Future Foods: Xin Guan, Qingzi Lei, Qiyang Yan, Xueliang Li, Jingwen Zhou, Guocheng Du, Jian Chen

You might also like

You are on page 1of 10

Future Foods 3 (2021) 100032

Contents lists available at ScienceDirect

Future Foods
journal homepage: www.elsevier.com/locate/fufo

Trends and ideas in technology, regulation and public acceptance of


cultured meat
Xin Guan a,b,c,d, Qingzi Lei a,b,c, Qiyang Yan a,b, Xueliang Li a,b, Jingwen Zhou a,b,c,d,
Guocheng Du a,b,∗, Jian Chen a,b,c,d,∗
a
Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
b
Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
c
National Engineering Laboratory for Cereal Fermentation Technology (NELCF), Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
d
Jiangsu Provisional Research Center for Bioactive Product Processing Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China

a r t i c l e i n f o a b s t r a c t

Keywords: As future foods, cultured meat is produced by culturing animal cells ex vivo rather than raising and slaughtering
Cultured meat animals. It is a promising way to address concerns about resource consumption, environmental pollution, public
Muscle tissue healthy that associated with conventional livestock production. In the past two years, dozens of cultured meat-
Large-scale manufacturing
related start-ups have been founded and millions of dollars have been raised, demonstrating the high business
Cost reduction
enthusiasm, broad market prospects and high profitability expected. However, currently many startups are yet
Animal component-free medium
Capital investment determining, optimizing steps of the process or trying to scale-up, so it is still a very long way before cultured meat
becomes comparable to the conventional meat. Here, we review the industry status of cultured meat around the
world from the geographical distribution, potential products, and funding landscape and, identify the advanced
technologies in large-scale manufacturing of cultured meat covering aspects from culture medium optimization,
bioreactor designation, scaffold fabrication and cost reduction. Moreover, we discuss the challenges related to
policy formulation and public acceptance of cultured meat.

1. Introduction (Cann, 2018; Jiang et al., 2020; Lee et al., 2020). Cultured meat is also
called lab-grown meat, which is produced by ex vivo culture of animal
The first challenge for humanity today is feeding 10 billion people cells by utilizing the operation techniques in the field of cell biology,
by 2050 (UNPD, 2019). As one of the most important food resources tissue and food engineering (Post, 2012). Generally, stem cells which
and nutrients for the clear majority of people, meat has already been have the potential of self-renewal and multiple differentiation are iso-
consumed over 300 million tons in 2014 and a 76% of growth by 2050 lated from an animal biopsy, and then expanded and differentiated ex
is predicted (Alexandratos and Bruinsma, 2012). The traditional way vivo to generate muscle fibers, fat or other cell types that make up muscle
of meat production is to breed, raise and slaughter whole animals, tissue. Subsequently, these cells are collected and assembled to form ed-
which takes a relatively long period, consumes large water and land ible meat end-products after food processing such as molding, coloring
resources, uses a large amount of fuel energy, and causes severe envi- and seasoning (Arshad et al., 2017; Datar and Betti, 2010; Kadim et al.,
ronment pollution. (FAO, 2006; Gerber et al., 2013; Herrero et al., 2011; 2015). Therefore, cultured meat technology intends to directly produce
Hoekstra and Mekonnen, 2012). In addition, the epidemics of African meat by ex vivo cell culture and no extensive animal raising and slaugh-
swine fever, avian flu and other animal diseases have brought more un- tering are required, making it a novel and promising meat production
certain factor to the conventional livestock production (Dixon et al., technology. Much more importantly, cultured meat technology would
2020; Normile, 2008; Taylor et al., 2019). Therefore, it is urgent to de- greatly shorten the meat production cycles, reduce the consumption of
velop high-efficient, eco-friendly and sustainable meat production strat- lands, water, and energy, and curb greenhouse gas emissions (Tuomisto
egy. and de Mattos, 2011).
As one of the “Top 10 Emerging Technologies of 2018″ published As early as the 1930s, British Minister Winston Churchill proposed
by the World Economic Forum, cultured meat has been considered as a the idea of eating meat by culturing individual tissues in medium rather
promising solution to mitigate concerns related to livestock agriculture than raising the entire animals (Churchill, 1932). This can be regarded


Corresponding authors at: Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China.
E-mail addresses: gcdu@jiangnan.edu.cn (G. Du), jchen@jiangnan.edu.cn (J. Chen).

https://doi.org/10.1016/j.fufo.2021.100032
Received 28 December 2020; Received in revised form 16 February 2021; Accepted 24 March 2021
2666-8335/© 2021 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/)
X. Guan, Q. Lei, Q. Yan et al. Future Foods 3 (2021) 100032

as the rudiment of cultured meat, but in terms of the science and tech- 3. Main production process of cultured meat
nology at that time, cultured meat was just a fantasy and vision of the
future. It was at the beginning of the 21st century that research on The production process of cultured meat is a set of tissue and food
cultured meat began and burgeoned. Since 2002, National Aeronautics engineering processes used to create a food product, which can be briefly
and Space Administration (NASA) funded research to produce cultured divided into four key steps: (1) acquisition of seed cells, (2) large-scale
turkey and golden fish meat, creating the first edible cultured turkey expansion of seed cells, (3) induced differentiation of seed cells into
strips (Edelman et al., 2005) and fish fillet (Benjaminson et al., 2002). In myofibers, adipocytes, or other mature cell types in muscle tissues, and
2013, professor Mark Post launched the world’s first cultured beef ham- (4) assembly and food processing of all cultured cells into meat products
burger after years of research, which is made up of more than 10,000 (Fig. 4).
muscle fibers and costs nearly $330,000 (Post, 2014). Although the high Step 1: Seed cells are animal cells that can vigorously proliferate
cost, the success of cultured meat technology triggered great attention and efficiently generate to myoblasts, adipocytes, and other cell types
of people from all walks of life, including scientists, entrepreneurs and that compose muscle tissue, usually referring to stem cells. Embryonic
investors. Since then, numerous companies have been founded to de- stem cells, induced pluripotent stem cells, muscle stem cells, and mes-
velop cultured meat products and have get access to capital, promoting enchymal stem cells are potential types of seed cells (Díaz-Flores et al.,
the explosion of cultured meat industry. 2006; Genovese et al., 2017; Kadim et al., 2015; Williams et al., 2012).
Because of the immature production process and high production In addition, some adult cells such as fibroblasts can also be operated to
cost, cultured meat is currently in its infancy, and researchers are fran- form myoblasts and lipoblasts by transdifferentiation or dedifferentia-
tically working on technological improvement to speed up the indus- tion (Boularaoui et al., 2018; Oki et al., 2008). In general, these cells can
trialization and commercialization process of cultured meat. This re- be efficiently obtained from a biopsy of living animal tissue after isola-
view summarizes current cultured meat-related star-ups and discusses tion via enzymatic digestion and mechanical disruption and purification
the challenges for the commercialization of cultured meat at technol- via flow sorting with specific surface markers (Ding et al., 2017). Al-
ogy, regulation and customer levels. Meanwhile, further research focus though various cell sources can be used to produce cultured meat, each
and potential strategies for the production and marketing of cultured cell type needs specific ex vivo expansion and differentiation strategy
meat are also put forward accordingly. according to their growth and development characteristics (Fish et al.,
2020; Stephens et al., 2018; Zhang et al., 2020).
Step 2: Once seed cells are collected, they need to be proliferated to
2. The blooming cultured meat industry achieve large cell numbers. The laboratory culture scale with flasks or
dishes is far from satisfying the market demand, so it is necessary to de-
Since the first cultured beef hamburger was produced in 2013, velop a large-scale bioreactor system (Post et al., 2020). Moreover, the
dozens of companies have get into the cultured meat industry and var- process should use economical serum-free medium, and monitor various
ious product species such as chicken, beef, pork, and seafood are in parameters such as pH, dissolved O2 and CO2 , concentration of impor-
the development process. By the end of 2020, approximately 60 early- tant nutrients and metabolic waste online (Allan et al., 2019). At the
stage companies have been set up in the worldwide, focusing on cul- same time, to efficiently utilize resources and keep low production cost,
tured meat end-products, raw materials or equipment along the value it is important to conduct the medium recycling with automatic removal
chain, and more than half of them launched within the past 2 years of toxic wastes and replenishment of nutrients based on the monitoring
(Choudhury et al., 2020; GFI, 2020). The geographical distribution feedback.
of these companies is in 19 countries across five continents, 37% in Step 3: When the desired number of cells is achieved, cells are then
North America, 25% in Asia, and 21% in Europe (Fig. 1). From 2016 induced to differentiate into myotubes, adipocytes, or other mature cell
to 2020, the total amount of capital invested in cultured meat compa- types in muscle tissues. The maturity level of generated final cells is
nies reaches approximately $460 million with over 3 quarters-more than a key evaluation index in this step because feature structure and nu-
$350 million-closing in 2019 and 2020 (Fig. 2A). Among them, Memphis trient content including proteins, fatty acids, and vitamins are signifi-
Meat has raised approximately $200 million to achieve the leader posi- cantly affected by cell maturity (Liu, 2019). It is worth noting that al-
tion and Mosa Meat is the second with received over $85 million. These though muscle stem cells are generally considered to have strong myo-
two companies account for more than half of total capital in the cultured genic differentiation potential, the diameter, length and protein content
meat industry (Fig. 2B). It is worth noting that nine companies have of ex vivo formed myofibers vary greatly based on the culture condi-
raised their Series A/B fundraising rounds in 2020, indicating growing tion and might be much lower than real muscle fibers (Braga et al.,
mainstream investor interest and gradually mature industry stage. 2017; Lamarche et al., 2021; Park et al., 2016). Therefore, it is impor-
In terms of the product interest, 28% of cultured meat companies tant to optimize the differentiation condition and increase the maturity
focus on cultured beef and pork, 12% of companies are interested in of differentiated cells based on the mechanism of in vivo muscle tissue
seafood, 10% of companies are interested in poultry, and 28% of com- development.
panies do not focus on final products but raw materials or equipment in Step 4: As the last step of cultured meat production, the obtained
the production process (Fig. 3). Interestingly, potential product choices mature cells are collected and processed including molding, color-
by companies show obvious regional differences, greatly influenced by ing, and seasoning, ultimately forming the cultured meat end-product
local dietary habits and customs. In specific, cultured fish maw will be (Zhao et al., 2019). Because conventional cell culture approach can only
launched by a Hong Kong company because in southern China fish maw form a two-dimensional (2D) thin cell layer, assembly of myofibers,
is often consumed in soup with credited properties of beautifying skin adipocytes, and maybe connective tissue cells are essential to form a
and anti-aging (Siu, 2019). Three companies in Europe and Japan intend piece of structured and marbleized meat (Stephens et al., 2018). More-
to produce cultured foie gras due to its unique position in their food cul- over, the molding procedure can be merged into Step 3 where various
ture (Southey, 2020; Webber, 2019). Likewise, cultured kangaroo will cell types are co-cultured in a biomimetic three-dimensional (3D) en-
be offered by an Australian company (Upshall, 2021). In addition, plenty vironment provided by the scaffold or hydrogel (Tuomisto, 2019). Be-
of business to business companies emerge to embrace the whole cul- sides, advances in 3D bio-printing technology makes it promising for
tured meat value chain, including low-cost cell culture medium, biore- creating muscle tissue with relative-large size and complex compos-
actor, scaffolding materials, and cell lines. So far there are at least three ite constructs through elaborate arrangement of cell-laden hydrogels
companies announcing to produce and sell cultured meat products in (Kang et al., 2016). At last, the final product can be achieved after food
2021, and more products will come to market in the next five years processing such as supplementation of heme proteins and seasoning sub-
(Fusaro, 2021; Lucas, 2020; Webber, 2019). stances.

2
X. Guan, Q. Lei, Q. Yan et al. Future Foods 3 (2021) 100032

Fig. 1. Geographic distribution of cultured meat companies. Fifty-eight companies in the field of cultured meat/fish are included, focusing on cultured meat end-
products, raw materials, or equipment along the value chain. More information on these companies is available in GFI’s company database (GFI, 2021).

4. Technical challenges for large-scale production of cultured ensure maximum safety, sustainability, controllability and accuracy of
meat cell culture process.
To date, some serum-free additives and medium are commercially
Although creating a cellular biomass can be achieved using existing available, such as the B27TM (Thermo Scientific) and XerumFreeTM
technology regardless of scale and cost, the product cannot be regarded (TNC Bio) additives and Essential 8TM (Life technologies) and
as equivalent to a piece of meat if not providing similar texture, flavor mTeSR1TM (Stemcell Technologies) medium. They have been demon-
and nutrition content. Therefore, currently the cultured meat industry is strated to work well to support the ex vivo growth and stemness mainte-
still in its infancy, indicating that each production procedure needs to be nance of various cell types such as neural stem cells, hematopoietic stem
optimized and many technology challenges need to be addressed before cells, mesenchymal stem cells (Hoang et al., 2020; Solis-Castro et al.,
cultured meat products are ready into the ordinary people’s table. 2020; Wilkinson et al., 2019). Florini and Roberts (1979) tested a serial
of hormones, glycoproteins, and growth factors and showed that the pro-
liferation effect of myoblasts in F12 basal medium supplemented with fe-
4.1. Animal component-free medium for cell culture tal globulin, insulin, and dexamethasone is comparable to the 10% horse
serum medium, but fetal globulin still derives from the FBS. The research
As a cell culture-based products, development of optimal culture group of Dr. Post compared the effects of 3 commercial serum-free me-
medium is one of the top priorities for cultured meat (Mizukami and dia with addition of 2 serum-replacements and several growth factors
Swiech, 2018). Specifically, the culture medium must not only promote on stimulating the cell proliferation of bovine myoblasts, and achieved
the efficient proliferation and differentiation of cells, but issues such as exponential cell expansion during 6 days of culture, albeit not to the
cost and food safety should also be taken into account. extent of serum-containing medium (Kolkmann et al., 2020). To date,
The first challenge is to remove the fetal bovine serum (FBS) and several cultured meat companies have publicly stated that they have
other animal-derived components from the culture medium. FBS is achieved the removal of animal serum in the production process, but no
an important and universal growth-promoting supplement in animal further details were disclosed (Fusaro, 2021; Lucas, 2020; Mosa, 2019).
cell medium, but it has high price, complicated and unclear compo- Because FBS is drawn from the fetus in pregnant cows during slaugh-
sition, large batch difference, and bring the risk of virus contamina- ter, containing hundreds of proteins and thousands of small molecule
tion (Fang et al., 2017; Gstraunthaler et al., 2013; Wessman and Lev- compounds, simulation of the entire components in FBS is extremely
ings, 1999). Besides, some components such as albumin, transferrin and difficult and high cost (Bettger and McKeehan, 1986). It is practical and
matrigel are also directly extracted from animals and therefore not sus- feasible to develop serum-free medium aiming to one certain cell type,
tainable. To in accord with the purpose of cultured meat which is avoid- achieving the precise nutrition supply in its proliferation and differenti-
ance of animal raising and slaughtering, all animal components should ation process. Benefit from a plenty of studies on the mechanism of mus-
be excluded in its production process. Furthermore, the development cle development in terms of the cell characteristics, signaling pathways,
of serum-free and animal component-free medium is fundamental to gene and protein expression, high-throughput screening of compounds

3
X. Guan, Q. Lei, Q. Yan et al. Future Foods 3 (2021) 100032

Fig. 2. Capital investment of cultured meat indus-


try. A) Total capital invested in cultured meat com-
panies over the years. B) Top ten companies regard-
ing the capital investment. Funding data are col-
lected from publicly available resources by Decem-
ber 31, 2020, not including companies that dedi-
cate cultured meat as part of business.

based on key pathways or targets will be one of effective approaches to environment or local environment in the bioreactor is inappropriate, it
develop cell-specific animal component-free medium (Bentzinger et al., may cause precocious cell differentiation or apoptosis.
2012; de Freitas Almeida et al., 2016; Tu et al., 2016). Various bioreactors can be applied for large-scale cultured meat
production and plenty of examples can be taken from the bioprocess-
4.2. Intelligent bioreactor for scale-up ing of microbial fermentation and genetic engineering pharmaceutical
(Moritz et al., 2015; Stephenson and Grayson, 2018; Xing et al., 2009).
Design and development of intelligent bioreactors for cultured meat As a classic bioreactor, the stirred tank reactor has been successfully ap-
is undoubtedly a technical challenge, whose expected scale would be the plied for the culture of different types of stem cells, such as mesenchymal
largest ever in the mammalian cell culture field. Taking the production stem cells (Grein et al., 2016), embryonic stem cells (Borys et al., 2020)
of one ton cultured meat as an example, the desired cell number is in and pluripotent stem cells (Manstein et al., 2019). However, for large-
the order of 1014 and the maximum accessible cell density in bioreactors volume stirred tank reactor the stirring intensity would be very high to
is 1~3 × 107 /mL, so one 10 m3 or ten 1 m3 bioreactors are the basic ensure the environment uniformity of the whole container, which might
configuration (Post et al., 2020). In vivo, the growth and development generate shear stress that exceed the endurance capacity of cells and
of cells are inseparable from the interaction environment of various or- lead to cell apoptosis (Allan et al., 2019). In contrast, bubble columns
gans, and likewise, the bioreactor should provide similar environment such as air-lift reactors have no stirring parts, and the driving force of liq-
for the cell ex vivo growth. In specific, it is necessary to artificially con- uid flow and mixing is the gravitational force acting on the entire reac-
struct the "heart" (mixing of nutrients), "lungs" (exchange of gases), and tor volume, achieving lower and more uniform shear stress distribution
"kidneys" (discharge of metabolic wastes) in bioreactors. If the overall (Nienow, 2015; Zhang et al., 2020). Li et al. (2020) designed a 300 m3

4
X. Guan, Q. Lei, Q. Yan et al. Future Foods 3 (2021) 100032

Fig. 3. Primary product interest of cultured meat companies.

air-lift reactor and estimated the optimal mass transfer, mixing and en- carded culture medium after removing toxic components and supple-
ergy dissipation rate with computational fluid dynamics for cultured menting nutrient ingredients (Taya and Kino-Oka, 2011). Therefore, it
meat production. Another popular bioreactor in the field of mammalian is necessary to monitor the change of main components in the medium
cell bio-manufacture is the perfusion reactor, which has the advantages and make timely adjustments of medium components, which depends on
in the flowing provision of nutriments and ejectment of metabolites. reliable process analysis technology. Apart from conventional monitor-
Cimetta et al. (2007) designed and developed a perfusion bioreactor ing indicators including dissolved O2 and CO2 , temperature, pH, glucose
with collagen sponges as the scaffold for the long-term culture of my- and lactic acid concentrations, cell-based indicators such as cell density,
oblasts and achieved improved cell viability, higher cell density, and morphology, characterized gene and protein expression is indispensable
uniform cell distribution throughout the entire 3D scaffold. However, (Xu et al., 2018). The location of sensor is worth to well considered to
it should be noted that different from the bioreactor designed for pro- ensure the accuracy and uniformity of collected data.
tein or compound production, the final product of cultured meat is the
muscle tissue cell itself, so the maintenance of morphology, character- 4.3. Degradable/edible scaffold for a 3D structure
istic, and function of cultured cells in the bioreactor become extremely
important. The goal of cultured meat is to manufacture a piece of meat that
Except for the large scale, real-time monitoring, high cell densities highly resembles the nutrition, appearance, texture and flavor of real
and low cost are expected to be achievable by bioreactors (Rafiq et al., muscle tissue. However, conventional cell culture methods can only
2013; Simaria et al., 2014). Cultivation of cells on micro-carriers is achieve a plane thin layer of cells, which is almost invisible (Post, 2014).
generally applied to achieve high density suspension culture of adher- The scaffold originated in the field of medical tissue engineering, which
ent cells. Micro-carriers are beads to which cells can adhere and grow are applied to provide an optimum microenvironment for tissue growth
in apposition. Because the bead surface is limited, new beads can be and regeneration (Ferreira et al., 2020; Handral et al., 2020; Jiao et al.,
added into the medium for bead-to-bead transfer of cells, achieving 2020). In general, a scaffold is a 3D porous network that mimics the
the scale-up of production (Bodiou et al., 2020). Although the pro- structure and function of extracellular matrix, on which cells adhere,
liferation and differentiation of myoblast, mesenchymal and pluripo- grow, and perform a function. The porous structure allows the input of
tent stem cells have been reported on micro-carriers, harvest of cells gases and nutritive materials and output of metabolic waste, promot-
from micro-carriers is a key procedure that should be designed carefully ing the maintenance of cell metabolism (Beale, 2014). Ideal scaffolds
(Bodiou et al., 2020). In addition, the coating, surface modification, and should have a relatively large specific surface area for cell adhesion and
size of micro-carriers should be tailored according to cell and bioreactor growth, flexible contraction and relaxation property, and good cell affin-
types (Moritz et al., 2015). Furthermore, medium recycling techniques ity and cell compatibility. At the same time, its digestibility, edibility,
can be applied to achieve the maximum use of medium (Arshad et al., safety, economy, and scalability should be considered specially for cul-
2017; Post et al., 2020). In specific, medium recycling is re-use of dis- tured meat (Browe and Freeman, 2019).

5
X. Guan, Q. Lei, Q. Yan et al. Future Foods 3 (2021) 100032

Fig. 4. Main production process of cultured meat. A muscle biopsy is performed on a living animal. Seed cells are isolated from a live animal biopsy and then
subjected to proliferation and myogenic or adipogenic differentiation. Cultured meat can be produced after food processing with ex vivo formed myofibers and
adipocytes.

Regarding the bio-based source of scaffold materials, usually they been reported to support the 3D culture of myoblasts and adipocytes
can be divided into three categories based on degradability and ed- (Ahmadi et al., 2015; Bodiou et al., 2020). As a part of the cultured
ibility: non-edible, non-edible but degradable, and edible scaffolds meat products, the texture, taste, thermal stability, cooking attribute
(Bodiou et al., 2020). As the constructing support for myofibers and and nutritional value of scaffolds are required for evaluated, as well
adipocytes of cultured meat, degradable or edible scaffolds are pre- as the economic and available for scalable production. Recently, a re-
ferred. Synthetic polymers, such as poly (lactic-co-glycolic acid) (PLGA) search group in Israel has reported the use of textured soy protein as
and polylactide (PLA), are popular degradable scaffold materials in the a scaffold to create a 3D cultured meat product with bovine satellite
field of tissue engineering (Liang et al., 2018; Patel et al., 2019). The cells, smooth muscle cells and endothelial cells, achieving meaty fla-
degradation products of these polymers are lactic acid and/or glycolic vor and sensorial attributes after cooking. Unfortunately, they provide
acid, which are safe in food. If degradable scaffolds are used, they should no information about the nutrition or the actual advantage on the final
remain stable during the cell growth procedure and be degraded spon- product quality properties by having endothelial cells (Ben-Arye et al.,
taneously or artificially before the acquisition of cultured meat prod- 2020).
ucts, so the degradation method and time should be well controlled to
prevent any cell damage or interferon of gene and protein expression.
4.4. Reduced cost for culture medium ingredients
Moreover, the residual amount of degradable material in the final prod-
uct should be detected although it might be considered as an ingredient.
Recently, cultured chicken, beef, and seafood products have been
In addition, physical methods or nontoxic reagents are preferred when
manufactured by some research groups on a pilot or larger scale, with an
stabilizing the 3D scaffolds with desired high porosity and excellent hy-
announced cost ranged from $66.4/kg to $2200.5/kg (Scipioni, 2020;
drophilic performance (Sadeghi et al., 2016).
Starostinetskaya, 2021). In contrast, the retail price of conventional
Alternatively, edible scaffolds that can be embedded in final cultured
meat is $2.1–3.9/kg for broiler chicken, $5.6–10.2/kg for beef cat-
meat products are another good choice. Native extracellular matrices in-
tle, and $2.7–7.1/kg for pork, far less expensive than current cultured
cluding collagen, gelatin, fibrin, etc., have a unique advantage as scaf-
meat cost (USDA, 2021). Therefore, cost reduction remains a high pri-
folds due to their nature characteristics for cell attachment and growth
ority in the development and optimization of cultured meat produc-
(Ali and Ahmed, 2018). MacQueen et al. (2019) developed a scaffold by
tion process. Throughout the production process of cultured meat, the
extruding gelatin microfibers and demonstrated that it supports the ad-
most significant cost driver can be attributed to the culture medium,
hesion, expansion, and maturation of muscle cells from cow and rabbit.
which contributes 55% to 95% of the production cost (Kolkmann et al.,
However, these native scaffolds are animal derived components, so the
2020).
use of them will fail to follow the fundamental goals of cultured meat.
In serum-free medium, cytokines are the most important components
Recently, recombinant expression of collagen or polypeptides has been
that support the cell growth and regulate the cell behavior, as well as
achieved by utilizing synthetic biology technology, showing promising
the most expensive components. For example, the cost compositions of
application to cultured meat (Fertala, 2020). In addition, natural mate-
Essential 8TM and DMEM with an addition of G-5 SupplementTM are
rials such as alginate and chitosan are also edible biomaterials and have
showed in Fig. 5. Essential 8TM is a commercial serum-free medium that

6
X. Guan, Q. Lei, Q. Yan et al. Future Foods 3 (2021) 100032

Fig. 5. Cost compositions of Essential 8TM and DMEM with an addition of G-5 SupplementTM . A) Essential 8TM medium. B) DMEM with an addition of G-5
SupplementTM .

has been used frequently for cultivation of various cell types from sev- et al. (2019) proved that the use of algal extracts as a substitute for
eral different species (Naung et al., 2019; Xiao et al., 2018). G-5 Sup- glucose and amino acids of basal culture media to support the growth
plement is a chemically defined serum-free supplement that includes 2 of C2C12 mouse myoblasts. Although protein hydrolysates are widely
kinds of cytokines bFGF and EGF which are proved to significantly pro- available, cheap, and have shown some benefit effects on cell growth,
mote the proliferation of muscle stem cells (de Freitas Almeida et al., their mechanism is not totally definite. Moreover, variable raw material
2016; Wei et al., 2011). It is obvious that in Essential 8TM 96% of the sources and not completely clear components might lead to great differ-
cost is attributed to cytokines and in DMEM with G-5 SupplementTM ences between batches of products (Lobo-Alfonso et al., 2008). There-
88% of the cost is Transferrin and cytokines, all of which are proteins. fore, future studies should focus on muscle-related cell types to investi-
Therefore, bringing down the cost of added proteins is the most critical gate the function and mechanism of protein hydrolysates in more depth
step in reducing the total medium costs. and improve their production process, quality control, and evaluation
The high price of cytokines is not due to the technical difficulty system accordingly.
in production, but their scientific research or medical attribute, which
possesses relatively low industrial scale but the highest industry stan- 5. Regulatory and social challenges for cultured meat
dards, including the purity, impurity component, endotoxin, and so on
(Choudhury et al., 2020). The production cost of many industrial-scale 5.1. Safety evaluation and regulatory policy
enzymes that are consumed in large quantities in the processing of food
and daily necessities like lipase, cellulase, and amylase are nearly $4 As a kind of brand new meat products, the safety evaluation and reg-
per gram, more than 104 -fold lower than the production cost of cy- ulatory policy of cultured meat need to be studied and formulated cau-
tokines (evaluated according to the retail price deducting 80% of profits) tiously. Regarding the regulatory status of global cultured meat industry,
(Specht, 2020). Therefore, it may be useful to develop the food-grade the European Union (EU) was the first initiator, the United States was
standard of cultured meat ingredients, so that low-cost cytokines are the most active promoter, and Israel and Singapore were positive partic-
available on the market. Moreover, it is often said in the fermentation ipants (Listek, 2020; Post et al., 2020). In EU, the EU Novel Foods Reg-
industry that greater scale creates lower cost per unit of output, so the ulation [(EU) 2015/2283], which came into force on 1 January 2018,
cytokine price inevitably decreases with the intensity of profession com- explicitly lists products produced through animal cell or tissue culture as
petition and expansion of production scales. new foods, clearing the legal hurdles for the marketing of cultured meat
Another way to reduce costs is by using protein hydrolysates that (EFSA, 2018). In United States, the Food and Drug Administration (FDA)
derived from plants or microorganisms as basic medium ingredients or and the US Department of Agriculture Food Safety and Inspection Ser-
even serum supplements. Protein hydrolysates are products obtained vice (USDA–FSIS) have introduced a joint regulation frame to regulate
after enzymatic, acid or alkali hydrolysis of proteins, and their main cultured meat after several discussions (FDA, 2019). In Australia, Food
components are peptides, with small amounts of amino acids, sug- Standards Australia New Zealand (FSANZ) which is a statutory author-
ars, lipids, minerals, and vitamins (Pasupuleti and Demain, 2010). ity to develop food standards for Australia and New Zealand announced
Plenty of functional hydrolysates have been identified and applied to that cultured meat could be included in their existing Food Standards
culture various animal cell types (Kim and Lee, 2009; Slivac et al., Code but specialized premarket approval is needed (FSANZ, 2017). In
2018; Taghizadeh et al., 2020). Jo et al. (2020) investigated and China, the supervision system and relevant regulations are being set up
found that whey protein hydrolysates prepared using Protamex and and improved for the cultured meat industry. Chinese researchers sug-
Alcalase can promote the proliferation and differentiation of MC3T3- gested that cultured meat should be regarded as a new food raw material
E1 osteoblasts. Hu et al. (2017) reported that yeast extracts can con- and managed according to the National Management Method for Safety
tribute to cell growth and antibody production of CHO cells. Okamoto Review of New Food Raw Materials (Wang et al., 2019). The security

7
X. Guan, Q. Lei, Q. Yan et al. Future Foods 3 (2021) 100032

of donor animals and the whole process of cultured meat production lot of capital investment and strong business competition will undoubt-
should be supervised strengthen, and an independent standard system edly facilitate the research process and shorten the timeline for the com-
and an objective regulatory system should be developed to assess the mercialization of cultured meat. Advanced technologies could speed up
food safety risk and nutrient composition. Just in December 2020, Sin- the development and optimization of animal component-free medium,
gapore regulators claimed to approve the application of publicly sell of a high-volume bioreactor, and edible scaffolds, and the production cost
cultured chicken product by the company called Eat Just, becoming the might be reduced significantly soon. However, there are fundamental
world’s first regulatory issued for cultured meat (Poinski, 2020). How- issues in terms of nutritive value, texture and flavor that need to be
ever, no specific information about the texture, flavor, and nutrients investigated deeply. More importantly, regulatory policy on the whole
including the amino acid composition, protein, fat, and mineral content process of production including plant construction, raw material pur-
of this product can be searched. chasing, manufacturing process, quality control of seed cells and final
The food safety risk of cultured meat involves in the chemical products should be formulated. Especially whether gene-editing, gene
safety, bio-safety, and nutrition security during the production process transfection or even transgenic technologies can be applied to cultured
(Wang et al., 2019). The medium components and extra additives for meat deserves to be widely discussed. Furthermore, media and corpo-
cell proliferation and differentiation, the materials of scaffolds, and the rate publicity for the sustainability, food safety, nutrition and health of
use of antibiotics and hormones should be in accord with relevant reg- cultured meat should be neutral and accurate. In general, with increas-
ulations (Chriki and Hocquette, 2020; Fujita et al., 2010; Lowe, 2006; ing demand of meat consumption and rapid development of cultured
Schnitzler et al., 2016). It is worth noting that for some additives it is meat industry, cultured meat will capture loyal consumers and possess
necessary to develop usage specification regarding food products due to significant market shares in the near future.
their first application in food area. In terms of the bio-safety of cultured
meat, the potential variation of genes and proteins of cells due to long-
Acknowledgements
term cultivation in vitro should be evaluated before premarket approval.
If gene modification is applied during the production process, sensitiza-
The authors acknowledge the Good Food Institute for providing cul-
tion, toxicity and tumorigenicity evaluation of the whole product should
tured meat company and product information.
be executed in strict rotation (Edelman et al., 2005; Mohorcich and
Reese, 2019). In a word, the evaluation criteria of food safety and policy
regulation aiming at all materials and products in the industrial chain Financial Support
of cultured meat need to be introduced as early as possible, which can
provide guidance to cultured meat research. This work was supported by the Natural Science Foundation of
Jiangsu Province (BK20202002), and the Consulting Project of the Chi-
nese Academy of Engineering (2020-XY-1722).
5.2. Public perception and acceptance

Issues of public perception and acceptance are always challenges in Declaration of Competing interest
emerging technologies and products, and cultured meat is no exception
(Bryant, 2020). Bryant and Barnett (2018) performed a systematic re- None.
view based on 14 studies about the consumer acceptance of cultured
meat and found conflicting responses in trying cultured meat. Cultured Author Contributions
meat is more acceptable to people who are familiar with it or after
being informed of the benefits of cultured meat for environment and Xin Guan drafted the manuscript, Qingzi Lei and Qiyang Yan col-
healthy. The consumer concerns focus on the uncertain food safety and lected the data with input and supervision of Xueliang Li, Jingwen Zhou,
price, such as unnaturalness, unhealthy, not tasty, and unexpected price. Guocheng Du and Jian Chen. All authors provided critical feedback and
Rolland et al. (2020) investigated the customer acceptance of cultured contributed to the final version of this manuscript.
meat after tasting and emphasized the effect of positive information in
improving acceptance and willingness to taste cultured meat. Surpris- References
ingly, 58% of the respondents were willing to pay a premium for cul-
tured meat of, on average, 37% above the price of regular meat. Ahmadi, F., Oveisi, Z., Samani, S.M., Amoozgar, Z., 2015. Chitosan based hydrogels: char-
Although cultured meat is generally targeted at meat eater, it may acteristics and pharmaceutical applications. Res. Pharm. Sci. 10 (1), 1–16.
Alexandratos, N., & Bruinsma, J. 2012. World agriculture towards 2030/2050: the 2012
attract some vegetarian-leaning individuals who don‘t eat meat due to revision. Retrieved from http://www.fao.org/3/a-ap106e.pdf.
compassion for animals, so the potential environmental and animal wel- Ali, A., Ahmed, S., 2018. Recent advances in edible polymer based hydrogels as a sustain-
fare benefits of cultured meat should be fairly estimated and propagan- able alternative to conventional polymers. J. Agric. Food Chem. 66 (27), 6940–6967.
doi:10.1021/acs.jafc.8b01052.
dized (Siegrist et al., 2018). A summary on cultured meat-related online Allan, S.J., De Bank, P.A., Ellis, M.J., 2019. Bioprocess design considerations for cultured
news comments in United States reveals more critical responses about meat production with a focus on the expansion bioreactor. Front. Sustain. Food Syst.
cultured meat such as “unnatural” and “unappealing” (Laestadius and 3, 9. doi:10.3389/fsufs.2019.00044.
Arshad, M.S., Javed, M., Sohaib, M., Saeed, F., Imran, A., Amjad, Z., Yildiz, F., 2017.
Caldwell, 2015). Therefore, except for striving to make technological Tissue engineering approaches to develop cultured meat from cells: a mini review.
breakthrough, the public science popularization about cultured meat Cogent Food Agric. 3 (1), 1320814. doi:10.1080/23311932.2017.1320814.
should be carried out through multiple channels. The development pur- Beale, R.G., 2014. Scaffold research — A review. J. Constr. Steel Res. 98, 188–200.
doi:10.1016/j.jcsr.2014.01.016.
pose, production technology, advantage and disadvantage of cultured
Ben-Arye, T., Shandalov, Y., Ben-Shaul, S., Landau, S., Zagury, Y., Ianovici, I., Lev-
meat, as well as the nutrition value, food safety and health effect of enberg, S., 2020. Textured soy protein scaffolds enable the generation of three-
products, should be popularized to the public without bias. Once con- -dimensional bovine skeletal muscle tissue for cell-based meat. Nat. Food 1 (4)
210-220.10.1038/s43016-020-0046-5.
sumers establish correct perception, they can choose whether to eat and
Benjaminson, M.A., Gilchriest, J.A., Lorenz, M., 2002. In vitro edible muscle pro-
when to eat cultured meat. tein production system (mpps): stage 1, fish. Acta Astronaut. 51 (12), 879–889.
doi:10.1016/S0094-5765(02)00033-4.
Bentzinger, C., Wang, Y.X., Rudnicki, M., 2012. Building muscle: molecular regulation of
6. Conclusion myogenesis. Cold Spring Harb. Perspect. Biol. 4. doi:10.1101/cshperspect.a008342.
Bettger, W., McKeehan, W., 1986. Mechanisms of cellular nutrition. Physiol. Rev. 66 (1),
1–35. doi:10.1152/physrev.1986.66.1.1.
As a novel food production technology, cultured meat might revolu- Bodiou, V., Moutsatsou, P., Post, M., 2020. Microcarriers for upscaling cultured meat pro-
tionize meat supply by a more efficient, green, and sustainable way. A duction. Front. Nutr.. 7 10.10.3389/fnut.2020.00010.

8
X. Guan, Q. Lei, Q. Yan et al. Future Foods 3 (2021) 100032

Borys, B.S., So, T., Roberts, E.L., Ferrie, L., Larijani, L., Abraham, B., Kallos, M.S., 2020. GFI. 2021. Alternative protein manufacturers and brands. Retrieved from
Large-scale expansion of feeder-free mouse embryonic stem cells serially passaged in https://gfi.org/resource/alternative-protein-company-database/.
stirred suspension bioreactors at low inoculation densities directly from cryopreser- Grein, T.A., Leber, J., Blumenstock, M., Petry, F., Weidner, T., Salzig, D., Czermak, P.,
vation. Biotechnol. Bioeng. 117 (5) 1316-1328.10.1002/bit.27279. 2016. Multiphase mixing characteristics in a microcarrier-based stirred tank bioreac-
Boularaoui, S.M., Abdel-Raouf, K.M.A., Alwahab, N.S.A., Kondash, M.E., Truskey, G.A., tor suitable for human mesenchymal stem cell expansion. Process Biochem. 51 (9)
Teo, J.C.M., Christoforou, N, 2018. Efficient transdifferentiation of human dermal 1109-1119.10.1016/j.procbio.2016.05.010.
fibroblasts into skeletal muscle. J. Tissue Eng. Regen. Med. 12 (2), E918–E936. Gstraunthaler, G., Lindl, T., Valk, J.v., 2013. A plea to reduce or replace fe-
doi:10.1002/term.2568. tal bovine serum in cell culture media. Cytotechnology 65 (5), 791–793.
Braga, M., Simmons, Z., Norris, K., Ferrini, M., Artaza, J., 2017. Vitamin D induces doi:10.1007/s10616-013-9633-8.
myogenic differentiation in skeletal muscle derived stem cells. Endocr. Connect. 6 Handral, H.K., Tay, S.H., Chan, W.W., Choudhury, D., 2020. 3D Printing of cultured meat
EC-17.10.1530/EC-17-0008. products. Crit. Rev. Food Sci. Nutr. doi:10.1080/10408398.2020.1815172.
Browe, D., Freeman, J., 2019. Optimizing C2 C12 myoblast differentiation using Herrero, M., Gerber, P., Vellinga, T., Garnett, T., Leip, A., Opio, C., McAllister, T.A., 2011.
polycaprolactone-polypyrrole copolymer scaffolds. J. Biomed. Mater. Res. Part A 107 Livestock and greenhouse gas emissions: the importance of getting the numbers right.
(1), 220–231. doi:10.1002/jbm.a.36556. Anim. Feed Sci. Technol. 166-67, 779–782. doi:10.1016/j.anifeedsci.2011.04.083.
Bryant, C.J., 2020. Culture, meat, and cultured meat. J. Anim. Sci. (8) 98. Hoang, V.T., Trinh, Q.-.M., Phuong, D.T.M., Bui, H.T.H., Hang, L.M., Ngan, N.T.H.,
doi:10.1093/jas/skaa172. Hoang, D.M., 2020. Standardized xeno- and serum-free culture platform enables large-
Bryant, C.J., Barnett, J., 2018. Consumer acceptance of cultured meat: a systematic review. scale expansion of high-quality mesenchymal stem/stromal cells from perinatal and
Meat Sci. 143, 8–17. doi:10.1016/j.meatsci.2018.04.008. adult tissue sources. Cytotherapy doi:10.1016/j.jcyt.2020.09.004.
Cann, O. 2018. These are the top 10 emerging technologies of 2018. Retrieved from Hoekstra, A.Y., Mekonnen, M.M., 2012. The water footprint of humanity. Proc. Natl. Acad.
https://www.weforum.org/agenda/2018/09/top-10-emerging-technologies-of-2018. Sci. U.S.A. 109 (9), 3232–3237. doi:10.1073/pnas.1109936109.
Choudhury, D., Tseng, T.W., Swartz, E., 2020. The business of cultured meat. Trends Hu, D., Zhao, L., Fan, L., Liu, X.-.P., Deng, X.-.C., Miu, S.-.W., Tan, W.-.S., 2017. Effects of
Biotechnol. 38 (6), 573–577. doi:10.1016/j.tibtech.2020.02.012. yeast extract on cell growth and antibody production in CHO cell culture. Biotechnol.
Chriki, S., Hocquette, J.-.F., 2020. The myth of cultured meat: a review. Front. Nutr. 7 Bull. 22 (6), 162–169.
(7). doi:10.3389/fnut.2020.00007. Jiang, G., Ameer, K., Kim, H., Lee, E.-.J., Ramachandraiah, K., Hong, G.-.P., 2020.
Churchill, W.S., 1932. Thoughts and Adventures. Thornton Butterworth, London. Strategies for sustainable substitution of livestock meat. Foods 9 (9), 1227.
Cimetta, E., Flaibani, M., Mella, M., Serena, E., Boldrin, L., De Coppi, P., Elvassore, N., doi:10.3390/foods9091227.
2007. Enhancement of viability of muscle precursor cells on 3D scaffold in a perfusion Jiao, Y., Li, C., Laijun, L., Wang, F., Liu, X., Mao, J., Wang, L., 2020. Construction and
bioreactor. Int. J. Artif. Organs. 30 (5) 415-428.10.1002/dat.20135. application of the textile-based tissue engineering scaffold: a review. Biomater. Sci. 8.
Datar, I., & Betti, M. 2010. Possibilities for an in vitro meat production system. Innov. Food doi:10.1039/D0BM00157K.
Sci. Emerg. Technol., 11(1), 13–22. doi:10.1016/j.ifset.2009.10.007. Jo, K., Hong, K.-.B., Suh, H.J., 2020. Effects of the Whey protein hydrolysates of various
de Freitas Almeida, C., Fernandes, S., Junior, A.F., Okamoto, O., Vainzof, M., 2016. Muscle protein enzymes on the proliferation and differentiation of 3T3-E1 osteoblasts. Prev.
satellite cells: exploring the basic biology to rule them. Stem Cells Int. 2016, 1–14. Nutr. Food Sci. 25, 71–77. doi:10.3746/pnf.2020.25.1.71.
doi:10.1155/2016/1078686. Kadim, I.T., Mahgoub, O., Baqir, S., Faye, B., Purchas, R., 2015. Cultured meat from muscle
Díaz-Flores, L., Madrid, J.F., Gutiérrez, R., Varela, H., Alvarez-Argüelles, H., 2006. Adult stem cells: a review of challenges and prospects. J. Integr. Agric. 14 (2), 222–233.
stem and transit-amplifying cell location. Histol. Histopathol. 21 (9), 995–1027. doi:10.1016/S2095-3119(14)60881-9.
doi:10.14670/HH-21.995. Kang, H.W., Lee, S.J., Ko, I.K., Kengla, C., Yoo, J.J., Atala, A., 2016. A 3D bioprinting sys-
Ding, S., Wang, F., Liu, Yan, Li, S., Zhou, G., Hu, P., 2017. Characterization and isolation tem to produce human-scale tissue constructs with structural integrity. Nat. Biotech-
of highly purified porcine satellite cells. Cell Death Discov. 3, 17003. doi:10.1038/cd- nol. 34 (3), 312–319. doi:10.1038/nbt.3413.
discovery.2017.3. Kim, S.H., Lee, G.M., 2009. Development of serum-free medium supplemented with
Dixon, L.K., Stahl, K., Jori, F., Vial, L., Pfeiffer, D.U., 2020. African swine fever epidemi- hydrolysates for the production of therapeutic antibodies in CHO cell cultures
ology and control. Ann. Rev. Anim. Biosci. 8, 221–246. doi:10.1146/annurev-ani- using design of experiments. Appl. Microbiol. Biotechnol. 83 (4), 639–648.
mal-021419-083741. doi:10.1007/s00253-009-1903-1.
Edelman, P.D., McFarland, D.C., Mironov, V.A., Matheny, J.G., 2005. Com- Kolkmann, A.M., Post, M.J., Rutjens, M.A.M., Essen, A.L.M.v., Moutsatsou, P, 2020.
mentary: in vitro-cultured meat production. Tissue Eng. 11 (5–6), 659–662. Serum-free media for the growth of primary bovine myoblasts. Cytotechnology 72,
doi:10.1089/ten.2005.11.659. 111–120. doi:10.1007/s10616-019-00361-y.
EFSA. 2018. Novel food. Retrieved from https://www.efsa.europa.eu/en/topics/topic/ Laestadius, L., Caldwell, M., 2015. Is the future of meat palatable? Perceptions of
novel-food. in vitro meat as evidenced by online news comments. Public Health Nutr. 1–11.
Fang, C.-.Y., Wu, C.-.C., Fang, C.-.L., Chen, W.-.Y., Chen, C.-.L., 2017. Long-term growth doi:10.1017/S1368980015000622.
comparison studies of FBS and FBS alternatives in six head and neck cell lines. PLoS Lamarche, É., AlSudais, H., Rajgara, R., Fu, D., Omaiche, S., Wiper-Bergeron, N., 2021.
One 12 (6), e0178960. doi:10.1371/journal.pone.0178960. SMAD2 Promotes Myogenin Expression and Terminal Myogenic Differentiation. De-
FAO. 2006. Livestock’s long shadow——Environmental issues and options. Retrieved from velopment 148 (3), dev.195495. doi:10.1242/dev.195495.
http://www.fao.org/3/a0701e/a0701e.pdf. Lee, H.J., Yong, H.I., Kim, M., Choi, Y.-.S., Jo, C., 2020. Status of meat alternatives and
FDA. 2019. Formal agreement between FDA and USDA regarding oversight of hu- their potential role in the future meat market - A review. Asian-australas. J. Anim.
man food produced using animal cell technology derived from cell lines of Sci. 33 (10), 1533–1543. doi:10.5713/ajas.20.0419.
USDA-amenable species. Retrieved from https://www.fda.gov/food/domestic- Li, X., Zhang, G., Zhao, X., Zhou, J., Du, G., Chen, J., 2020. A conceptual air-lift reactor
interagency-agreements-food/formal-agreement-between-fda-and-usda-regarding- design for large scale animal cell cultivation in the context of in vitro meat production.
oversight-human-food-produced-using-animal-cell. Chem. Eng. Sci. 211, 10 1016/j.ces.2019.115269.
Ferreira, F.V., Otoni, C.G., De France, K.J., Barud, H.S., Lona, L.M.F., Cranston, E.D., Liang, C., Luo, Y., Yang, G., Xia, D., Liu, L., Zhang, X., Wang, H., 2018. Graphene ox-
Rojas, O.J, 2020. Porous nanocellulose gels and foams: breakthrough status in ide hybridized nHAC/PLGA scaffolds facilitate the proliferation of MC3T3-E1 cells.
the development of scaffolds for tissue engineering. Mater. Today 37, 126–141. Nanoscale Res. Lett. 13 (1), 15. doi:10.1186/s11671-018-2432-6.
doi:10.1016/j.mattod.2020.03.003. Listek, V. 2020. The Cultured Meat Revolution: singapore and Israel One
Fertala, A. 2020. Three decades of research on recombinant collagens: reinvent- Step Closer to Commercializing Lab Grown Chicken. Retrieved from
ing the wheel or developing new biomedical products? Bioengineering, 7, 155. https://3dprint.com/276467/the-cultured-meat-revolution-singapore-and-israel-one-
doi:10.3390/bioengineering7040155. step-closer-to-commercializing-lab-grown-chicken/.
Fish, K., Rubio, N., Stout, A., Yuen, J., Kaplan, D., 2020. Prospects and challenges Liu, W.Y., 2019. A review on the genetic regulation of myogenesis and muscle develop-
for cell-cultured fat as a novel food ingredient. Trends Food Sci. Technol. 98. ment. Am. J. Biochem. Biotechnol. 15, 1–12. doi:10.3844/ajbbsp.2019.1.12.
doi:10.1016/j.tifs.2020.02.005. Lobo-Alfonso, J., Price, P., Jayme, D., 2008. Benefits and Limitations of Protein Hy-
Florini, J.R., Roberts, S.B., 1979. A serum-free medium for the growth of muscle cells in drolysates as Components of Serum-Free Media for Animal Cell Culture Applications.
culture. In vitro 15 (12), 983–992. doi:10.1007/BF02619157. In: Pasupuleti, V., Demain, A. (Eds.), Protein Hydrolysates in Biotechnology. Springer,
FSANZ. 2017. Australia New Zealand Food Standards Code – Standard 1.5.1 – Novel foods. Dordrecht doi:10.1007/978-1-4020-6674-0_4.
Retrieved from https://www.legislation.gov.au/Details/F2017C00324. Lowe, K.C., 2006. Blood substitutes: from chemistry to clinic. J. Mater. Chem. 16 (43),
Fujita, H., Endo, A., Shimizu, K., Nagamori, E., 2010. Evaluation of Serum-free differen- 4189–4196. doi:10.1039/b604923k.
tiation conditions for C2 C12 myoblast cells assessed as to active tension generation Lucas, A. 2020. Singapore issues first regulatory approval for lab-grown meat
capability. Biotechnol. Bioeng. 107 (5), 894–901. doi:10.1002/bit.22865. to Eat Just. Retrieved from https://www.cnbc.com/2020/12/01/singapore-issues-
Fusaro, D. 2021. Future meat technologies reduces cultured chicken breast cost first-regulatory-approval-for-lab-grown-meat-to-eat-just.html.
to $7.50. Retrieved from https://www.foodprocessing.com/industrynews/2021/ MacQueen, L.A., Alver, C.G., Chantre, C.O., Ahn, S., Cera, L., Gonzalez, G.M., Parker, K.K.,
future-meat-chicken-7-50/. 2019. Muscle tissue engineering in fibrous gelatin: implications for meat analogs. NPJ
Genovese, N.J., Domeier, T.L., Telugu, B.P.V.L., Roberts, R.M, 2017. Enhanced develop- Sci. Food 3 (1), 20. doi:10.1038/s41538-019-0054-8.
ment of skeletal myotubes from porcine induced pluripotent stem cells. Sci. Rep. 7, Manstein, F., Halloin, C., Zweigerdt, R., 2019. Human pluripotent stem cell
41833. doi:10.1038/srep41833. expansion in stirred tank bioreactors. Methods Mol. Biol. 1994, 79–91.
Gerber, P.J., Steinfeld, H., Henderson, B., Mottet, A., Opio, C., Dijkman, J., Tempio, G. doi:10.1007/978-1-4939-9477-9_7.
2013. Tackling climate change through livestock: a global assessment of emissions and Mizukami, A., Swiech, K., 2018. Mesenchymal stromal cells: from discovery
mitigation opportunities. Retrieved from http://www.fao.org/3/i3437e/i3437e.pdf. to manufacturing and commercialization. Stem Cells Int. 2018, 4083921.
GFI. (2020). 2019 State of the Industry Report Cultivated Meat. Retrieved from doi:10.1155/2018/4083921.
https://gfi.org/resource/cultivated-meat-eggs-and-dairy-state-of-the-industry-report/. Mohorcich, J., Reese, J., 2019. Cell-cultured meat: lessons from GMO adoption and resis-
tance. Appetite 143, 9. doi:10.1016/j.appet.2019.104408.

9
X. Guan, Q. Lei, Q. Yan et al. Future Foods 3 (2021) 100032

Moritz, M., Verbruggen, S., Post, M., 2015. Alternatives for large-scale pro- Starostinetskaya, A. 2021. This startup makes lab-grown chicken for less
duction of cultured beef: a review. J. Integr. Agric. 14 (2), 208–216. than $10 per serving. Retrieved from https://vegnews.com/2021/2/lab-
doi:10.1016/s2095-3119(14)60889-3. grown-chicken-less-than-10-per-serving.
Mosa. 2019. Growth Medium without Fetal Bovine Serum (FBS). Retrieved from Stephens, N., Di Silvio, L., Dunsford, I., Ellis, M., Glencross, A., Sexton, A.,
https://mosameat.com/blog/growth-medium-without-fetal-bovine-serum-fbs. 2018. Bringing cultured meat to market: technical, socio-political, and regula-
Naung, N.Y., Duncan, W., Silva, R., Coates, D., 2019. Localization and characterization tory challenges in cellular agriculture. Trends Food Sci. Technol. 78, 155–166.
of human palatal periosteum stem cells in serum-free, xeno-free medium for clinical doi:10.1016/j.tifs.2018.04.010.
use. Eur. J. Oral Sci. 127 (2), 99–111. doi:10.1111/eos.12603. Stephenson, M., Grayson, W., 2018. Recent advances in bioreactors for cell-based thera-
Nienow, A.W., 2015. Mass transfer and mixing across the scales in animal cell culture. In: pies. F1000Res 7. doi:10.12688/f1000research.12533.1.
Animal Cell Culture. Springer, pp. 137–167. Taghizadeh, M., Niazi, A., Moghadam, A., Afsharifar, A., 2020. The potential applica-
Normile, D., 2008. Driven to extinction. Science 319, 1606–1609. tion of the protein hydrolysates of three medicinal plants: cytotoxicity and functional
Okamoto, Y., Haraguchi, Y., Sawamura, N., Asahi, T., Shimizu, T., 2019. Mammalian cell properties. J. Food Sci. 85. doi:10.1111/1750-3841.15379.
cultivation using nutrients extracted from microalgae. Biotechnol. Prog. 36 (2), e2941. Taya, M., Kino-Oka, M., 2011. Bioreactors for animal cell cultures. Compr. Biotechnol.
doi:10.1002/btpr.2941. (Second Ed.) 2, 373–382.
Oki, Y., Watanabe, S., Endo, T., Kano, K., 2008. Mature adipocyte-derived dedifferentiated Taylor, R.A., Condoleo, R., Simons, R.R.L., Gale, P., Kelly, L.A., Snary, E.L, 2019.
fat cells can transdifferentiate into skeletal myocytes in vitro. Biochem. Biophys. Res. The risk of infection by African swine fever virus in European swine through
Commun. 377 (3), 780–785. doi:10.1247/csf.08038. boar movement and legal trade of pigs and pig meat. Front. Vet. Sci. 6, 486.
Park, S.-.Y., Yun, Y., Lim, J.-.S., Kim, M.-.J., Kim, S.-.Y., Kim, J.-.E., Kim, I.-.S., 2016. doi:10.3389/fvets.2019.00486.
Stabilin-2 modulates the efficiency of myoblast fusion during myogenic differentiation Tu, M., Levin, J., Hamilton, A., Borodinsky, L., 2016. Calcium signaling in
and muscle regeneration. Nat. Commun. 7, 10871. doi:10.1038/ncomms10871. skeletal muscle development, maintenance and regeneration. Cell Calcium 59.
Pasupuleti, V, Demain, A, 2010. Protein Hydrolysates in Biotechnology[M]. Springer. doi:10.1016/j.ceca.2016.02.005.
Patel, K.H., Dunn, A.J., Talovic, M., Haas, G.J., Marcinczyk, M., Elmashhady, H., Tuomisto, H.L., 2019. The eco-friendly burger could cultured meat improve
Garg, K., 2019. Aligned nanofibers of decellularized muscle ECM support the environmental sustainability of meat products? EMBO Rep. 20 (1), 6.
myogenic activity in primary satellite cells in vitro. Biomed. Mater. 14 (3) doi:10.15252/embr.201847395.
035010.10.1088/1748-605X/ab0b06. Tuomisto, H.L., de Mattos, M.J.T., 2011. Environmental impacts of cultured meat produc-
Poinski, M. 2020. Eat Just lands first regulatory approval for cell-based meat. Retrieved tion. Environ. Sci. Technol. 45 (14), 6117–6123. doi:10.1021/es200130u.
from https://www.fooddive.com/news/eat-just-lands-first-regulatory-approval-for- UNPD. 2019. World Population Prospects 2019: highlights. Retrieved from
cell-based-meat/589907/. https://www.un.org/development/desa/publications/world-population-prospects-
Post, M.J., 2012. Cultured meat from stem cells: challenges and prospects. Meat Sci. 92 2019-highlights.html.
(3), 297–301. doi:10.1016/j.meatsci.2012.04.008. Upshall, E. 2021. Producer of lab-grown kangaroo meat Vow raises $6 m in
Post, M.J., 2014. Cultured beef: medical technology to produce food. J. Sci. Food Agric. seed funding. Retrieved from https://www.foodbev.com/news/producer-of-cell-
94 (6), 1039–1041. doi:10.1002/jsfa.6474. based-kangaroo-vow-raises-6m-in-seed-funding/?__cf_chl_jschl_tk__=977cf126dc0fe00cf-
Post, M.J., Levenberg, S., Kaplan, David L., Genovese, Nicholas, Fu, J., Bryant, C.J., Mout- 044333a49a8a0edadff3f66-1613448050-0-AaoAyIH5kQmASDJG1XZNtCWkRf212XGe-
satsou, P., 2020. Scientific, sustainability and regulatory challenges of cultured meat. cHepgnCzX_1muRSNs44dh46F1RmTMk8OMKdd47kL-hSTu_5bpc-ll0AmfS6WvE-
Nat. Food 1, 403–415. doi:10.1038/s43016-020-0112-z. bED5m9lcHK5y_3i1w67GMQHYxEO_EyOT4Yq7-_NZArbcRMjafXYMmZTddxapg-
Rafiq, Q.A., Brosnan, K.M., Coopman, K., Nienow, A.W., Hewitt, C.J., 2013. Culture of hu- 2fRMUu4Zx7r-7wKqojhb5OSbcz53byi5UgHBOsaL1rKBr_N72kOowtFCJQ3uFJVMj-
man mesenchymal stem cells on microcarriers in a 5 l stirred-tank bioreactor. Biotech- 3wCCUIzVhlPhRZ9dOXUl08rK8NgRuQdC55pg0dcjSbc2j1DSjkz1raNaG6BRs1ZD2UY-
nol. Lett. 35 (8), 1233–1245. doi:10.1007/s10529-013-1211-9. H3JPkmT2-4RSGb7PKwQlznh0Rt3Eu4L0q3BXdFF0KouhDemyjW_w8qRUvKkKeKm-
Rolland, N.C.M., Markus, C.R., Post, M.J, 2020. The effect of information content on AwX4KJtAQeFyuikgPiPdgDJm26bYajxIrmt9Eu88TnTOaqFjgnpKWQ.
acceptance of cultured meat in a tasting context. PLoS One 15 (10), e0240630. USDA. 2021. Meat Price Spreads. Retrieved from https://www.ers.usda.gov/data-
doi:10.1371/journal.pone.0240630. products/meat-price-spreads/.
Sadeghi, A., Nokhasteh, S., Molavi, M., Khorsand-Ghayeni, M., Naderi-Meshkin, H., Wang, T., Zhou, J., Zhao, X., Zhang, G., LI, X., Du, G., Sun, X., 2019. Research progress
Mahdizadeh, A., 2016. Surface modification of electrospun PLGA scaffold with on lab-grown meat risk prevention and safetymanagement norms. Food Forment. Ind.
collagen for bioengineered skin substitutes. Mater. Sci. Eng. C 66, 130–137. 45 (11), 254–258.
doi:10.1016/j.msec.2016.04.073. Webber, J. 2019. Lab-Grown Foie Gras Will Be in Restaurants By 2021. Retrieved from
Schnitzler, A.C., Verma, A., Kehoe, D.E., Jing, D.H., Murrell, J.R., Der, K.A., https://www.livekindly.co/lab-grown-foie-gras-restaurants/.
Rook, M.S., 2016. Bioprocessing of human mesenchymal stem/stromal cells for Wei, Y., Li, Y., Chen, C., Stoelzel, K., Kaufmann, A.M., Albers, A.E., 2011. Human skeletal
therapeutic use: current technologies and challenges. Biochem. Eng. J. 108, 3–13. muscle-derived stem cells retain stem cell properties after expansion in myosphere
doi:10.1016/j.bej.2015.08.014. culture. Exp. Cell Res. 317 (7), 1016–1027. doi:10.1016/j.yexcr.2011.01.019.
Scipioni, J. 2020. This restaurant will be the first ever to serve lab-grown chicken (for $23). Wessman, S.J., Levings, R.L., 1999. Benefits and risks due to animal serum used in cell
Retrieved from https://www.cnbc.com/2020/12/18/singapore-restaurant-first-ever- culture production. Dev. Biol. Stand. 99, 3–8.
to-serve-eat-just-lab-grown-chicken.html?&qsearchterm=cultured%20meat. Wilkinson, A.C., Ishida, R., Kikuchi, M., Sudo, K., Morita, M., Crisostomo, R.V., Ya-
Siegrist, M., Sütterlin, B., Hartmann, C., 2018. Perceived naturalness and evoked mazaki, S., 2019. Long-term ex vivo haematopoietic-stem-cell expansion allows non-
disgust influence acceptance of cultured meat. Meat Sci. 139, 213–219. conditioned transplantation. Nature 571, 117–121. doi:10.1038/s41586-019-1244-x.
doi:10.1016/j.meatsci.2018.02.007. Williams, L.A., Davis-Dusenbery, B.N., Eggan, K.C., 2012. SnapShot: directed differenti-
Simaria, A.S., Hassan, S., Varadaraju, H., Rowley, J., Warren, K., Vanek, P., Farid, S.S., ation of pluripotent stem cells. Cell 149 (5), 1174. doi:10.1016/j.cell.2012.05.015,
2014. Allogeneic cell therapy bioprocess economics and optimization: single-use cell e1171.
expansion technologies. Biotechnol. Bioeng. 111 (1), 69–83. doi:10.1002/bit.25008. Xiao, J., Yang, D., Li, Q., Tian, W., Guo, W., 2018. The establishment of a chemically
Siu, E. 2019. World’s first cultivated fish maw shows the importance of global defined serum-free culture system for human dental pulp stem cells. Stem Cell Res.
markets. Retrieved from https://www.gfi-apac.org/blog/worlds-first-cultivated-fish- Ther. 9 (1), 191. doi:10.1186/s13287-018-0928-8.
maw-shows-the-importance-of-global-markets/. Xing, Z.Z., Kenty, B.N., Li, Z.J., Lee, S.S., 2009. Scale-up analysis for a CHO cell
Slivac, I., Logarusic, M., Bojanic, K., Srcek, V., Radošević, K., 2018. Protein hydrolysates culture process in large-scale bioreactors. Biotechnol. Bioeng. 103 (4), 733–746.
from hempseed oil-cake as cell culture media supplement. J. Biotechnol. 280, S30. doi:10.1002/bit.22287.
doi:10.1016/j.jbiotec.2018.06.094. Xu, S., Jiang, R., Mueller, R., Hoesli, N., Kretz, T., Bowers, J., Chen, H., 2018. Probing
Solis-Castro, O.O., Boissonade, F.M., Rivolta, M.N., 2020. Establishment and neural differ- lactate metabolism variations in large-scale bioreactors. Biotechnol. Prog. 34 (3), 756–
entiation of neural crest-derived stem cells (NCSCs) from human dental pulp in serum- 766. doi:10.1002/btpr.2620.
free conditions. Stem Cells Transl Med 9 (11), 1462–1476. doi:10.1002/sctm.20-0037. Zhang, G., Zhao, X., Li, X., Du, G., Zhou, J., Chen, J., 2020. Challenges and possibili-
Southey, F. 2020. ‘Ethical foie-gras’: cultured meat start-up taps duck egg cells ties for bio-manufacturing cultured meat. Trends Food Sci. Technol. 97, 443–450.
to recreate French delicacy. Retrieved from https://www.foodnavigator.com/ doi:10.1016/j.tifs.2020.01.026.
Article/2020/05/14/Ethical-foie-gras-Cultured-meat-start-up-taps-duck-egg-cells-to- Zhao, X., Zhang, G., Li, X., Sun, X., Zhou, J., Du, G., Chen, J., 2019. Commercial production
recreate-French-delicacy. of artificial meat. Food Ferment. Ind. (11) 248–253.
Specht, L. (2020). An analysis of culture medium costs and production volumes for culti-
vated meat. Retrieved from.

10

You might also like