botany. 65:1939–1946. doi:10.1093/jxb/eru098.
  • Rommens CM. 2004. All-native DNA transformation: a new approach to plant genetic engineering. Trends in Plant Science. 9:457–464.
  • Rommens CM, Haring MA, Swords K, Davies HV. 2007. The intragenic approach as a new extension to traditional plant breeding. Trends in Plant Science. 12:397–403. doi:10.1016/j.tplants.2007.08.001.
  • Rommens CM, Humara JM, Ye J, Yan H, Richael C, Zhang L, Perry R, Swords K. 2004. Crop improvement through modification of the plant’s own genome. Plant Physiology. 135:421–431.
  • Royal Society Te Apārangi. 2019. Gene editing: scenarios in the primary industries; [accessed 12 January 2023]. https://www.royalsociety.org.nz/assets/Uploads/Gene-Editing-Scenarios-Primary-industries-DIGITAL.pdf.
  • Rozas P, Kessi-Pérez EI, Martínez C. 2022. Genetically modified organisms: adapting regulatory frameworks for evolving genome editing technologies. Biological Research. 55: Article 31. doi:10.1186/s40659-022-00399-x.
  • Rozin P. 2005. The meaning of “natural”. Psychological Science. 16:652–658.
  • Rozin P. 2006. Naturalness judgments by lay Americans: process dominates content in judgments of food or water acceptability and naturalness. Judgment and Decision Making. 1:91–97.
  • Ruse M. 1975. Charles Darwin’s theory of evolution: an analysis. Journal of the History of Biology. 8:219–241. http://www.jstor.org/stable/4330635.
  • Sadowski J, Kole C. 2016. Genetics, genomics and breeding of vegetable brassicas. Boca Raton: CRC Press. 450. doi:10.1201/b10880
  • Salamini F, Ozkan H, Brandolini A, Schäfer-Pregl R, Martin W. 2002. Genetics and geography of wild cereal domestication in the near east. Nature Reviews Genetics. 3:429–441. doi:10.1038/nrg817.
  • Sánchez-León S, Gil-Humanes J, Ozuna CV, Giménez MJ, Sousa C, Voytas DF, Barro F. 2018. Low-gluten, nontransgenic wheat engineered with CRISPR/Cas9. Plant Biotechnology Journal. 16:902–910. doi:10.1111/pbi.12837.
  • Sandhu KS, Merrick LF, Sankaran S, Zhang Z, Carter AH. 2022a. Prospectus of genomic selection and phenomics in cereal, legume and oilseed breeding programs. Frontiers in Genetics. 12: Article 829131. doi:10.3389/fgene.2021.829131.
  • Sandhu KS, Shiv A, Kaur G, Meena MR, Raja AK, Vengavasi K, Mall AK, Kumar S, Singh PK, Singh J, et al. 2022b. Integrated approach in genomic selection to accelerate genetic gain in sugarcane. Plants. 11: Article 2139. doi:10.3390/plants11162139.
  • Sandler R. 2004. An aretaic objection to agricultural biotechnology. Journal of Agricultural and Environmental Ethics. 17:301–317. doi:10.1023/B:JAGE.0000033078.05859.08.
  • Sanford JC. 1990. Biolistic plant transformation. Physiologia Plantarum. 79:206–209. doi:10.1111/j.1399-3054.1990.tb05888.x.
  • Sattler MC, Carvalho CR, Clarindo WR. 2016. The polyploidy and its key role in plant breeding. Planta. 243:281–296. doi:10.1007/s00425-015-2450-x.
  • Sauer NJ, Mozoruk J, Miller RB, Warburg ZJ, Walker KA, Beetham PR, Schöpke CR, Gocal GFW. 2016. Oligonucleotide-directed mutagenesis for precision gene editing. Plant Biotechnology Journal. 14:496–502. doi:10.1111/pbi.12496.
  • Saurabh S, Vidyarthi AS, Prasad D. 2014. RNA interference: concept to reality in crop improvement. Planta. 239:543–564. doi:10.1007/s00425-013-2019-5.
  • Sawai S, Ohyama K, Yasumoto S, Seki H, Sakuma T, Yamamoto T, Takebayashi Y, Kojima M, Sakakibara H, Aoki T, et al. 2014. Sterol side chain reductase 2 is a key enzyme in the biosynthesis of cholesterol, the common precursor of toxic steroidal glycoalkaloids in potato. The Plant Cell. 26:3763–3774. doi:10.1105/tpc.114.130096.
  • Schaart JG, van de Wiel CCM, Lotz LAP, Smulders MJM. 2016. Opportunities for products of new plant breeding techniques. Trends in Plant Science. 21:438–449. doi:10.1016/j.tplants.2015.11.006.
  • Schaart JG, Visser RG. 2009. Novel plant breeding techniques. Consequences of new genetic modification-based plant breeding techniques in comparison to conventional plant breeding. Wageningen (NT): Wageningen University and Research Center; COGEM Research Report Number 2009-02. The Netherlands Commission on Genetic Modification; [accessed 6 January 2023]. https://edepot.wur.nl/137009.
  • Schmidt CW. 2005. Genetically modified foods: breeding uncertainty. Environmental Health Perspectives. 113:A526–A533. doi:10.1289/ehp.113-a526.
  • Schouten HJ, Krens FA, Jacobsen E. 2006. Cisgenic plants are similar to traditionally bred plants. EMBO Reports. 7:750–753. doi:10.1038/sj.embor.7400769.
  • Schuermann D, Molinier J, Fritsch O, Hohn B. 2005. The dual nature of homologous recombination in plants. TRENDS in Genetics. 21:172–181. doi:10.1016/j.tig.2005.01.002.
  • Senthil-Kumar M, Mysore KS. 2011. New dimensions for VIGS in plant functional genomics. Trends in Plant Science. 16:656–665. doi:10.1016/j.tplants.2011.08.006.
  • Séralini GE, Mesnage CE, Gress R, Defarge S, Malatesta N, Hennequin M, de Vendômois D, S J. 2014. Republished study: long-term toxicity of a roundup herbicide and a roundup-tolerant genetically modified maize. Environmental Sciences Europe. 26: Article 14. http://www.enveurope.com/content/26/1/14.
  • Shan Q, Zhang Y, Chen K, Zhang K, Gao C. 2015. Creation of fragrant rice by targeted knockout of the OsBADH2 gene using TALEN technology. Plant Biotechnology Journal. 13:791–800. doi:10.1111/pbi.12312.
  • Shen C, Yin X-C, Jiao B-Y, Li J, Jia P, Zhang X-W, Cheng X-H, Ren J-X, Lan H-D, Hou W-B, et al. 2022. Evaluation of adverse effects/events of genetically modified food consumption: a systematic review of animal and human studies. Environmental Sciences Europe. 34: Article 8. doi:10.1186/s12302-021-00578-9.
  • Shen J-X, Fu T-D, Yang G-S, Tu J-X, Ma C-Z. 2006. Prediction of heterosis using QTLs for yield traits in rapeseed (Brassica napus L.). Euphytica. 151:165–171. doi:10.1007/s10681-006-9137-0.
  • Shew AM, Nalley LL, Snell HA, Nayga RM, Dixon BL. 2018. CRISPR versus GMOs: public acceptance and valuation. Global Food Security. 19:71–80. doi:10.1016/j.gfs.2018.10.005.
  • Shi J, Gao H, Wang H, Lafitte HR, Archibald RL, Yang M, Hakimi SM, Mo H, Habben JE. 2017. ARGOS8 variants generated by CRISPR-Cas9 improve maize grain yield under field drought stress conditions. Plant Biotechnology Journal. 15:207–216. doi:10.1111/pbi.12603.
  • Shukla V, Doyon Y, Miller J, DeKelver RC, Moehle EA, Worden SE, Mitchell JC, Arnold NL, Gopalan S, Meng X, Choi VM, et al. 2009. Precise genome modification in the crop species Zea mays using zinc-finger nucleases. Nature. 459:437–441. doi:10.1038/nature07992.
  • Siegrist M. 2008. Factors influencing public acceptance of innovative food technologies and products. Trends in Food Science & Technology. 19:603–608. doi:10.1016/j.tifs.2008.01.017.
  • Singh RK, Prasad M. 2022. Biomolecules of mushroom: a recipe of human wellness. Critical Reviews in Biotechnology. 42:913–930. doi:10.1080/07388551.2021.1964431.
  • Sjöberg L. 2000. Perceived risk and tampering with nature. Journal of Risk Research. 3:353–367. doi:10.1080/13669870050132568.
  • Smirnova OG, Ibragimova SS, Kochetov AV. 2012. Simple database to select promoters for plant transgenesis. Transgenic Research. 21:429–437. doi:10.1007/s11248-011-9538-2.
  • Smyth SJ, Lassoued R. 2019. Agriculture R&D implications of the CJEU’s gene-specific mutagenesis ruling. Trends in Biotechnology. 37:337–340. doi:10.1016/j.tibtech.2018.09.004.
  • Snape J, Riggs T. 1975. Genetical consequences of single seed descent in the breeding of self-pollinating crops. Heredity. 35:211–219. doi:10.1038/hdy.1975.85.
  • Song G-q, Sink KC, Walworth AE, Cook MA, Allison RF, Lang GA. 2013. Engineering cherry rootstocks with resistance to Prunus necrotic ring spot virus through RNAi-mediated silencing. Plant Biotechnology Journal. 11:702–708. doi:10.1111/pbi.12060.
  • Songstad DD, Petolino JF, Voytas DF, Reichert NA. 2017. Genome editing of plants. Critical Reviews in Plant Sciences. 36:1–23. doi:10.1080/07352689.2017.1281663.
  • Sopory S, Munshi M. 1996. Anther culture. In: Jain SM, Sopory SK, Veilleux RE, editors. In vitro haploid production in higher plants, vol. 23. Dordrecht: Kluwer; p. 145–176. doi:10.1007/978-94-017-1860-8_9
  • Sprague GF. 1946. The experimental basis for hybrid maize. Biological Reviews. 21:101–120. doi:10.1111/j.1469-185X.1946.tb00317.x.
  • Sprink T, Wilhelm R, Hartung F. 2022. Genome editing around the globe: an update on policies and perceptions. Plant Physiology. 190:1579–1587. doi:10.1093/plphys/kiac359.
  • Stenseth NC, Andersson L, Hoekstra HE. 2022. Gregor Johann Mendel and the development of modern evolutionary biology. Proceedings of the National Academy of Sciences. 119(30):Article 119. doi:10.1073/pnas.2201327119.
  • Stoskopf NC, Tomes DT, Christie BR. 2019. Plant breeding: theory and practice. London and New York: Routledge Taylor and Francis Group.
  • Stupar RM, Gardiner JM, Oldre AG, Haun WJ, Chandler VL, Spinger NM. 2008. Gene expression analyses in maize inbreds and hybrids with varying levels of heterosis. BMC Plant Biology. 8: Article 33. doi:10.1186/1471-2229-8-33.
  • Sun Y, Jiao G, Liu Z, Zhang X, Li J, Guo X, Du W, Du J, Francis F, Zhao Y, Xia L. 2017. Generation of high-amylose rice through CRISPR/Cas9-mediated targeted mutagenesis of starch branching enzymes. Frontiers in Plant Science. 8: Article 298. doi:10.3389/fpls.2017.00298.
  • Svitashev S, Young JK, Schwartz C, Gao H, Falco SC, Cigan AM. 2015. Targeted mutagenesis, precise gene editing, and site-specific gene insertion in maize using Cas9 and guide RNA. Plant Physiology. 169:931–945. doi:10.1104/pp.15.00793.
  • Szymajda M, Napiórkowska B, Korbin M, Żurawicz E. 2015. Studies on the interspecific crossing compatibility among three Prunus species and their hybrids. Horticultural Science. 42(2):70–82. doi:10.17221/273/2014-HORTSCI.
  • Tagliabue G. 2016. The meaningless pseudo-category of “GMOs”. EMBO Reports. 17:10–13. doi:10.15252/embr.201541385.
  • Tait J. 2001. More faust than frankenstein: The European debate about the precautionary principle and risk regulation for genetically modified crops. Journal of Risk Research. 4:175–189. doi:10.1080/13669870010027640.
  • Tak S, Kaushik RA, Nath A. 2016. Studies on heterosis in interspecific hybrids of cucumis. The Bioscan. 11:3155–3159.
  • The Royal Society. 2016. Is it safe to eat GM crops?; [accessed 1 January 2023]. https://royalsociety.org/topics-policy/projects/gm-plants/is-it-safe-to-eat-gm-crops/.
  • Tirado R, Johnston P. 2010. Food security: GM crops threaten biodiversity. Science. 328:170–171.
  • Tourism NZ. 2009. Pure as celebrating 10 years of 100% Pure New Zealand; [accessed 27 April 2022]. https://www.tourismnewzealand.com/media/1544/pure-as-celebrating-10-years-of-100-pure-new-zealand.pdf.
  • Townsend J, Wright D, Winfrey R, Fu F, Maeder ML, Joung JK, Voytas DF. 2009. High-frequency modification of plant genes using engineered zinc-finger nucleases. Nature. 459:442–445. doi:10.1038/nature07845.
  • Tozer KN, Carswell K, Griffiths WM, Crush JR, Cameron CA, Chapman DF, Popay A, King W. 2017. Growth responses of diploid and tetraploid perennial ryegrass (Lolium perenne) to soil-moisture deficit, defoliation and a root-feeding invertebrate. Crop and Pasture Science. 68:632–642. doi:10.1071/CP17154.
  • Trethowan RM, Reynolds MP, Ortiz-Monasterio JI, Ortiz R. 2007. The genetic basis of the green revolution in wheat production. Plant Breeding Reviews. 28:39–58.
  • Tripathi JN, Ntui VO, Ron M, Muiruri SK, Britt A, Tripathi L. 2019. CRISPR/cas9 editing of endogenous banana streak virus in the B genome of Musa spp. overcomes a major challenge in banana breeding. Communications Biology. 2: Article 46. doi:10.1038/s42003-019-0288-7.
  • Turnbull C, Lillemo M, Hvoslef-Eide TA. 2021. Global regulation of genetically modified crops amid the gene edited crop boom – A review. Frontiers in Plant Science. 12: Article 630396. doi:10.3389/fpls.2021.630396.
  • United States Patent. 2009. Tomato plants. Patent No.: US 7,612.261 B2; [accessed 25 February 2023]. https://patentimages.storage.googleapis.com/17/d5/0d/dcba5fd7f54abf/US7612261.pdf.
  • USDA. 2022. Agricultural Biotechnology Glossary; [accessed 27 December 2022]. https://www.usda.gov/topics/biotechnology/biotechnology-glossary#:~:text = Genetic%20engineering%3A%20Manipulation%20of%20an,to%20as%20recombinant%20DNA%20techniques.
  • Vanblaere T, Szankowski I, Schaart J, Schouten H, Flachowsky H, Broggini GAL, Gessler C. 2011. The development of a cisgenic apple plant. Journal of Biotechnology. 154:304–311.
  • Vanderzwaag DL, Fuller SD, Myers RA. 2002. Canada and the precautionary principle/approach in ocean and coastal management: wading and wandering in tricky currents. Ottawa Law Review. 34:117–158.
  • van Heerwaarden J, Doebley J, Briggs WH, Glaubitz JC, Goodman MM, de JS, Gonzalez J, RossIbarra J. 2011. Genetic signals of origin, spread, and introgression in a large sample of maize landraces. Proceedings of the National Academy of Sciences. 108:1088–1092. doi:10.1073/pnas.1013011108.
  • Vanlay M, Samnang S, Jung H-J, Choe P, Kang KK, Nou I-S. 2022. Interspecific and intraspecific hybrid rootstocks to improve horticultural traits and soil-borne disease resistance in tomato. Genes. 13: Article 1468. doi:10.3390/genes13081468.
  • Van Tuyl JM, Lim K-B. 2003. Interspecific hybridisation and polyploidisation as tools in ornamental plant breeding. Acta Horticulturae. 612:13–22. doi:10.17660/ActaHortic.2003.612.1.
  • Vega Rodríguez A, Rodríguez-Oramas C, Sanjuán Velázquez E, Hardisson de la Torre A, Rubio Armendáriz C, Carrascosa Iruzubieta C. 2022. Myths and realities about genetically modified food: a risk-benefit analysis. Applied Sciences. 12: Article 2861. doi:10.3390/app12062861.
  • Veillet F, Perrot L, Chauvin L, Kermarrec M-P, Guyon-Debast A, Chauvin J-E, Nogué F, Mazier M. 2019. Transgene-free genome editing in tomato and potato plants using Agrobacterium-mediated delivery of a CRISPR/Cas9 cytidine base editor. International Journal of Molecular Sciences. 20: Article 402. doi:10.3390/ijms20020402.
  • Verma N, Giri SK, Singh G, Gill R, Kumar A. 2022. Epigenetic regulation of heat and cold stress responses in crop plants. Plant Gene. 17: Article 100351. doi:10.1016/j.plgene.2022.100351.
  • Vida G, Gál M, Uhrin A, Veisz O, Syed MH, Flavell AJ, Wang Z, Bedő Z. 2009. Molecular markers for the identification of resistance genes and marker-assisted selection in breeding wheat for leaf rust resistance. Euphytica. 170:67–76. doi:10.1007/s10681-009-9945-0.
  • Virmani SS, Aquino RC, Khush GS. 1982. Heterosis breeding in rice (Oryza sativa L.). Theoretical and Applied Genetics. 63:373–380. doi:10.1007/BF00303911.
  • Waltz E. 2015. USDA approves next-generation GM potato. Nature Biotechnology. 33:12–13. doi:10.1038/nbt0115-12.
  • Waltz E. 2016. CRISPR-edited crops free to enter market, skip regulation. Nature Biotechnology. 34: Article 582. doi:10.1038/nbt0616-582.
  • Waltz E. 2018. With a free pass, CRISPR-edited plants reach market in record time. Nature Biotechnology. 36:6–7. doi:10.1038/nbt0118-6b.
  • Waltz E. 2019. Appetite grows for biotech foods with health benefits. Nature Biotechnology. 37:573–575. doi:10.1038/d41587-019-00012-9.
  • Waltz E. 2022. GABA-enriched tomato is first CRISPR-edited food to enter market. Nature Biotechnology. 40:9–11. doi:https://doi.org/10.1038/d41587-021-00026-2.
  • Wang F, Wang C, Liu P, Lei C, Hao W, Gao Y, Liu Y-G, Zhao K. 2016. Enhanced rice blast resistance by CRISPR/Cas9-targeted mutagenesis of the ERF transcription factor gene OsERF922. PLoS One. 11: Article e0154027. doi:10.1371/journal.pone.0154027.
  • Wang Y, Cheng X, Shan Q, Zhang Y, Liu J, Gao C, Qiu J-L. 2014. Simultaneous editing of three homoeoalleles in hexaploid bread wheat confers heritable resistance to powdery mildew. Nature Biotechnology. 32:947–951. doi:10.1038/nbt.2969.
  • Wehrs M, Tanjore D, Eng T, Lievense J, Pray TR, Mukhopadhyay A. 2019. Engineering robust production microbes for large-scale cultivation. Trends in Microbiology. 27:524–537. doi:10.1016/j.tim.2019.01.006.
  • Weinhold B. 2006. Epigenetics: The science of change. Environmental Health Perspectives. 114: Article 3. doi:10.1289/ehp.114-a160.
  • Wesseler J, Bonanno A, Drabik D, Materia VC, Malaguti L, Meyer M, Venus TJ. 2015. Overview of the agricultural input sector in the EU. Directorate-General for Internal Policies. Policy Department B: Structural and Cohesion Policies. Agriculture and Rural Development. doi:10.2861/49815.
  • West JD, Bergstrom CT. 2021. Misinformation in and about science. Proceedings of the National Academy of Sciences 118: Article e1912444117. https://www.pnas.org/doi/abs/10.1073pnas.1912444117.
  • West-Eberhard MJ. 1989. Phenotypic plasticity and the origins of diversity. Annual Review of Ecology and Systematics. 20:249–278. https://www.jstor.org/stable/2097092.
  • Wheatley MS, Yang Y. 2021. Versatile applications of the CRISPR/Cas toolkit in plant pathology and disease management. Phytopathology. 111:1080–1090. doi:10.1094/PHYTO-08-20-0322-IA.
  • Whelan AI, Lema MA. 2015. Regulatory framework for gene editing and other new breeding techniques (NBTs) in Argentina. GM Crops & Food. 6:253–265. doi:10.1080/21645698.2015.1114698.
  • Wiener JB. 2001. Precaution in a multi-risk world. Duke Law School public law and legal theory working paper series, Working Paper No. 23. Durham, North Carolina; [accessed 26 February 2023]. https://papers.ssrn.com/sol3/papers.cfm?abstract_id = 293859.
  • Wilkins J. 2007. The dimensions, modes and definitions of species and speciation. Biology & Philosophy. 22:247–266. doi:10.1007/s10539-006-9043-9.
  • Wolter F, Schindele P, Puchta H. 2019. Plant breeding at the speed of light: the power of CRISPR/Cas to generate directed genetic diversity at multiple sites. BMC Plant Biology. 19: Article 176. doi:10.1186/s12870-019-1775-1.
  • Wu J, Chen C, Xian G, Liu D, Lin L, Yin S, Sun Q, Fang Y, Zhang H, Wang Y. 2020. Engineering herbicide-resistant oilseed rape by CRISPR/Cas9-mediated cytosine base-editing. Plant Biotechnology Journal. 18:1857–1859. doi:10.1111/pbi.13368.
  • Yamagishi H, Bhat SR. 2014. Cytoplasmic male sterility in Brassicaceae crops. Breeding Science. 64:38–47. doi:10.1270/jsbbs.64.38.
  • Yang J, Zimmerly S, Perlman PS, Lambowitz AM. 1996. Efficient integration of an intron RNA into double-stranded DNA by reverse splicing. Nature. 381:332–335. doi:10.1038/381332a0.
  • Yilmaz S, Nyerges A, van der Oost J, Church GM, Claassens NJ. 2022. Towards next-generation cell factories by rational genome-scale engineering. Nature Catalysis. 5:751–765. doi:10.1038/s41929-022-00836-w.
  • Yin X, Biswal AK, Dionora J, Perdigon KM, Balahadia CP, Mazumdar S, Chater C, Lin HC, Coe RA, Kretzschmar T, Gray JE. 2017. CRISPR-Cas9 and CRISPR-Cpf1 mediated targeting of a stomatal developmental gene EPFL9 in rice. Plant Cell Reports. 36:745–757. doi:10.1007/s00299-017-2118-z.
  • Young ND. 1999. Cautiously optimistic vision for marker-assisted breeding. Molecular Breeding. 5:505–510. doi:10.1023/A:1009684409326.
  • Younis A, Hwang YJ, Lim KB. 2014. Exploitation of induced 2n-gametes for plant breeding. Plant Cell Reports. 33:215–223. doi:10.1007/s00299-013-1534-y.
  • Zachos FE. 2016. Species concepts in biology: historical development, theoretical foundations and practical relevance. Springer International Publishing Switzerland. 227. doi: 10.1007/978-3-319-44966-1
  • Zafar SA, Zaidi SSEA, Gaba Y, Singla-Pareek SL, Dhankher OP, Li X, Mansoor S, Pareek A. 2020. Engineering abiotic stress tolerance via CRISPR/ Cas-mediated genome editing. Journal of Experimental Botany. 71:470–479. doi:10.1093/jxb/erz476.
  • Zaidi SS, Vanderschuren H, Qaim M, Mahfouz MM, Kohli A, Mansoor S, Tester M. 2019. New plant breeding technologies for food security. Science. 363:1390–1391. https://www.science.org/doi/abs/10.1126science.aav6316.
  • Zanoni U, Dudley JW. 1989. Comparison of different methods of identifying inbreds useful for improving elite maize hybrids. Crop Science. 29:577–582. doi:10.2135/cropsci1989.0011183X002900030005x.
  • Zetsche B, Gootenberg JS, Abudayyeh OO, Slaymaker IM, Makarova KS, Essletzbichler P, Volz SE, Joung J, Van Der Oost J, Regev A, Koonin EV. 2015. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell. 163:759–771. doi:10.1016/j.cell.2015.09.038.
  • Zeven AC. 1998. Landraces: a review of definitions and classifications. Euphytica. 104:127–139.
  • Zhang H, Zhang J, Wei P, Zhang B, Gou F, Feng Z, Mao Y, Yang L, Zhang H, Xu N, Zhu J-K. 2014. The CRISPR/Cas9 system produces specific and homozygous targeted gene editing in rice in one generation. Plant Biotechnology Journal. 12:797–807. doi:10.1111/pbi.12200.
  • Zhu T, Mettenburg K, Peterson DJ, Tagliani L, Baszczynski CL. 2000. Engineering herbicide-resistant maize using chimeric RNA/DNA oligonucleotides. Nature Biotechnology. 18:555–558. doi:10.1038/75435.
  • Zhu X, Liu X, Liu T, Wang Y, Ahmed N, Li Z, Jiang H. 2021. Synthetic biology of plant natural products: from pathway elucidation to engineered biosynthesis in plant cells. Plant Communications. 2: Article 100229. doi:10.1016/j.xplc.2021.100229
  • Zsögön A, Čermák T, Naves E, Notini MM, Edel KH, Weinl S, Freschi L, Voytas DF, Kudla J, Peres LEP. 2018. De novo domestication of wild tomato using genome editing. Nature Biotechnology. 36:1211–1216. doi:10.1038/nbt.4272.